CN109294996B - RFFT2 cell - Google Patents

RFFT2 cell Download PDF

Info

Publication number
CN109294996B
CN109294996B CN201811153192.4A CN201811153192A CN109294996B CN 109294996 B CN109294996 B CN 109294996B CN 201811153192 A CN201811153192 A CN 201811153192A CN 109294996 B CN109294996 B CN 109294996B
Authority
CN
China
Prior art keywords
cells
polypeptide
cell
rfft2
impact
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
CN201811153192.4A
Other languages
Chinese (zh)
Other versions
CN109294996A (en
Inventor
焦顺昌
张嵘
周子珊
解佳森
王海燕
李营营
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.)
Beijing Dingcheng Taiyuan Biotechnology Co ltd
Original Assignee
Beijing Dingcheng Taiyuan Biotechnology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beijing Dingcheng Taiyuan Biotechnology Co ltd filed Critical Beijing Dingcheng Taiyuan Biotechnology Co ltd
Priority to CN201811153192.4A priority Critical patent/CN109294996B/en
Publication of CN109294996A publication Critical patent/CN109294996A/en
Application granted granted Critical
Publication of CN109294996B publication Critical patent/CN109294996B/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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • 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
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • 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
    • C12N2800/80Vectors containing sites for inducing double-stranded breaks, e.g. meganuclease restriction sites
    • 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/10Vectors comprising a non-peptidic targeting moiety

Abstract

The invention provides an RFFT2 cell, and belongs to the technical field of biology. The invention provides a T cell with integrated attack and defense, high accuracy and high killing property. The cell is prepared by the following method: the PBMC cells are loaded with polypeptide causing tumor mutation, and then polypeptide impact is carried out on the PBMC cells loaded with the polypeptide; performing expanded culture after impact to obtain FF cells; so that the tumor-mutated polypeptide is used as an antigen to directly stimulate FF cells to screen precise polypeptide; culturing PBMC cells, and performing polypeptide impact by using accurate polypeptide in the culture process; continuing culturing after impacting to obtain RFF cells; screening to obtain specific cells capable of identifying the precision polypeptide; obtaining a TCR gene from a specific cell; culturing PBMC (peripheral blood mononuclear cell), knocking out the original TCR gene, and transferring the obtained TCR gene to prepare an RFFT cell; and (3) carrying out immunosuppressive signal molecule blocking on the obtained RFFT cells by using a monoclonal antibody medicine to obtain the RFFT2 cells.

Description

RFFT2 cell
Technical Field
The invention relates to the technical field of biology, in particular to an RFFT2 cell for cellular immunotherapy.
Background
Tumor cell immunotherapy is an emerging tumor therapy model, which collects immune cells from a patient, cultures and expands in vitro, and then transfuses them back into the patient to stimulate and enhance the body's autoimmune function to treat tumors. Tumor cell immunotherapy is the fourth method of tumor treatment following surgery, radiation therapy and chemotherapy.
When the normal or bioengineered human body cells are transplanted or input into the body of a patient, the newly input cells can replace damaged cells or have stronger immune killing function, thereby achieving the purpose of treating diseases.
The bioengineered cells are modified by special methods during in vitro culture process, and can effectively kill tumor cells in patients. For example, chinese patent application CN201210194280.5 provides a human cytokine-induced killer cell. Chinese patent application CN201510034781.0 provides a tumor cell specific polyclonal T cell. The chinese patent application CN201510013987.5 provides an anti-tumor T cell and a preparation method thereof. Chinese patent application CN201711060030.1 provides a CAR-T cell for treating AIDS-associated lymphoma, and a preparation method and application thereof. CN201610824893.0 provides a double-antigen specific T cell regulated by an antibody, a preparation method and an application thereof.
In the prior art, T cells are generally presented through DC cells to generate specific killing T cells, or viruses are used as vectors to induce the specific killing of the T cells through a lentivirus transfection technology. But the effect is not good because of unclear tumor antigen and the obstacle of tumor microenvironment immunosuppression; or the effect is poor because of the simple and thin specific cells directly facing the complex tumor microenvironment, or the target is single and only effective for individual tumors.
Disclosure of Invention
The invention aims to provide a novel TCR-T cell (named as RFFT2 cell) which is used for cellular immunotherapy and realizes the precision, specificity and safety of killing.
The invention provides RFFT2 cells for cellular immunotherapy, wherein the preparation method of the RFFT2 cells comprises the following steps:
s1) loading the polypeptide causing tumor mutation to the PBMC cells, and then carrying out one-time polypeptide impact on the PBMC cells loaded with the polypeptide;
s2) expanding culture after impact to obtain FF cells;
s3) so that the tumor mutated polypeptide acts as an antigen to directly stimulate the FF cells to screen for the precise polypeptide;
s4), culturing PBMC cells, and performing multiple polypeptide impacts by using the accurate polypeptide in the culture process;
s5) continuing culturing after impact to obtain RFF cells;
s6) stimulating the obtained RFF cells by taking the precision polypeptide as an antigen, and screening to obtain specific cells capable of identifying the precision polypeptide;
s7) obtaining a TCR beta chain CDR3 region sequence of a specific cell through sequencing, and obtaining a TCR gene through amplifying the TCR beta chain CDR3 region sequence;
s8) culturing the PBMC cell, knocking out the original TCR gene in the cell, and transferring the TCR gene which is obtained in the step S7 and can be specifically combined with the precise polypeptide to prepare the RFFT cell;
s9) performing immunosuppressive signal molecule blocking on the RFFT cells obtained in the step S8 with a monoclonal antibody to obtain RFFT2 cells, wherein the immunosuppressive signal molecule comprises: PD-1, Tim-3, LAG3, CTLA-4, BTLA, VISTA, CD160, TIGIT, 2B4(CD 244).
Further, in step S2, the expanding culturing after the impacting to obtain the FF cell composition comprises:
culturing the PBMC cells after the polypeptide impact in a cell culture device pre-paved with a cell stimulating factor OKM-25 for 5 days;
transferring to a cell culture device containing a culture solution OKM-100+ 12% FBS for continuous culture till the 10 th day;
transferring the cells to a cell culture device containing a culture solution OKM-200+ 5% FBS, and continuing culturing for 14-21 days.
Further, in step S4, the polypeptide impact is repeated 3-4 times.
Furthermore, in the polypeptide impact, polypeptide impact is carried out by using polypeptide solution with the concentration of 10 to 100 mu g/mL.
Further, the impact time of the polypeptide impact is 1-4 h.
Further, the tumor-causing mutant polypeptide is synthesized by the following method:
1) exon sequencing
Sequencing the whole exon of the tumor cell;
comparing the sequencing result of the whole exon with the genome of a normal cell, and screening out a mutant amino acid site;
2) epitope prediction
Taking the mutated amino acid site as the center, extending 10 amino acids to two sides to obtain a section of polypeptide with 21 amino acids as a potential antigen epitope;
the potential epitope is considered as the epitope when IC50 is less than 1000 nM;
3) synthetic polypeptides
The epitope peptide is synthesized by a polypeptide solid phase synthesis method.
Further, the tumor cells are derived from engineering cell lines, including H1299, H226, H358, H1563, H2228, A549, Renca, LLC mouse Lewis lung cancer cells, CRL-6323B16F1, CRL-25394T 1, U14 mouse cervical cancer cells, BV-2 mouse glioma cells and G422 mouse glioma cells.
Further, prior to step S8, a TCR gene knockout CRISPR lentiviral vector was constructed to infect CD8+ T cells sorted by magnetic beads from PBMC cells for preparing RFFT cells.
Further, the step S9 includes the steps of resuspending the RFFT cells obtained in the step S8 in a culture solution OKM-200+ 5% FBS, adding a single drug of inhibitory signal molecules with a final concentration of 50-200 μ g/mL, and sealing at 37 ℃ for 1h to obtain the RFFT2 cells.
The invention has the following beneficial effects:
1) the invention provides a TCR-T cell (named as RFFT2 cell) for cellular immunotherapy, which is a super T cell with both attack and defense. The effective accurate polypeptide is screened out and subjected to secondary polypeptide impact on PBMC cells. It is noted that increased amounts of polypeptide shock stimulation were used in the second precision polypeptide shock. Then, on the basis of two times of stimulation, the TCR-T technology is combined for third stimulation, common T cells are transformed into T cells with specific killing effect, and the killing efficiency and the precision are high.
2) In the invention, the combination body is externally closed, cultured in vitro and amplified in vitro simultaneously, so that the precision, specificity and safety of searching and killing are realized, more tumor species are covered, and the adaptability of the T cells with high-precision killing effect to a complex tumor microenvironment is improved.
3) Compared with the killing effect of directly transfecting and inducing T cells by using lentiviruses, the method is simple and convenient and has high safety.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below, and it is obvious that the drawings in the following description are only some embodiments of the present invention, and for those skilled in the art, other drawings can be obtained according to these drawings without creative efforts.
Figure 1 shows antigen loading efficiency detection;
FIG. 2 shows the results of a precision polypeptide screen;
FIG. 3 shows the flow detection of specific cell ratios;
FIG. 4 shows TCR distribution of specific cells;
FIG. 5 shows the flow assay of the knockout of an existing TCR on a cell;
FIG. 6 shows inhibitory target in vitro occlusion;
FIG. 7 shows the killing of target cells by RFFT2 cells according to an embodiment of the invention;
FIG. 8 shows the detection of cytokine release by RFFT2 cells according to an embodiment of the present invention;
figure 9 shows the tumor-bearing mouse survival curve.
Detailed Description
The technical solutions in the embodiments of the present invention will be clearly and completely described below with reference to the drawings in the embodiments of the present invention, and it is obvious that the described embodiments are only a part of the embodiments of the present invention, and not all of the embodiments. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
Tumor cell immunotherapy is an emerging mode of tumor therapy. In the aspect of tumor cell immunotherapy, the existing LAK, DC, CIK, DC-CIK cells have been proved to be basically ineffective. The present invention provides a novel T cell useful for cellular immunotherapy. The invention modifies T cells, provides the TCR-T cells, has strong lethality and high accuracy, and covers various tumors.
The embodiment of the invention provides a TCR-T cell (named as RFFT2 cell) for cellular immunotherapy, and the preparation method comprises the following steps:
s1) loading the polypeptide causing tumor mutation to the PBMC cells, and then carrying out one-time polypeptide impact on the PBMC cells loaded with the polypeptide;
s2) expanding culture after impact to obtain FF cells;
s3) so that the tumor mutated polypeptide acts as an antigen to directly stimulate the FF cells to screen for the precise polypeptide;
s4), culturing PBMC cells, and performing multiple polypeptide impacts by using the accurate polypeptide in the culture process;
s5) continuing culturing after impact to obtain RFF cells;
s6) stimulating the obtained RFF cells by taking the precision polypeptide as an antigen, and screening to obtain specific cells capable of identifying the precision polypeptide;
s7) obtaining a TCR beta chain CDR3 region sequence of a specific cell through sequencing, and obtaining a TCR gene through amplifying the TCR beta chain CDR3 region sequence;
s8) knocking out the original TCR gene in the peripheral blood T cell of the patient, and transferring the TCR gene which is obtained in the step S7 and can be specifically combined with the precise polypeptide to prepare an RFFT cell;
s9) performing immunosuppressive signal molecule blocking on the RFFT cells obtained in the step S8 with a monoclonal antibody to obtain RFFT2 cells, wherein the immunosuppressive signal molecule comprises: PD-1, Tim-3, LAG3, CTLA-4, BTLA, VISTA, CD160, TIGIT, 2B4(CD 244).
Herein, the definition of "RFFT 2" cells refers to the following meanings:
r-precise polypeptide secondary impact technology;
FF-mixed polypeptide technology;
T-TCR-T technology;
2-in vitro closed protection technology.
That is, the "RFFT 2" cell herein is a novel T cell prepared by an RFFT2 scheme composed of a combined polypeptide mixing technique, an accurate polypeptide secondary impact technique, a TCR-T (T cell receptor (TCR) chimeric T cell) technique, and an in vitro blocking protection technique, and can be used for tumor immunotherapy. As the name implies, "FF" cells are T cells derived from the FF protocol. "RFF" cells are T cells derived from the RFF protocol. "RFFT" cells are T cells derived from the RFFT protocol.
The embodiments of the present invention provide a TCR-T cell for use in cellular immunotherapy. In the RFFT2 scheme of the embodiment of the invention, firstly, the mixed polypeptide is used for directly impacting PBMC cells loaded with the polypeptide to perform first stimulation; secondly, screening the accurate polypeptide, taking the screened accurate polypeptide as an antigen to directly stimulate FF cells, and performing secondary stimulation; followed by a third stimulation by TCR-T technique. T cells are transformed through three times of stimulation, and the transformed T cells achieve the precision, specificity and safety of searching and killing. Meanwhile, the in vitro culture and in vitro sealing protection technology are combined, so that the adaptability of the modified T cells to the in vivo complex immune environment is improved.
Prior to step S1, the tumorigenic polypeptide may be synthesized by: 1) sequencing exons; 2) predicting the epitope; 3) and (3) synthesizing the polypeptide.
1) Exon sequencing
Carrying out whole exon sequencing by using tumor cells, and then analyzing sequencing information by using software to obtain MHC type information on one hand; on the other hand, the whole exon sequencing result is compared with the genome of normal cells, and the mutated amino acid site is selected.
In exon sequencing, tumor cells may be derived from engineered cell lines, or may be derived from a patient, such as patient peripheral blood or tumor tissue.
In exon sequencing, the engineered cell lines include H1299, H226, H358, H1563, H2228, A549, Renca, LLC mouse Lewis lung cancer cells, CRL-6323B16F1, CRL-25394T 1, U14 mouse cervical cancer cells, BV-2 mouse glioma cells, G422 mouse glioma cells. The engineering cell line is a cell line which is obtained by modifying or recombining genetic materials of host cells by adopting a genetic engineering technology or a cell fusion technology and has unique characteristics of stable heredity.
2) Epitope prediction
In the prediction of epitope, 10 amino acids are extended from the site of mutated amino acid as the center to both sides, and the 21 amino acid polypeptide is used as "potential epitope". The IC50 of potential epitopes was analyzed using prediction software (recommended software: NetMHCpan3.0, PickPocket, Artificial Neural Networks (ANN)). A potential epitope is considered to be an "epitope" if IC50 < 1000 nM.
3) Synthetic polypeptides
The epitope peptide is synthesized by a polypeptide solid phase synthesis method.
In step S1, the PBMC cells loaded with the polypeptide are directly stimulated by polypeptide-impacting with the polypeptide-mutagenic polypeptide for the first stimulation.
Specifically, the loading of the PBMC cells with the polypeptide causing tumor mutation and then performing polypeptide impact on the PBMC cells loaded with the polypeptide can specifically be as follows:
1) preparing a polypeptide solution: dissolving polypeptides in RPMI 1640+ 10% FBS (fetal bovine serum) or OKM100+ 12% FBS, wherein the final concentration of each polypeptide is 10-100 mug/mL, preferably 50 mug/mL, for later use;
2) resuscitating PBMC 1 day ahead, blowing cells, sucking 15mL, counting and centrifuging;
3) resuspending PBMCs with the formulated polypeptide solution;
4) impact into a cell culture plate;
5)375%CO2and impacting for 1-4h, preferably 4h to obtain impacted PBMC cells for later use.
In step S2, the expanding culture after the impact to obtain FF cells may specifically be:
1) stimulation factor OKM-25 pre-plated, 40. mu.L of OKM-25+4mL PBS (phosphate buffered saline), 2 mL/dish (4.5 cm)2) Room temperature 4h, 4 ℃ for standby;
2) transferring the impacted PBMC to a cell culture plate or a culture bottle pre-paved with OMK-25;
3) shaking evenly, 5% CO at 37 ℃2Culturing, and recording as day 0;
4) observing the condition of the co-cultured cells, on day 5, according to the cell density, the co-cultured cells were transferred to a large flask supplemented with fresh medium OKM-100+ 12% FBS, 20mL at 75cm2In a culture bottle;
5) on the 7 th day of co-culture, 20mL of fresh OKM-100+ 12% FBS was added;
6) co-culture on day 10 in OKM-200+ 5% FBS medium, the co-cultured cells were cultured from 75cm2Transfer to 175cm in the bottle2In a big bottle;
7) beating 25mL of culture solution OKM-200+ 5% FBS, transferring into a large bottle, and repeating for 2 times; make up to 200mL with medium OKM-200+ 5% FBS.
8) FF cell composition can be obtained after culturing for 14-21 days. And extracting the T cells from the FF cell composition by centrifugation to obtain FF cells.
In step S3, T cells are isolated and extracted from the FF cell composition such that the tumor mutated polypeptide acts as an antigen to directly stimulate the T cells to screen for a precise polypeptide.
The screening criteria for the precise polypeptide were:
polypeptide as antigen with FF cells as baseline; the two are independently repeated, the high detection value is a high base line, and the low detection value is a low base line;
the difference between the two baselines is a system error, and during data analysis, the detection values > low baseline, > high baseline and > high baseline + system error are respectively marked. And (5) the detection value is greater than the high baseline and the system error is the effective accurate polypeptide.
In step S3, the step of directly stimulating T cells with the polypeptide as an antigen to screen precise polypeptide may specifically be:
1) centrifuging the obtained FF cell composition at 1500rpm for 5min to collect T cells, adding 10mL PBS to resuspend the cells and counting, centrifuging at 1500rpm for 5min, collecting T cells, resuspending with 1640+ 10% FBS +200U/mL IL2, and adjusting the count to 1 × 106Per mL;
2) t cells were plated in 96-well flat bottom plates using a row gun, 200μ L/well, cell number 2X 105A plurality of; then respectively adding 10 mu L of 1mg/mL mutant polypeptide with the final concentration of 50 mu g/mL, and arranging 3 compound holes on each polypeptide;
3) setting a positive control: t cells +100ng/mL OKT3(CD3 monoclonal antibody); negative control: RPMI 1640+ 10% FBS +200U/mL IL2 (Interleukin 2); two T cell controls were used as background release detection, first T cell addition, and last T cell addition, respectively; taking the difference of two background releases as a system error;
4)37℃、5%CO2after 24h of stimulation, centrifugation is carried out at 1500rpm for 10min, and 140 microliter of supernatant is transferred to a new 96-well plate;
5) centrifuging 96-well plate at 1500rpm for 10min, and collecting sample for ELISA (enzyme-linked immunosorbent assay) (or storing at-80 deg.C);
6) screening of the precise polypeptide:
based on the accurate polypeptide screening standard, the detection value is greater than the high baseline and the system error is the effective accurate polypeptide.
In step S4, PBMC cells are cultured and a second stimulation is performed by polypeptide shock with the precision polypeptide during the culturing process.
Compared to step S1, an additional amount of precision polypeptide impact is used in step S4. That is, the PBMC cells are subjected to multiple polypeptide shock stimuli in this step. Specifically, the polypeptide impact can be repeated for 3-4 times.
During the culture process, the culture protocol is similar to the expanded culture of the PBMC cells loaded with the polypeptide in the step S2. Firstly culturing for a period of time in a device pre-paved with cell stimulating factors OKM-25, and then sequentially transferring to an OKM-100+ 12% FBS culture medium and an OKM-200+ 5% FBS culture medium for continuous culture.
Step S4 may specifically include the following steps:
culturing the PBMC cells in a cell culture device pre-paved with a cell stimulating factor OKM-25, and performing polypeptide impact on the accurate polypeptide obtained in the step S3 after culturing for a period of time;
and then transferring the cells to a cell culture device (culture plate or culture bottle) containing a culture solution OKM-100+ 12% FBS for continuous culture, and performing polypeptide impact on the precise polypeptides obtained in the step S3 every 3-4 days.
The impact time per polypeptide impact may be 1-4 h. Ordinary T cells are transformed into RFF cells with more accurate killing capacity through the impact stimulation of the added accurate polypeptide.
In step S5, the culture is continued after the impact, and RFF cells are obtained. A suitable medium may be OKM200+ 5% FBS. Transferring the polypeptide after impacting to a device containing culture solution OKM200+ 5% FBS for continuous culture at 37 ℃ and 5% CO2And culturing to obtain the T cell, namely the RFF cell, obtained by secondary impact of the accurate polypeptide.
In step S6, the RFF cells obtained by stimulating the precise polypeptide as an antigen, staining CD8, CD137 and IFN-gamma of the stimulated cells, sorting the cells by a flow cytometer, and screening to obtain specific cells capable of identifying the precise polypeptide.
In step S7, the sequence of the CDR3 region of the TCR β chain of the specific cell is obtained by sequencing. The TCR gene is obtained by amplifying the CDR3 region sequence of the TCR beta chain.
The method specifically comprises the following steps:
1) extracting a genome from the cell sorted in step S6;
2) performing TCR sequencing analysis on the genome, and determining a high-frequency TCR sequence according to TCR distribution frequency;
3) extracting mRNA of PBMC, reverse transcribing to obtain DNA, designing primer according to the sequence of high frequency TCR, and amplifying to obtain TCR gene.
In step S8, the original TCR gene of the cell can be knocked out using CRISPR technology.
In step S8, the original TCR gene and surface immunosuppressive signal molecule in the peripheral blood T cell are knocked out, and the TCR gene obtained in step S7 and capable of specifically binding to the precision polypeptide is transferred to obtain RFFT1 cell. T cells were engineered by a third stimulation using TCR-T technology.
In step S8, a CRISPR lentiviral vector for knocking out the original TCR gene and a TCR gene expression vector for expressing the TCR gene determined in step S7 can be constructed, respectively. Which infected CD8+ T cells sorted by magnetic beads from PBMC cells.
Specifically, CD8+ T cells sorted by PBMC cells through magnetic beads are infected by CRISPR lentiviral vectors for knocking out original TCR genes, and the original TCR is knocked out; then, the cells are infected with the TCR gene expression vector expressing the TCR gene determined in step S7, and the TCR gene determined in step S7 is transferred. Infected CD8+ T cells were then resuspended in OKM100+ 12% FBS and plated on pre-plated plates of stimulation factor OKM-25 for continued culture to obtain RFFT cells.
The construction process of the CRISPR lentiviral vector for knocking out the original TCR gene is as follows:
1) finding out CDS region of mRNA of TCR gene on pubmed, analyzing conservation region of TCR, and predicting knockout target of conservation region;
2) designing a forward primer and a reverse primer required for synthesizing the sgRNA, and carrying out amplification reaction on the forward primer and the reverse primer 1: 1, treating at 95 ℃ for 5-60 min after mixing, and then slowly cooling to form a DNA sequence of the sgRNA;
3) performing double enzyme digestion on a CRISPR lentiviral expression vector, connecting the CRISPR lentiviral expression vector with double-stranded DNA corresponding to sgRNA, transferring the double-stranded DNA into a clone competent cell, and after 12h, selecting a single clone for sequencing, and reserving the clone with correct sequencing;
4) and (3) extracting CRISPR lentiviral vector plasmids carrying DNA sequences corresponding to the sgRNAs, and packaging the viruses.
The construction process of the TCR gene expression vector capable of expressing the TCR gene determined in step S7 is as follows: and (4) performing virus packaging based on the determined TCR gene. And will not be described in detail herein.
In step S9, blocking the RFFT cells obtained in step S8 with a monoclonal antibody to obtain RFFT2 cells, wherein the immunosuppressive signal molecules include: PD-1, Tim-3, LAG3, CTLA-4, BTLA, VISTA, CD160, 2B4(CD 244). Through the antibody drug in-vitro sealing protection technology, T cells are protected, and the adaptability of the T cells to in-vivo complex immune microenvironment is improved.
In one example, the RFFT cells obtained in step S8 were resuspended in OKM-200+ 5% FBS, and then a single drug of inhibitory signal molecule was added to a final concentration of 150. mu.g/mL, and blocked at 37 ℃ for 1h to obtain RFFT2 cells.
For example, the detailed operation of step S9 may specifically be:
1) centrifuging at 1000rpm for 5min, and collecting cultured RFFT cells;
2) washed once with PBS, centrifuged at 1000rpm for 5min, resuspended TCR-T in OKM-200+ 5% FBS and adjusted to 1X 107/mL;
3) Adding single-resistant drug (such as PD-1 antibody and the like) of inhibitory signal molecules, wherein the final concentration is 50-200 mu g/mL, preferably 150 mu g/mL, and blocking at 0-37 ℃ for 1-4h, preferably 37 ℃ for 1 h.
The embodiment of the invention provides a novel T cell (RFFT2 cell) for tumor immunotherapy, which is transformed into a TCR-T cell with good accuracy, high killing property, capability of covering various tumors and strong adaptability to a complex immune microenvironment by combining a mixed polypeptide technology, an accurate polypeptide secondary impact technology, a TCR-T technology and an in vitro sealing protection technology.
The following is a further detailed description of RFFT2 cells for cellular immunotherapy as described in the examples of the invention.
(one) sequencing of all exons
1) Performing whole exon sequencing using the engineered cell line;
2) sequencing information was analyzed using software: on one hand, obtaining MHC type information; on the other hand, comparing the sequencing result of the whole exon with the genome of a normal cell, and screening out a mutation site;
(II) epitope prediction
1) Taking the mutated amino acid site as the center, extending 10 amino acids to both sides, and taking the 21 amino acid polypeptide as the potential antigen epitope;
2) the IC50 of potential epitopes was analyzed using prediction software (recommendation software: NetMHCpan3.0, PickPocket, and Artificial Neural Networks (ANN)), and if IC50 is less than 1000nM, the potential epitope is considered as an "epitope";
(III) Synthesis of Polypeptides
The epitope peptide synthesis method adopts a polypeptide solid phase synthesis method
1) Anchoring: anchoring the first amino acid to the solid phase resin;
2) deprotection: protected amino acid the protecting group of the amino group is removed using an alkaline solvent;
3) and (3) activation: activating the amino acid carboxyl group to be linked using an activating agent;
4) and (3) bonding: the activated carboxyl group reacts with the naked amino group of the previous amino acid to form a peptide
5) And (5) repeating the steps 2-4 to completely synthesize the whole epitope peptide chain.
(IV) PBMC Loading mutant Polypeptides
1) Preparing a polypeptide solution: dissolving polypeptides by 1640+ 10% FBS or OKM100+ 12% FBS, wherein the final concentration of each polypeptide is 50 mug/mL for later use;
2) resuscitating PBMC 1 day ahead, blowing cells, sucking 15mL, counting and centrifuging;
3) resuspending the PBMC in the prepared polypeptide solution and placing into a cell culture plate for impact;
4)375%CO2impacting for 4h for standby;
(V) expanded culture of PBMC after polypeptide impact
1) Stimulation factor OKM-25 pre-plated, 40. mu.L of OKM-25+4mL PBS, 2 mL/dish (4.5 cm)2) Room temperature 4h, 4 ℃ for standby;
2) transferring the impacted PBMC to a culture flask pre-paved with OMK 25;
3) shaking evenly, 5% CO at 37 ℃2Culturing, and recording as day 0;
4) co-cultured cells were observed, and on day 5, the cells were transferred to a large flask, supplemented with fresh medium OKM-100+ 12% FBS, 20mL at 75cm, depending on cell density2In a culture bottle;
5) on the 7 th day of co-culture, 20mL of fresh OKM-100+ 12% FBS was added;
6) co-culture on day 10 in OKM-200+ 5% FBS medium, the co-cultured cells were cultured from 75cm2The one in the flask was transferred to 175cm2In a big bottle;
7) beating 25mL of culture solution OKM-200+ 5% FBS, transferring into a large bottle, and repeating for 2 times; make up to 200mL with medium OKM-200+ 5% FBS.
8) Culturing for 14-21 days to obtain FF cell composition.
(VI) the polypeptide is used as an antigen to directly stimulate T cells to screen precise polypeptides:
1) the T cells in the FF cell composition are the T cells obtained by the FF protocol (FF cells). Centrifuging to collect the obtained FF cell composition, centrifuging at 1500rpm for 5min to collect FF cells, adding 10mL PBS to resuspend the cells and counting, centrifuging at 1500rpm for 5min, collecting T-cells, resuspending with 1640+ 10% FBS +200U/mL IL2, and adjusting the counting to 1 × 106cells/mL;
2) FF cells were plated on 96-well flat-bottom plates with a row gun at 200. mu.L/well and 2X 10 cells/well5cells; then respectively adding 10 mu L of 1mg/mL mutant polypeptide with the final concentration of 50 mu g/mL, and arranging 3 compound holes on each polypeptide;
3) setting a positive control: FF cells +100ng/mL OKT 3; negative control: 1640+ 10% FBS +200U/mLIL 2; two FF cell controls are used as background release detection, namely a first FF cell addition and a last FF cell addition;
4)37℃、5%CO2after 24h of stimulation, centrifugation is carried out at 1500rpm for 10min, and 140 microliter of supernatant is transferred to a new 96-well plate;
5) the 96-well plate is centrifuged again at 1500rpm for 10min, and samples are taken for ELISA detection (or the samples are stored at-80 ℃).
(VII) accurate polypeptide evaluation standard:
1) if the positive control and the negative control are normal, the data is credible;
2) the polypeptide is used as an antigen, and T-cells is used as a baseline;
3) each group of experiments comprises two baselines, a high baseline and a low baseline, the difference of the two baselines is a system error, and when data are analyzed, detection values > low baseline, > high baseline and > high baseline + system error are respectively marked; and (5) the detection value is greater than the high baseline and the system error is the effective accurate polypeptide.
(VIII) preparation of RFF cells from the selected precision polypeptide
1) PBMC were cultured (stimulated) with FF cell composition culture protocolFactor OKM-25 Pre-plating, 40. mu.L of OKM-25+4mLPBS, 2 mL/dish (4.5 cm)2) Room temperature 4h, 4 ℃ for standby; the culture conditions were 37 ℃ and 5% CO2) By day 3, re-impacting the precision polypeptide;
2) take 2X 107Adding polypeptide with the final concentration of 50 mu g/mL into the T cells, and impacting for 4 hours;
3) after impacting for 4h, the height is changed to 25cm2Culture flask supplemented with OKM100+ 12% FBS, 5% CO at 37 deg.C2Culturing, transferring to 75cm according to cell growth2The cell density in the culture flask was kept as high as 1X 106Per mL;
4) repeating the precision polypeptide shock, i.e., repeating steps 2) and 3), at days 7, 10, and 14 of culture;
5) cells entered 175cm2When the cells are cultured in a culture bottle, the culture medium is OKM200+ 5% FBS, and the cells are cultured for 10-21 days to obtain the T cells, namely the RFF cells, obtained by secondary impact of the precise polypeptides.
(nine) culture and isolation of mutant antigen-specific T cells
1) Stimulating RFF cells by directly taking the precise polypeptide as an antigen for 12-72 hours for later use;
2) the stimulated cells were stained with CD8, CD137, IFN-. gamma.and sorted by flow cytometry, selecting CD8+ CD137+, or CD8+ IFN-. gamma. + cells.
(Ten) TCR frequency detection of CD8+ T cells and cloning of high frequency TCR
1) Extracting the genome from the cells obtained by sorting in the step nine;
2) carrying out TCR sequencing analysis on the genome, and determining a high-frequency TCR sequence according to TCR distribution frequency;
3) extracting mRNA of PBMC, reverse transcribing to obtain DNA, designing primer according to the sequence of high frequency TCR, and amplifying to obtain TCR gene.
(eleventh) construction of TCR knockout CRISPR Lentiviral vectors
1) Finding out CDS region of mRNA of TCR gene on pubmed, analyzing conservation region of TCR, and predicting knockout target of conservation region;
2) designing a forward primer and a reverse primer required for synthesizing the sgRNA, and carrying out amplification reaction on the forward primer and the reverse primer 1: 1, treating at 95 ℃ for 5-60 min after mixing, and then slowly cooling to form a DNA sequence of the sgRNA;
3) performing double enzyme digestion on a CRISPR lentiviral expression vector, connecting the CRISPR lentiviral expression vector with double-stranded DNA corresponding to sgRNA, transferring the double-stranded DNA into a clone competent cell, and after 12h, selecting a single clone for sequencing, and reserving the clone with correct sequencing;
4) and (3) extracting CRISPR lentiviral vector plasmids carrying DNA sequences corresponding to the sgRNAs, and packaging the viruses.
(twelve) TCR-T
1) Recovering PBMCs, and sorting CD8+ cells by magnetic beads for later use;
2) infecting a CD8+ T cell by the CRISPR lentiviral vector obtained in the step eleven, and knocking out the original TCR at the same time;
3) after infection, culturing CD8+ T cells in a culture medium for 3 days, transferring the TCR gene expression vector constructed in the step ten to infect the cells so as to transfer the TCR gene constructed in the step ten;
4) infected CD8+ T cells were resuspended in OKM100+ 12% FBS and plated on pre-plating with stimulation factor OKM-25, recorded as day 0;
5) observing cell condition and cell density, and on day 5, transferring the co-cultured cells to a large culture flask, and supplementing fresh culture solution OKM-100+ 12% FBS;
6) cells were removed from 75cm2The one in the flask was transferred to 175cm2The post-large bottle culture solution is OKM-200+ 5% FBS;
7) and when the cells are cultured for 14-21 days, the TCR-T cells, namely the RFFT cells, of which the immunosuppressive signal molecules are knocked out can be harvested.
(thirteen) in vitro blocking of immunosuppressive Signal molecules
1) Centrifuging at 1000rpm for 5min, and collecting cultured RFFT cells;
2) washed once with PBS, centrifuged at 1000rpm for 5min, resuspended TCR-T in OKM-200+ 5% FBS and adjusted to 1X 107/mL;
3) Adding single drug of inhibitory signal molecule, such as PD-1 antibody, etc., to a final concentration of 150 μ g/mL, and blocking at 37 deg.C for 1 h.
(fourteen) tumor-bearing mouse survival experiments
1) And (4) inoculating the tumor cell line to an NSG mouse to make an ectopic tumor-bearing animal model. Will be 5X 105Tumor cells expressing specific antigens were suspended in 100. mu.L of physiological saline and injected subcutaneously into the right flank of 30 NSG mice, respectively, and the mice were numbered.
2) Growth of tumor to 100-120mm3The cells were returned to the left and right groups, and the animal model was randomly divided into three groups of 5 mice each, one group given placebo physiological saline, and one group given 1X 10T cells (control group) without any genetic manipulation according to the tumor volume7One group was given to RFFT2 cells at 1X 107And 7 days after the first injection of the cells, performing the second injection, 7 days after the third injection of the cells, continuously observing for 60 days, counting survival data, and drawing a survival curve.
Results of the experiment
1. Mutation site and epitope prediction
Table 1 shows the sequencing detected mutation sites and the epitope prediction results, and the underlined are the mutated amino acids.
TABLE 1 epitope prediction
Figure BDA0001818382540000131
Figure BDA0001818382540000141
2. Antigen load efficiency detection
Synthesizing a predicted mutant antigen according to the table 1, labeling biotin, and detecting the distribution of the biotin on the cell surface by using PE-labeled affinity streptomycin after the antigen is loaded on PBMC so as to detect the efficiency of extracting polypeptide antigen by the PBMC; the results are shown in figure 1-antigen loading efficiency assay: a is a detection result without loading the marker polypeptide, b is a detection result with loading the biotin polypeptide, and the result shows that: the loading efficiency of PBMC was 54.4% (FL2-H subset 54.4% indicates that the PI stain stained 54.4% of the cells).
3. Screening for accurate Polypeptides
As shown in FIG. 2, 12 polypeptides were used to stimulate cultured FF cells and effective polypeptides were detected by detecting IFN-. gamma.secretion, and the results are shown in FIG. 2: the release amount of IFN-gamma caused by the polypeptides No. 2, No. 4, No. 6 and No. 10 is greater than high baseline and system error, and the polypeptide belongs to effective and accurate polypeptide.
RFF cell-specific cell proportion detection
The screened polypeptide No. 6 is stimulated for multiple times on the basis of FF cells, and after culture, the proportion of T cells specific to the precise polypeptide is detected in a flow mode, and the result is shown in figure 3, and specific T cells are shown in a box: compared with a control group without treatment, the RFF cells are subjected to multiple impact stimulation, the ratio of cells releasing IFN-gamma caused by the No. 6 polypeptide is obviously higher than that of cells (FF cells) without multiple impact stimulation, and the specific T cell ratio can be improved by accurate multiple stimulation of the polypeptide; sorting of CD8+ IFN-. gamma. + (boxed) cells was also performed by flow cytometry.
5. Identification and cloning of high frequency TCR
The cells obtained by sorting were subjected to genome extraction and sequencing of TCR, the distribution of TCR is shown in fig. 4 (top 20 of high frequency distribution), and the distribution frequency of TCR9 is higher, which indicates that this TCR is closely related to the mutant antigen, and the TCR was amplified according to the TCR sequence.
TABLE 2 sequence case of CDR3 of TCR beta chain
Figure BDA0001818382540000151
6. Knockout of original TCR
The original TCR on PBMC was knocked out using CRISPR technique, and the results are shown in figure 5.
Known TCR-. beta.s:
amino acids:
Figure BDA0001818382540000152
the horizontal line is the CDR3 sequence, the sequence to be replaced
TCR-. beta.after replacement:
Figure BDA0001818382540000153
the horizontal line is the replaced CDR3 sequence
7. Detection of inhibitory target in vitro blocking efficiency
The inhibitory signal molecule antibody is marked by fluorescence, and the blocking condition of the PD-1 antibody to the cells is detected by flow. As a result, as shown in FIG. 6, the proportion of cells in which fluorescence signals could be detected was 96.9%.
Killing of target cells by RFFT2 cells
As shown in fig. 7, RFF cells, RFFT cells, and RFFT2 cells all had a certain killing effect on target cells compared to the control group, and the killing efficiency of RFF cells, RFFT cells, and RFFT2 cells on target cells derived from mutant epitopes was measured, and the results were shown in 20: 1 and 40: 1 (effector cells: target cells), the difference from the Mock group was significant. Wherein the killing efficiency of the RFFT2 cells on the tumor is more than that of the RFFT cells and more than that of the RFF cells.
Detection of cytokine Release by RFFT2 cells
When tumor cells are co-cultured with effector cells, because the effector cells can recognize the mutant antigens on the tumor cells, a series of cytokines are produced, IFN-gamma is one of the most important cytokines in the anti-tumor effect, and FIG. 8 is the detection of IFN-gamma released when cells and tumor cells are co-cultured in different culture modes. The results show that: after co-culture with tumor cells, RFF protocol cells, RFFT protocol cells and RFFT2 cells all produced significant amounts of IFN- γ compared to IFN- γ produced by effector cells themselves (effector cells only), and in particular RFFT and RFFT2 cells released more IFN- γ, consistent with the results of the killing experiment: t cells expressing specific TCR, combined with blocking of inhibitory targets, can more effectively improve anti-tumor capacity.
10. Experiment on survival of tumor-bearing mice
The results are shown in fig. 9, that reinfusion of RFFT2 cells of the present example had a significant effect on the improvement in survival of tumor-bearing mice. A P value less than 0.01(P value 0.0017) indicates statistical significance.
11. Clinical cases
The administration process comprises the following steps:
the first course of treatment: RFFT2 cells were administered once a month, 1X 10 in number 92 times for each cell;
the second course of treatment: RFFT2 cells at a rate of 1X 10 cells/half year 92 cells in total.
TABLE 3
Numbering Sex Age (age) Disease diagnosis Progression-free survival time after completion of administration
1 Woman 76 Ovarian cancer 2017.4-Up to now
2 Woman 71 Cervical cancer 2017.4-Up to now
3 For male 71 Stomach cancer 2017.3-Up to now
4 For male 77 Lung cancer 2017.9-Up to now
5 For male 65 Lung cancer 2017.1-Up to now
The above description is only for the specific embodiments of the present invention, but the scope of the present invention is not limited thereto, and any person skilled in the art can easily conceive of the changes or substitutions within the technical scope of the present invention, and all the changes or substitutions should be covered within the scope of the present invention. Therefore, the protection scope of the present invention shall be subject to the protection scope of the claims.

Claims (7)

1. An RFFT2 cell for cellular immunotherapy, wherein the RFFT2 cell is prepared by a method comprising the following steps:
s1) Synthesis of a tumor-mutagenizing polypeptide
1) Exon sequencing
Sequencing the whole exon of the tumor cell;
comparing the sequencing result of the whole exon with the genome of a normal cell, and screening out a mutant amino acid site;
2) epitope prediction
Taking a mutated amino acid site as a center, extending 10 amino acids to two sides, and taking a section of polypeptide with 21 amino acids as a potential antigen epitope;
the potential epitope is considered as the epitope when IC50 is less than 1000 nM;
3) synthetic polypeptides
Synthesizing antigen epitope peptide by using a polypeptide solid phase synthesis method, and marking the antigen epitope peptide as a polypeptide causing tumor mutation;
the PBMC cells are loaded with the polypeptide causing the tumor mutation, and then the PBMC cells loaded with the polypeptide are subjected to polypeptide impact for one time;
s2) expanding culture after impact to obtain FF cells;
s3) so that the tumor mutated polypeptide acts as an antigen to directly stimulate the FF cells to screen for the precise polypeptide; wherein the accurate polypeptide evaluation standard is as follows: setting two independent repeats by taking FF cells as a baseline, wherein a high detection value is a high baseline, and a low detection value is a low baseline; the difference between the two baselines is the systematic error; the experimental group with the detection value of more than the high baseline and the system error is the effective accurate polypeptide;
s4), culturing PBMC cells, and performing polypeptide impact for multiple times by using the accurate polypeptide obtained in the step S3 in the culture process, wherein the polypeptide impact is repeated for 3-4 times;
s5) continuing culturing after impact to obtain RFF cells;
s6) stimulating the obtained RFF cells by taking the precise polypeptide as an antigen, and screening specific cells capable of identifying the precise polypeptide;
s7) obtaining a TCR beta chain CDR3 region sequence of the specific cell through sequencing, and obtaining a TCR gene through amplifying the TCR beta chain CDR3 region sequence;
s8) culturing PBMC cells, knocking out the original TCR gene in the cells, transferring the TCR gene which is obtained in the step S7 and can be specifically combined with the precise polypeptide, and culturing to obtain RFFT cells;
s9) carrying out immunosuppressive signal molecule blocking on the RFFT cells obtained in the step S8 by using a monoclonal antibody, and culturing to obtain the RFFT2 cells, wherein the immunosuppressive signal molecule comprises: PD-1, Tim-3, LAG3, CTLA-4, BTLA, VISTA, CD160, TIGIT, 2B4(CD 244).
2. The RFFT2 cells for cellular immunotherapy according to claim 1, wherein in step S2, the post-impact expansion culture to obtain FF cell composition comprises:
culturing the PBMC cells after the polypeptide impact in a cell culture device pre-paved with a cell stimulating factor OKM-25 for 5 days;
transferring to a cell culture device containing a culture solution OKM-100+ 12% FBS for continuous culture till the 10 th day;
transferring the cells to a cell culture device containing a culture solution OKM-200+ 5% FBS, and continuing culturing for 14-21 days.
3. The RFFT2 cells for cellular immunotherapy, according to claim 1, wherein in step S4, the polypeptide impact is performed every 3-4 days.
4. The RFFT2 cell for cellular immunotherapy, according to claim 1, wherein in the polypeptide impact, the polypeptide impact is performed with a polypeptide solution with a concentration of 10 μ g/mL-100 μ g/mL.
5. The RFFT2 cell for cellular immunotherapy, according to claim 1, wherein the tumor cell is derived from an engineered cell line comprising H1299, H226, H358, H1563, H2228, A549, Renca, LLC mouse Lewis lung cancer cell, CRL-6323B16F1, CRL-25394T 1, U14 mouse cervical cancer cell, BV-2 mouse glioma cell, G422 mouse glioma cell.
6. The RFFT2 cell for cellular immunotherapy, according to claim 1, wherein in step S8, PBMC cells are de-infected with a CRISPR lentiviral vector for knockout of an original TCR and a CRISPR lentiviral vector for knockout of a surface immunosuppressive signal molecule, and the knockout of the original TCR and the removal of the immunosuppressive signal molecule are performed simultaneously.
7. The RFFT2 cells for cellular immunotherapy, according to claim 1, wherein the blocking of the RFFT cells obtained in step S8 with a monoclonal antibody against immunosuppressive signaling molecules to obtain RFFT2 cells comprises:
and (3) resuspending the RFFT cells obtained in the step S8 by using a culture solution OKM-200+ 5% FBS, adding a single drug of an inhibitory signal molecule with the final concentration of 50-200 mu g/mL, and sealing at 37 ℃ for 1h to obtain the RFFT2 cells.
CN201811153192.4A 2018-09-30 2018-09-30 RFFT2 cell Active CN109294996B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811153192.4A CN109294996B (en) 2018-09-30 2018-09-30 RFFT2 cell

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811153192.4A CN109294996B (en) 2018-09-30 2018-09-30 RFFT2 cell

Publications (2)

Publication Number Publication Date
CN109294996A CN109294996A (en) 2019-02-01
CN109294996B true CN109294996B (en) 2020-11-06

Family

ID=65161378

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811153192.4A Active CN109294996B (en) 2018-09-30 2018-09-30 RFFT2 cell

Country Status (1)

Country Link
CN (1) CN109294996B (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015533473A (en) * 2012-07-12 2015-11-26 ペルシミューン,インコーポレイテッド Individual cancer vaccine and adaptive immune cell therapy
CN104926944B (en) * 2015-05-22 2018-04-13 北京康爱瑞浩生物科技股份有限公司空港分公司 Multitarget composite antigen load C D8+The preparation method and its usage of cytotoxic T lymphocyte
CN108456247A (en) * 2017-02-20 2018-08-28 上海恒润达生生物科技有限公司 Target the T cell receptor and application thereof of NY-ESO-1
CN107630005A (en) * 2017-09-19 2018-01-26 深圳市北科生物科技有限公司 Express T cell and its application of PLAC1 specificity TCRs

Also Published As

Publication number Publication date
CN109294996A (en) 2019-02-01

Similar Documents

Publication Publication Date Title
CN106074601A (en) Method and composition for cellular immunotherapy
US20100254958A1 (en) Antigen-Specific T-Cell Preparations from Bone Marrow
Schaafsma et al. VISTA targeting of T-cell quiescence and myeloid suppression overcomes adaptive resistance
US20230138309A1 (en) Methods of isolating t-cells and t-cell receptors from tumor by single-cell analysis for immunotherapy
CN110093376B (en) Construction method of LRFFT1 cells
CN109136284B (en) AFFT2 cell
CN109294998B (en) RFF1 cell
CN109294982B (en) RFF2 cell
CN109294996B (en) RFFT2 cell
CN110129371B (en) Construction method of RFFT2 cells
CN110129372B (en) Construction method of RFFT1 cells
CN110129374B (en) Construction method of RFFT (radiofrequency FT) cells
CN110157745B (en) Construction method of HAFFT1 cell
CN109280643B (en) RFFT cell
JP2022512538A (en) Anti-LMP2 TCR-T cell therapy for the treatment of EBV-related cancers
CN109294995B (en) RFFT1 cell
CN109136278B (en) MRFFT1 cell
CN109679917B (en) LRFFT2 cell
CN109294997B (en) LRFFT1 cell
CN105031632B (en) A kind of dendritic cell vaccine and the preparation method and application thereof
CN110129269B (en) Construction method of RFF2 cells
CN110093374B (en) Construction method of MRFFT1 cell
CN110408657B (en) Construction method of AFFT1 cells
CN109790224A (en) Tumor-antigen peptide and its application derived from CACNA1H
CN110093373B (en) Construction method of AFFT2 cells

Legal Events

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