CN114277056A - Preparation method of renal cancer antigen CA9 protein specific cytotoxic T lymphocyte - Google Patents

Preparation method of renal cancer antigen CA9 protein specific cytotoxic T lymphocyte Download PDF

Info

Publication number
CN114277056A
CN114277056A CN202111598162.6A CN202111598162A CN114277056A CN 114277056 A CN114277056 A CN 114277056A CN 202111598162 A CN202111598162 A CN 202111598162A CN 114277056 A CN114277056 A CN 114277056A
Authority
CN
China
Prior art keywords
cells
sisocs1
iapa
cell
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202111598162.6A
Other languages
Chinese (zh)
Inventor
段训威
肖桂清
杨槟煌
王礼兴
陈怀宇
吴文林
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.)
Quanzhou Normal University
Original Assignee
Quanzhou Normal University
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 Quanzhou Normal University filed Critical Quanzhou Normal University
Priority to CN202111598162.6A priority Critical patent/CN114277056A/en
Publication of CN114277056A publication Critical patent/CN114277056A/en
Pending legal-status Critical Current

Links

Images

Landscapes

  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The invention provides a preparation method of a renal cancer antigen CA9 protein specific cytotoxic T lymphocyte, belonging to the technical field of immune cell therapy. The method of the invention comprises the following steps: preparing a rAAV2-siSOCS1-CA9 recombinant virus vector carrying target genes of an immune suppression negative control gene siSOCS1 and CA 9; separating and culturing DC cells in mononuclear cells; infecting the recombinant virus vector of rAAV2-siSOCS1-CA9, inducing the DC cell to mature, and obtaining iAPA-CA9-DC cell; the iAPA-CA9-DC cells and T cells are mixed and cultured to obtain iAPA-CA9-DC/CTL cells with antigen specificity, namely, renal carcinoma protein CA9 specific cytotoxic T lymphocytes activated by dendritic cells. The method of the invention cultures and expands a large amount of tumor antigen specific T cells to achieve the purpose of treating cancer.

Description

Preparation method of renal cancer antigen CA9 protein specific cytotoxic T lymphocyte
Technical Field
The invention belongs to the technical field of immune cell therapy, and particularly relates to a preparation method of a renal cancer antigen CA9 protein specific cytotoxic T lymphocyte. The cytotoxic T lymphocyte is activated by dendritic cells modified by rAAV2 carrying an immune negative regulatory gene SOCS1 gene and a carbonic anhydrase IX gene, has CA9 tumor antigen specificity, and can be used for immune cell therapy against renal cell carcinoma.
Background
The most common space occupying lesion in adult kidney, Renal Cell Carcinoma (RCC), accounts for approximately 2% of all tumors and increases worldwide by 1.5-5.9% every year. The first diagnosis of some patients is late stage or has far-end metastasis, and kidney cancer has multiple drug-resistant genes which are not sensitive to radiotherapy and chemotherapy and are worse after recovery, the median survival time is less than 12 months, and the 5-year survival rate is less than 10%. At present, surgical resection is the best treatment method, but the surgical resection of micro and metastatic lesions is difficult to perform, and the recurrence rate of postoperative patients is as high as 20-40%; in addition, some patients are effectively treated by cytokines such as IL-2, IFN-alpha and the like, but the toxic effect of the drugs is very obvious.
The occurrence and development of RCC are related to the functional deficiency of T lymphocytes in patients. Particularly, the humoral immunity and the cellular immunity of the RCC patients are generally low at the late stage, and the RCC cells cannot be recognized and killed by T cells. Because the number of Dendritic Cells (DC) of a tumor-bearing host is small, RCC antigen can not be effectively presented, and the RCC cells secrete cytokines and adhesion molecules such as VEGF, IL-10, TGF-beta, PGE2 and the like, so that the mature differentiation of the dendritic cells is stopped at a juvenile stage or is transformed to other endothelial cells, and the low expression of MHC-II molecules and certain co-stimulatory molecules causes the DC to seriously reduce or even lose the capability of presenting antigen and activating T cells.
At present, in vitro experiments prove that lymphocytes for separating peripheral blood of a patient have good reaction capability on various foreign antigens. Therefore, the adoptive immune cell therapy (DC/CTL) of the tumor is expected to be a better RCC treatment method, the action mechanism is that the DC loads RCC antigen in vitro, activates specific CTL through the interaction of MHC/antigen peptide-TCR, and is returned to an organism to start anti-RCC immune response so as to eliminate RCC. At present, DC is loaded by tumor cell lysate, but the DC has the defects that the types of non-tumor related antigens are multiple, autoimmune diseases are easy to induce, and the immunogenicity is low due to multiple antigens; the other stimulating inducer adopts tumor antigen peptide to sensitize DC, which has good specificity and can be immunized for many times, but has the defects that the antigen peptide is generally expensive in price and short in half-life period, and good CTL can be obtained only by immunization for many times.
As the research on the loaded DC is deepened, researchers find that CTL can be promoted to fully exert the anti-tumor effect by preparing the transgenic DC and inducing the CTL in vitro. Compared with tumor whole antigen and antigen lysate, the DC/CTL prepared by tumor transgenosis has better specificity and stronger anti-tumor immune response; compared with tumor antigen peptide, the tumor transgene can be expressed continuously, and the preparation cost of DC/CTL is effectively reduced. At present, rAAV has the characteristics of strong infection capacity, good safety, low immunogenicity, long-term in vivo expression of foreign genes and the like, is regarded as one of the most promising gene transfer vectors, and is widely applied to gene therapy and vaccine research in the world. The tumor cell targeted therapy technology (ACTL) based on DC is that DC is transduced by rAAV which is reconstructed by gene recombination to carry specific tumor-associated antigenic determinant gene, and CTL which specifically kills tumor cells is stimulated to generate. Research shows that in the aspect of CTL response capacity, rAAV with antigen is superior to protein load, the efficiency of DC transfection is up to more than 90%, and the rAAV has the function of up-regulating DC.
The method for preparing the DC/CTL by the tumor transgenosis is safe and has few side effects, and has incomparable high targeting property for RCC of tiny and metastatic lesions by a traditional means. However, in practice, the antigen-loaded DC/CTL alone often fails to elicit a sufficiently strong anti-tumor immune response, and thus the therapeutic effect is not as desirable as expected. This is because in order to avoid the attack of the host immune system, tumors develop various escape mechanisms, and in the antigen presentation stage, factors such as low immunogenicity of tumor antigens, reduced expression of MHC molecules, deletion of coordinative stimulatory molecules, secretion of suppressive cytokines, and the like may exist. Therefore, certain methods must be used to modify and treat DCs to enhance their antigen presentation and T cell immune priming capabilities.
Cytokine signaling inhibitory factor-1 (SOCS 1) is a negative regulator of JAK-STAT signaling pathway, negatively regulates cytokine signaling pathway and TLR signaling pathway, and can inhibit intracellular signaling of IL, IFN and other factors. By silencing or interfering withSOCS1The expression of the gene can promote DC maturation and change the tumor tolerance state. Chen et al propose enhancing DC function by inhibiting the inhibition of immune-presentation attutors (iAPA). The iAPA is a novel tumor immune cell treatment technology, and the core is that small interfering ribonucleic acid (siRNA) is adopted to specifically block key negative regulatory factors in DC, so that the gene expression of the iAPA is silenced, an organism is induced to break through the immune tolerance of the iAPA to the tumor, the antigen specific immune response is enhanced, and the clinical curative effect of tumor immunotherapy is further improved.
In addition, carbonic anhydrase IX (CAIX or CA 9) is a glycoprotein distributed on cell and nuclear membranes. Oosterwi JK et al first obtained murine CA9 monoclonal antibody, and immunohistochemistry was performed on most types of RCC using monoclonal antibody, with 88% of the antigen expressed and most tissues not. The gene is located at 9p12-13, 10898bp in total, is homologous with MN and G250 genes, has the same gene conformation, expresses proteins distributed on cell membranes and nuclear membranes, a mature body is glycoprotein encoded by 459 amino acids, wherein 38 amino acids (aal-37) form an N-terminal Signal Peptide (SP), 377 amino acids (aa 38-414) form an extracellular region, 20 amino acids (aa 415-434) form a transmembrane region, and 25 amino acids (aa 435-459) form the C-terminal of a cell. The extracellular region is divided into two parts: a proteoglycan domain (PG) and a carbonic anhydrase domain (CA). Wherein the CA region is located in the cell membrane and comprises 257 amino acids (aal 35-391), and 59 amino acids (aa 53-111) between SP and CA have 38% homology with the human keratin adhesion region. Therefore, the tumor associated antigen CA9 can be used as an RCC tumor marker.
Based on the technical research results, the method for killing RCC cells by immunity is a very promising therapeutic means, and the key point is the selection of DC stimulating antigens and the optimization of the method, so that the DC is enhanced, and T cell specific immunity is started. The ACTL technology endows the DC/CTL with high targeting property, the iAPA technology can effectively solve the self tolerance problem of an organism and obviously improve the antigen presenting capability of the DC, and if the ACTL technology and the iAPA technology can be jointly used, the antigen specific immune response of the DC/CTL is expected to be obviously enhanced. In the research, rAAV2 is adopted to mediate RNAi to down-regulate SOCS1 gene expression, and the combination of CA9 and S-Flagellin stimulates DC cells to mature, induces specific DC/CTL to specifically kill anti-CA 9 positive RCC. The significance lies in that the targeting property of CTL to RCC is effectively enhanced, the damage to autologous cells is avoided, and clinically, the iAPA technology is applied, so that the growth of tumors can be effectively inhibited, the cells are prevented from being transferred remotely, and the life cycle of a patient is prolonged.
Disclosure of Invention
The invention aims to provide a preparation method of a renal cancer antigen CA9 protein specific cytotoxic T lymphocyte aiming at the defects of the prior art. Under the premise of collecting a small amount of peripheral blood of a patient, the invention cultures and expands a large amount of tumor antigen specific T cells to specifically kill the tumor cells.
In order to achieve the purpose, the invention adopts the following technical scheme:
a preparation method of a renal cancer antigen CA9 protein specific cytotoxic T lymphocyte comprises the following steps:
(1) preparing a rAAV2-siSOCS1-CA9 recombinant virus vector carrying siSOCS1 and CA9 target genes;
(2) infecting, differentiating and inducing DC cells to mature by the rAAV2-siSOCS1-CA9 recombinant virus vector to obtain iAPA-CA9-DC cells;
(3) the iAPA-CA9-DC cells and T cells are mixed and cultured to obtain iAPA-CA9-DC/CTL cells, namely the dendritic cell activated CA9 tumor antigen specific cytotoxic T lymphocytes.
The target gene sequences carrying the target siSOCS1 and CA9 in the step (1) are sequentially as follows: a U6 promoter sequence, a SOCS1 siRNA sequence, a SOCS1 siRNA reverse sequence, a CMV promoter sequence, a CA9 protein gene sequence, an IRES sequence, a Flagellin Flagellin gene sequence; the sequence of the SOCS1 siRNA is shown as SEQ ID NO.1, the reverse sequence of SOCS1 siRNA is shown as SEQ ID NO.2, the sequence of SOCS1 siRNA and the reverse sequence of SOCS1 siRNA are complementary in reverse direction, and both ends of a gene segment are respectively provided with BamH 1/HindIII enzyme cutting sites; the CA9 protein gene sequence, the IRES sequence and the Flagellin Flagellin gene sequence are respectively shown as SEQ ID NO.3, SEQ ID NO.4 and SEQ ID NO. 5; the target gene is assembled with an adenovirus vector to obtain the rAAV2-siSOCS1-CA9 recombinant virus vector.
Infecting, differentiating and inducing mature DC cells by the rAAV2-siSOCS1-CA9 recombinant virus vector in the step (2) to obtain iAPA-CA9-DC cells; the method specifically comprises the following steps:
1) according to whole blood: physiological saline: the lymphocyte separation liquid is in a ratio of 1:1:1, the centrifugal force is 650g, the centrifugal time is 15min, the speed of rise is 4, the speed of fall is 0, a mononuclear cell layer above the lymphocyte separation liquid is extracted after centrifugation, and washing is carried out twice;
2) adjustment of monocyte density to 5X 106cell/mL, as per 106 cell/cm2The cell number of (2) was added to a culture flask at 37 ℃ with 5% CO2Culturing for 2-4h in a concentration incubator, slightly shaking the culture bottle, shaking the non-adherent cells, sucking away the upper non-adherent cells, centrifugally collecting, and freezing;
3) the original flask was supplemented with Lymphocyte serum-free medium KBM581 (KBM 581) medium containing 2 IU/mL of DC cell culture factor (Novoprotein), and incubated at 37 ℃ with 5% CO2Culturing in a concentration incubator;
4) after overnight culture, gently shaking the culture bottle at the 2 nd day, shaking the non-adherent cells, sucking away the upper layer of the non-adherent cells, centrifugally collecting, and freezing again;
5) adding the 581 culture medium containing the DC cell culture factor into the DC cell culture bottle again, and continuously culturing for 3 d; culturing to 5d, completely replacing the culture medium with KBM581 culture medium containing 2 IU/mL DC cell maturation factor, and continuing culturing for 48 h; collecting suspension cells, calculating the total amount of the cells, infecting by using the rAAV2-siSOCS1-CA9 recombinant virus vector according to MOI 100, adding the rAAV2-siSOCS1-CA9 recombinant virus vector to infect for 2h, changing the solution, and continuously culturing for 48h to obtain the iAPA-CA9-DC cells.
In the step (3), the specific steps of mixed culture of the iAPA-CA9-DC cells and the T cells are as follows:
1) adding stimulating factor CD3 Monoclonal antibody 50 ng/ml, CD 2850 ng/mL of monoclonal antibody and IL-21000U/mL of monoclonal antibody stimulate the proliferation of T cells;
2) after the T cells are cultured for 48h by stimulating proliferation, iAPA-CA9-DC cells and the T cells are mixed and cultured according to the proportion of 1:5, and the cell density is adjusted to 1 multiplied by 106cell/mL, adding KBM581 culture medium containing IL 2; iAPA-CA9-DC/CTL cells were obtained.
A cancer therapeutic agent comprising the above-mentioned renal cancer antigen CA9 protein-specific cytotoxic T lymphocyte.
The application of the kidney cancer antigen CA9 protein specific cytotoxic T lymphocyte in cancer treatment.
The application of the cancer treatment drug containing the kidney cancer antigen CA9 protein specific cytotoxic T lymphocyte in cancer treatment.
Advantageous effects
The DC cell is infected by rAAV2-siSOCS1-CA9 recombinant virus vector with siSOCS1 and CA9 target genes, the DC cell is subjected to gene modification to obtain a renal cancer antigen CA9 protein specific DC cell, the CA9 specificity is transferred to the T cell by the DC iAPA-CA9-DC cell of the in vitro antigen presenting cell, and a large number of CA9 specific cytotoxic T lymphocyte iAPA-CA9-DC/CTL cells are obtained through amplification, so that the aim of treating cancer is fulfilled.
Drawings
FIG. 1 shows the construction of rAAV2-siSOCS1-CA 9.
FIG. 2 flow cytometry detected the expression of CA9 in transfected DCs. A. A control group; B. rAAV2-siSOCS1-CA9 transfection group.
Figure 3 flow cytometry detected hla.a 2402/CA9 tetramer specificity of CTLs. A. A control group; B. rAAV2-siSOCS1-CA9 transfection group
FIG. 4. iAPA-CA9-DC/CTL cell proliferation potency was compared with that of T cell.
FIG. 5. iAPA-CA9-DC/CTL cell and T cell phenotypic changes. A. CD (compact disc)3 +/CD4 +;B. CD3 +/CD8 +;C. CD3 +/CD56 +. *P < 0.05, **P < 0.01 vs T cells。
Detailed Description
In order to explain the technical contents of the invention, the objects and effects achieved by the invention in detail, the following description is given with reference to the embodiments.
Example 1 preparation of Kidney cancer antigen CA9 protein-specific cytotoxic T lymphocytes
The preparation method of the kidney cancer antigen CA9 protein specific cytotoxic T lymphocyte comprises the following steps:
(1) preparing rAAV2-siSOCS1-CA9 recombinant virus vector carrying siSOCS1 and CA9 target genes:
the target gene sequence in the recombinant virus vector is as follows: u6 promoter-SOCS 1 siRNA and SOCS1 siRNA reverse sequence-CMV promoter-CA 9 gene-IRES sequence-Flagellin Flagellin gene sequence, rAAV2-siSOCS1-CA9 construction scheme as shown in figure 1. The sequence of the SOCS1 siRNA is shown as SEQ ID NO.1, the reverse sequence of SOCS1 siRNA is shown as SEQ ID NO.2, the sequence of SOCS1 siRNA and the reverse sequence of SOCS1 siRNA are complementary in reverse direction, and both ends of a gene segment are respectively provided with BamH 1/HindIII enzyme cutting sites; the CA9 protein gene sequence, the IRES sequence and the Flagellin Flagellin gene sequence are respectively shown as SEQ ID NO.3, SEQ ID NO.4 and SEQ ID NO. 5; the target gene is assembled with an adeno-associated virus vector type 2 (AAV 2) to obtain the rAAV2-siSOCS1-CA9 recombinant virus vector. The assembly of the target gene and the adeno-associated virus vector is completed by Hemanshun Biotechnology Ltd.
(2) Isolation, differentiation, maturation and infection of DC cells
Mixing whole blood, physiological saline and lymphocyte separation solution at a ratio of 1:1:1, centrifuging at a centrifugal force of 650g for 15min, and increasing and decreasing at a speed of 4 and 0. After centrifugation, the mononuclear cell layer above the lymphocyte separation solution is extracted and washed twice.
Differentiation and Induction of DC cells and preparation of iAPA-CA 9-DC: adjustment of monocyte density to 5X 106cell/ml according to 106 cell/cm2The cell number of (2) was added to a culture flask at 37 ℃ with 5% CO2Culturing for 2h in a concentration incubator, slightly shaking the culture bottle, shaking the non-adherent cells, sucking away the upper layer of the non-adherent cells, centrifugally collecting, and freezing. KBM581 medium containing 2 IU/mL IL-2, 50 ng/mL rmGM-CSF, and 25 ng/mL rmIL-4 was added to the original flask at 37 deg.C with 5% CO2Culturing in a concentration incubator. After overnight culture, gently shaking the culture bottle at 2d, shaking the non-adherent cells, sucking away the upper layer of non-adherent cells, centrifugally collecting, and freezing again. Adding KBM581 culture medium containing 2 IU/mL IL-2, 50 ng/mL rmGM-CSF and 25 ng/mL rmIL-4 into the DC cell culture bottle again, and continuing to culture for 3 d; when the culture reached 5d, the medium was completely changed to KBM581 medium containing 1.2. mu.g/mL of LPS, and the culture was continued for 48 hours. Collecting suspension cells, calculating the total amount of the cells, infecting rAAV2-siSOCS1-CA9 recombinant virus vectors according to MOI 100, adding rAAV2-siSOCS1-CA9 recombinant virus vectors, infecting for 2h, changing into fresh KBM581 culture medium containing 2 IU/mL IL-2, 50 ng/mL rmGM-CSF and 25 ng/mL rmIL-4, continuously culturing for 48h to obtain iAPA-CA9-DC cells, and identifying the recombinant virus infection condition of rAAV2-siSOCS1-CA9 by flow. The results are shown in FIG. 2, and the results in FIG. 2 show that the rAAV2-siSOCS1-CA9 recombinant virus-infected DC cell CA9 is successfully expressed compared with the control group DC cell which is not infected with the rAAV2-siSOCS1-CA9 recombinant virus. The results in FIG. 2 show that the rAAV2-siSOCS1-CA9 recombinant viral vector successfully transfects DC cells, and the iAPA-CA9-DC ratio is as high as 83.4% (FIG. 2B).
(3) Preparation of renal carcinoma antigen CA9 protein specific cytotoxic T lymphocyte.
Adding stimulating factor CD3 Monoclonal antibody 50 ng/ml, CD 2850 ng/mL of monoclonal antibody and IL-21000U/mL of monoclonal antibody stimulate the proliferation of T cells. After the T cells are stimulated and cultured for 48h, the iAPA-CA9-DC cells and the T cells are harvested and mixed and cultured according to the proportion of 1:5, and the cell density is adjusted to be 1 multiplied by 106cell/mL, 1000U/mLIL2 in KBM581, iAPA-CA9-DC/CTL cells were obtained. And (4) performing fluid infusion operation according to the cell growth condition, and periodically sampling and tracking the cell proliferation condition, the survival rate and the phenotype.
The results in fig. 3 show that the positive rate of hla. a. x 2402/CA9 tetramers of the T cells obtained by iAPA-CA9-DC cell stimulation, i.e., iAPA-CA9-DC/CTL cells (fig. 3B), was significantly different from the control group (fig. 3A).
FIG. 4 iAPA-CA9-DC/CTL comparison with T cell proliferation potency. The result shows that the proliferation capacity of the iAPA-CA9-DC/CTL cell is obviously higher than that of the T cell.
The results in FIG. 5 show that compared with the killing activity of T cells on target cells CA9-293FT, iAPA-CA9-DC/CTL cells have significantly enhanced specific cell killing toxicity of iAPA-CA9-DC/CTL cells on target cells CA9-293FT compared with non-specific CTL in a control group.
SEQUENCE LISTING
<110> quan State college of education
<120> preparation method of renal cancer antigen CA9 protein specific cytotoxic T lymphocyte
<130> 5
<160> 5
<170> PatentIn version 3.3
<210> 1
<211> 19
<212> DNA
<213> SEQ ID NO.1
<400> 1
cacgcacttc cgcacattc 19
<210> 2
<211> 67
<212> DNA
<213> SEQ ID NO.2
<400> 2
cgggatccgc acgcacttcc gcacattctt caagagagaa tgtgcggaag tgcgtgtttt 60
taagctt 67
<210> 3
<211> 1380
<212> DNA
<213> SEQ ID NO.3
<400> 3
atggctcccc tgtgccccag cccctggctc cctctgttga tcccggcccc tgctccaggc 60
ctcactgtgc aactgctgct gtcactgctg cttctggtgc ctgtccatcc ccagaggttg 120
ccccggatgc aggaggattc ccccttggga ggaggctctt ctggggaaga tgacccactg 180
ggcgaggagg atctgcccag tgaagaggat tcacccagag aggaggatcc acccggagag 240
gaggatctac ctggagagga ggatctacct ggagaggagg atctacctga agttaagcct 300
aaatcagaag aagagggctc cctgaagtta gaggatctac ctactgttga ggctcctgga 360
gatcctcaag aaccccagaa taatgcccac agggacaaag aaggggatga ccagagtcat 420
tggcgctatg gaggcgaccc gccctggccc cgggtgtccc cagcctgcgc gggccgcttc 480
cagtccccgg tggatatccg cccccagctc gccgccttct gcccggccct gcgccccctg 540
gaactcctgg gcttccagct cccgccgctc ccagaactgc gcctgcgcaa caatggccac 600
agtgtgcaac tgaccctgcc tcctgggcta gagatggctc tgggtcccgg gcgggagtac 660
cgggctctgc agctgcatct gcactggggg gctgcaggtc gtccgggctc ggagcacact 720
gtggaaggcc accgtttccc tgccgagatc cacgtggttc acctcagcac cgcctttgcc 780
agagttgacg aggccttggg gcgcccggga ggcctggccg tgttggccgc ctttctggag 840
gagggcccgg aagaaaacag tgcctatgag cagttgctgt ctcgcttgga agaaatcgct 900
gaggaaggct cagagactca ggtcccagga ctggacatat ctgcactcct gccctctgac 960
ttcagccgct acttccaata tgaggggtct ctgactacac cgccctgtgc ccagggtgtc 1020
atctggactg tgtttaacca gacagtgatg ctgagtgcta agcagctcca caccctctct 1080
gacaccctgt ggggacctgg tgactctcgg ctacagctga acttccgagc gacgcagcct 1140
ttgaatgggc gagtgattga ggcctccttc cctgctggag tggacagcag tcctcgggct 1200
gctgagccag tccagctgaa ttcctgcctg gctgctggtg acatcctagc cctggttttt 1260
ggcctccttt ttgctgtcac cagcgtcgcg ttccttgtgc agatgagaag gcagcacaga 1320
aggggaacca aagggggtgt gagctaccgc ccagcagagg tagccgagac tggagcctag 1380
<210> 4
<211> 594
<212> DNA
<213> SEQ ID NO.4
<400> 4
taaattccgg cccctctccc tccccccccc ctaacgttac tggccgaagc cgcttggaat 60
aaggccggtg tgcgtttgtc tatatgttat tttccaccat attgccgtct tttggcaatg 120
tgagggcccg gaaacctggc cctgtcttct tgacgagcat tcctaggggt ctttcccctc 180
tcgccaaagg aatgcaaggt ctgttgaatg tcgtgaagga agcagttcct ctggaagctt 240
cttgaagaca aacaacgtct gtagcgaccc tttgcaggca gcggaacccc ccacctggcg 300
acaggtgcct ctgcggccaa aagccacgtg tataagatac acctgcaaag gcggcacaac 360
cccagtgcca cgttgtgagt tggatagttg tggaaagagt caaatggctc tcctcaagcg 420
tattcaacaa ggggctgaag gatgcccaga aggtacccca ttgtatggga tctgatctgg 480
ggcctcggtg cacatgcttt acatgtgttt agtcgaggtt aaaaaaacgt ctaggccccc 540
cgaaccacgg ggacgtggtt ttcctttgaa aaacacgatg ataatatggc caca 594
<210> 5
<211> 1492
<212> DNA
<213> SEQ ID NO.5
<400> 5
atggcacaag tcattaatac aaacagcctg tcgctgttga cccagaataa cctgaacaaa 60
tcccagtccg ctctgggcac cgctatcgag cgtctgtctt ccggtctgcg tatcaacagc 120
gcgaaagacg atgcggcagg tcaggcgatt gctaaccgtt ttaccgcgaa catcaaaggt 180
ctgactcagg cttcccgtaa cgctaacgac ggtatctcca ttgcgcagac cactgaaggc 240
gcgctgaacg aaatcaacaa caacctgcag cgtgtgcgtg aactggcggt tcagtctgct 300
aacagcacca actcccagtc tgacctcgac tccatccagg ctgaaatcac ccagcgcctg 360
aacgaaatcg accgtgtatc cggccagact cagttcaacg gcgtgaaagt cctggcgcag 420
gacaacaccc tgaccatcca ggttggtgcc aacgacggtg aaactatcga tatcgatctg 480
aagcagatca actctcagac cctgggtctg gatacgctga atgtgcaaca aaaatataag 540
gtcagcgata cggctgcaac tgttacagga tatgccgata ctacgattgc tttagacaat 600
agtactttta aagcctcggc tactggtctt ggtggtactg accagaaaat tgatggcgat 660
ttaaaatttg atgatacgac tggaaaatat tacgccaaag ttaccgttac ggggggaact 720
ggtaaagatg gctattatga agtttccgtt gataagacga acggtgaggt gactcttgct 780
ggcggtgcga cttccccgct tacaggtgga ctacctgcga cagcaactga ggatgtgaaa 840
aatgtacaag ttgcaaatgc tgatttgaca gaggctaaag ccgcattgac agcagcaggt 900
gttaccggca cagcatctgt tgttaagatg tcttatactg ataataacgg taaaactatt 960
gatggtggtt tagcagttaa ggtaggcgat gattactatt ctgcaactca aaataaagat 1020
ggttccataa gtattaatac tacgaaatac actgcagatg acggtacatc caaaactgca 1080
ctaaacaaac tgggtggcgc agacggcaaa accgaagttg tttctattgg tggtaaaact 1140
tacgctgcaa gtaaagccga aggtcacaac tttaaagcac agcctgatct ggcggaagcg 1200
gctgctacaa ccaccgtttt aaaacccgct gcagaaaatt gatgctgctt tggcacaggt 1260
tgacacgtta cgttctgacc tgggtgcggt acagaaccgt ttcaactccg ctattaccaa 1320
cctgggcaac accgtaaaca acctgacttc tgcccgtagc cgtatcgaag attccgacta 1380
cgcgaccgaa gtttccaaca tgtctcgcgc gcagattctg cagcaggccg gtacctccgt 1440
tctggcgcag gcgaaccagg ttccgcaaaa cgtcctctct ttactgcgtt aa 1492

Claims (8)

1. The preparation method of the kidney cancer antigen CA9 protein specific cytotoxic T lymphocyte is characterized by comprising the following steps:
(1) preparing a rAAV2-siSOCS1-CA9 recombinant virus vector carrying siSOCS1 and CA9 target genes;
(2) infecting, differentiating and inducing DC cells to mature by the rAAV2-siSOCS1-CA9 recombinant virus vector to obtain iAPA-CA9-DC cells;
(3) the iAPA-CA9-DC cell and the T cell are mixed and cultured to obtain the iAPA-CA9-DC/CTL cell, namely the kidney cancer antigen CA9 protein specific cytotoxic T lymphocyte.
2. The culture method according to claim 1, wherein: the target gene sequences carrying the target siSOCS1 and CA9 in the step (1) are sequentially as follows: a U6 promoter sequence, a SOCS1 siRNA sequence, a SOCS1 siRNA reverse sequence, a CMV promoter sequence, a CA9 protein gene sequence, an IRES sequence, a Flagellin Flagellin gene sequence; the sequence of the SOCS1 siRNA is shown as SEQ ID NO.1, the reverse sequence of the SOCS1 siRNA is shown as SEQ ID NO.2, the sequence of the SOCS1 siRNA and the reverse sequence of the SOCS1 siRNA are reversely complementary; the target gene is assembled with AAV2 as the adeno-associated virus 2 vector to obtain rAAV2-siSOCS1-CA9 recombinant virus vector.
3. The method of claim 1, wherein: in the step (2), the rAAV2-siSOCS1-CA9 recombinant virus vector is used for infecting, differentiating and inducing the DC cells to mature, and the rAAV2-siSOCS1-CA9 recombinant virus vector is used for infecting the DC cells according to MOI 100.
4. The method of claim 1, wherein: in the step (3), the mixed culture of the iAPA-CA9-DC cells and the T cells comprises the following specific steps:
1) adding stimulating factor CD3Monoclonal antibody 50 ng/ml, CD2850 ng/mL of monoclonal antibody and IL-21000U/mL of monoclonal antibody stimulate the proliferation of T cells;
2) after the T cells are cultured for 48h by stimulating proliferation, iAPA-CA9-DC cells and the T cells are mixed and cultured according to the proportion of 1:5, and the cell density is adjusted to 1 multiplied by 106cell/mL, KBM581 media supplemented with 1000U/mL IL 2; iAPA-CA9-DC/CTL cells were obtained.
5. A renal cancer antigen CA9 protein-specific cytotoxic T lymphocyte prepared by the method of claim 1.
6. A cancer therapeutic agent comprising the renal cancer antigen CA9 protein-specific cytotoxic T lymphocyte of claim 5.
7. The renal cancer antigen CA9 protein-specific cytotoxic T lymphocyte of claim 5, for use in biological fields of non-disease diagnosis and treatment.
8. The use of the cancer therapeutic agent comprising the renal cancer antigen CA9 protein-specific cytotoxic T lymphocyte of claim 6 in the fields of non-disease diagnosis and therapeutic biology.
CN202111598162.6A 2021-12-24 2021-12-24 Preparation method of renal cancer antigen CA9 protein specific cytotoxic T lymphocyte Pending CN114277056A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111598162.6A CN114277056A (en) 2021-12-24 2021-12-24 Preparation method of renal cancer antigen CA9 protein specific cytotoxic T lymphocyte

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111598162.6A CN114277056A (en) 2021-12-24 2021-12-24 Preparation method of renal cancer antigen CA9 protein specific cytotoxic T lymphocyte

Publications (1)

Publication Number Publication Date
CN114277056A true CN114277056A (en) 2022-04-05

Family

ID=80874938

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111598162.6A Pending CN114277056A (en) 2021-12-24 2021-12-24 Preparation method of renal cancer antigen CA9 protein specific cytotoxic T lymphocyte

Country Status (1)

Country Link
CN (1) CN114277056A (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104004712A (en) * 2014-04-24 2014-08-27 深圳市合一康生物科技有限公司 Preparation method of antigen-specific cytotoxicity T lymphocytes
WO2019016360A1 (en) * 2017-07-21 2019-01-24 Cellectis Engineered immune cells resistant to tumor microoenvironment
CN111560351A (en) * 2020-04-14 2020-08-21 山东省齐鲁细胞治疗工程技术有限公司 CA9 tumor antigen specific cytotoxic T lymphocyte

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104004712A (en) * 2014-04-24 2014-08-27 深圳市合一康生物科技有限公司 Preparation method of antigen-specific cytotoxicity T lymphocytes
WO2019016360A1 (en) * 2017-07-21 2019-01-24 Cellectis Engineered immune cells resistant to tumor microoenvironment
CN111560351A (en) * 2020-04-14 2020-08-21 山东省齐鲁细胞治疗工程技术有限公司 CA9 tumor antigen specific cytotoxic T lymphocyte

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
汪忠镐著: "《汪忠镐血管外科学》", vol. 2010, 浙江科学技术出版社, pages: 205 *

Similar Documents

Publication Publication Date Title
US9844508B2 (en) Tumor vaccine and method for producing the same
CN112830974B (en) Chimeric antigen receptor, carrier, human dendritic cell, cell line, solid tumor treatment drug, preparation method and application
WO2003073998A2 (en) Local production and/or delivery of anti-cancer agents by stromal cell precursors
CN109468283A (en) A kind of immunocyte of the dual Chimeric antigen receptor gene modification based on CD19 and BCMA and its application
JPWO2007040105A1 (en) Method for producing T cell population
CN109456945A (en) A kind of application for improving DC cell HPV viruse antigen and offering DC cell after the gene modification method and gene modification of efficiency
EA010434B1 (en) Process for producing cytotoxic lymphocyte
CN113416260B (en) Claudin18.2-targeted specific chimeric antigen receptor cell and preparation method and application thereof
WO2017071173A1 (en) Tumor therapeutic agent modified by il-12/cd62l fusion protein and preparation method and use thereof
EA012520B1 (en) Process for producing cytotoxic lymphocytes
CN112226463B (en) Plasmid combination and application thereof in preparation of modified immune cells
CN104001185A (en) Preparation method of specific dendritic cell vaccine of CEA positive tumor
CN104524560A (en) Dendritic cell tumor vaccine and preparation method and application thereof
CN116478932A (en) Genetically modified immune cells and uses thereof
CN114277056A (en) Preparation method of renal cancer antigen CA9 protein specific cytotoxic T lymphocyte
CN114934071B (en) CAR vector expressing immune regulation factor and application thereof
CN112972666B (en) Preparation method of personalized gene modified tumor DC vaccine
CN109963862B (en) Polypeptides and uses thereof
CN111560351A (en) CA9 tumor antigen specific cytotoxic T lymphocyte
CN112795584B (en) Nucleic acid resisting GCC, preparation method thereof, immune cell with nucleic acid and application thereof
CN112029730A (en) Genetically modified mesenchymal stem cell and application thereof
Huang et al. GP96 and SMP30 protein priming of dendritic cell vaccination induces a more potent CTL response against hepatoma
JP7068702B2 (en) The adjuvant composition and its use
CN110139875B (en) COL14A 1-derived tumor antigen polypeptide 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