CN111249307A - T cell with effect of improving immune response and preparation method and application thereof - Google Patents

T cell with effect of improving immune response and preparation method and application thereof Download PDF

Info

Publication number
CN111249307A
CN111249307A CN201811464461.9A CN201811464461A CN111249307A CN 111249307 A CN111249307 A CN 111249307A CN 201811464461 A CN201811464461 A CN 201811464461A CN 111249307 A CN111249307 A CN 111249307A
Authority
CN
China
Prior art keywords
cells
cell
1bbl
cd40l
immune response
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
CN201811464461.9A
Other languages
Chinese (zh)
Inventor
冷启彬
张朔
黄狄文
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.)
Guangzhou Bright Medical Technology Co ltd
Original Assignee
Guangzhou Bright Medical Technology 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 Guangzhou Bright Medical Technology Co ltd filed Critical Guangzhou Bright Medical Technology Co ltd
Priority to CN201811464461.9A priority Critical patent/CN111249307A/en
Publication of CN111249307A publication Critical patent/CN111249307A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants

Abstract

The invention relates to a T cell with an effect of improving immune response and a preparation method and application thereof. The T cell with the effect of improving the immune response is prepared by integrating a foreign gene 4-1BBL on an antigen-specific T cell chromosome for overexpression. The T cell with the effect of improving the immune response has the advantages of fast self proliferation and high expression quantity of IFN-gamma, and can obviously promote the proliferation and the expression level of the IFN-gamma of the surrounding antigen-specific T cell without carrying foreign genes, play a role in guiding the general in the aspect of activating the immune response capability of the surrounding antigen-specific T cell, and play a vital application in treating tumors, chronic viral infection and bacterial infection.

Description

T cell with effect of improving immune response and preparation method and application thereof
Technical Field
The invention relates to the technical field of T cells, in particular to a T cell with an effect of improving immune response and a preparation method and application thereof.
Background
T cell therapy techniques currently used to improve immunity include expanded T cell therapy techniques, CAR-T cell therapy techniques, and TCR-T cell therapy techniques. The first technique is simply to expand a patient's specific cells in large numbers for treatment, with no substantial change in T cell function. CAR-T and TCR-T cell therapy techniques are to express artificially modified or screened T cell receptors to enhance the recognition ability to tumors or viruses, thereby achieving the purpose of eliminating viruses and tumors. None of these three existing T cell therapy techniques can enhance the function of other immune cells in the body that are not engineered and therefore are not effective in the treatment of chronic viral infections and solid tumors.
Tumors can be said to be the first leading killer of today's society affecting human health, with almost millions of people dying from cancer each year. The current treatment means for tumors include chemotherapy and radiotherapy besides early surgical excision. Immunotherapy has advanced dramatically in recent years, including immune checkpoint inhibitors and adoptive cell therapy. The former mainly includes PD-L1 and CTLA-4 antibody, the latter mainly refers to TCR-T and CAR-T technologies. Wherein the former can induce the body immune response to the utmost extent by inhibiting the inhibitory signal pathway of immune cells, thereby eliminating the tumor. However, tumor antigens are generally low in affinity, and even if immunosuppressive signals are suppressed, strong immune responses are difficult to stimulate, so that the tumor antigens only play a certain role in treating individual tumors. In the latter, T cells with tumor antigen recognition ability are obtained mainly by screening TCR-T, then TCR sequences of the T cells are obtained, then the patient-derived T cells also express the TCR capable of recognizing the tumor antigen through gene modification technology, and finally a large amount of cells are obtained through in vitro culture and returned to treat patients. However, the affinity of this TCR to tumor antigens naturally occurring in the body is also low and it is difficult to provoke an effective immune response. CAR-T is a chimeric antigen receptor which has high affinity to tumor antigens and is expressed on T cells of patients through a genetic modification technology, and finally, a large number of cells are obtained through in vitro culture and are returned to treat the patients. The advantages are that the receptor has high affinity to antigen, gets rid of the restriction of MHC molecules, can directly recognize tumor antigen, and greatly improves the killing ability to tumor cells. The disadvantage is that it may cause a cytokine storm in the patient and is neurotoxic. Cytokine storms, often manifested as high fever, low blood pressure, shock, etc., occur at rates in excess of 60% to 70%, with the more severe cytokine storms occurring at rates of about 20% to 30%. Drugs such as glucocorticoids, IL-6 antibodies, etc. are generally required for control. The nervous system toxicity is mainly manifested by encephaledema, intracranial pressure rise, epilepsy, change of consciousness state and the like, the occurrence mechanism of the toxicity is not clear at present, and the reports that patients have gone to the world due to the CAR-T-induced neurotoxicity appear at times abroad. Moreover, CAR-T charges are close to the daily value and are difficult to popularize among patients. The most important point is that it is difficult to find a receptor molecule which is specifically expressed only on tumor cells, and the therapeutic effect on solid tumors is general, and only CAR-19 and CAR-20 have good curative effect in leukemia patients.
Chronic viral infections include mainly HIV and HBV. Immunity against HIV infection: HIV mainly infects human CD4+T cells, leading to severe immunodeficiency. According to the '2005 global AIDS transmission report' of the United nations, the name is: since the confirmation of HIV viral infection in 1981, 6500 million of the accumulated infected population has caused 2800 million deaths, and 3800 million of the existing AIDS patients; 280 million patients die in 2005, with about 400 million new infected population; at present, the number of people actually infected in China exceeds 100 thousands, and the Chinese medicine enters a rapid growth period. CD4 for AIDS+T cells are destroyed resulting in an immune deficiency, so patients are often accompanied by a broad spectrum of opportunistic viral, parasitic and cellular infections. Disruption of CD4 by HIV infection+T cells, the patient's immunity against HIV infection is always in an abnormal immunosuppressive state. In the early stages of infection, HIV infects CD4+T cells undergo extensive replication, at which point B cells can induce a strong immune response. But also requires CD4 for humoral immunity+Th cells assist, and thus the immune response of B cells is subsequently suppressed. Later HIV reduces replication and the immune response continues to be suppressed. There is no very effective method for treating and preventing AIDS because of the resistance to infectionThe key regulation and control cells of the infection immunity are infected and destroyed, so that the anti-infection adaptive immune response cannot be effectively induced. Immunity against HBV infection: human Hepatitis B Virus (HBV) infection is a worldwide public health problem. Due to the presence of MHC polymorphisms and susceptibility patterns, as well as mutations or polymorphisms of other key genes, there are approximately 1.2 million asymptomatic carriers in china that are easily transformed into HBV chronic patients when immunity is reduced. Chronic infection with HBV can last for ten years, and liver tissues of patients are repeatedly damaged, and gradually develop into cirrhosis and liver cancer. The long-term progression of chronic hepatitis b is the result of both escape of HBV from anti-infective immunity and immune tolerance of the host immune system to HBV. Typical chronic infection with HBV can be divided into four stages with time of infection: an immune tolerance phase, an immune response phase, an inactive carrier phase after HBeAg turns negative, and a second acute attack phase. The root cause of chronic infection is that HBV can effectively escape anti-infection immunity, the immune system of a patient can not eliminate HBV infection all the time, and currently, no effective treatment means for HBV exists.
Intracellular bacterial infections such as tubercle bacillus, listeria, etc. Macrophages can phagocytose intracellular bacteria, but because most of the intracellular bacteria can escape phagocytosis and killing, the macrophages cannot lyse the bacteria, and instead cause the concealment and diffusion of the bacteria. Because specific antibodies cannot neutralize intracellular bacteria, intracellular bacterial infection is primarily dependent on cellular immunity. Wherein CD4+INF-gamma secreted by Th cells can activate the bactericidal function of macrophages, and CD8CTL can effectively kill target cells, so that intracellular bacteria are released and phagocytosed by the macrophages after the intracellular bacteria are conditioned by antibodies. Tubercle bacillus can suppress immune responses in a variety of ways: inhibit the early phagocyte formation process of phagocytes and, in addition, inhibit the release of antigen from phagosomes, thereby failing to effectively induce MHC-I presentation of antigen and activate CD8CTL immune response.
In the present situation, immune checkpoint inhibitor therapy against tumors is only effective for individual tumors because T cells of the body have low affinity for tumor antigens and even if the inhibitory response against T cells is suppressed, the body hardly has a strong immune response against tumor antigens. Importantly, it is difficult to find such receptor molecules that are specifically expressed only on tumor cells, and the therapeutic effect on solid tumors is poor. Viral infection and bacterial infection can only prolong the life cycle of patients by inhibiting the replication of viruses through medicaments, and no good radical treatment means exists. The fundamental reason is the body's naturally occurring CD8+T cells have too low an affinity for antigens to effectively recognize tumor cells and virus-infected target cells and thus effectively eliminate tumors and viruses.
In conclusion, there is no effective treatment for tumor, chronic viral infection (such as HIV, HBV), bacterial infection (such as tubercle bacillus, superbacteria) and the treatment cost is expensive.
Accordingly, it would be desirable to provide a T cell that is capable of enhancing the T cell immune response of an organism (e.g., a patient).
Disclosure of Invention
Based on this, the main object of the present invention is to provide a T cell with an immune response-enhancing effect, particularly a CD8 with an immune response-enhancing effect+T cells. The T cell can recognize and kill target cells and simultaneously stimulate immune responses of other antigen-specific T cells.
The purpose of the invention is realized by the following technical scheme:
a T cell with the function of improving immune response is prepared by integrating a foreign gene 4-1BBL on an antigen-specific T cell chromosome for overexpression. The antigen mentioned here includes viruses, bacteria, fungi, tumors, etc., and may be a polypeptide, a protein, or a substance capable of inducing a T cell immune response by gene expression in cells and the above listed microorganisms.
Another objective of the invention is to provide a method for preparing T cells with the efficacy of improving immune response.
The technical purpose is realized by the following technical scheme:
a method for preparing T cells with the effect of improving immune response comprises the following steps:
obtaining antigen specific T cells and activating the antigen specific T cells with specific antigens;
constructing a viral particle carrying the foreign gene 4-1BBL, transfecting said activated antigen-specific T cells with said viral particle, thereby integrating the foreign gene 4-1BBL into the chromosome of said activated antigen-specific T cells.
In some embodiments, the viral particle is constructed using a retrovirus.
In some of these embodiments, the retrovirus is Migr 1.
In some of these embodiments, the viral particle is constructed from a lentivirus.
In some of these embodiments, the antigen-specific T cell is a CD8+ T cell.
In some of these embodiments, the specific antigen is a polypeptide recognized by antigen-specific T cells. The specific antigen refers to polypeptides which are derived from tumors, viruses, bacteria and the like and can be recognized by T cells, and polypeptides which are derived from tumors, viruses, bacteria and the like and are expressed by genes. Such as N4.
It is still another object of the present invention to provide a use of the above-mentioned T cell having an immune response-enhancing effect for promoting an immune response function of an immune cell adjacent to the T cell. Such as T cells, DC cells, macrophages, NK cells, B cells, and the like. The immune response function includes promotion of cytokine secretion, antigen presentation, killing function and the like.
In some embodiments, the use is co-culturing the immune cells with the T cells that enhance the efficacy of an immune response.
In some embodiments, it is a further object of the present invention to provide a use of the above-mentioned T cell having an efficacy of enhancing an immune response for treating tumors, chronic viral infections, bacterial infections, fungal infections.
Compared with the prior art, the invention has the following beneficial effects:
the invention integrates exogenous gene 4-1BBL to the chromosome of the antigen specific T cell to carry out over-expression to prepare the T cell with the effect of improving immune response. The T cell with the effect of improving the immune response has the advantages of fast self proliferation and high expression quantity of IFN-gamma, and can obviously promote the proliferation and IFN-gamma expression level of the peripheral antigen-specific T cells without carrying foreign genes (for example, the antigen-specific T cells which are co-cultured with the T cell and do not carry foreign genes are returned into tumor tissues and then the antigen-specific T cells in the tumor tissues), play a role in activating the immune response capacity of the peripheral antigen-specific T cells, and play a role in treating tumors, chronic viral infection and bacterial infection.
Drawings
FIG. 1 is an electrophoretogram showing the amplification result of example 1;
FIGS. 2 to 7 are maps of foreign gene-carrying retrovirus plasmids constructed in example 1;
FIG. 8 is a schematic diagram of the operation route of example 2;
FIGS. 9 and 10 show the detection of OT-I CD8 by flow cytometry and RT-PCR in example 2+Expression results of T cells CD40L and 4-1 BBL;
FIG. 11 is a schematic view of the operational flow and the result chart of example 3;
FIG. 12 is a schematic view of the operational flow and the result chart of example 4;
FIG. 13 is a schematic view of the operational flow and the result chart of example 5;
FIG. 14 is a flowchart showing the operation of example 6;
FIG. 15 and FIG. 16 are graphs showing the results of the flow assay in example 6;
FIG. 17 is a schematic view of the operational procedure and a result chart of example 7.
Detailed Description
The present invention will be described in further detail with reference to specific examples. In order to clearly understand the technical contents of the present invention, the following examples are given in detail. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, generally followed by conventional conditions, such as Sambrook et al, molecular cloning: the conditions described in the Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or according to the manufacturer's recommendations. The various reagents used in the examples are commercially available.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. The terminology used in the description of the invention herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used herein, the term "and/or" includes any and all combinations of one or more of the associated listed items.
Example 1 preparation of a retrovirus expression plasmid carrying a foreign Gene
1. Constructing clone: (1) total RNA from mouse spleen cells and DC cells (dendritic cells) was extracted and reverse-transcribed to give a cDNA library. (2) The cDNA library is used as a template, the sequences of CD40L and 4-1BBL are amplified by PCR, the sequence of IERS is amplified by PCR by using Migr1 as a vector, and the 2A sequence synthesized by the company is used as a template for PCR amplification of the 2A sequence. The method comprises the following steps:
primers used for PCR amplification include:
Figure BDA0001889363430000051
the PCR amplification system is as follows:
Figure BDA0001889363430000052
Figure BDA0001889363430000061
the procedure used for PCR amplification was:
Figure BDA0001889363430000062
the sequences of CD40L (SEQ ID No.19) and 4-1BBL (SEQ ID No.20) obtained by PCR amplification (see FIG. 1) are respectively shown as follows:
SEQ ID No.19:
ATGATAGAAACATACAGCCAACCTTCCCCCAGATCCGTGGCAACTGGACTTCCAGCGAGCATGAAGATTTTTATGTATTTACTTACTGTTTTCCTTATCACCCAAATGATTGGATCTGTGCTTTTTGCTGTGTATCTTCATAGAAGATTGGATAAGGTCGAAGAGGAAGTAAACCTTCATGAAGATTTTGTATTCATAAAAAAGCTAAAGAGATGCAACAAAGGAGAAGGATCTTTATCCTTGCTGAACTGTGAGGAGATGAGAAGGCAATTTGAAGACCTTGTCAAGGATATAACGTTAAACAAAGAAGAGAAAAAAGAAAACAGCTTTGAAATGCAAAGAGGTGATGAGGATCCTCAAATTGCAGCACACGTTGTAAGCGAAGCCAACAGTAATGCAGCATCCGTTCTACAGTGGGCCAAGAAAGGATATTATACCATGAAAAGCAACTTGGTAATGCTTGAAAATGGGAAACAGCTGACGGTTAAAAGAGAAGGACTCTATTATGTCTACACTCAAGTCACCTTCTGCTCTAATCGGGAGCCTTCGAGTCAACGCCCATTCATCGTCGGCCTCTGGCTGAAGCCCAGCAGTGGATCTGAGAGAATCTTACTCAAGGCGGCAAATACCCACAGTTCCTCCCAGCTTTGCGAGCAGCAGTCTGTTCACTTGGGCGGAGTGTTTGAATTACAAGCTGGTGCTTCTGTGTTTGTCAACGTGACTGAAGCAAGCCAAGTGATCCACAGAGTTGGCTTCTCATCTTTTGGCTTACTCAAACTCTGA
SEQ ID No.20:
ATGGACCAGCACACACTTGATGTGGAGGATACCGCGGATGCCAGACATCCAGCAGGTACTTCGTGCCCCTCGGATGCGGCGCTCCTCAGAGATACCGGGCTCCTCGCGGACGCTGCGCTCCTCTCAGATACTGTGCGCCCCACAAATGCCGCGCTCCCCACGGATGCTGCCTACCCTGCGGTTAATGTTCGGGATCGCGAGGCCGCGTGGCCGCCTGCACTGAACTTCTGTTCCCGCCACCCAAAGCTCTATGGCCTAGTCGCTTTGGTTTTGCTGCTTCTGATCGCCGCCTGTGTTCCTATCTTCACCCGCACCGAGCCTCGGCCAGCGCTCACAATCACCACCTCGCCCAACCTGGGTACCCGAGAGAATAATGCAGACCAGGTCACCCCTGTTTCCCACATTGGCTGCCCCAACACTACACAACAGGGCTCTCCTGTGTTCGCCAAGCTACTGGCTAAAAACCAAGCATCGTTGTGCAATACAACTCTGAACTGGCACAGCCAAGATGGAGCTGGGAGCTCATACCTATCTCAAGGTCTGAGGTACGAAGAAGACAAAAAGGAGTTGGTGGTAGACAGTCCCGGGCTCTACTACGTATTTTTGGAACTGAAGCTCAGTCCAACATTCACAAACACAGGCCACAAGGTGCAGGGCTGGGTCTCTCTTGTTTTGCAAGCAAAGCCTCAGGTAGATGACTTTGACAACTTGGCCCTGACAGTGGAACTGTTCCCTTGCTCCATGGAGAACAAGTTAGTGGACCGTTCCTGGAGTCAACTGTTGCTCCTGAAGGCTGGCCACCGCCTCAGTGTGGGTCTGAGGGCTTATCTGCATGGAGCCCAGGATGCATACAGAGACTGGGAGCTGTCTTATCCCAACACCACCAGCTTTGGACTCTTTCTTGTGAAACCCGACAACCCATGGGAATGA。
cloning and constructing an enzyme digestion system of a required vector and a PCR product:
MIGR1-CD40L reaction System:
Figure BDA0001889363430000071
both were digested at 37 ℃ overnight to ensure full cleavage.
MIGR1-4-1BBL reaction System:
Figure BDA0001889363430000072
both were digested at 37 ℃ overnight to ensure full cleavage.
Migr1-CD40L-IRES-4-1BBL reaction system:
Figure BDA0001889363430000073
the cleavage was carried out overnight at 37 ℃ to ensure complete cleavage.
Migr1-CD40L-P2A-4-1BBL reaction system:
Figure BDA0001889363430000074
Figure BDA0001889363430000081
the digestion was carried out overnight at 37 ℃ to ensure full cutting, followed by addition of 0.2ul CIP for 1h to prevent self-ligation of the vector.
Migr1-4-1BBL-IRES-CD40L reaction system:
Figure BDA0001889363430000082
both were digested at 37 ℃ overnight to ensure full cleavage.
Migr1-4-1BBL-TA2-CD40L reaction system:
Figure BDA0001889363430000083
the construction of the above clones respectively adopts the means of enzyme digestion connection and gene recombination: firstly, cDNA is taken as a template for PCR amplification to obtain CD40L and 4-1BBL fragments, then the XhoI and EcoRI enzyme cutting sites are utilized to respectively construct CD40L (SEQ ID No.1 and SEQ ID No.2 amplification) and 4-1BBL fragments (SEQ ID No.3 and SEQ ID No.4 amplification) on a Migr1 vector under the action of T4 ligase, and thus plasmids Migr1-CD40L and Migr1-4-1BBL are obtained. Secondly, plasmid Migr1-CD40L is subjected to double enzyme digestion by using restriction enzymes NcoI and SalI-HF to obtain a linearized vector, and then a 4-1BBL fragment (SEQ ID No.5 and SEQ ID No.6 are amplified) is constructed on Migr1-CD40L under the action of recombinase, so that a retrovirus expression plasmid Migr1-CD40L-IRES-4-1BBL is obtained. A linearized vector is obtained by singly digesting plasmid Migr1-CD40L with restriction enzyme EcoRI, and a P2A fragment (amplified by SEQ ID No.7 and SEQ ID No. 8) and a 4-1BBL fragment (amplified by SEQ ID No.9 and SEQ ID No. 10) are constructed on Migr1-CD40L under the action of recombinase, so that a retrovirus expression plasmid Migr1-CD40L-P2A-4-1BBL is obtained. A linearized vector is obtained by double digestion of plasmid Migr1-4-1BBL with restriction enzymes EcoRI and SalI-HF, and IRES fragments (amplified by SEQ ID No.11 and SEQ ID No. 12) and CD40L fragments (amplified by SEQ ID No.13 and SEQ ID No. 14) are constructed on Migr1-4-1BBL under the action of recombinase, so that a retrovirus expression plasmid Migr1-4-1BBL-IRES-CD40L is obtained. A linearized vector is obtained by using restriction enzyme EcoRI to singly cut plasmid Migr1-4-1BBL, and a T2A fragment (amplified by SEQ ID No.15 and SEQ ID No. 16) and a CD40L fragment (amplified by SEQ ID No.17 and SEQ ID No. 18) are constructed on the Migr1-4-1BBL under the action of recombinase, so that a retrovirus expression plasmid Migr1-4-1BBL-T2A-CD40L is obtained.
Referring to the above procedures, the resulting retrovirus plasmid carrying a foreign gene includes a retrovirus expression plasmid carrying a foreign gene CD40L alone (Migr1-CD40L, see FIG. 2), a retrovirus expression plasmid carrying a foreign gene 4-1BBL alone (Migr1-4-1BBL, see FIG. 3), a retrovirus expression plasmid carrying both a foreign gene 4-1BBL and CD40L together (Migr1-4-1BBL-T2A-CD40L, see FIG. 7, Migr1-4-1BBL-IRES-CD40L, see FIG. 6, Migr1-CD40L-P2A-4-1BBL, see FIG. 5, Migr1-CD40L-IRES-4-1BBL, see FIG. 4).
2. Packaging the virus:
the retroviral vector carrying the exogenous gene and a helper plasmid (helper plasmid) are transfected into 293T cells cultured in advance by utilizing lipo2000 transfection reagents respectively, cell supernatants are collected at 48h, 72h and 96h respectively, and the cells are stored at minus 80 ℃ after being subpackaged.
+Example 2 expression of CD40L and 4-1BBL on the surface of OT-I CD8T cells
A.293T was passaged and plated and the retrovirus packaged, as in example 1.
b. 6-8 weeks old OT-I mice were harvested, splenocytes isolated and activated by addition of N4.
c.24h later transduction of activated OT-I CD8 with different retroviruses carrying foreign genes+T cell:
treatment ① transduction of activated OT-I CD8 with retrovirus carrying foreign Gene CD40L alone+A T cell;
treatment ② transduction of activated OT-I CD8 with retrovirus carrying exogenous gene 4-1BBL alone+A T cell;
treatment ③ transduction of activated OT-I CD8 with retrovirus carrying exogenous Gene 4-1BBL-2A-CD40L+A T cell;
treatment ④ transduction of activated OT-I CD8 with retrovirus carrying exogenous Gene 4-1BBL-IRES-CD40L+A T cell;
treatment ⑤ transduction of activated OT-I CD8 with retrovirus carrying foreign Gene CD40L-2A-4-1BBL+A T cell;
treatment ⑥ transduction of activated OT-I CD8 with retrovirus carrying foreign Gene CD40L-IRES-4-1BBL+T cells.
d. After another 24h, the cells were split into two parts, one for flow detection and one for RT-PCR detection. Please refer to fig. 8.
Obtained by the above operations: CD8 capable of expressing foreign gene CD40L+T cell, CD8 capable of expressing exogenous gene 4-1BBL+T-cells, CD8 expressing both 4-1BBL and CD40L+T cells.
Transduction of N4-activated OT-I CD8 after packaging of different plasmids into retroviruses+T cells and detection of OT-I CD8 by flow cytometry and RT-PCR+Expression of T cells CD40L and 4-1 BBL. See fig. 9 and 10 for results.
From the data, it was found that both 4-1BBL and CD40L were efficiently expressed after transduction of the cells.
(1) In the double-protein expression vector, when 4-1BBL is in front of CD40L, namely the CD40L protein expression level of the two groups of cells transduced by Migr1-4-1BBL-T2A-CD40L and Migr1-4-1BBL-IRES-CD40L is obviously higher than that of the two groups transduced by Migr1-CD40L-P2A-4-1BBL and Migr1-CD40L-IRES-4-1BBL, and even higher than that of the group transduced by Migr1-CD40L, wherein the OT-I CD8 transduced by Migr1-4-1BBL-T2A-CD40L is higher than that transduced by the group transduced by Migr1-CD40L+The expression level of T cell CD40L was highest, and the cells transduced with the empty vectors Migr1 and Migr1-4-1BBL showed little expression.
(2) Cells transduced with Migr1-4-1BBL alone expressed 4-1BBL the highest, followed by Migr1-4-1BBL-T2A-CD40L and Migr1-4-1BBL-IRES-CD40L transduced cells, but the two groups of transduced cells were significantly lower after expression of 4-1BBL than Migr1-4-1BBL transduced cells. The expression of the two groups of transduced cells, namely Migr1-4-1BBL-T2A-CD40L and Migr1-4-1BBL-IRES-CD40L, is stronger than that of the two groups of transduced cells, namely, CD40L and 4-1BBL, than that of the two groups of transduced cells, namely, Migr1-CD40L-P2A-4-1BBL and Migr1-CD40L-IRES-4-1BBL, and the proportion of CD8+ T cells expressing the two proteins is the highest.
(3) Meanwhile, RNA of each group of cells is extracted, and the expression levels of mRNA of 4-1BBL and CD40L in the cells transduced by different viruses are detected by an RT-PCR technology. First, the transcription level of CD40L in each group of cells, Migr1 and Migr1-4-1BBL were almost not expressed compared to the blank control, and were consistent with the protein expression level. The gene transcription level of the cell which is transduced with Migr1-CD40L alone is basically consistent with the protein expression, and the transcription level of the cell CD40L transduced with Migr1-4-1BBL-T2A-CD40L is obviously higher than that of the other groups, which is consistent with the protein expression. The higher level of transcription of cells transduced by Migr1-CD40L-P2A-4-1BBL than that transduced by Migr1-CD40L should be caused by high efficiency of viral transduction. The low protein expression may be caused by low cleavage efficiency of intracellular hydrolase, so that the fusion proteins CD40L and 4-1BBL cannot be cut effectively, and the expression of CD40L is not high in flow detection. Next, the transcript levels of 4-1BBL, Migr1 and Migr1-CD40L transduced cells were compared to the blank control. The transcriptional levels of 4-1BBL from cells transduced with Migr1-4-1BBL alone and Migr1-CD40L-IRES-4-1BBL were consistent with protein expression levels. In contrast, the cells transduced with Migr1-CD40L-P2A-4-1BBL and Migr-4-1BBL-T2A-CD40L had a high transcription level, which may result in low protein expression of 4-1BBL being detected due to inefficient cleavage of the fusion protein by hydrolytic enzymes.
+Example 3 detection of cell proliferation after expression of CD40L and 4-1BBL on the surface of OT-I CD8T cells
a. Collecting 6-8 week old OT-I mice, separating spleen cells, adding N4 for stimulating culture.
Activated OT-I CD8 after 24h+T cells were evenly divided and activated CD8 was transduced with different viruses obtained in example 1 separately+T cells.
c. After 24h, the CD8 is respectively added+T cells were collected and counted under the microscope. The operational flow is shown in fig. 11A.
The detection result is shown in FIG. 11B. Overexpression of CD40L promoted cell proliferation compared to controls, but was not statistically different. The over-expression of 4-1BBL can effectively promote the cell proliferation, and the over-expression of 4-1BBL and CD40L has stronger auxiliary effect, which indicates that 4-1BBL and CD40L have an additive effect when promoting the cell proliferation.
+Example 4 detection of immature DC after expression of CD40L and 4-1BBL on the surface of OT-I CD8T cells LOW LOW LOW(CD80CD40MHC-II) helper activation
a. On day-3, the femurs and tibias of 6-8 weeks old C57BL/6 female mice were harvested, bone marrow cells were isolated, and DCs were cultured by adding GM-CSF and IL 4.
b. On day 0, 6-8 weeks old OT-I mice were harvested, spleen cells were isolated, and N4 was added to stimulate culture.
c.24h after transduction of activated OT-I CD8 with different viruses obtained in example 1+T cells.
d. After about 24 hours, OT-I splenocytes and 5 days of cultured DCs were collected and counted, respectively.
e. OT-I splenocytes and DCs were mixed at a ratio of 1:2, 10ng/ml IL15 was added, mixed well and plated on a round bottom 96 well plate for culture.
f. After the co-culture for 18h, the expression of the surface membrane protein of the DCs is detected by flow. See figure 12 for operation and results.
The first is the expression of CD40 on the surface of DCs (dendritic cells), which has been confused by the fact that Tgm overexpresses CD40L, but does not promote upregulation of CD40 on the surface of DCs, but instead causes downregulation of CD40 expression, and appears to be negatively correlated with the expression of Tgm surface CD 40L.
Expression of trans-4-1 BBL promotes the upregulation of CD40L on the surface of DCs. However, the expression of CD40L was much higher than that of Migr1-4-1BBL transduced cells, although the two groups of Migr1-4-1BBL-T2A-CD40L and Migr1-4-1BBL-IRES-CD40L transduced cells also expressed 4-1BBL, so that the expression of DCs surface CD40 was also down-regulated in these two experiments.
Moreover, overexpression of 4-1BBL also promoted upregulation of CD80 on the surface of DCs, but did not promote MHC-II expression, while high expression of CD40L still acted negatively, i.e., downregulated MHC-II expression, presumably in relation to downregulation of CD 40. This example demonstrates that Tgm transduced with Migr1-4-1BBL is effective in promoting further maturation of DCs.
+Example 5 detection of unmodified OT- +Proliferation-assisting action of ICD8T cells
a. On day-5, C57BL/6 female mice of 6-8 weeks old were collected, bone marrow cells in femur and tibia were isolated, and dendritic cells derived from bone marrow cells were cultured by adding GM-CSF (granulocyte colony stimulating biological factor) and IL4 (recombinant human interleukin-4), and the culture medium was changed once a day at intervals.
b. On day 0, 6-8 week-old CD45.2-OT-I mice were harvested, spleens were separated, and N4 was added to stimulate culture.
c.24h after transduction of activated OT-I CD8 with different viruses obtained in example 1+T cells, and adding dendritic cells cultured for 6 days to 800ng/ml CPG (adenine guanine nucleotide) stimulated further maturation and incubation continued for 16 h.
d. Day 2, CPDCs in G-stimulated culture were collected and counted, and the desired number of cells were added to the N4-loaded peptide and incubated for 2.5h at 37 ℃ in an incubator. Simultaneously taking 6-8 weeks old CD45.2+OT-I mice and spleen cells were isolated and resuspended in serum-free RPMI1640 medium. The desired number of cells was diluted to 10 millions/ml in RPMI1640 medium and transferred to 50ml tube, which was added with 7ml at the maximum, and CFSE (fluorescent dye CFDA-SE) was added to 1/2500 for a final dilution of 2. mu.M. Coating with tinfoil paper, standing at room temperature for 7min while shaking, adding 5 times volume of precooled 20% FBS RPMI1640 culture medium, and centrifuging at 320g and 4 deg.C for 10 min.
e. The supernatant was discarded and the cells were washed twice with 1 × PBS and re-counted.
f. And virus-transduced CD45.2-OT-I splenocytes and peptide-loaded DCs were collected and recounted with CD45.2+OT-I splenocytes are mixed evenly according to a certain proportion, are spread in a round bottom 96-hole cell culture plate for culture, and 10ng/ml IL15 is added.
g. After 36h of co-culture, flow detection was performed. The operation is shown in FIG. 13A. See fig. 13B, 13C for results.
The present inventors found that only Tgm overexpressing CD40L promoted proliferation of Tum (bystander T cells or flanking T cells) compared to cells transduced with the empty vector virus, but without statistical differences. Cells overexpressing 4-1BBL alone were more effective at promoting cell proliferation than cells overexpressing CD40L alone, but were still not statistically different from the empty vector control. Migr1-4-1BBL-T2A-CD40L transduced cells, i.e., cells overexpressing both 4-1BBL and CD40L, were found from the data to significantly promote cell proliferation, but were still not statistically different compared to the empty vector control, but were significantly different compared to cells overexpressing only one of the costimulatory molecules alone. From the previous data it is known that the highest proportion of cells co-expressing both proteins in such virally transduced cells, presumably providing stronger signals when both proteins are present on one cell at the same time, more efficiently promoting proliferation of other cells, suggesting that cells transduced with CD40L and 4-1BBL may promote cell proliferation mainly by direct contact with other cells, rather than by secretion of cytokines, since expression of IFN- γ by Migr1-4-1BBL-T2A-CD40L transduced cells is much lower than by Migr1-4-1BBL transduced cells, but further validation remains.
Example 6 detection of unmodified OT- Assistant effects of the cellular immune function of ICD8+ T cells
a. Collecting CD45.2-OT-I mice with the age of 6-8 weeks, separating spleens, and adding N4 for stimulating culture.
After 24h the activated OT-I CD8 was transduced with the different viruses obtained in example 1, respectively+T cells.
c. Taking CD45.2 of 6-8 weeks old after 24h+OT-I mice, spleen cells were isolated and resuspended by counting.
d. Collecting and recounting virus-transduced CD45.2-OT-I splenocytes obtained in step b, and comparing the collected cells with the CD45.2 cells obtained in step c+OT-I splenocytes were mixed at a ratio of 1:1, plated in round bottom 96 well cell culture plates, and added with 10ng/ml IL15 and 50ng/ml N4.
e. And (5) co-culturing for about 72 hours, and detecting by flow.
Operation please refer to fig. 14. See fig. 15 and fig. 16 for results.
Regardless of CD8+Whether T cells overexpress costimulatory molecules, IFN- γ is produced when activated by N4. But not overexpressing CD8 of costimulatory molecules+T cells or T cells overexpressing only CD40L produce only low levels of IFN- γ, while CD8 overexpressing 4-1BBL+T cells can help produce high levels of IFN-gamma. Cells overexpressing 4-1BBL are effective in promoting more CD8+T cells express INF-gamma and can effectively improve the average expression level of IFN-gamma.
By comparing Migr1-4-1BBL-2A-CD40L and Migr1-4-1BBL transduced cells, it was found that over-expressing high levels of CD40L also increased the proportion of IFN- γ expressing CD8+ T cells, but did not effectively enhance the average IFN- γ expression level, i.e., the expression of IFN- γ per cell was low. Furthermore, a comparison of cells transduced with Migr1-4-1BBL and Migr1-4-1BBL-IRES-CD40L and Migr1-4-1BBL-2A-CD40L led to the conclusion that the higher the expression level of 4-1BBL, the higher the expression level of IFN-. gamma.i.e.the expression of IFN-. gamma.positively correlated with the expression of 4-1 BBL.
In combination with the above data, it was speculated that CD40L may be more likely to promote IFN-. gamma.+CD8+Proliferation of T cells to increase IFN-gamma+Proportion of CD8+ T cells, but not CD8+The expression of IFN-gamma of T cells, while 4-1BBL can not only promote the proliferation of CD8+ T cells, but also effectively stimulate the high expression of IFN-gamma.
Tgm over-expressing CD40L can also increase IFN-gamma+The proportion of Tum was comparable to the control, but the helper effect was minimal compared to cells overexpressing 4-1 BBL. Consistent with previous data, cells overexpressing 4-1BBL significantly stimulated the secretion of Tum IFN-. gamma.and this effect was still positively correlated with the expression level of 4-1 BBL.
+Example 7 Back infusion of therapeutic MO5 after expression of CD40L and 4-1BBL on the surface of OT-I CD8T cells
a. The MO5 cells were revived and passed through 3-4 passages.
b. Cells were harvested and counted on day 1, resuspended in sterile 1 × PBS and diluted to 4 millions/ml. MO5 cells were inoculated subcutaneously by syringe into the backs of 6-8 week old C57BL/6 mice, 50. mu.l per mouse (0.2million), and the mice were randomized into four groups.
c. Tumor sizes were recorded every 3 days starting on day 7.
d. On the 10 th day, 6-8 weeks old OT-I mice were harvested, spleen cells were isolated, and N4 was added to stimulate culture.
e.24h post-transduction of activated OT-I CD8 with the different viruses of example 1+T cells.
f. Cells were collected and counted on day 12 separately and finally diluted to 10 millions/ml with sterile 1 × PBS. The OT-I splenocytes from step e were injected via the mouse tail vein into the periphery of 200. mu.l/mouse (2 million).
Operation is shown in figure 17A. The results are shown in FIGS. 17B and 17C.
According to the results shown: it has been shown in the above in vitro experiments that overexpression of CD40L is far from expected in development of cell function, whereas overexpression of 4-1BBL is effective in stimulating cell proliferation and functional development. This is also true in vivo, where cells overexpressing CD40L alone are barely able to inhibit tumor growth, while cells overexpressing 4-1BBL are effective in inhibiting tumor cell proliferation. Why are Migr1-4-1BBL transduced cells strongest in antitumor effect? From in vitro experimental data, it is clear that Tgm over-expressing 4-1BBL alone is less able to promote cell proliferation than cells over-expressing both 4-1BBL and CD40L, but is able to effectively stimulate CD8+Mean expression levels of T cell IFN- γ, suggesting CD8 in anti-tumor immunity+The killing ability of T cells is dependent on CD8+The function of the T cells develops, not the number.
In conclusion, the invention integrates the exogenous gene 4-1BBL into the chromosome of the antigen-specific T cell to carry out overexpression so as to prepare the T cell with the effect of improving immune response. The T cell with the effect of improving the immune response not only has fast self proliferation and high expression quantity of IFN-gamma, but also can obviously promote the proliferation and IFN-gamma expression level of the peripheral antigen specific T cell without carrying the foreign gene (for example, the antigen specific T cell which is co-cultured with the T cell and does not carry the foreign gene is returned to the tumor tissue and then is in the tumor tissue), and plays a role of guiding the general in the aspect of activating the immune response capacity of the peripheral antigen specific T cell.
The technical features of the embodiments described above may be arbitrarily combined, and for the sake of brevity, all possible combinations of the technical features in the embodiments described above are not described, but should be considered as being within the scope of the present specification as long as there is no contradiction between the combinations of the technical features.
The above-mentioned embodiments only express several embodiments of the present invention, and the description thereof is more specific and detailed, but not construed as limiting the scope of the invention. It should be noted that, for a person skilled in the art, several variations and modifications can be made without departing from the inventive concept, which falls within the scope of the present invention. Therefore, the protection scope of the present patent shall be subject to the appended claims.
Sequence listing
<110> Guangzhou Baorui medical technology, Inc
<120> T cell with immune response improving effect and preparation method and application thereof
<160>20
<170>SIPOSequenceListing 1.0
<210>1
<211>33
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>1
tctctcgagg ccaccatgat agaaacatac agc 33
<210>2
<211>33
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>2
ggcggaattc tcagagtttg agtaagccaa aag 33
<210>3
<211>31
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>3
tctctcgagg ccaccatgga ccagcacaca c 31
<210>4
<211>27
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>4
ggcgaattct cattcccatg ggttgtc 27
<210>5
<211>27
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>5
aatatggcca caaccatgga ccagcac 27
<210>6
<211>33
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>6
aagcttggct gcaggtcatt cccatgggtt gtc 33
<210>7
<211>24
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>7
tttggcttac tcaaactcag aagg 24
<210>8
<211>19
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>8
aagtgtgtgc tggtccatg 19
<210>9
<211>25
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>9
atggaccagc acacacttga tgtgg 25
<210>10
<211>31
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>10
ggagggagag gggcgtcatt cccatgggtt g 31
<210>11
<211>34
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>11
gacaacccat gggaatgacc cctctccctc cccc 34
<210>12
<211>29
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>12
ctatcatggt tgtggccata ttatcatcg 29
<210>13
<211>35
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>13
atggccacaa ccatgataga aacatacagc caacc 35
<210>14
<211>40
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>14
ataagcttgg ctgcaggtca gagtttgagt aagccaaaag 40
<210>15
<211>34
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>15
ccgacaaccc atgggaaaga aggaagcgtg gaag 34
<210>16
<211>30
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>16
tgtttctatc atggggccgg gattttcctc 30
<210>17
<211>29
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>17
ggccccatga tagaaacata cagccaacc 29
<210>18
<211>38
<212>DNA
<213> Artificial Sequence (Artificial Sequence)
<400>18
ggagggagag gggcgtcaga gtttgagtaa gccaaaag 38
<210>19
<211>783
<212>DNA
<213> mouse (Mus musculus)
<400>19
atgatagaaa catacagcca accttccccc agatccgtgg caactggact tccagcgagc 60
atgaagattt ttatgtattt acttactgtt ttccttatca cccaaatgat tggatctgtg 120
ctttttgctg tgtatcttca tagaagattg gataaggtcg aagaggaagt aaaccttcat 180
gaagattttg tattcataaa aaagctaaag agatgcaaca aaggagaagg atctttatcc 240
ttgctgaact gtgaggagat gagaaggcaa tttgaagacc ttgtcaagga tataacgtta 300
aacaaagaag agaaaaaaga aaacagcttt gaaatgcaaa gaggtgatga ggatcctcaa 360
attgcagcac acgttgtaag cgaagccaac agtaatgcag catccgttct acagtgggcc 420
aagaaaggat attataccat gaaaagcaac ttggtaatgc ttgaaaatgg gaaacagctg 480
acggttaaaa gagaaggact ctattatgtc tacactcaag tcaccttctg ctctaatcgg 540
gagccttcga gtcaacgccc attcatcgtc ggcctctggc tgaagcccag cagtggatct 600
gagagaatct tactcaaggc ggcaaatacc cacagttcct cccagctttg cgagcagcag 660
tctgttcact tgggcggagt gtttgaatta caagctggtg cttctgtgtt tgtcaacgtg 720
actgaagcaa gccaagtgat ccacagagtt ggcttctcat cttttggctt actcaaactc 780
tga 783
<210>20
<211>930
<212>DNA
<213> mouse (Mus musculus)
<400>20
atggaccagc acacacttga tgtggaggat accgcggatg ccagacatcc agcaggtact 60
tcgtgcccct cggatgcggc gctcctcaga gataccgggc tcctcgcgga cgctgcgctc 120
ctctcagata ctgtgcgccc cacaaatgcc gcgctcccca cggatgctgc ctaccctgcg 180
gttaatgttc gggatcgcga ggccgcgtgg ccgcctgcac tgaacttctg ttcccgccac 240
ccaaagctct atggcctagt cgctttggtt ttgctgcttc tgatcgccgc ctgtgttcct 300
atcttcaccc gcaccgagcc tcggccagcg ctcacaatca ccacctcgcc caacctgggt 360
acccgagaga ataatgcaga ccaggtcacc cctgtttccc acattggctg ccccaacact 420
acacaacagg gctctcctgt gttcgccaag ctactggcta aaaaccaagc atcgttgtgc 480
aatacaactc tgaactggca cagccaagat ggagctggga gctcatacct atctcaaggt 540
ctgaggtacg aagaagacaa aaaggagttg gtggtagaca gtcccgggct ctactacgta 600
tttttggaac tgaagctcag tccaacattc acaaacacag gccacaaggt gcagggctgg 660
gtctctcttg ttttgcaagc aaagcctcag gtagatgact ttgacaactt ggccctgaca 720
gtggaactgt tcccttgctc catggagaac aagttagtgg accgttcctg gagtcaactg 780
ttgctcctga aggctggcca ccgcctcagt gtgggtctga gggcttatct gcatggagcc 840
caggatgcat acagagactg ggagctgtct tatcccaaca ccaccagctt tggactcttt 900
cttgtgaaac ccgacaaccc atgggaatga 930

Claims (10)

1. A T cell having an effect of enhancing an immune response, which is prepared by integrating a foreign gene 4-1BBL into an antigen-specific T cell chromosome and overexpressing the same.
2. The method for preparing T cells having an immune response-enhancing effect according to claim 1, comprising the steps of:
obtaining antigen specific T cells and activating the antigen specific T cells with specific antigens;
constructing a viral particle carrying the foreign gene 4-1BBL, transfecting said activated antigen-specific T cell with said viral particle, thereby integrating the foreign gene 4-1BBL into the chromosome of said activated antigen-specific T cell.
3. The method of claim 2, wherein the viral particle is constructed using a retrovirus.
4. The method of claim 3, wherein the retrovirus is Migr 1.
5. The method of claim 2, wherein the viral particle is constructed using lentivirus.
6. The method of any one of claims 2 to 5, wherein the antigen-specific T cell is CD8+T cells.
7. The method of any one of claims 2 to 5, wherein the specific antigen is a polypeptide recognized by an antigen-specific T cell.
8. Use of the T cell having an immune response-enhancing effect according to claim 1 for promoting an immune response function of an immune cell adjacent to the T cell.
9. The use of claim 8, wherein said use is co-culturing said immune cells with said T cells that enhance the efficacy of an immune response.
10. Use of the T cell of claim 1 for enhancing immune response in the treatment of tumors, chronic viral infections, bacterial, fungal infections.
CN201811464461.9A 2018-12-03 2018-12-03 T cell with effect of improving immune response and preparation method and application thereof Pending CN111249307A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811464461.9A CN111249307A (en) 2018-12-03 2018-12-03 T cell with effect of improving immune response and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811464461.9A CN111249307A (en) 2018-12-03 2018-12-03 T cell with effect of improving immune response and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN111249307A true CN111249307A (en) 2020-06-09

Family

ID=70942860

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811464461.9A Pending CN111249307A (en) 2018-12-03 2018-12-03 T cell with effect of improving immune response and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN111249307A (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100178276A1 (en) * 2007-03-30 2010-07-15 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred t lymphocytes

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100178276A1 (en) * 2007-03-30 2010-07-15 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred t lymphocytes

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BRENDA DE KEERSMAECKER等: "The combination of 4-1BBL and CD40L strongly enhances the capacity of dendritic cells to stimulate HIV-specific T cell responses", 《JLB》 *
ERHAO ZHANG等: "Prospects for chimeric antigen receptor-modified T cell therapy for solid tumors", 《 MOLECULAR CANCER》 *
MARK B. GEYER等: "Review:current clinical applications of chimeric antigen receptor (CAR) modified T cells", 《CYTOTHERAPY》 *
OLADAPO O. YEKU等: "Armored CAR T-cells: utilizing cytokines and pro-inflammatory ligands to enhance CAR T-cell anti-tumour efficacy", 《BIOCHEM. SOC. TRANS.》 *
王扬等: "利用MigR1质粒构建CD19-CAR逆转录病毒载体优化其对原代T细胞的转染效率", 《中华血液学杂志》 *

Similar Documents

Publication Publication Date Title
EP3699268A1 (en) Universal car-t cell, preparation method therefor and application thereof
WO2018014384A1 (en) Tcr-/pd-1- double negative t-cell and construction method thereof
JP6175103B2 (en) Methods for increasing immune response
Zhen et al. Synergistic antitumor effect on bladder cancer by rational combination of programmed cell death 1 blockade and CRISPR-Cas9-mediated long non-coding RNA urothelial carcinoma associated 1 knockout
Chen et al. Enhancement of CTLs induced by DCs loaded with ubiquitinated hepatitis B virus core antigen
CN114592010A (en) NK-CAR-MbIL-15 cell and preparation method and application thereof
WO2023123195A1 (en) Engineered immune cell target gene of which can be regulated, preparation method therefor, and use thereof
CN107217041B (en) DC cell with high antigen presentation and antigen-specific T cell, and preparation method and application thereof
CN111254119B (en) T cell with effect of improving immune response and preparation method and application thereof
CN111249307A (en) T cell with effect of improving immune response and preparation method and application thereof
CN110819594A (en) Mesenchymal stem cell for continuously over-expressing IFN-gamma and application thereof
CN114702569B (en) PD-L1 related vaccine and application thereof
CN113736742B (en) Application of PRTN3 gene as target for activating cytotoxic immune cells in tumor immunotherapy
CN114369621B (en) Gene biological agent and preparation method and application thereof
CN113337505B (en) Application of artificially constructed antisense nucleotide fragment Ri111 in polarization of Th1 cells
CN115820742A (en) Method for improving transduction efficiency of NK cell lentivirus
CN117431271A (en) Recombinant expression vector for constructing CAR-T cells and CAR-T cells
US20100247502A1 (en) Tumor/b-cell hybrid cells and uses thereof
CN117305249A (en) IL21-NKG2D CAR-T cell and preparation method and application thereof
WO2018032619A1 (en) Applications of soluble protein baff in b cell in-vitro culture and proliferation
CN117417894A (en) Tumor infiltration lymphocyte for knocking out Roquin-1 and/or Regnase-1 genes and application thereof
Hu et al. Thomas B. Huffaker, Soh-Hyun Lee, William W. Tang, Jared A. Wallace, Margaret Alexander, Marah C. Runtsch, Dane K. Larsen, Jacob Thompson, Andrew G. Ramstead, Warren P. Voth
CN117304343A (en) Preparation and application of GPC3 targeted CAR-NK cells
CN115944650A (en) Application of tumor infiltrating cells in preparation of antitumor drugs and model construction method
CN112608903A (en) Lymphocyte and culture system and culture method 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
RJ01 Rejection of invention patent application after publication
RJ01 Rejection of invention patent application after publication

Application publication date: 20200609