CN111166876B - Immunopotentiator combination, encoding nucleic acid and application thereof - Google Patents
Immunopotentiator combination, encoding nucleic acid and application thereof Download PDFInfo
- Publication number
- CN111166876B CN111166876B CN201811343570.5A CN201811343570A CN111166876B CN 111166876 B CN111166876 B CN 111166876B CN 201811343570 A CN201811343570 A CN 201811343570A CN 111166876 B CN111166876 B CN 111166876B
- Authority
- CN
- China
- Prior art keywords
- cells
- leu
- ser
- ala
- thr
- 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
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/39—Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/0005—Vertebrate antigens
- A61K39/0011—Cancer antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/04—Immunostimulants
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/54—Interleukins [IL]
- C07K14/5443—IL-15
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70578—NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/715—Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
- C07K14/7155—Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/515—Animal cells
- A61K2039/5154—Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/53—DNA (RNA) vaccination
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/54—Medicinal preparations containing antigens or antibodies characterised by the route of administration
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
- A61K2039/55516—Proteins; Peptides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
- A61K2039/55522—Cytokines; Lymphokines; Interferons
- A61K2039/55527—Interleukins
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Immunology (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Molecular Biology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Toxicology (AREA)
- Zoology (AREA)
- Gastroenterology & Hepatology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- General Chemical & Material Sciences (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Epidemiology (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Oncology (AREA)
- Cell Biology (AREA)
- Communicable Diseases (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
The invention provides an immunopotentiator combination, which comprises Bcl2, CD40L, IL15 and IL15 receptor alpha (IL 15 Ra). In particular, fusion proteins comprising the combination, nucleic acids encoding the proteins of the components of the composition are provided. The composition comprises amino acid sequences shown as SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 3 and SEQ ID NO 4. Vaccine compositions comprising the compositions and routes of delivery of the compositions are also provided. The immunopotentiator compositions of the present invention can provide protective immunity from pathogen infection and can be used for the prevention and/or treatment of a variety of tumors.
Description
Technical Field
The invention relates to the field of medical products, in particular to an immunopotentiator combination, coding nucleic acid and application thereof.
Background
The immune response of the body is first to capture antigen by Antigen Presenting Cells (APC), processed and processed to present antigen information to lymphocytes, and then to initiate a series of specific immune responses. Dendritic Cells (DC) are the APC considered to be the strongest in function at present, and the most important characteristic of the DC is that the DC can stimulate the proliferation and the activation of Naive T cells (nasal T cells) and is a central link for starting, regulating and maintaining specific immune response. In the anti-tumor immunity of the body, the cellular immunity mediated by T cells plays an important role.
Researches find that the tumor patients have the characteristics of reduced DC number and functional defects, and the number and the function of tumor tissues and DC infiltration around the tumor tissues have close relation with the occurrence, the development, the metastasis and the prognosis of tumors. The tumor densely infiltrated by the DC has high differentiation degree and better prognosis; whereas tumors that are mildly infiltrated by DCs are often associated with low differentiation and malignant progression. Tumor cells have high-level Fas expression, can induce apoptosis of lymphocytes expressed by FasL, and can secrete immunosuppressive cytokines such as TGF-beta, IL-10 and the like, so that the antigen presenting capability is reduced, and immune attack is avoided.
In recent years, it has become clear that the immune system does recognize tumor antigens, but despite the presence of tumor antigens, T cells are assured to remain quiescent. Based on this phenomenon, there is a theory: antigen presenting cells in the patient, which fail to correctly recognize the tumor antigen, present it to T lymphocytes, and elicit a tumor-specific immune response. In recent years, increasing the number of antigen presenting cells, and improving the ability of antigen presenting cells, especially DC cells, to take up, transport, present antigen, and stimulate T cells, are a major focus in current tumor immunization research.
Numerous studies have shown that CD40-CD 40L-mediated signaling can induce APC activation. The latter plays an important role in the activation of tumor-specific immune killer T cells: provides an antigen stimulating signal and a second signal, and the two signals act synergistically to start immune cascade reaction and exert the tumor immune function. Meanwhile, the research finds that the CD40L can also maintain the activation and survival of DC cells and the expansion of CD8+ T cells, and can antagonize the inhibitory effect of IL-10 cytokines on the differentiation, maturation and function of the DC cells.
The CD40L molecule can interact with DC surface CD40 to activate DC, and promote APC co-stimulatory molecule expression and cytokine secretion. Activated DCs are the bridge between tumor cells and tumor-specific immune killer T cells, and can present tumor antigens to and activate T cells, producing tumor-specific killer T cells. Thus, activation of DCs via the CD40-CD40L signaling molecule pathway can enhance the presentation of poorly immunogenic tumor cell antigens, inducing tumor-specific immunity.
IL-15 is a cytokine with a structure similar to that of IL-2, and is widely expressed in various cells and tissues, such as monocytes, macrophages, dc cells, fibroblasts, and the like. IL-15 activates downstream JAK1, JAK3 by binding to IL-15 receptor α, leading to phosphorylation of downstream STAT3 and STAT5 and activation of signaling pathways, inducing phosphorylation of BCL2, MAP kinase pathway, lck and syk, leading to proliferation and maturation of cells.
IL-15 is capable of regulating T cell and NK cell activation and proliferation, and maintaining the survival of memory T cells in the absence of antigen stimulation. It has been demonstrated that IL-15 inhibits apoptosis by inducing BCL2L1/BCL-x (L) in rodent lymphocytes. Similarly, il-15 was also found to inhibit T lymphocyte apoptosis in humans by inducing BCL2 and/or Bcl-xL.
Recently, researchers have constructed a DNA tumor vaccine expressing recombinant Il-15 protein using NDV as a vector. Preclinical results show that this tumor vaccine has shown potential to control melanoma growth in a mouse model. Similarly, a recombinant vaccinia virus expressing influenza A protein and Il-15 could promote cross protection of CD4+ T cells. Another recombinant Brucella DNA vaccine containing the Il-15 gene was shown to enhance CD8+ T cell immune response in mice.
In recent years, there have been a number of reports of tumor treatment using antigen-loaded DC vaccines, and from the data reported so far, DC vaccines appear to represent a new and very promising approach for improved tumor immunotherapy. However, the use of DC vaccines alone generally does not lead to the desired improvement in immunotherapeutic effects and does not lead to satisfactory clinical results.
Disclosure of Invention
Therefore, the aim of the present method is to provide a safe and effective means for DC tumor vaccines, compositions enhancing DC vaccine function, in particular for the treatment of tumors and/or infectious diseases.
In particular, the object of the present invention is solved by providing a vaccine/immunomodulator combination comprising an RNA vaccine comprising at least one RNA or DNA (comprising at least one open reading frame encoding at least one antigen), or a DNA vaccine, more even an antigenic polypeptide vaccine, as a vaccine, and BCL2, CD40L, IL-15 and IL-15Ra as modulators. In addition, the vaccine/modulator combination may be in the form of a mixture comprising nucleic acids or polypeptides of the combination, or a viral vector expressing the vaccine/modulator combination, or other forms including the vaccine/inhibitor component. In addition, the object is solved by a combination of an RNA or polypeptide vaccine and an inhibitor for use in a method for treating tumors or infectious diseases.
In the present invention, "antigen" refers to a substance that can be recognized by the immune system and is capable of eliciting an antigen-specific immune response by forming antibodies or/and antigen-specific T cells. In general, an antigen can be a protein or polypeptide that comprises at least one antigenic epitope and can be presented by the Major Histocompatibility Complex (MHC) to the surface of a T cell. In the present invention, the antigen may be a product of translation of mRNA or a product of transcription and translation of DNA.
A vaccine is typically understood to provide one or more antigens, preferably immunogens, for prophylactic or therapeutic substances. For example, a vaccine comprises the antigen, or a nucleic acid encoding the antigen, which may be DNA or RNA; or cells expressing the antigen, such as DC cells or PBMC cells. The antigen or immunogen may be derived from any material suitable for vaccination.
The antigenic nucleic acid molecules of the present invention encode immunogenic peptides of bacteria, viruses, fungi, or other pathogens including, but not limited to, human hepatitis viruses including HAV, HBV, HCV, cytomegalovirus CMV, human immunodeficiency virus HIV, EB virus, dengue virus, human Papilloma Virus (HPV), respiratory syncytial virus, rhinoviruses, human T-lymphotropic virus type I (HTLV-1), influenza, bovine Leukemia Virus (BLV), pertussis, polio, measles, mumps, rubella, smallpox, shingles, anthrax, tetanus, rotavirus, rabies, fowl pox, meningococcus, anthrax, encephalitis, pneumococci, staphylococci, neisseria, escherichia coli, shigella, leishmania, respiratory syncytial virus, parainfluenza, adenovirus, varicella, flavivirus, mycobacterium tuberculosis, and malaria, among others.
In one embodiment, the antigenic nucleic acid molecule encodes a tumor antigen. In this case, the tumor antigen may be expressed on the surface, cytoplasm, or nucleus of the tumor cell. The tumor antigen may also be selected from proteins that are overexpressed in tumor cells compared to normal cells. Tumor antigens can be further divided into tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs). Tumor Associated Antigens (TAAs) are a class of antigenic molecules found in both tumor and normal cells, including: embryonic proteins, glycoprotein antigens, squamous cell antigens, and the like. Tumor-associated antigens are not specific to tumor cells, and normal cells can also be synthesized in minute quantities and highly expressed when tumor cells proliferate. Tumor specific antigens refer to neoantigens that are expressed only on the surface of tumor cells and not on normal cells. Such antigens may be present in tumors of different individual consent tissue types, e.g. melanoma specific antigens encoded by human malignant melanoma genes may be present in melanoma cells of different individuals, but not expressed by normal melanoma cells. TSA can also be common to tumors of different histological types, for example, mutated Ras gene products can be commonly found in lung cancer, digestive tract tumors, etc., but because its amino acid sequence is inconsistent with the expression product of normal proto-oncogene Ras, it can be recognized by the immune system of the body to stimulate the immune response of the body. In general, such antigens, which are produced by mutation, are called neoantigens (neoantigen). These antigens are all recognized by cytotoxic T lymphocytes and cells presenting the antigen can be killed by T lymphocytes.
<xnotran> , : TDO2, MAGEC1, HMOX1, WT1, LY6H, AIM2, IDO1, CHI3L1, IL13RA2, LCK, GFAP, KIF20A, CNTN2, MUC16, PEG10, TNC, SOX11, IGF2BP3, S100A8, AKAP4, TTK, CHI3L2, PTHLH, CDC45, PMEL, TOP2A, PTTG1, NRCAM, HMMR, MUC4, LY6K, SOX10, FOSL1, PRAME, FOLR1, BIRC5, KIF2C, ITGAV, ART4, PROM1, CT83, S100A9, PPIB, S100A12, STAT1, EPCAM, ROR1, MLANA, KAAG1, KLK3, NT5E, PTPRZ1, SPAG4, MET, RGS1, CSPG4, PDCD1LG2, MUC1, CD274, PSCA, WDHD1, FABP7, PLIN2, PTGER2, HAVCR2, TPD52, ABCC3, TSSK6, ERBB2, CCDC110, TERT, CTAG2, SEC61G, ADORA2A, AURKB, ACPP, EPHX1, PTGS1, EZH2, PTGS2, PLK4, DDX43, PA2G4, PAX8, IDH1, SFMBT1, EPHA2, NAPSA, LPGAT1, NUF2, SPAG5, STAT3, MELK, ST8SIA1, EBAG9, KIFC3, CEACAM5, RPL19, SYCP2, DSE, ANKRD30A, TRAPPC1, RGS5, MGAT5, KRT19, B3GALNT1, CAGE1, AGER, ACRBP, LAG3, NELFA, RAB38, CCND1, SART1, UBE2V1, SLAMF7, KCNMA1, MUM1, HSPH1, GUCY1A3, AKAP13, SQSTM1, BCAN, CCNB1, TP53, SUGT1, AURKA, RAN, LY6D, NLGN4X, SART3, PRKDC, FOXP3, HBEGF, PIK3R1, SLC1A3, PCNA, KIF1C, BSG, ATP2A3, SPAG9, RPSA, NFYC, LRRC8A, IQGAP1, LY6E, TRIOBP, ART1, BAGE, BIRC7, CA9, CCDC54, DCT, IDO2, MAGED4, SOX2, SYCP1, TYR, 5T4, BAGE2, BORIS, CALR3, CSAG2, CSH1, </xnotran> CTAG1A, CTAG1B, CTAGE1, E6, FMR1NB, GAGE1, GAGE2A, GAGE3, GAGE4, GAGE5, GAGE6, GAGE7, GAGE8, GRDX, HNPRL, IGHD, MAGEA1, MAGEA10, MAGEA12, MAGEA2, MAGEA3, MAGEA4, MAGEA6, MAGEA9, MAGEB1, MAGEB2, MAGEC2, PAGE4, PAP, SAGE1, SPANXB1, SPO11, SSX1, SSX2, SSX4, brachyury/TFT, TTR, TYRP1, VSIR, XAGE1B, XAGE1E, ENAH, AKR1B10, GPC3, AFP, FLVCR1, FGF19, PSPH, ABL2, CCT3, SMYD3, TMEM106C, ZNF260, EPCAM, ICK, PHF20L1, ANXA3, ZNF623, CASK, FAM122B, IRS1, OTUD6B, TMEM68, VASH2, FGF3, TERF1, TSHZ2, TTC13, UTP14A.
Furthermore, tumor antigens may also include individual tumor-specific neoantigens which are produced by genetic mutations in tumor cells. The mutated gene may be any gene in a cell, and its expression product may be expressed on the cell surface or inside the cell.
In order to prevent instability of RNA and degradation of multiple pathways, nucleic acid molecules can be optimized according to the various natural degradation pathways of RNA that are known. For example, the terminal structure is crucial for the stability of mRNA. For example, at the 5' end of a naturally occurring mRNA, there is a modified guanosine nucleotide called a 5' cap structure, and the 3' end has an adenosine nucleotide (i.e., poly-A tail) structure of about 200 to 300 bases in length, and UTR sequences at the 5' and 3' ends, such as those of human beta-globin.
The immune modulator combination provided by the invention comprises BCL2 protein, CD40L protein, IL-15 protein and Il-15Ra protein. Wherein the CD40L molecule has an amino acid sequence comprising SEQ ID NO: 1; an Il-15 molecule having a sequence comprising SEQ ID NO: 2; an Il-15Ra molecule having a sequence comprising SEQ ID NO: 3; a BCL2 molecule having a sequence comprising SEQ ID NO: 4.
In one embodiment, nucleic acid molecules encoding CD40L, nucleic acid molecules encoding IL-15, and nucleic acid molecules encoding IL-15Ra are presented. The compositions of the invention may be encoded by separate nucleic acid molecules, or by one or two nucleic acid molecules, and the coding regions of the components of the compositions may be encoded by the nucleic acid molecules as set forth in SEQ ID NOs: 7 (i) may be linked by a nucleic acid sequence encoding a self-cleaving polypeptide sequence.
In one embodiment, a nucleic acid molecule encoding CD40L is provided having a nucleotide sequence comprising SEQ ID NO: 6; nucleic acid molecules encoding IL-15 and IL-15Ra are provided having a nucleotide sequence comprising SEQ ID NO:5, which molecule encodes both an IL-15 and an IL-15Ra molecule; nucleic acid molecules encoding BCL2 proteins are provided having a nucleic acid sequence comprising SEQ ID NO: 8.
The vaccine/modulator compositions of the present invention can be delivered to host DC cells in vivo by methods known in the art. In one embodiment, the vaccine/modulator compositions of the present invention may be introduced by viral vectors such as adenovirus (AdV), adeno-associated virus (AAV), retrovirus, lentivirus, herpes simplex virus, and the like. In addition, the vaccine/modulator compositions of the present invention may also be introduced by transfection of liposomal nanoparticles into host cells. In one embodiment, the vaccine/modulator compositions of the present invention can be introduced into the subject's own DC cells by electroporation, using the DC cells as a vector. In one embodiment, the vaccine/modulator composition of the present invention may be introduced into autologous PBMC cells or allogeneic PBMC cells of a subject by electroporation, and the autologous PBMC cells or allogeneic PBMC cells are used as vectors for introduction into the subject.
Drawings
FIG. 1 is a graph showing the survival of DC cells transfected with nucleic acid molecules encoding proteins of the present composition, which shows significantly higher survival rates than control groups.
FIG. 2 is a graph showing the phenotype of DC cells transfected with nucleic acid molecules encoding proteins of the compositions of the present invention, wherein the CD80, CD86, and CD83 on the surface of mature DC cells (mDC-survivin vs. mDC-survivin/regulator nucleic acid) are significantly upregulated, as compared to Immature Dendritic Cells (iDC), indicating significant mature DC cell characteristics; the phenotype of the DC cells transfected with the survivin/modulator nucleic acid molecule is consistent with the phenotype of DC cells transfected with survivin antigen alone, as compared to DC cells transfected with survivin antigen alone.
FIG. 3 is a graph of the effect of immune modulator combinations on T cell responses, whereby DC cells transfected with survivin/modulator nucleic acid molecules are capable of eliciting a stronger T cell response than groups transfected with survivin antigen alone, and whereby the DC cells stimulated to have a higher proportion of TNF-a and IFN-r expression.
Detailed Description
The following detailed description of specific embodiments of the invention, which are intended to be illustrative only, and the invention is described in further detail. These examples should not be construed as limiting the invention thereto.
The first embodiment is as follows: preparation of DNA and mRNA encoding antigens and immunodetection Point inhibitors
1. Preparation of DNA and mRNA constructs
A DNA sequence encoding mRNA for tumor antigen survivin protein, a DNA sequence encoding BCL2, CD40L, IL-15, and IL-15Ra in the present invention, and a subsequent in vitro transcription reaction were constructed for this example. Constructs were made by codon optimization to introduce a high GC sequence to stabilize the synthesized mRNA, followed by a 3' utr sequence of human-derived β -globin, followed by a segment of polyadenylic acid (including but not limited to the 64 adenosine or longer poly-a-sequence used in this example).
2. In vitro transcription
The corresponding DNA plasmid prepared according to example 1 was first linearized using the speI endonuclease and mRNA prepared by in vitro transcription using T7 RNA polymerase using the linearized plasmid as template. The prepared mRNA was then purified by lithium chloride precipitation.
The second embodiment: effect of immunosuppressant composition mRNA on dendritic cell phenotype and survival
1. In vitro induction culture of DC cells
Sterile extracting 50ml of healthy human venous blood, separating peripheral blood mononuclear cells by using lymphocyte separation liquid in an ultraclean workbench, adding the mononuclear cells into an AIM-V culture medium, and putting the AIM-V culture medium into a 37 ℃ and 5% CO2 incubator for incubation so as to adhere the mononuclear cells to the wall. After 2h, nonadherent cells were removed, adherent cells were added to iDC medium (GM-CSF was added to AIM-V medium to a final concentration of 800U/mL, IL-4 was added to 500U/mL), and cultured in a 5% CO2 incubator at 37 ℃ for 6 days. Half of the cell culture medium was transferred to a centrifuge tube, and 500g of the medium was centrifuged to collect cells, the supernatant was removed, and an equal volume of fresh mDC medium (the formulation of the fresh mDC medium: 1600U/mL GM-CSF and 1000U/mL IL-4, TNF-a (5 ng/mL), IL-1 beta (5 ng/mL), IL-6 (150 ng/mL) and prostaglandin E2 (PGE 2) (1 ug/mL)) was added thereto, and after resuspension of the cells, the cells were added to a flask and cultured for 8 to 18 hours to induce maturation of the DC cells.
2. Transfection of DC cells with immunosuppressant compositions
On the day of transfection, DC cells were digested into cell suspension with nonenzymatic cell-digesting agents, centrifuged, washed twice with PBS, resuspended in PBS, and adjusted to a cell density of 25-30X 10 6 DCs/ml. According to each 10 6 Transfection of DC cells to 4ug of mRNA, mixing of DC cellsAnd the modulator nucleic acid molecules (BCL 2, CD40L and IL-15-IRES-IL15 Ra) mRNA described herein, the cell-mRNA mixture was added to an electric rotor, and the antigen mRNA was transfected into DC cells using an ECM630 electrotransfer. The cells after the electroporation were resuspended in cytokine-free AIM-V medium and the cell density was adjusted to 1X 10 6 DCs/ml were seeded into 96 well cell culture plates at a volume of 200ul per well, placed at 37 ℃ in 5% CO 2 And continuing culturing in the cell culture box. GFP mRNA was transfected into DC cells under the same conditions as the control group. The number of DC cells in the plates was recorded daily for 5 consecutive days.
3. Determination of transfection efficiency
24 hours after transfection, the proportion of DC cells expressing green fluorescent protein to all DC cells was analyzed by flow cytometry.
4. Identification of DC cell phenotype
Using direct immunofluorescence labeling, transfected DC cells were centrifuged and the cells were resuspended in FACS buffer (2% FBS in PBS) at a cell concentration of 1X 10 6 cells/ml, 100ul of transfected DC cell suspension was added to the flow cell tube, and 5ul of the corresponding antibodies CD80, CD83, CD86, and isotype control were added. Staining for 30min at 4 ℃ in dark. 3ml of FACS Buffer was added to each tube to wash the cells, the supernatant was discarded, 500ul of FACS Buffer was added, and the expression of CD80, CD83, and CD86 was detected by flow analysis.
As shown in FIG. 1, DC cells transfected with the immunomodulator composition exhibited better cell viability than the untransfected DC cell control.
As shown in FIG. 2, there was no significant difference in the stable expression of the cell surface molecules CD80, CD83, CD86 in DC cells transfected with the immunomodulator composition compared to the untransfected DC cell control.
Example three: effect of immunomodulator compositions on T cell response
1. In vitro induction culture of DC cells
Sterile extracting 50ml of healthy human venous blood, separating peripheral blood mononuclear cells by using lymphocyte separation liquid in an ultraclean workbench, adding the mononuclear cells into an AIM-V culture medium, and putting the AIM-V culture medium into a 37 ℃ and 5% CO2 incubator for incubation so as to adhere the mononuclear cells to the wall. After 2h, nonadherent cells were removed, adherent cells were added to iDC medium (GM-CSF was added to AIM-V medium to a final concentration of 800U/mL, IL-4 was added to 500U/mL), and cultured in a 5% CO2 incubator at 37 ℃ for 6 days. Half of the cell culture medium was transferred to a centrifuge tube, and 500g of the medium was centrifuged to collect cells, the supernatant was removed, and an equal volume of fresh mDC medium (the formulation of the fresh mDC medium: 1600U/mL GM-CSF and 1000U/mL IL-4, TNF-a (5 ng/mL), IL-1 beta (5 ng/mL), IL-6 (150 ng/mL) and prostaglandin E2 (PGE 2) (1 ug/mL)) was added thereto, and after resuspension of the cells, the cells were added to a flask and cultured for 8 to 18 hours to induce maturation of the DC cells.
2. Transfection of DC cells with immunosuppressant compositions
On the day of transfection, DC cells were digested into cell suspensions using non-enzymatic cell digestion reagents, centrifuged, washed twice with PBS, resuspended in PBS, and adjusted to a cell density of 25-30X 10 6 DCs/ml. According to each 10 6 Transfection of DC cells with 4ug of mRNA ratio, mixing DC cells with antigen mRNA and the regulator composition of the invention (Bcl 2, CD40L, IL15, and IL15 Ra) mRNA combination, adding the cell-mRNA mixture to an electric rotor, and transfecting the antigen mRNA into the DC cells using an ECM630 electric rotor. The cells after the electroporation were resuspended in a cytokine-free 1640 medium, and the cell density was adjusted to 2X10 5 DCs/ml were placed in a 5% CO2 cell incubator at 37 ℃ for further 6 hours.
3. Recovering overnight MNC cells at 2x10 6 The cells were seeded in 96-well plates at a concentration of/ml for T lymphocyte activation. The test grouping case is: a MNC blank control group, a MNC + antigen mRNA-DC vaccine group, a MNC + antigen/immunomodulator composition mRNA-DC vaccine group and a MNC + PMA/Ionomycin positive control group; according to grouping conditions, DC cells loaded with corresponding mRNA are added into different holes, and the MNC is DC = 10; the concentration of Anti-CD3/Anti-CD28 in the positive control is 1 mu g/ml or the concentration of PMA/Ionomycin is 50ng/ml and 1ug/ml; culturing at 37 ℃ for 10 to 12 days.
4. Adding 2 mu M monensin or 3 mu g/ml Brefeldin A into the cell culture solution 5-8h before collecting cells, and fully and uniformly mixing; (Monensin and Brefeldin A should not exceed 12h in cytosol as blockers of protein transport).
5. The cells were transferred to a flow tube, stained with fluorescently labeled antibodies to CD3, CD4, CD8, fixed and permeabilized, and stained intracellularly with fluorescently labeled antibodies to TNF-a and IFN-r.
6. The ratio of TNF-a + and IFN-r + cells in lymphocytes was measured by flow cytometry.
As shown in FIG. 3, the use of either antigen mRNA alone or antigen/immunomodulator composition mRNA can elicit an anti-tumor specific immune response in T lymphocytes. In the control group transfected with mRNA for survivin antigen alone, the proportion of IFN-r positive T lymphocytes was 0.35%, whereas in the group transfected with mRNA for the antigen/immunomodulator composition, the proportion of IFN-r positive T lymphocytes was 1.96% which was 5.60 times that of the control group; in the control group transfected with mRNA for survivin antigen alone, the proportion of T lymphocytes positive to TNF-a was 1.86%, whereas in the group transfected with mRNA for the antigen/immunomodulator composition, the proportion of T lymphocytes positive to TNF-a was 3.26%, which was 1.75 times that of the control group, and was significantly different. The experimental result shows that the immunomodulator composition can obviously enhance the antigen presenting and activating capacities of DC cells on T lymphocytes, better stimulate the T lymphocytes and generate stronger anti-tumor specific immune response.
Sequence listing
<120> an immunomodulator composition, nucleic acid encoding same and use thereof
<141> 2018-11-12
<160> 8
<170> SIPOSequenceListing 1.0
<210> 1
<211> 261
<212> PRT
<213> Homo sapiens
<400> 1
Met Ile Glu Thr Tyr Asn Gln Thr Ser Pro Arg Ser Ala Ala Thr Gly
1 5 10 15
Leu Pro Ile Ser Met Lys Ile Phe Met Tyr Leu Leu Thr Val Phe Leu
20 25 30
Ile Thr Gln Met Ile Gly Ser Ala Leu Phe Ala Val Tyr Leu His Arg
35 40 45
Arg Leu Asp Lys Ile Glu Asp Glu Arg Asn Leu His Glu Asp Phe Val
50 55 60
Phe Met Lys Thr Ile Gln Arg Cys Asn Thr Gly Glu Arg Ser Leu Ser
65 70 75 80
Leu Leu Asn Cys Glu Glu Ile Lys Ser Gln Phe Glu Gly Phe Val Lys
85 90 95
Asp Ile Met Leu Asn Lys Glu Glu Thr Lys Lys Glu Asn Ser Phe Glu
100 105 110
Met Gln Lys Gly Asp Gln Asn Pro Gln Ile Ala Ala His Val Ile Ser
115 120 125
Glu Ala Ser Ser Lys Thr Thr Ser Val Leu Gln Trp Ala Glu Lys Gly
130 135 140
Tyr Tyr Thr Met Ser Asn Asn Leu Val Thr Leu Glu Asn Gly Lys Gln
145 150 155 160
Leu Thr Val Lys Arg Gln Gly Leu Tyr Tyr Ile Tyr Ala Gln Val Thr
165 170 175
Phe Cys Ser Asn Arg Glu Ala Ser Ser Gln Ala Pro Phe Ile Ala Ser
180 185 190
Leu Cys Leu Lys Ser Pro Gly Arg Phe Glu Arg Ile Leu Leu Arg Ala
195 200 205
Ala Asn Thr His Ser Ser Ala Lys Pro Cys Gly Gln Gln Ser Ile His
210 215 220
Leu Gly Gly Val Phe Glu Leu Gln Pro Gly Ala Ser Val Phe Val Asn
225 230 235 240
Val Thr Asp Pro Ser Gln Val Ser His Gly Thr Gly Phe Thr Ser Phe
245 250 255
Gly Leu Leu Lys Leu
260
<210> 2
<211> 162
<212> PRT
<213> Homo sapiens
<400> 2
Met Arg Ile Ser Lys Pro His Leu Arg Ser Ile Ser Ile Gln Cys Tyr
1 5 10 15
Leu Cys Leu Leu Leu Asn Ser His Phe Leu Thr Glu Ala Gly Ile His
20 25 30
Val Phe Ile Leu Gly Cys Phe Ser Ala Gly Leu Pro Lys Thr Glu Ala
35 40 45
Asn Trp Val Asn Val Ile Ser Asp Leu Lys Lys Ile Glu Asp Leu Ile
50 55 60
Gln Ser Met His Ile Asp Ala Thr Leu Tyr Thr Glu Ser Asp Val His
65 70 75 80
Pro Ser Cys Lys Val Thr Ala Met Lys Cys Phe Leu Leu Glu Leu Gln
85 90 95
Val Ile Ser Leu Glu Ser Gly Asp Ala Ser Ile His Asp Thr Val Glu
100 105 110
Asn Leu Ile Ile Leu Ala Asn Asn Ser Leu Ser Ser Asn Gly Asn Val
115 120 125
Thr Glu Ser Gly Cys Lys Glu Cys Glu Glu Leu Glu Glu Lys Asn Ile
130 135 140
Lys Glu Phe Leu Gln Ser Phe Val His Ile Val Gln Met Phe Ile Asn
145 150 155 160
Thr Ser
<210> 3
<211> 267
<212> PRT
<213> Homo sapiens
<400> 3
Met Ala Pro Arg Arg Ala Arg Gly Cys Arg Thr Leu Gly Leu Pro Ala
1 5 10 15
Leu Leu Leu Leu Leu Leu Leu Arg Pro Pro Ala Thr Arg Gly Ile Thr
20 25 30
Cys Pro Pro Pro Met Ser Val Glu His Ala Asp Ile Trp Val Lys Ser
35 40 45
Tyr Ser Leu Tyr Ser Arg Glu Arg Tyr Ile Cys Asn Ser Gly Phe Lys
50 55 60
Arg Lys Ala Gly Thr Ser Ser Leu Thr Glu Cys Val Leu Asn Lys Ala
65 70 75 80
Thr Asn Val Ala His Trp Thr Thr Pro Ser Leu Lys Cys Ile Arg Asp
85 90 95
Pro Ala Leu Val His Gln Arg Pro Ala Pro Pro Ser Thr Val Thr Thr
100 105 110
Ala Gly Val Thr Pro Gln Pro Glu Ser Leu Ser Pro Ser Gly Lys Glu
115 120 125
Pro Ala Ala Ser Ser Pro Ser Ser Asn Asn Thr Ala Ala Thr Thr Ala
130 135 140
Ala Ile Val Pro Gly Ser Gln Leu Met Pro Ser Lys Ser Pro Ser Thr
145 150 155 160
Gly Thr Thr Glu Ile Ser Ser His Glu Ser Ser His Gly Thr Pro Ser
165 170 175
Gln Thr Thr Ala Lys Asn Trp Glu Leu Thr Ala Ser Ala Ser His Gln
180 185 190
Pro Pro Gly Val Tyr Pro Gln Gly His Ser Asp Thr Thr Val Ala Ile
195 200 205
Ser Thr Ser Thr Val Leu Leu Cys Gly Leu Ser Ala Val Ser Leu Leu
210 215 220
Ala Cys Tyr Leu Lys Ser Arg Gln Thr Pro Pro Leu Ala Ser Val Glu
225 230 235 240
Met Glu Ala Met Glu Ala Leu Pro Val Thr Trp Gly Thr Ser Ser Arg
245 250 255
Asp Glu Asp Leu Glu Asn Cys Ser His His Leu
260 265
<210> 8
<211> 239
<212> PRT
<213> Homo sapiens
<400> 8
Met Ala His Ala Gly Arg Thr Gly Tyr Asp Asn Arg Glu Ile Val Met
1 5 10 15
Lys Tyr Ile His Tyr Lys Leu Ser Gln Arg Gly Tyr Glu Trp Asp Ala
20 25 30
Gly Asp Val Gly Ala Ala Pro Pro Gly Ala Ala Pro Ala Pro Gly Ile
35 40 45
Phe Ser Ser Gln Pro Gly His Thr Pro His Pro Ala Ala Ser Arg Asp
50 55 60
Pro Val Ala Arg Thr Ser Pro Leu Gln Thr Pro Ala Ala Pro Gly Ala
65 70 75 80
Ala Ala Gly Pro Ala Leu Ser Pro Val Pro Pro Val Val His Leu Thr
85 90 95
Leu Arg Gln Ala Gly Asp Asp Phe Ser Arg Arg Tyr Arg Arg Asp Phe
100 105 110
Ala Glu Met Ser Ser Gln Leu His Leu Thr Pro Phe Thr Ala Arg Gly
115 120 125
Arg Phe Ala Thr Val Val Glu Glu Leu Phe Arg Asp Gly Val Asn Trp
130 135 140
Gly Arg Ile Val Ala Phe Phe Glu Phe Gly Gly Val Met Cys Val Glu
145 150 155 160
Ser Val Asn Arg Glu Met Ser Pro Leu Val Asp Asn Ile Ala Leu Trp
165 170 175
Met Thr Glu Tyr Leu Asn Arg His Leu His Thr Trp Ile Gln Asp Asn
180 185 190
Gly Gly Trp Asp Ala Phe Val Glu Leu Tyr Gly Pro Ser Met Arg Pro
195 200 205
Leu Phe Asp Phe Ser Trp Leu Ser Leu Lys Thr Leu Leu Ser Leu Ala
210 215 220
Leu Val Gly Ala Cys Ile Thr Leu Gly Ala Tyr Leu Gly His Lys
225 230 235
<210> 4
<211> 2316
<212> RNA
<213> Artificial Sequence (Artificial Sequence)
<400> 4
agaccggccu cgagcagcug aagcuugcau gccugcaggu cgacucuaga ggauccgcca 60
ccaugagaau uucgaaacca cauuugagaa guauuuccau ccagugcuac uuguguuuac 120
uucuaaacag ucauuuucua acugaagcug gcauucaugu cuucauuuug ggcuguuuca 180
gugcagggcu uccuaaaaca gaagccaacu gggugaaugu aauaagugau uugaaaaaaa 240
uugaagaucu uauucaaucu augcauauug augcuacuuu auauacggaa agugauguuc 300
accccaguug caaaguaaca gcaaugaagu gcuuucucuu ggaguuacaa guuauuucac 360
uugaguccgg agaugcaagu auucaugaua caguagaaaa ucugaucauc cuagcaaaca 420
acaguuuguc uucuaauggg aauguaacag aaucuggaug caaagaaugu gaggaacugg 480
aggaaaaaaa uauuaaagaa uuuuugcaga guuuuguaca uauuguccaa auguucauca 540
acacuucuug agcggccgcc gcccgcccca cgacccgcag cgcccgaccg aaaggagcgc 600
acgaccccau cauccaauuc cgcccccccc cccuaacguu acuggccgaa gccgcuugga 660
auaaggccgg ugugcguuug ucuauauguu auuuuccacc auauugccgu cuuuuggcaa 720
ugugagggcc cggaaaccug gcccugucuu cuugacgagc auuccuaggg gucuuucccc 780
ucucgccaaa ggaaugcaag gucuguugaa ugucgugaag gaagcaguuc cucuggaagc 840
uucuugaaga caaacaacgu cuguagcgac ccuuugcagg cagcggaacc ccccaccugg 900
cgacaggugc cucugcggcc aaaagccacg uguauaagau acaccugcaa aggcggcaca 960
accccagugc cacguuguga guuggauagu uguggaaaga gucaaauggc ucuccucaag 1020
cguauucaac aaggggcuga aggaugccca gaagguaccc cauuguaugg gaucugaucu 1080
ggggccucgg ugcacaugcu uuacaugugu uuagucgagg uuaaaaaacg ucuaggcccc 1140
ccgaaccacg gggacguggu uuuccuuuga aaaacacgau gauaauaugg ccacaacguc 1200
gacgccacca uggcuccuag gagagccaga ggguguagga cacugggacu gccagcucug 1260
cugcugcugc ugcugcugag accuccagcu acaaggggaa ucaccugccc uccuccuaug 1320
agcguggagc acgccgacau uugggugaag agcuacagcc uguacagccg ggagcgcuac 1380
auuugcaaca gcggcuucaa gaggaaggcc ggaacaagcu cucucaccga gugcgugcug 1440
aacaaggcca ccaacguggc ccauuggaca accccuagcc ugaagugcau cagggaccca 1500
gcacuggugc accagagacc agcuccuccu agcacaguga ccacagccgg agugacaccu 1560
cagccagaaa gccugagccc uagcggaaaa gaaccagccg ccucuagccc cagcagcaau 1620
aauaccgccg ccacaacagc cgcuauugug ccaggaagcc agcugaugcc uagcaagagc 1680
ccuagcaccg gcacaacaga gaucagcagc cacgagagca gccacggaac accuagccag 1740
accacagcca agaauuggga gcugaccgcc agcgccagcc accagccucc aggaguguac 1800
ccucagggac acagcgauac caccguggcc aucucuacca gcacagugcu gcugugcgga 1860
cugucagcug ugucccugcu ggcuugcuac cugaagagca gacagacccc uccucuggcc 1920
agcguggaaa uggaggcuau ggaggcccug ccagugacuu ggggaaccuc uagcagagac 1980
gaggaccugg agaauugcag ccaccaccug uaggaauucg cuggagccuc gguagccguu 2040
ccuccugccc gcugggccuc ccaacgggcc cuccuccccu ccuugcaccg gcccuuccug 2100
gucuuuggcu ggagccucgg uagccguucc uccugcccgc ugggccuccc aacgggcccu 2160
ccuccccucc uugcaccggc ccuuccuggu cuuugaaaaa aaaaaaaaaa aaaaaaaaaa 2220
aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 2280
aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaa 2316
<210> 5
<211> 946
<212> RNA
<213> Artificial Sequence (Artificial Sequence)
<400> 5
gagacaagcu ugcaugccug caggucgacu cuagaggauc caccggucgc caccaugauc 60
gaaacauaca accaaacuuc uccccgaucu gcggccacug gacugcccau cagcaugaaa 120
auuuuuaugu auuuacuuac uguuuuucuu aucacccaga ugauuggguc agcacuuuuu 180
gcuguguauc uucauagaag guuggacaag auagaagaug aaaggaaucu ucaugaagau 240
uuuguauuca ugaaaacgau acagagaugc aacacaggag aaagauccuu auccuuacug 300
aacugugagg agauuaaaag ccaguuugaa ggcuuuguga aggauauaau guuaaacaaa 360
gaggagacga agaaagaaaa cagcuuugaa augcaaaaag gugaucagaa uccucaaauu 420
gcggcacaug ucauaaguga ggccagcagu aaaacaacau cuguguuaca gugggcugaa 480
aaaggauacu acaccaugag caacaacuug guaacccugg aaaaugggaa acagcugacc 540
guuaaaagac aaggacucua uuauaucuau gcccaaguca ccuucuguuc caaucgggaa 600
gcuucgaguc aagcuccauu uauagccagc cucugccuaa agucccccgg uagauucgag 660
agaaucuuac ucagagcugc aaauacccac aguuccgcca aaccuugcgg gcaacaaucc 720
auucacuugg gaggaguauu ugaauugcaa ccaggugcuu cgguguuugu caaugugacu 780
gauccaagcc aagugagcca uggcacuggc uucacguccu uuggcuuacu caaacucuga 840
agcggccgcg gauccccggg uaccgagcuc gaauucuuaa uuaaaaaaaa aaaaaaaaaa 900
aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaa 946
<210> 6
<211> 552
<212> RNA
<213> Encephalomyocarditis virus
<400> 6
acguuacugg ccgaagccgc uuggaauaag gccggugugc guuugucuau auguuauuuu 60
ccaccauauu gccgucuuuu ggcaauguga gggcccggaa accuggcccu gucuucuuga 120
cgagcauucc uaggggucuu uccccucucg ccaaaggaau gcaaggucug uugaaugucg 180
ugaaggaagc aguuccucug gaagcuucuu gaagacaaac aacgucugua gcgacccuuu 240
gcaggcagcg gaacccccca ccuggcgaca ggugccucug cggccaaaag ccacguguau 300
aagauacacc ugcaaaggcg gcacaacccc agugccacgu ugugaguugg auaguugugg 360
aaagagucaa auggcucucc ucaagcguau ucaacaaggg gcugaaggau gcccagaagg 420
uaccccauug uaugggaucu gaucuggggc cucggugcac augcuuuaca uguguuuagu 480
cgagguuaaa aaacgucuag gccccccgaa ccacggggac gugguuuucc uuugaaaaac 540
acgaugauaa ua 552
<210> 7
<211> 842
<212> RNA
<213> Artificial Sequence (Artificial Sequence)
<400> 7
gagaccggcc ucgagcagcu gaagcuuccu gcaggucgac ucuagagcca ccauggcgca 60
cgcugggaga acaggguacg auaaccggga gauagugaug aaguacaucc auuauaagcu 120
gucgcagagg ggcuacgagu gggaugcggg agaugugggc gccgcgcccc cgggggccgc 180
ccccgcaccg ggcaucuucu ccucccagcc cgggcacacg ccccauccag ccgcaucccg 240
ggacccgguc gccaggaccu cgccgcugca gaccccggcu gcccccggcg ccgccgcggg 300
gccugcgcuc agcccggugc caccuguggu ccaccugacc cuccgccagg ccggcgacga 360
cuucucccgc cgcuaccgcc gcgacuucgc cgagaugucc agccagcugc accugacgcc 420
cuucaccgcg cggggacgcu uugccacggu gguggaggag cucuucaggg acggggugaa 480
cugggggagg auuguggccu ucuuugaguu cggugggguc augugugugg agagcgucaa 540
ccgggagaug ucgccccugg uggacaacau cgcccugugg augacugagu accugaaccg 600
gcaccugcac accuggaucc aggauaacgg aggcugggau gccuuugugg aacuguacgg 660
ccccagcaug cggccucugu uugauuucuc cuggcugucu cugaagacuc ugcucaguuu 720
ggcccuggug ggagcuugca ucacccuggg ugccuaucug ggccacaagu gauuaauuaa 780
aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 840
aa 842
Claims (1)
1. Use of an immunopotentiator composition for the preparation of a dendritic cell reagent for increasing the proportion of IFN-. Gamma.and/or TNF-. Alpha.positive cells in T lymphocytes, wherein said immunopotentiator composition comprises nucleic acid molecules encoding a Bcl2 protein, a CD40L protein, an IL15 receptor alpha protein and a survivin antigen, wherein the amino acid sequence of CD40L is represented by SEQ ID NO. 1, the amino acid sequence of IL15 is represented by SEQ ID NO. 2, the amino acid sequence of IL15 receptor alpha is represented by SEQ ID NO. 3, and the amino acid sequence of Bcl2 is represented by SEQ ID NO. 4.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201811343570.5A CN111166876B (en) | 2018-11-13 | 2018-11-13 | Immunopotentiator combination, encoding nucleic acid and application thereof |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201811343570.5A CN111166876B (en) | 2018-11-13 | 2018-11-13 | Immunopotentiator combination, encoding nucleic acid and application thereof |
Publications (2)
Publication Number | Publication Date |
---|---|
CN111166876A CN111166876A (en) | 2020-05-19 |
CN111166876B true CN111166876B (en) | 2022-10-11 |
Family
ID=70622134
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN201811343570.5A Active CN111166876B (en) | 2018-11-13 | 2018-11-13 | Immunopotentiator combination, encoding nucleic acid and application thereof |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN111166876B (en) |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2024041618A1 (en) * | 2022-08-25 | 2024-02-29 | 广州百吉生物制药有限公司 | Engineered immune cell co-expressing cd40l, preparation therefor, and use thereof |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR20030084826A (en) * | 2003-09-30 | 2003-11-01 | 국립암센터 | Dendritic cell pulsing allogeneic tumor-associated antigen, method for the preparation thereof and vaccine composition containing same for treating cancer |
CN101460054A (en) * | 2006-04-07 | 2009-06-17 | 阿戈斯治疗公司 | Mature dendritic cell compositions and methods for culturing same |
CN103405759A (en) * | 2013-07-23 | 2013-11-27 | 蔡建辉 | Method for preparing tumor-specific DC vaccine by applying CD34+ cells of umbilical cord blood |
CN107929727A (en) * | 2017-08-15 | 2018-04-20 | 北京启辰生生物科技有限公司 | A kind of preparation method of new dendritic cell vaccine |
-
2018
- 2018-11-13 CN CN201811343570.5A patent/CN111166876B/en active Active
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR20030084826A (en) * | 2003-09-30 | 2003-11-01 | 국립암센터 | Dendritic cell pulsing allogeneic tumor-associated antigen, method for the preparation thereof and vaccine composition containing same for treating cancer |
CN101460054A (en) * | 2006-04-07 | 2009-06-17 | 阿戈斯治疗公司 | Mature dendritic cell compositions and methods for culturing same |
CN103405759A (en) * | 2013-07-23 | 2013-11-27 | 蔡建辉 | Method for preparing tumor-specific DC vaccine by applying CD34+ cells of umbilical cord blood |
CN107929727A (en) * | 2017-08-15 | 2018-04-20 | 北京启辰生生物科技有限公司 | A kind of preparation method of new dendritic cell vaccine |
Non-Patent Citations (4)
Title |
---|
Bcl-2 Controls Dendritic Cell Longevity In Vivo;Adam Nopora等;《The Journal of Immunology》;20021231;第169卷;摘要 * |
Characterization of Interleukin-15-Transpresenting Dendritic Cells for Clinical Use;J.M.J.Van den Bergh等;《Journal of Immunology Research》;20170713;第2017卷;摘要 * |
Cytokine Maturation Followed by CD40L mRNA Electroporation Results in a Clinically Relevant Dendritic Cell Product Capable of Inducing a Potent Proinflammatory CTL Response;David M. Calderhead等;《J Immunother》;20081031;第31卷(第8期);摘要、第732页左栏第2段至右栏第2段、表1 * |
Increased function and survival of IL-15-transduced human dendritic cells are mediated by up-regulation of IL-15Rα and Bcl-2;Irina L. Tourkova等;《Journal of Leukocyte Biology》;20021130;第72卷;摘要、RESULTS和DISCUSSION部分 * |
Also Published As
Publication number | Publication date |
---|---|
CN111166876A (en) | 2020-05-19 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN110464841B (en) | Immunity-enhancing pharmaceutical composition and application thereof | |
US20220062398A1 (en) | Long chain antigen containing interepitope sequence that promotes antigen presentation to t cells | |
Schaft et al. | Generation of an optimized polyvalent monocyte-derived dendritic cell vaccine by transfecting defined RNAs after rather than before maturation | |
Chiriva‐Internati et al. | Efficient generation of cytotoxic T lymphocytes against cervical cancer cells by adeno‐associated virus/human papillomavirus type 16 E7 antigen gene transduction into dendritic cells | |
Jeras et al. | In vitro preparation and functional assessment of human monocyte-derived dendritic cells—potential antigen-specific modulators of in vivo immune responses | |
Dullaers et al. | Side-by-side comparison of lentivirally transduced and mRNA-electroporated dendritic cells: implications for cancer immunotherapy protocols | |
KR20040025690A (en) | Improved transfection of eukaryotic cells with linear polynucleotides by electroporation | |
WO2018206577A1 (en) | Interferon primed plasmacytoid dendritic cells | |
Koya et al. | Potent maturation of monocyte-derived dendritic cells after CD40L lentiviral gene delivery | |
JP2021518767A (en) | Human DC cell amplification method and human DC cell resource bank | |
US20180078627A1 (en) | Method for antigen loading of dendritic cells and vaccine | |
Pincha et al. | Identity, potency, in vivo viability, and scaling up production of lentiviral vector-induced dendritic cells for melanoma immunotherapy | |
Riedl et al. | Overexpression of CCL-21/secondary lymphoid tissue chemokine in human dendritic cells augments chemotactic activities for lymphocytes and antigen presenting cells | |
Mason et al. | RNA-loaded CD40-activated B cells stimulate antigen-specific T-cell responses in dogs with spontaneous lymphoma | |
CN113101363B (en) | Circular RNA vaccine and application thereof | |
Sabado et al. | Dendritic cell vaccines | |
CN111166876B (en) | Immunopotentiator combination, encoding nucleic acid and application thereof | |
Zhou et al. | Comparative analysis of cytotoxic T lymphocyte response induced by dendritic cells pulsed with recombinant adeno-associated virus carrying α-fetoprotein gene or cancer cell lysate | |
Michiels et al. | Induction of antigen-specific CD8+ cytotoxic T cells by dendritic cells co-electroporated with a dsRNA analogue and tumor antigen mRNA | |
CN107574149B (en) | Maturation promoting method of dendritic cells and application thereof | |
JP2002069001A (en) | Cell vaccine consisting mainly of dendritic cell | |
EP2575867A1 (en) | Novel interferon-alpha-producing bone marrow dendritic cells | |
WO2018032619A1 (en) | Applications of soluble protein baff in b cell in-vitro culture and proliferation | |
Hu et al. | Induction of antigen-specific cytotoxic T-cell response by dendritic cells generated from ecto-mesenchymal stem cells infected with an adenovirus containing the MAGE-D4a gene | |
CN116970562B (en) | Preparation method of antigen-specific T cells and application of antigen-specific T cells in immunotherapy |
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 |