CN116814688B - Vector carrying bispecific promoter, construction method and application thereof - Google Patents

Vector carrying bispecific promoter, construction method and application thereof Download PDF

Info

Publication number
CN116814688B
CN116814688B CN202310794540.0A CN202310794540A CN116814688B CN 116814688 B CN116814688 B CN 116814688B CN 202310794540 A CN202310794540 A CN 202310794540A CN 116814688 B CN116814688 B CN 116814688B
Authority
CN
China
Prior art keywords
vector
promoter
virus
bispecific
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202310794540.0A
Other languages
Chinese (zh)
Other versions
CN116814688A (en
Inventor
宋灿
王振宇
冉杰
田甜
于杨
郭平
顾雨春
吴理达
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.)
Chengnuo Regenerative Medical Technology Beijing Co ltd
Original Assignee
Chengnuo Regenerative Medical Technology Beijing 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 Chengnuo Regenerative Medical Technology Beijing Co ltd filed Critical Chengnuo Regenerative Medical Technology Beijing Co ltd
Priority to CN202310794540.0A priority Critical patent/CN116814688B/en
Publication of CN116814688A publication Critical patent/CN116814688A/en
Application granted granted Critical
Publication of CN116814688B publication Critical patent/CN116814688B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention discloses a carrier carrying a bispecific promoter, a construction method and application thereof, and provides a carrier of the bispecific promoter, a construction method of the carrier of the bispecific promoter, a host cell comprising the carrier of the bispecific promoter, a virus and a preparation method thereof, and a related pharmaceutical composition which can be specifically expressed in specific tumor cells to realize accurate killing of the tumor cells, and meanwhile, the carrier of the bispecific promoter in the invention is not expressed or is rarely expressed in non-tumor cells, so that the probability of accidental injury of normal cells is reduced, the safety is improved, and the pharmaceutical composition has wide market application prospect.

Description

Vector carrying bispecific promoter, construction method and application thereof
Technical Field
The invention belongs to the technical field of biology, and relates to a vector carrying a bispecific promoter, a construction method and application thereof.
Background
Gene medicine using adeno-associated virus (AAV) as carrier has the characteristics of durable gene expression and low immunogenicity, and has been widely used in the fields of monogenic genetic diseases, ophthalmic diseases, nervous system diseases, etc. However, AAV has weak infection specificity on tumor cells, so that normal cells can be infected while tumor cells are infected, and the application of AAV in the field of tumor treatment is limited.
The Tumor specific promoter (Tumor-specific promoter) holds promise for the application of AAV in the Tumor field. Tumor specific promoters refer to promoter sequences that are specifically expressed in tumor cells. These promoters have higher activity than normal cells or are activated only in tumor cells. The application of the tumor specific promoter can achieve various purposes. First, they can be used for tumor gene therapy, and by combining a therapeutic gene with a tumor specific promoter, the therapeutic gene is expressed only in tumor cells, thereby improving the specificity and safety of the therapy. Second, tumor specific promoters can also be used in tumor immunotherapy to promote immune cells in tumor-specific anti-tumor responses locally by combining immune-related genes with tumor specific promoters. In addition, the tumor specific promoter can also be used for tumor diagnosis, and the visualization of tumor cells or the development of molecular probes can be realized by combining a reporter gene with the tumor specific promoter.
Disclosure of Invention
The invention provides a carrier carrying a dual-specificity promoter, a construction method and application thereof, which have killing function when two specific promoters are simultaneously started, and improve the accuracy and safety of tumor treatment in order to make up for the defects of the prior art and solve the specific problems in the prior art.
The invention provides a vector of a bispecific promoter, wherein the bispecific promoter is two specific promoters.
The carrier comprises: the bispecific promoters are linked to the genes of interest, respectively.
The gene of interest comprises at least one of the following combinations: a combination of Casp1 acts and GSDMD, a combination of GSDMD C and GSDMD N.
The two components of each combination of the target genes can be respectively called target gene 1 and target gene 2.
The term "vector" includes biological or chemical parts of a nucleic acid sequence that can be introduced into an appropriate host cell for replication or expression of the nucleic acid sequence. Vectors include non-viral vectors and viral vectors. In some embodiments of the invention, the non-viral vector may be selected from the group consisting of nanoparticles, novel biological materials, naked DNA, phage, transposons, plasmids, cosmids, artificial chromosomes. Plasmids, other cloning and expression vectors, their nature, and methods of construction that can be used in accordance with the present invention will be apparent to those skilled in the art.
Further, the two promoters of the bispecific promoter may be referred to as specific promoter 1, specific promoter 2, respectively.
Further, the structure of the vector of the bispecific promoter comprises a specific promoter 1, a target gene 1, a specific promoter 2 and a target gene 2 which are connected in sequence.
Further, the specific promoter and the target gene are connected with a special sticky end to ensure the accuracy of connection.
Further, the specific promoter is a cancer-associated specific promoter.
Further, the cancer includes squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, peritoneal cancer, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, liver cancer, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine cancer, salivary gland cancer, kidney cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic cancer, anal cancer, penile cancer, scrotal cancer, esophageal cancer, biliary tract tumor, and cancer of the head and neck.
Further, the two promoters corresponding to the bispecific promoter are any two of PCA3, PEG3, human telomerase reverse transcriptase promoter, survivin promoter, medkine promoter, carcinoembryonic antigen promoter, MUC-1 promoter and probasin gene promoter.
Further, the vectors include plasmids, lentiviral vectors, retroviral vectors, adenoviral vectors, adeno-associated viral vectors, linear polynucleotides.
Further, the vector includes an AAV viral core plasmid.
Further, the vector of the bispecific promoter comprises a vector with the structure AAV-PCA3-Casp1act-PEG 3-GSDMD.
Further, the vector of the bispecific promoter comprises a Casp1 act nucleotide sequence as shown in SEQ ID NO. 1.
Further, the vector of the bispecific promoter comprises a GSDMD nucleotide sequence as shown in SEQ ID NO. 2.
Further, the vector of the bispecific promoter comprises the PCA3 nucleotide sequence as shown in SEQ ID NO. 3.
Further, the vector of the bispecific promoter comprises a PEG3 nucleotide sequence as shown in SEQ ID NO. 4.
Further, the vector structure of the bispecific promoter comprises the nucleotide sequences of SEQ ID NO. 3, SEQ ID NO. 1, SEQ ID NO. 4 and SEQ ID NO. 2 which are connected in sequence.
In certain embodiments, the structure of the vector of the bispecific promoter is AAV-PCA3-Casp1act-PEG3-GSDMD, meaning that the PCA3 promoter, casp1act, PEG3 promoter, GSDMD are linked in sequence in an AAV virus. In certain specific embodiments, the PCA3 promoter, casp1act, PEG3 promoter, GSDMD are linked in sequence in the AAV virus comprising the nucleotide sequence of SEQ ID NO:3, SEQ ID NO:1, SEQ ID NO:4, SEQ ID NO:2, linked in sequence. In certain embodiments, the structure of the vector of the bispecific promoter is AAV-PEG3-GSDMD-PCA3-Casp 1act, which means that the PEG3 promoter, GSDMD, PCA3 promoter, casp1act are linked in sequence in AAV virus. In certain specific embodiments, sequentially ligating a PEG3 promoter, GSDMD, PCA3 promoter, casp1act in the AAV virus comprises sequentially ligating the nucleotide sequences of SEQ ID NO. 4, SEQ ID NO. 2, SEQ ID NO. 3, SEQ ID NO. 1.
In certain embodiments, the vector may comprise other suitable "regulatory elements" or "regulatory sequences" in addition to the promoter, including but not limited to enhancers; a transcription factor; a transcription terminator; high efficiency RNA processing signals such as splicing and polyadenylation signals (polyA); sequences that stabilize cytoplasmic mRNA, such as the Woodchuck Hepatitis Virus (WHV) post-transcriptional regulatory element (WPRE); sequences that enhance translation efficiency (i.e., kozak consensus sequences); a sequence that enhances protein stability; and, if desired, sequences that enhance secretion of the encoded product. In certain embodiments, examples of the polyA include SV40, bovine growth hormone (bGH), and TK polyA. In certain embodiments, examples of such enhancers include the alpha fetoprotein enhancer, the TTR minimal promoter/enhancer, the LSP (TH binding globulin promoter/alpha 1-microglobulin/specific library protein (bikunin) enhancer), and other enhancers.
The invention also provides a construction method of the vector of the bispecific promoter, which is used for constructing the vector, and comprises the following steps:
Obtaining a gene of interest comprising at least one of the following combinations: a combination of Casp1 acts and GSDMD, a combination of GSDMD C and GSDMD N;
obtaining a bispecific promoter;
and assembling the combination of the target genes and the bispecific promoter on a vector to obtain the vector.
Further, the two components of each combination of the genes of interest may be called the gene of interest 1 and the gene of interest 2, respectively.
Further, the two promoters of the bispecific promoter may be referred to as specific promoter 1, specific promoter 2, respectively.
Further, the structure of the vector of the bispecific promoter comprises a specific promoter 1, a target gene 1, a specific promoter 2 and a target gene 2 which are connected in sequence.
Further, the specific promoter and the target gene are connected with a special sticky end to ensure the accuracy of connection.
Further, the two promoters of the bispecific promoter comprise any two of PCA3, PEG3, human telomerase reverse transcriptase promoter, survivin promoter, medkine promoter, carcinoembryonic antigen promoter, MUC-1 promoter and probasin gene promoter.
Further, the vectors include plasmids, lentiviral vectors, retroviral vectors, adenoviral vectors, adeno-associated viral vectors, linear polynucleotides.
Further, the vector is an AAV viral core plasmid.
In certain embodiments, the AAV virus comprises an AAV viral core plasmid or an AAV viral core plasmid engineered plasmid, the structure of which comprises an AAV capsid and a vector genome packaged in the capsid.
The term "vector genome" refers to an engineered viral or non-viral core vector, generally referred to as a plasmid, lentiviral vector, retroviral vector, adenoviral vector, core genetic material in an adeno-associated viral vector, linear polynucleotide, and the like. The "vector genome" may be, under appropriate conditions, the core genetic material of a non-proviral vector or the core genetic material of a non-proviral vector. In certain specific embodiments, the vector genome may be a vector of the bispecific promoters described herein. In certain specific embodiments, the vector genome comprises a viral core plasmid or a viral core plasmid engineered plasmid.
In certain embodiments, an AAV viral vector is provided. An "AAV virus" is a dnase resistant viral particle comprising two elements, an AAV capsid and a vector genome comprising at least non-AAV coding sequences packaged within the AAV capsid. The source of AAV capsids can be any of several tens of naturally occurring and available adeno-associated viruses, as well as engineered AAV.
The term "AAV capsid" or "AAV vector intermediate" refers to an assembled AAV capsid lacking the desired genomic sequences packaged therein. It may also be referred to as an "empty" capsid. The capsid may contain no gene sequence of interest or only partially packaged genomic sequence insufficient to effect expression of the gene product. The "empty" capsids are nonfunctional to transfer the gene of interest to the host cell.
Further, the construction method comprises any one of the following steps:
a. amplifying the combination of the target genes, amplifying two promoters of the dual-specificity promoter, respectively connecting the target genes with the specificity promoter, and cloning the connecting object to the carrier to obtain the carrier comprising the dual-specificity promoter and the target genes;
b. Amplifying the target gene and two promoters of the specific promoter, and then directly cloning the target gene and the two promoters on the vector to obtain the vector comprising the bispecific promoter and the target gene.
Further, the specific promoter and the target gene are connected with a special sticky end to ensure the accuracy of connection.
The invention also provides a host cell comprising the vector as described above.
Further, the host cells include prokaryotic cells and eukaryotic cells.
Further, the prokaryotic cells include bacteria, actinomycetes, cyanobacteria, mycoplasma, chlamydia, rickettsia.
Further, the eukaryotic cells include mammalian cells, insect cells, plant cells, yeast cells.
Further, the mammalian cells include fibroblasts, 293T cells.
In one embodiment, the host cell is a cell capable of engineering culture. The term "host cell" may refer to a packaging cell line in which the vector is produced from a production plasmid. In a certain embodiment, the term "host cell" may refer to any target cell in which transgene expression is desired. Thus, "host cell" refers to a prokaryotic or eukaryotic cell containing an exogenous or heterologous nucleic acid sequence that has been introduced into the cell by any means (e.g., electroporation, calcium phosphate precipitation, microinjection, transformation, viral infection, transfection, liposome delivery, membrane fusion techniques, high-speed DNA coated aggregates, viral infection, and protoplast fusion).
In a particular embodiment, the term "host cell" refers to a culture of cells of various mammalian species used for in vitro evaluation of the compositions described herein. In other embodiments herein, the term "host cell" refers to a cell used to produce and package a viral vector or recombinant virus. In another embodiment, the term "host cell" is intended to refer to a target cell of an individual treated in vivo for a disease or condition as described herein. In certain embodiments, the term "host cell" is a prostate cell.
The invention also provides a virus comprising the vector as described above.
The carrier generation process may comprise, for example, the following method steps: cell culture initiation, cell passage, cell inoculation, transfection of cells with plasmid DNA, replacement of the post-transfection medium with serum-free medium, and harvesting of the vector-containing cells and medium.
Further, the viruses include lentiviruses, retroviruses, adenoviruses, adeno-associated viruses, herpesviruses, poxviruses, papovaviruses, baculoviruses, papillomaviruses.
Further, the virus is an adeno-associated virus.
The invention also provides a preparation method of the virus, which comprises the following steps:
Transfecting the vector as described above into a host cell; and amplifying and expressing the transfected host cells to obtain the virus.
Further, the viruses include lentiviruses, retroviruses, adenoviruses, adeno-associated viruses, herpesviruses, poxviruses, papovaviruses, baculoviruses, papillomaviruses.
Further, the virus is an adeno-associated virus.
Further, the means by which the vector is transformed into a host cell include electroporation, calcium phosphate precipitation, microinjection, transformation, viral infection, transfection, liposome delivery, membrane fusion techniques, high-speed DNA coated aggregates, viral infection, and protoplast fusion.
Further, the host cells include prokaryotic cells and eukaryotic cells.
Further, the prokaryotic cells include bacteria, actinomycetes, cyanobacteria, mycoplasma, chlamydia, rickettsia.
Further, the eukaryotic cells include mammalian cells, insect cells, plant cells, yeast cells.
Further, the mammalian cells include fibroblasts, 293T cells.
The invention also provides a pharmaceutical composition comprising the vector as described above, the host cell as described above, and the virus as described above.
Further, the pharmaceutical composition further comprises pharmaceutically acceptable auxiliary ingredients.
Further, the adjunct ingredients include solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, gums.
Further, the pharmaceutical composition is in the form of a lipid particle, liposome, vesicle, nanosphere or nanoparticle or the like.
In certain embodiments, the pharmaceutical composition is engineered into a non-viral vector, such as a plasmid or other genetic element, which is admixed with a carrier, diluent, excipient, and/or the like to form the pharmaceutical composition. In certain embodiments, the pharmaceutical composition is engineered into a viral vector that is mixed with a carrier, diluent, excipient, and/or the like to form the pharmaceutical composition. In certain embodiments, the pharmaceutical composition is engineered into a vector genome that is packaged into an AAV capsid to form an adeno-associated virus, which is mixed with a carrier, diluent, excipient, and/or the like to form the pharmaceutical composition. In certain embodiments, the vector genome may be packaged into different viruses.
The term "adjunct ingredients" encompasses any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, gums, and the like. The use of such media and medicaments for pharmaceutically active substances is well known in the art. Supplementary active ingredients may also be incorporated into the compositions. By "pharmaceutically acceptable" is meant molecular entities and compositions that do not produce allergic or similar untoward reactions when administered to a host. Conventional and pharmaceutically acceptable routes of administration include, but are not limited to, direct delivery to the desired organ (e.g., liver (optionally via hepatic artery), lung, heart, eye, kidney), oral, inhalation, intranasal, intrathecal, intratracheal, intraarterial, intraocular, intravenous, intramuscular, subcutaneous, intradermal, and other parental routes of administration. The routes of administration may be combined, as desired.
Any suitable route of administration (e.g., oral, inhalation, intranasal, intratracheal, intraarterial, intraocular, intravenous, intramuscular, intraperitoneal, and other parental routes) may be selected. Thus, the pharmaceutical compositions may be formulated for any suitable route of administration, for example, in the form of a liquid solution or suspension (e.g., for intravenous administration, for oral administration, etc.). Alternatively, the pharmaceutical composition may be in solid form (e.g., in the form of a tablet or capsule, e.g., for oral administration). In some embodiments, the pharmaceutical composition may be in the form of a powder, drops, spray, or the like.
The invention also provides the use of the vector as defined above, the method of construction as defined above, or the host cell as defined above in the preparation of a virus or plasmid.
Further, the viruses include lentiviruses, retroviruses, adenoviruses, adeno-associated viruses, herpesviruses, poxviruses, papovaviruses, baculoviruses, papillomaviruses.
Further, the virus is an adeno-associated virus.
The invention also provides the application of the vector, the construction method, the host cell, the virus or the preparation method in preparing medicines for treating tumors.
Further, the cancer includes squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, peritoneal cancer, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, liver cancer, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine cancer, salivary gland cancer, kidney cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic cancer, anal cancer, penile cancer, scrotal cancer, esophageal cancer, biliary tract tumor, and cancer of the head and neck.
The terms "comprises," "comprising," "includes," "including," and variations thereof include other components, elements, integers, steps, and the like. In contrast, the term "consisting of … …" and variations thereof does not include other components, elements, integers, steps, etc.
The invention has the beneficial effects that:
The invention provides a vector of a double-specific promoter, which has the function of being capable of specifically expressing in tumor cells, and expressing two necessary target genes under the condition that two promoters are started, wherein the interaction of the two target genes plays a role in killing the tumor cells; meanwhile, the vector of the bispecific promoter is not expressed and rarely expressed in non-tumor cells, and if one promoter is abnormally expressed in the non-tumor cells, the vector does not play a role in killing cells, so that the probability of accidental injury of normal cells is reduced, the safety is improved, and the vector has a wide market application prospect.
Drawings
FIG. 1 is a graph of the expression of different promoters in Fibroblasts (FB);
FIG. 2 is a graph of the expression of different promoters in normal prostate cells (RWPE-1);
FIG. 3 is a graph of the expression of different promoters in prostate cancer cells (C4-2);
FIG. 4 is a graph showing the killing of different plasmid in prostate cancer cells (C4-2) cells;
FIG. 5 is a graph of killing by different plasmids in normal prostate cells (RWPE-1);
FIG. 6 is a graph of in vivo experimental effectiveness of AAV, wherein A is a graph of tumor effect after different plasmids are inoculated into a control group of mice and an experimental group of mice, and B is a graph of tumor volume after inoculation;
FIG. 7 is a graph showing the statistical weight of control mice and experimental mice after inoculation with different plasmids;
FIG. 8 is a biochemical test diagram of mouse blood, wherein A is a graph of transglutaminase, B is a graph of glutamic pyruvic transaminase, C is a graph of alkaline phosphatase, and D is a graph of glutamic oxaloacetic transaminase.
Detailed Description
The following describes the embodiments of the present invention in further detail with reference to examples.
Example 1 specific promoter verification
1. Experimental materials
① Prostate cancer cell C4-2, normal prostate cell Rwpe-1, fibroblast FB, 293T cell;
② PEI transfection reagent.
2. Experimental method
1) Vector construction
Using EGFP as a control, the SFFV-EGFP gene (control), AAV-SFFV-Casp1act-GSDMD, PCA3-Casp1 act-PEG3-GSDMD were synthesized and ligated into AAV vectors via cleavage sites 5'-PacI and 3' -EcoRI, respectively.
The nucleotide sequences of the elements are as follows:
casp1 act nucleotide sequence (SEQ ID NO: 1):
ATGAACCCAGCTATGCCCACATCCTCAGGCTCAGAAGGGAATGTCAAGCTTTGCTCCCTAGAAGAAGCTCAAAGGATATGGAAACAAAAGTCGGCAGAGATTTATCCAATAATGGACAAGTCAAGCCGCACACGTCTTGCTCTCATTATCTGCAATGAAGAATTTGACAGTATTCCTAGAAGAACTGGAGCTGAGGTTGACATCACAGGCATGACAATGCTGCTACAAAATCTGGGGTACAGCGTAGATGTGAAAAAAAATCTCACTGCTTCGGACATGACTACAGAGCTGGAGGCATTTGCACACCGCCCAGAGCACAAGACCTCTGACAGCACGTTCCTGGTGTTCATGTCTCATGGTATTCGGGAAGGCATTTGTGGGAAGAAACACTCTGAGCAAGTCCCAGATATACTACAACTCAATGCAATCTTTAACATGTTGAATACCAAGAACTGCCCAAGTTTGAAGGACAAACCGAAGGTGATCATCATCCAGGCCTGCCGTGGTGACAGCCCTGGTGTGGTGTGGTTTAAAGATGGAAGCGGAGCTACTAACTTCAGCCTGCTGAAGCAGGCTGGAGACGTGGAGGAGAACCCTGGACCTATGGCTATTAAGAAAGCCCACATAGAGAAGGATTTTATCGCTTTCTGCTCTTCCACACCAGATAATGTTTCTTGGAGACATCCCACAATGGGCTCTGTTTTTATTGGAAGACTCATTGAACATATGCAAGAATATGCCTGTTCCTGTGATGTGGAGGAAATTTTCCGCAAGGTTCGATTTTCATTTGAGCAGCCAGATGGTAGAGCGCAGATGCCCACCACTGAAAGAGTGACTTTGACAAGATGTTTCTACCTCTTCCCAGGACATTAA
GSDMD nucleotide sequence (SEQ ID NO: 2):
ATGGGGTCGGCCTTTGAGCGGGTAGTCCGGAGAGTGGTCCAGGAGCTGGACCATGGTGGGGAGTTCATCCCTGTGACCAGCCTGCAGAGCTCCACTGGCTTCCAGCCCTACTGCCTGGTGGTTAGGAAGCCCTCAAGCTCATGGTTCTGGAAACCCCGTTATAAGTGTGTCAACCTGTCTATCAAGGACATCCTGGAGCCGGATGCCGCGGAACCAGACGTGCAGCGTGGCAGGAGCTTCCACTTCTAC GATGCCATGGATGGGCAGATACAGGGCAGCGTGGAGCTGGCAGCCCCAGGACAGGCAAAGATCGCAGGCGGGGCCGCGGTGTCTGACAGCTCCAGCACCTCAATGAATGTGTACTCGCTGAGTGTGGACCCTAACACCTGGCAGACTCTGCTCCATGAGAGGCACCTGCGGCAGCCAGAACACAAAGTCCTGCAGCAGCTGCGCAGCCGCGGGGACAACGTGTACGTGGTGACTGAGGTGCTGCAGACACAGAAGGAGGTGGAAGTCACGCGCACCCACAAGCGGGAGGGCTCGGGCCGGTTTTCCCTGCCCGGAGCCACGTGCTTGCAGGGTGAGGGCCAGGGCCATCTGAGCCAGAAGAAGACGGTCACCATCCCCTCAGGCAGCACCCTCGCATTCCGGGTGGCCCAGCTGGTTATTGACTCTGACTTGGACGTCCTTCTCTTCCCGGATAAGAAGCAGAGGACCTTCCAGCCACCCGCGACAGGCCACAAGCGTTCCACGAGCGAAGGCGCCTGGCCACAGCTGCCCTCTGGCCTCTCCATGATGAGGTGCCTCCACAACTTCCTGACAGATGGGGTCCCTGCGGAGGGGGCGTTCACTGAAGACTTCCAGGGCCTACGGGCAGAGGTGGAGACCATCTCCAAGGAACTGGAGCTTTTGGACAGAGAGCTGTGCCAGCTGCTGCTGGAGGGCCTGGAGGGGGTGCTGCGGGACCAGCTGGCCCTGCGAGCCTTGGAGGAGGCGCTGGAGCAGGGCCAGAGCCTTGGGCCGGTGGAGCCCCTGGACGGTCCAGCAGGTGCTGTCCTGGAGTGCCTGGTGTTGTCCTCCGGAATGCTGGTGCCGGAACTCGCTATCCCTGTTGTCTACCTGCTGGGGGCACTGACCATGCTGAGTGAAACGCAGCACAAGCTGCTGGCGGAGGCGCTGGAGTCGCAGACCCTGTTGGGGCCGCTCGAGCTGGTGGGCAGCCTCTTGGAGCAGAGTGCCCCGTGGCAGGAGCGCAGCACCATGTCCCTGCCCCCCGGGCTCCTGGGGAACAGCTGGGGCGAAGGAGCACCGGCCTGGGTCTTGCTGGACGAGTGTGGCCTAGAGCTGGGGGAGGACACTCCCCACGTGTGCTGGGAGCCGCAGGCCCAGGGCCGCATGTGTGCACTCTACGCCTCCCTGGCACTGCTATCAGGACTGAGCCAGGAGCCCCACTAAPCA3 Nucleotide sequence (SEQ ID NO: 3):
TGTCAACATAGTGTGACGGGAAGGAGCATGATGAGACAGAAGGAAGGTTTAAACTGGGAAATCTGAGAAATGGTATGGTTGTATGTGGGTTGGCATTCTTGCATGATGGGAGTGGCCACCTGCTTTCATATTCTGAAGTCAGAGTGTTCCAGACAGAAGAAATAGCAAGTGCCGAGAAGCTGGCATCAGAAAAACAGAG GGGAGATTTGTGTGG
PEG3 nucleotide sequence (SEQ ID NO: 4):
GAAAGAGAAAGAGAATGGGACGGCATGTGACTGCCTGATGAAGTTGGCGTGCTTGCTCAAAAGTTCTGCGAGATTGACGGCTCTCTGGATTTGAGCCAAGGACACGCCTGGGAAGCCACGGTGACCTCACAAGGCCCGGAATCTCCGCGAGAATTTCAGTGTTGTTTTCCTCTCTCCACCTTTCTCAGGGACTTCCGAAACTCCGCCTCTCCGGTGACGTCAGCATAGCGCTGCGTCAGACTATAAACTCCCGGGTGATCGTGTTGGCGCAGATTGACTCAGTTCGCAGCTTGTGGAAGATTACATGCGAGACCCCGCGCGACTCCGCATCCCTTTGCCGGGACAGCCTTTGCGACAGCCCGTGAGACATCACGTCCCCGAGCCCCACGCCTGAGGGCGACATGAACGCGCTGGCCTTGAGAGCAATCCGGACCCACGACCGCTTTTGGCAAACCGAACCGGAC
EGFP nucleotide sequence (SEQ ID NO: 5):
ATGGTGAGCAAGGGCGAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCGAGCTGGACGGCGACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGCGATGCCACCTACGGCAAGCTGACCCTGAAGTTCATCTGCACCACCGGCAAGCTGCCCGTGCCCTGGCCCACCCTCGTGACCACCCTGACCTACGGCGTGCAGTGCTTCAGCCGCTACCCCGACCACATGAAGCAGCACGACTTCTTCAAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTCTTCAAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCGACACCCTGGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAGGACGGCAACATCCTGGGGCACAAGCTGGAGTACAACTACAACAGCCACAACGTCTATATCATGGCCGACAAGCAGAAGAACGGCATCAAGGTGAACTTCAAGATCCGCCACAACATCGAGGACGGCAGCGTGCAGCTCGCCGACCACTACCAGCAGAACACCCCCATCGGCGACGGCCCCGTGCTGCTGCCCGACAACCACTACCTGAGCACCCAGTCCGCCCTGAGCAAAGACCCCAACGAGAAGCGCGATCACATGGTCCTGCTGGAGTTCGTGACCGCCGCCGGGATCACTCTCGGCATGGACGAGCTGTACAAG
2) AAV virus packaging and concentration
A. About 2X 10 7 293FT cells were plated onto T175 flasks (25 ml DMEM medium) one day in advance and incubated in a 37℃incubator with 5% CO 2 for about 16-24 hours;
B. Preparation of PEI-DNA mixture: preparing a 1.5ml centrifuge tube and a 15ml centrifuge tube, uniformly mixing 10.5ug of AAV virus core plasmid, 10.5ug of capsid plasmid, 30ug of Helper plasmid and 3ml of Opti-MEM in the 50ml centrifuge tube, uniformly mixing 150ul PEI (100 uM concentration) and 3ml of Opti-MEM in the 15ml centrifuge tube, incubating for 5 minutes at room temperature, fully mixing the two, and incubating for 20 minutes at room temperature;
C. Carefully sucking the supernatant of the 293T cells, carefully cleaning the supernatant once by using 3ml of DPBS, sucking the DPBS, adding 18ml of Opti-MEM culture medium into 50ml of a centrifuge tube in the step 2, repeatedly blowing and beating for a plurality of times, and then adding the mixture into a 293FT cell culture bottle;
D. After the dish in the step 3 is placed in a 37 ℃ incubator containing 5% CO 2 for incubation for about 6 hours, the whole supernatant is sucked away, and 25ml of DMEM complete medium is added;
E. collecting the supernatant respectively at 48 hr and 72 hr, mixing, and filtering with 0.45um filter membrane;
F. adding 12.5mL 5 XPEG 8000 precipitated virus, mixing completely, and standing overnight at 4deg.C;
G. Centrifuging at 4000g for 30min at 4 ℃ on the next day;
H. The supernatant was discarded and 500uL PBS was added to resuspend virus;
I. Viral titers were determined using qPCR.
3) AAV infection of target cells
A. The day before infection, C4-2, rwpe-1, FB cells were plated into 6-well plates, and 6X 10 5(C4-2、RWPE-1)、3×105 (FB) per well was added to the 6-well plates;
B. various viruses were added at MOI 1000 on the day of infection;
C. Culturing in a cell culture box.
4) Promoter start detection
Cells expressing green fluorescent protein were observed by confocal microscopy.
3. Experimental results
After AAV viruses carrying different promoters infect different cells, the results show that constitutive promoter SFFV can normally promote the expression of the downstream EGFP gene in three cells (FIG. 1, FIG. 2 and FIG. 3), while tumor specific promoters PCA3 and PEG3 show poor promoter capacity in normal cells (FIG. 1 and FIG. 2), but can effectively promote the expression of EGFP in prostate cancer cells (FIG. 3). At the same time, the results show that CEA specific promoter has a small leakage initiation in the normal prostate cell line RWPE-1 (FIG. 2).
Example 2 in vitro verification of the killing Capacity of specific promoter-initiated killer Gene against tumor cells
1. Experimental materials
① Prostate cancer cell C4-2, normal prostate cell Rwpe-1; ② Live Tox Red dead cell indicator reagent.
2. Experimental method
① C4-2 RWPE-1 cells were seeded into 12-well plates at 4X 10 5/well one day prior to cell transfection;
② The culture medium is discarded from each hole on the day of cell transfection, 750ul of complete culture medium is replaced, and the cells are placed in a cell culture box for culturing for 30min;
③ Diluting 0.75ug plasmid with 38uL serum-free DMEM medium, diluting 2.25uL Ployjet reagent with 38uL serum-free DMEM medium, immediately adding diluted Polyjet into diluted solution, mixing, and standing at room temperature for 15min;
④ Adding the mixture obtained by ③ into a corresponding hole, adding a1 XC.LIVE Tox Red dead cell indicator reagent, uniformly mixing the mixture with the Chinese character '8', adding the mixture into the corresponding hole, immediately placing the mixture into CellCyte to collect fluorescent signals (indicating dead cells), collecting 4 fields of view in each hole, and collecting the fluorescent signals once every 3 hours;
⑤ After the collection is completed, the fluorescence areas of all the holes are separated, and the death condition of the cells is counted.
3. Experimental results
The structure showed that cells transfected with AAV-SFFV-Casp1act-GSDMD and AAV-PCA3-Casp1 act-PEG3-GSDMD showed massive death in C4-2 cells compared to control AAV empty vector (FIG. 4); in the prostate normal cell RWPE-1, only the plasmid AAV-SFFV-Casp1act-GSDMD transfected with the constitutive promoter is dead, and the cells are not obviously dead after AAV empty vector and specific promoter AAV-PCA3-Casp1 act-PEG3-GSDMD are transfected (FIG. 5). The double-specific promoter can improve the safety on the premise of ensuring the effectiveness by controlling the expression of the killer genes.
Example 3 in vitro verification of the efficacy and safety of specific promoter-initiated killer genes for tumor cell killing
1. Experimental materials
① Nude mice; ② Prostate cancer cells C4-2.
2. Experimental method
A. Subcutaneous tumor formation in mice
① Digesting C4-2 tumor cells, and adjusting the cell concentration to 2X 10 7 cells/mL
② Selecting a right armpit needle of a mouse, passing forward, slightly moving the needle slightly before injection, and ensuring the needle is positioned under the skin;
③ After the whole needle is slowly inserted into the skin, 100 mu L of cell suspension is slowly injected, and obvious bulge can be seen;
④ After the injection is finished, slowly rotating to withdraw the needle head, so as to avoid liquid leakage as much as possible;
⑤ Tumor volume measurement is performed after subcutaneous tumor formation, and tumor volume calculation is performed according to the formula:
Volume = long diameter x short diameter x 1/2
Mice were divided into control (Con) and experimental (AAV) groups according to tumor volume;
B. Mice dosing and tumor volume and body weight monitoring
① 100UL of AAV virus (1X 10 10, experimental group) was injected by tail vein, and 100uL of PBS was injected in control group;
② Tumor volume, mouse body weight were measured and recorded every 3 to 4 days;
③ The biochemical detection of the mouse blood is carried out by each blood biochemical analyzer.
C. Biochemical detection of mouse blood and organ coefficients
① After 4 weeks of administration, the eyesockets of all mice are subjected to blood sampling, serum is collected, and the contents of glutamic pyruvic transaminase, glutamic oxaloacetic transaminase, alkaline phosphatase and transglutaminase in the blood of the mice are detected by a blood biochemical analyzer;
② After completion of blood collection, the mice were euthanized, and the main organs (heart, liver, spleen, lung, kidney, brain) were collected and organ coefficients were calculated.
3. Experimental results
The results of the mice experiments showed that AAV-treated groups can significantly inhibit tumor growth (fig. 6), and that the mice had no significant differences in body weight from the control group (fig. 7). The double-promoter start killing gene AAV virus can effectively inhibit the growth of tumor and has no obvious toxic or side effect.
AAV-PCA3-CASP1-PEG3-GSDMD showed no significant difference in the contents of glutamic-pyruvic transaminase, glutamic-oxaloacetic transaminase, alkaline phosphatase and glutaminyl transferase in blood (FIG. 8), and also showed no significant difference in the organ coefficients of heart, liver, spleen, lung, kidney and brain (Table 1). The 5AAV-PCA3-CASP1-PEG3-GSDMD was shown to be not significantly toxic to mice.
TABLE 1 organ coefficients of major organs
The above description of the embodiments is only for the understanding of the method of the present invention and its core ideas. It should be noted that it will be apparent to those skilled in the art that several improvements and modifications can be made to the present invention without departing from the principle of the invention, and these improvements and modifications will fall within the scope of the claims of the invention.

Claims (32)

1. A vector of a bispecific promoter, which is characterized in that the bispecific promoter is two specific promoters of PCA3 and PEG 3;
The carrier comprises: the double-specificity promoters are respectively connected with target genes;
The gene of interest comprises at least one of the following combinations: a combination of Casp1 acts and GSDMD, a combination of GSDMD C and GSDMD N;
the two components in each combination of the target genes can be respectively called a target gene 1 and a target gene 2; the two promoters of the bispecific promoter can be respectively called a specific promoter 1 and a specific promoter 2; the two promoters are two different promoters;
the structure of the vector of the bispecific promoter comprises a specific promoter 1, a target gene 1, a specific promoter 2 and a target gene 2 which are connected in sequence;
the Casp1 act nucleotide sequence is shown as SEQ ID NO. 1.
2. The vector of claim 1, wherein the vector comprises a plasmid, a lentiviral vector, a retroviral vector, an adenoviral vector, an adeno-associated viral vector, a linear polynucleotide.
3. The vector of claim 1, wherein the vector of the bispecific promoter comprises a vector of the structure AAV-PCA3-Casp1 act-PEG 3-GSDMD.
4. The vector of claim 3, wherein the vector of the bispecific promoter comprises the nucleotide sequence GSDMD as set forth in SEQ ID NO. 2.
5. The vector of claim 3, wherein the vector of the bispecific promoter comprises the PCA3 nucleotide sequence as shown in SEQ ID No. 3.
6. The vector of claim 3, wherein the vector of the bispecific promoter comprises the PEG3 nucleotide sequence shown in SEQ ID No. 4.
7. The vector of claim 3, wherein the vector structure of the bispecific promoter comprises the nucleotide sequence of SEQ ID NO. 3, SEQ ID NO. 1, SEQ ID NO. 4, SEQ ID NO. 2, linked in sequence.
8. A method of constructing a vector of a bispecific promoter, wherein the method of constructing is for constructing the vector of any one of claims 1-7, the method of constructing comprising:
Obtaining a gene of interest comprising at least one of the following combinations: a combination of Casp1 acts and GSDMD, a combination of GSDMD C and GSDMD N;
Obtaining bispecific promoters PCA3 and PEG3;
and assembling the combination of the target genes and the bispecific promoter on a vector to obtain the vector.
9. The method of claim 8, wherein the vector comprises a plasmid, a lentiviral vector, a retroviral vector, an adenoviral vector, an adeno-associated viral vector, or a linear polynucleotide.
10. The method of claim 9, wherein the vector is an AAV viral core plasmid.
11. The construction method according to claim 8, characterized in that the construction method comprises any one of the following:
a. amplifying the combination of the target genes, amplifying a bispecific promoter, connecting the target genes with the specific promoter, and cloning the connector onto the vector to obtain the vector comprising the bispecific promoter and the target genes;
b. amplifying the target gene and the specific promoter, and then directly cloning the amplified target gene and the specific promoter onto the vector to obtain the vector comprising the bispecific promoter and the target gene.
12. A host cell comprising the vector of any one of claims 1-7.
13. The host cell of claim 12, wherein the host cell comprises a prokaryotic cell, a eukaryotic cell.
14. The host cell of claim 13, wherein the prokaryotic cell comprises bacteria, actinomycetes, cyanobacteria, mycoplasma, chlamydia, rickettsia.
15. The host cell of claim 13, wherein the eukaryotic cell comprises a mammalian cell, an insect cell, a plant cell, a yeast cell.
16. The host cell of claim 15, wherein the mammalian cell comprises a fibroblast cell, a 293T cell.
17. A virus comprising the vector of any one of claims 1-7.
18. The virus of claim 17, wherein the virus comprises a lentivirus, retrovirus, adenovirus, adeno-associated virus, herpes virus, poxvirus, papovavirus, baculovirus, papillomavirus.
19. The virus of claim 18, wherein the virus is an adeno-associated virus.
20. A method of producing a virus according to any one of claims 17 to 19, comprising the steps of:
transfecting the vector of any one of claims 1-7 into a host cell; and
Amplifying and expressing the transfected host cells to obtain the virus.
21. The method of claim 20, wherein the virus comprises a lentivirus, retrovirus, adenovirus, adeno-associated virus, herpes virus, poxvirus, papovavirus, baculovirus, papilloma virus.
22. The method of claim 21, wherein the virus is an adeno-associated virus.
23. The method of claim 22, wherein the host cell comprises a prokaryotic cell, a eukaryotic cell.
24. The method of claim 23, wherein the prokaryotic cell comprises a bacterium, actinomycete, cyanobacteria, mycoplasma, chlamydia, rickettsia.
25. The method of claim 23, wherein the eukaryotic cell comprises a mammalian cell, an insect cell, a plant cell, a yeast cell.
26. The method of claim 25, wherein the mammalian cells comprise fibroblasts, 293T cells.
27. A pharmaceutical composition comprising the vector of any one of claims 1-7, the host cell of any one of claims 12-16, the virus of any one of claims 17-19.
28. The pharmaceutical composition of claim 27, further comprising a pharmaceutically acceptable auxiliary ingredient.
29. Use of the vector of any one of claims 1-7, the construction method of any one of claims 8-11, or the host cell of any one of claims 12-16 in the preparation of a virus or plasmid.
30. The use according to claim 29, wherein the virus comprises lentivirus, retrovirus, adenovirus, adeno-associated virus, herpes virus, poxvirus, papovavirus, baculovirus, papillomavirus.
31. The use of claim 30, wherein the virus is an adeno-associated virus.
32. Use of the vector of any one of claims 1-7, the construction method of any one of claims 8-11, the host cell of any one of claims 12-16, the virus of any one of claims 17-19, or the preparation method of any one of claims 20-26 in the manufacture of a medicament for the treatment of prostate cancer, liver cancer.
CN202310794540.0A 2023-06-30 2023-06-30 Vector carrying bispecific promoter, construction method and application thereof Active CN116814688B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310794540.0A CN116814688B (en) 2023-06-30 2023-06-30 Vector carrying bispecific promoter, construction method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310794540.0A CN116814688B (en) 2023-06-30 2023-06-30 Vector carrying bispecific promoter, construction method and application thereof

Publications (2)

Publication Number Publication Date
CN116814688A CN116814688A (en) 2023-09-29
CN116814688B true CN116814688B (en) 2024-06-07

Family

ID=88121797

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310794540.0A Active CN116814688B (en) 2023-06-30 2023-06-30 Vector carrying bispecific promoter, construction method and application thereof

Country Status (1)

Country Link
CN (1) CN116814688B (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101899420A (en) * 2010-01-16 2010-12-01 韩从辉 Method for preparing dual-regulated oncolytic adenovirus for expressed superantigen gene of targeted prostate tumor
CN102191245A (en) * 2010-03-15 2011-09-21 浙江东方基因生物制品有限公司 Method and reagent for detecting tumor cell in circulatory blood by applying tumor peculiar promoter to express reporter gene
CN102985094A (en) * 2010-06-30 2013-03-20 东源生物医药科技(上海)有限公司 Recombinant tumor vaccine and method of producing such vaccine
RU2013114741A (en) * 2013-04-02 2014-10-20 Общество С Ограниченной Ответственностью "Ива" UNIVERSAL CANCER-SPECIFIC PROMOTORS FOR EXPRESSION OF TRANSGENES IN TUMOR CELLS
CN111485000A (en) * 2020-05-06 2020-08-04 浙江大学 Method for establishing cell apoptosis model mediated by porcine GSDMD protein
CN112592900A (en) * 2020-12-15 2021-04-02 华中农业大学 Packaging method for constructing oncolytic adeno-associated virus oAAVs for expressing pyroptosis protein and application of packaging method
CN116218844A (en) * 2022-09-07 2023-06-06 吉满生物科技(上海)有限公司 Bidirectional promoter, over-expression vector thereof, lentiviral expression plasmid and application

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012000188A1 (en) * 2010-06-30 2012-01-05 Tot Shanghai Rd Center Co., Ltd. Recombinant tumor vaccine and method of producing such

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101899420A (en) * 2010-01-16 2010-12-01 韩从辉 Method for preparing dual-regulated oncolytic adenovirus for expressed superantigen gene of targeted prostate tumor
CN102191245A (en) * 2010-03-15 2011-09-21 浙江东方基因生物制品有限公司 Method and reagent for detecting tumor cell in circulatory blood by applying tumor peculiar promoter to express reporter gene
CN102985094A (en) * 2010-06-30 2013-03-20 东源生物医药科技(上海)有限公司 Recombinant tumor vaccine and method of producing such vaccine
RU2013114741A (en) * 2013-04-02 2014-10-20 Общество С Ограниченной Ответственностью "Ива" UNIVERSAL CANCER-SPECIFIC PROMOTORS FOR EXPRESSION OF TRANSGENES IN TUMOR CELLS
CN111485000A (en) * 2020-05-06 2020-08-04 浙江大学 Method for establishing cell apoptosis model mediated by porcine GSDMD protein
CN112592900A (en) * 2020-12-15 2021-04-02 华中农业大学 Packaging method for constructing oncolytic adeno-associated virus oAAVs for expressing pyroptosis protein and application of packaging method
CN116218844A (en) * 2022-09-07 2023-06-06 吉满生物科技(上海)有限公司 Bidirectional promoter, over-expression vector thereof, lentiviral expression plasmid and application

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Isolation and Characterization of the Promoter of the Human Prostate Cancer-specific DD3 Gene;Gerald W. Verhaegh et al;THE JOURNAL OF BIOLOGICAL CHEMISTRY;第275卷(第48期);摘要,第37497页倒数第3段 *
Targeting gene expression selectively in cancer cells by using the progression-elevated gene-3 promoter;Zhao-Zhong Su et al;BIOLOGICAL SCIENCES;第102卷(第04期);摘要 *
虞先浚主编.胰腺癌的诊断和治疗 新技术、新理念、新策略.复旦大学出版社,2020,第136页. *

Also Published As

Publication number Publication date
CN116814688A (en) 2023-09-29

Similar Documents

Publication Publication Date Title
JP4860886B2 (en) Double-stranded parvovirus vector
Xiao et al. Gene transfer by adeno-associated virus vectors into the central nervous system
JP2022506515A (en) Vector system for expressing regulatory RNA
CN107043774A (en) A kind of chimeric strong promoter and application thereof
CN112226461B (en) CD4 positive cell specific gene transfer vector and application thereof
US5576201A (en) Retroviral vector particles for transducing non-proliferating cells
Ma et al. Non-classical nuclear localization signal peptides for high efficiency lipofection of primary neurons and neuronal cell lines
JP2021502822A (en) Non-human papillomavirus for gene delivery in vitro and in vivo
CN116814688B (en) Vector carrying bispecific promoter, construction method and application thereof
US5580766A (en) Retroviral vector particles for transducing non-proliferating cells
CN116949041A (en) Artificially designed mRNA UTR nucleotide sequence and its use
CN114395017A (en) Preparation method and application of SARS-CoV-2 virus-like particle
CN114908089B (en) Construction method and application of 3' UTR
Al Abbar et al. Production of lentiviral vector with polycistronic transcripts for reprogramming of mouse fibroblast cells
WO2024027632A1 (en) Novel plasmid backbone to reduce dna impurities in raav preparation
CN116732039B (en) Sequence combination for promoting RNA sequence to be cyclized and translated directly in cells and application thereof
CN116970650B (en) Envelope protein combination, targeting virus vector containing envelope protein combination and preparation method of targeting virus vector
WO2024083013A1 (en) Human mitochondrial dynamin like gtpase and use thereof
JP4766297B2 (en) A vector comprising a non-virus-derived enhancer, cPPT, and CTS
CN117904201A (en) Viral gene therapy vector for eliminating hepatitis B virus and application thereof
KR101242114B1 (en) The method for screening transgenic animals generated by ultrasound image-guided gene delivery technique using in vivo imaging of reporter gene expression
CN117964721A (en) Virus-like particle and construction method and application thereof
CN117867023A (en) Visual retinoic acid level detection method based on adeno-associated virus vector
WO2024015881A2 (en) Compositions, systems, and methods for targeted transcriptional activation
EP1626091B1 (en) Duplexed parvovirus vectors

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