CN114010666A - Application of oncolytic virus, PARP inhibitor and PD-1 antibody in preparation of antitumor drugs - Google Patents

Application of oncolytic virus, PARP inhibitor and PD-1 antibody in preparation of antitumor drugs Download PDF

Info

Publication number
CN114010666A
CN114010666A CN202111233544.9A CN202111233544A CN114010666A CN 114010666 A CN114010666 A CN 114010666A CN 202111233544 A CN202111233544 A CN 202111233544A CN 114010666 A CN114010666 A CN 114010666A
Authority
CN
China
Prior art keywords
oncolytic virus
ohsv
olaparib
antibody
cells
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.)
Granted
Application number
CN202111233544.9A
Other languages
Chinese (zh)
Other versions
CN114010666B (en
Inventor
蔡宇伽
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.)
Shanghai Jiaotong University
Original Assignee
Shanghai Jiaotong University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Jiaotong University filed Critical Shanghai Jiaotong University
Priority to CN202111233544.9A priority Critical patent/CN114010666B/en
Publication of CN114010666A publication Critical patent/CN114010666A/en
Application granted granted Critical
Publication of CN114010666B publication Critical patent/CN114010666B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/763Herpes virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • 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
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16621Viruses as such, e.g. new isolates, mutants or their genomic sequences

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Mycology (AREA)
  • Toxicology (AREA)
  • Endocrinology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention provides an application of an oncolytic virus, a PARP inhibitor and a PD-1 antibody in preparing an anti-tumor medicament, wherein miRT124 and hGMCSF are inserted into a type I herpes simplex virus to obtain an oncolytic virus oHSV, so that the safety of the oncolytic virus is improved, and the immune response of an organism is enhanced; through the combined use of the oncolytic virus oHSV and the Olaparib, the PD-1 expression quantity of the CD4+ T cell and the CD8+ T cell can be increased by combining the oncolytic virus oHSV and the Olaparib, so that an immune checkpoint inhibitor, namely a PD-1 antibody is introduced, the curative effect of a treatment scheme in breast cancer is further improved, the size of an in-situ tumor of the breast cancer of a mouse is further inhibited by combining the oHSV, the Olaparib and the PD-1 antibody, on the basis of the combined use of the oHSV, the Olaparib and the PD-1 antibody, the number of lung metastasis foci is further reduced, and the survival period of the mouse is remarkably prolonged.

Description

Application of oncolytic virus, PARP inhibitor and PD-1 antibody in preparation of antitumor drugs
Technical Field
The invention belongs to the field of biological medicine, and particularly relates to application of oncolytic virus, PARP (para-amyloid peptide) inhibitor and PD-1 antibody in preparation of antitumor drugs
Background
Oncolytic viruses with unique oncolytic mechanisms offer a new option for the treatment of tumors, especially for some tumors that do not respond to conventional treatment, wherein the replication of the oncolytic virus in tumor cells can kill the tumor cells by lysis, and furthermore, the oncolytic virus and tumor-associated antigens released by lysis can induce immune response to further kill the tumor.
Due to tumor heterogeneity and complexity of tumor microenvironment and the influence of the immune system of the body, it is difficult to achieve good therapeutic effect in some tumors by using oncolytic virus alone, for example, the first FDA approved oncolytic virus T-VEC cannot improve the overall survival rate of melanoma patients, and only 16.3% of patients receiving treatment have tumor volume that continuously shrinks within 6 months. This greatly affects the indications for oncolytic viruses.
The oncolytic virus can be combined with other medicines to realize advantage complementation, thereby improving the tumor treatment effect. For example, Amgen company and Michelia nobilis cooperate to carry out clinical results of the combination treatment of the T-VEC and the CTLA-4 antibody on the advanced melanoma stage I, which shows that the overall response rate of the combination treatment is remarkably higher than that of single drug treatment. In addition, the literature reports that histone deacetylase inhibitors, anti-tumor angiogenesis inhibitors and the like can synergistically enhance the anti-tumor treatment effect of herpes simplex virus oncolytic viruses.
Chinese patent 201711478207.X discloses the combined use of M1 oncolytic virus of alphavirus and PARP inhibitor, and the function of killing tumor cells is improved at a cellular level. The patent discloses that oncolytic virus and PARP inhibitors kill tumor cells in vitro, in-vivo experiments are not carried out, namely, the tumor cells are verified in an animal model which can best reflect the actual clinical situation, the using time of the using number and the using time of the using number of the oncolytic virus and the using time of the using number and the using time of the PARP inhibitors are not researched, in addition, the change of the body immunity situation is not researched after the oncolytic virus and the PARP inhibitors are used, and the problems are solved in the scheme. And the patent document does not mention the technical scheme of the combined use with the PD-1 antibody.
Disclosure of Invention
An object of the present invention is to provide an anti-tumor pharmaceutical composition, which comprises an oncolytic virus selected from the group consisting of herpes simplex virus type I and a PARP inhibitor.
As one embodiment of the present invention, the PARP inhibitor is Olaparib, which has the formula C24H23FN4O3(ii) a The structural formula of Olaparib is shown as formula I:
Figure BDA0003316740380000021
the PARP inhibitor adopted by the invention is Olaparib which can simultaneously inhibit the activities of PARP1 and PARP2, and the Olaparib is an approved medicament for clinical treatment, and the safety and reliability of the invention are verified. The composition further comprises a PD-1 antibody. The PD-1 antibody is a monoclonal or polyclonal antibody directed against human PD-1 and murine PD-1.
The oncolytic virus is prepared by inserting miRT124 and hGMCSF into type I herpes simplex virus. The oncolytic virus is prepared by a method comprising the following steps:
s1, amplifying sequences at two ends of HSV-1 and ICP34.5 genes respectively;
s2, connecting sequences at two ends of hGM-CSF, CMV-GFP, 4xmIRT124 and ICP34.5 and plasmid pCMV-GFP by using a homologous recombination method to form a complete donor plasmid which is marked as pULL34.5-hGM-CSF-lox 2272-CMV-eGFP-lox2272-4xmIRT 124;
s3, designing sgRNA aiming at an ICP34.5 region, connecting the sgRNA to a Lenti-CRISPR-V2 vector in an enzyme digestion connection mode, and marking as Lenti-CRISPR-ICP 34.5;
s4, co-transfecting HEK293T cells with donor plasmid pULL34.5-hGM-CSF-lox 2272-CMV-eGFP-lox2272-4xmIRT124 and Lenti-CRISPR-ICP34.5 plasmid targeting ICP34.5, and collecting culture supernatant after the cells are completely diseased; adding the supernatant into Vero cells, observing fluorescent plaques after several hours, and picking the fluorescent plaques; obtaining HSV-1 inserted with hGMCSF, CMV-GFP and 4xmIRT124, and marking as oHSV-GFP;
s5, amplifying a Cre fragment by using a plasmid with the Cre fragment as a template, and connecting the Cre fragment to a pCCL-PGK-eGFP vector by using an enzyme digestion connection method, wherein the pCCL-PGK-eGFP vector is marked as pCCL-PGK-eGFP;
s6, transfecting the pCCL-PGK-Cre plasmid prepared in the step S5 to a HEK293T cell, adding the oHSV-GFP prepared in the step S4 in a dosage of 1.0 MOI, and collecting a culture supernatant after the cell is completely diseased; adding the supernatant into Vero cells, observing non-fluorescent plaques, and selecting the non-fluorescent plaques; HSV-1 with hGMCSF and 4xmIRT124 inserted was obtained and designated oHSV.
Preferably, step S4 specifically includes the following steps:
co-transfecting HEK293T cells with a donor plasmid pULL34.5-hGM-CSF-lox 2272-CMV-eGFP-lox2272-4xmIRT124 and a Lenti-CRISPR-ICP34.5 plasmid targeting ICP34.5, adding HSV-1 at the dose of MOI (1.0) after 24 hours, and collecting culture supernatant after the cells are completely diseased; adding the supernatant into Vero cells by a 10-fold dilution method, observing fluorescent plaques after 48 hours, and selecting the fluorescent plaques; the HSV-1 inserted with hGMCSF, CMV-GFP and 4xmIRT124 is obtained by multiple rounds of purification by the method and is marked as oHSV-GFP.
Preferably, step S5 specifically includes the following steps:
transfecting HEK293T cells with the pCCL-PGK-Cre plasmid prepared in the step S5, adding the oHSV-GFP prepared in the step S4 in a dosage of MOI (1.0) after 24 hours, and collecting culture supernatant after the cells are completely diseased; adding the supernatant into Vero cells, observing no fluorescent plaque after 48 hours, and selecting the non-fluorescent plaque; after multiple rounds of purification by the method, HSV-1 inserted with hGMCSF and 4xmIRT124 is obtained and is marked as oHSV.
Further, the dose of the oncolytic virus is 10 × 107-100×107The dosage of PFU and the Olaparib is 100-1000 mg/kg.
Further, the dosage of the PD-1 antibody is 450 to 750 mu g.
The application of the anti-tumor medicine composition in preparing anti-tumor medicines or preparations also belongs to the protection scope of the invention.
The tumor is liver cancer, colorectal cancer, bladder cancer, breast cancer, cervical cancer, prostatic cancer, glioma, melanoma, pancreatic cancer, nasopharyngeal carcinoma, lung cancer or gastric cancer.
It is another object of the present invention to provide an anti-tumor drug system comprising an oncolytic virus selected from the group consisting of herpes simplex virus type I and a PARP inhibitor.
The PARP inhibitor is a substance for inhibiting PARP activity, a substance for degrading PARP, a gene tool for reducing PARP level, or any combination thereof.
As one embodiment of the present invention, the PARP inhibitor is Olaparib, which has the formula C24H23FN4O3(ii) a The structural formula of Olaparib is shown as formula I:
Figure BDA0003316740380000031
the preparation method of the oncolytic virus comprises the following steps:
s1, amplifying sequences at two ends of HSV-1 and ICP34.5 genes respectively;
s2, connecting sequences at two ends of hGM-CSF, CMV-GFP, 4xmIRT124 and ICP34.5 and plasmid pCMV-GFP by using a homologous recombination method to form a complete donor plasmid which is marked as pULL34.5-hGM-CSF-lox 2272-CMV-eGFP-lox2272-4xmIRT 124;
s3, designing sgRNA aiming at an ICP34.5 region, connecting the sgRNA to a Lenti-CRISPR-V2 vector in an enzyme digestion connection mode, and marking as Lenti-CRISPR-ICP 34.5;
s4, co-transfecting a donor plasmid pULL34.5-hGM-CSF-lox 2272-CMV-eGFP-lox2272-4xmIRT124 and a Lenti-CRISPR-ICP34.5 plasmid targeting ICP34.5 into HEK293T cells, adding HSV-1 at the dose of MOI (1.0) after 24 hours, and collecting culture supernatant after the cells are completely diseased; adding the supernatant into Vero cells by a 10-fold dilution method, observing fluorescent plaques after 48 hours, and selecting the fluorescent plaques; obtaining HSV-1 inserted with hGMCSF, CMV-GFP and 4xmIRT124 through multi-round purification by the method, and marking as oHSV-GFP;
s5, amplifying a Cre fragment by using a plasmid with the Cre fragment as a template, and connecting the Cre fragment to a pCCL-PGK-eGFP vector by using an enzyme digestion connection method, wherein the pCCL-PGK-eGFP vector is marked as pCCL-PGK-eGFP;
s6, transfecting the pCCL-PGK-Cre plasmid prepared in the step S5 into HEK293T cells, adding the oHSV-GFP prepared in the step S4 in a dosage of MOI (equal to 1.0) after 24 hours, and collecting culture supernatant after the cells are completely diseased; adding the supernatant into Vero cells, observing no fluorescent plaque after 48 hours, and selecting the non-fluorescent plaque; after multiple rounds of purification by the method, HSV-1 inserted with hGMCSF and 4xmIRT124 is obtained and is marked as oHSV.
The total dose of the oncolytic virus is 10 multiplied by 107-100×107The total dosage of PFU and the Olaparib is 100-1000 mg/kg.
Preferably, the total dose of the oncolytic virus is 10 x107-100×107The total dosage of PFU and the Olaparib is 100-1000 mg/kg.
The anti-tumor drug system also comprises a PD-1 antibody, and the dosage of the PD-1 antibody is 450-750 mu g.
Preferably, the total amount of the PD-1 antibody is 450. mu.g and 750. mu.g, wherein the PD-1 antibody is a monoclonal or polyclonal antibody directed against human PD-1 and murine PD-1.
The application of the anti-tumor medicine system in the preparation of anti-tumor medicines also belongs to the protection scope of the invention.
Compared with the prior art, the invention has the beneficial effects that:
1) the invention provides an oncolytic virus inserted with miRT124 and hGMCSF, which improves the safety of the oncolytic virus and enhances the immune response of an organism;
2) the inventor proves that PARP inhibitors such as Olaparib can enhance the replication of the oncolytic virus through a series of experiments, thereby enhancing the curative effect of the oncolytic virus;
3) the inventor finds that the expression quantity of PD-1 of CD4+ T cells and CD8+ T cells can be increased when the oncolytic virus oHSV and Olaparib are used in combination, so that an immune checkpoint inhibitor, namely a PD-1 antibody is introduced, the curative effect of a treatment scheme in breast cancer is further improved, and the combination of oHSV and Olaparib reduces the number of lung metastases of the breast cancer of a mouse;
4) the oHSV, the Olaparib and the PD-1 antibody are used in a combined manner, the size of the breast cancer in situ of the mouse is inhibited, and on the basis of the combined use of the oHSV and the Olaparib, the number of lung metastasis foci is further reduced, so that the survival period of the mouse is remarkably prolonged;
5) the OHSV, the Olaparib and the PD-1 antibody are used together to activate the immune system of the body, so that the number of CD45 positive cells is increased; the up-regulation of PD-1 expression levels without altering the number of CD4+ and CD8+ T cells is also responsible for the enhanced therapeutic efficacy of the combination with immune checkpoint inhibitors.
Drawings
FIG. 1 is a graph showing the statistical analysis of the number of lung metastases of breast cancer in mice when oncolytic virus and Olaparib are used in combination;
FIG. 2 is a graph showing the analysis of the size results of mouse breast cancer in situ tumors when oncolytic virus, Olaparib and PD-1 antibody are used in combination;
FIG. 3 is a graph showing the quantitative results of lung metastases of mouse breast cancer when oncolytic virus, Olaparib and PD-1 antibody are used in combination;
FIG. 4: graphs for the number of mouse CD45+ cells when oncolytic virus, Olaparib, and PD-1 antibodies were used in combination;
FIG. 5: results of analysis of expression levels of mouse CD4+ T cell PD-1 when oncolytic virus, Olaparib and PD-1 antibody were combined;
FIG. 6: results of analysis of expression levels of mouse CD8+ T cell PD-1 when oncolytic virus, Olaparib and PD-1 antibody were combined;
FIG. 7: results analysis of the significantly prolonged survival of mice using oncolytic virus, Olaparib in combination with PD-1 antibody.
Detailed Description
The embodiments of the present invention are described below with reference to specific embodiments, and other advantages and effects of the present invention will be easily understood by those skilled in the art from the disclosure of the present specification. The invention is capable of other and different embodiments and of being practiced or of being carried out in various ways, and its several details are capable of modification in various respects, all without departing from the spirit and scope of the present invention.
In the following examples, PARP inhibitors refer to the compound Olaparib (Chinese name: Olaparib) and related compounds capable of inhibiting PARP binding activity, catalytic activity; the molecular formula of Olaparib is C24H23FN4O3(ii) a The structural formula is as follows:
Figure BDA0003316740380000061
PD-1 antibodies are monoclonal or polyclonal antibodies directed against human PD-1 and murine PD-1.
Herpes simplex virus type I (HSV-1) is from the university of Ourhees, Denmark.
The technical solution and the technical effect of the present invention will be further described in detail with reference to the following examples.
Example 1
Preparation of oncolytic Virus oHSV
The oncolytic virus oHSV is obtained by modifying a herpes simplex virus I (HSV-1), and the preparation method comprises the following specific steps:
1) amplifying sequences at two ends of HSV-1 and ICP34.5 genes respectively by using a PCR method; among them, HSV-1 (GenBank: JQ 780693.1); the upstream base sequence of ICP34.5 is shown as SEQ ID NO.1 in the sequence table; the downstream base of ICP34.5 is shown as SEQ ID NO.2 in the sequence table;
2) then, the sequences at the two ends of hGM-CSF, CMV-GFP, 4xmIRT124 and ICP34.5 and the plasmid pCMV-GFP are connected by using a homologous recombination method to form a complete donor plasmid which is marked as pULL34.5-hGM-CSF-lox 2272-CMV-eGFP-lox2272-4xmIRT 124; wherein the sequence of hGM-CSF is shown as SEQ ID NO.3 in the sequence table; the sequence of the CMV-GFP is shown as SEQ ID NO.4 in the sequence table;
3) designing sgRNA (shown as SEQ ID NO.6 in a sequence table) aiming at an ICP34.5 region (shown as SEQ ID NO.5 in the sequence table), carrying out enzyme digestion through Esp3I, and connecting T4DNA ligase to a Lenti-CRISPR-V2 vector, wherein the vector is marked as Lenti-CRISPR-ICP 34.5;
4) co-transfecting HEK293T cells with a donor plasmid pULL34.5-hGM-CSF-lox 2272-CMV-eGFP-lox2272-4xmIRT124 and a Lenti-CRISPR-ICP34.5 plasmid targeting ICP34.5, adding HSV-1 at the dose of MOI (1.0) after 24 hours, and collecting culture supernatant after the cells are completely diseased; adding the supernatant into Vero cells by a 10-fold dilution method, observing fluorescent plaques after 48 hours, and selecting the fluorescent plaques; obtaining HSV-1 inserted with hGMCSF, CMV-GFP and 4xmIRT124 through multi-round purification by the method, and marking as oHSV-GFP;
5) using a plasmid with a Cre fragment as a template, amplifying the Cre fragment, and connecting the Cre fragment to a pCCL-PGK-eGFP vector by a BamHI and XhoI double enzyme digestion and T4DNA ligase connection method, and marking the pCCL-PGK-eGFP vector as pCCL-PGK-eGFP;
6) transfecting HEK293T cells with pCCL-PGK-Cre plasmids, adding oHSV-GFP in a dose of MOI (equal to 1.0) after 24 hours, and collecting culture supernatant after the cells are completely diseased; adding the supernatant into Vero cells, observing no fluorescent plaque after 48 hours, and selecting the non-fluorescent plaque; after multiple rounds of purification by the method, HSV-1 inserted with hGMCSF and 4xmIRT124 is obtained and is marked as oHSV.
Example 2
Test experiment for combined use of oncolytic virus oHSV and Olaparib
(1) Experimental materials:
6-week-old BALB/C mice, a mouse breast cancer cell line 4T1, RPMI1640 medium, oncolytic virus oHSV, Olaparib.
(2) The experimental method comprises the following steps:
A. will be 5X105The 4T1 cells were injected in situ into the right fat pad of the mouse;
B. when the tumor size reaches 100mm3At the time of treatment, therapy was initiated, with oncolytic virus injected every other day in situ at a dose of 5X107PFU, 5 total injections; the Olaparib is injected intraperitoneally every day, the using amount of each time is 50mg/kg, and the total injection is 10 times;
C. on the second day after the fifth oncolytic virus injection, the in situ tumor is removed;
D. on the seventh day after the removal of the orthotopic tumor, the mice were sacrificed, the lungs were taken out, and the number of lung metastases was observed and recorded under a microscope;
(3) the experimental results are as follows:
the results in FIG. 1 show that OHSV combined with Olaparib can inhibit the number of metastasis in the lung of breast cancer in mice.
Example 3
Oncolytic viruses oHSV, Olaparib were used in combination with PD-1 antibodies in the test experiment.
(1) Experimental materials:
6-week-old BALB/C mice, a mouse breast cancer cell line 4T1, an RPMI1640 medium, an oncolytic virus, Olaparib, DNase, collagenase, hyaluronidase, an anti-mouse PD-1 antibody, an anti-mouse CD45 antibody, an anti-mouse CD4 antibody, and an anti-mouse CD8 antibody.
(2) The experimental method comprises the following steps:
E. will be 5X105The 4T1 cells were injected in situ into the right fat pad of the mouse;
F. when the tumor size reaches 100mm3At the time of treatment, therapy was initiated, with oncolytic virus injected every other day in situ at a dose of 5X107PFU, 5 injections in total, total oncolytic virus dose 25X 107PFU,; injecting the Olaparib intraperitoneally every day, wherein the dosage of each injection is 50mg/kg, the total injection is 10 times, and the total dosage of the Olaparib is 10 mg; the anti-mouse PD-1 antibody is injected into the abdominal cavity of the second day after the third injection of the oncolytic virus and the removal of the in-situ tumor for the fifth time respectively, the using amount is 200 mug each time, and the total injection is 3 times;
G. on the second day after the fifth injection of the oncolytic virus, removing the in situ tumor, digesting the in situ tumor by using DNase, collagenase and hyaluronidase, then incubating by using anti-mouse PD-1 antibody, anti-mouse CD45 antibody, anti-mouse CD4 antibody and anti-mouse CD8 antibody, and finally detecting related indexes by using a flow cytometer;
H. the short-term experiment was conducted as follows: on the seventh day after the removal of the orthotopic tumor, the mice were sacrificed, the lungs were taken out, and the number of lung metastases was observed and recorded under a microscope;
survival experiments were conducted as follows: after removal of the orthotopic tumor, the mice were not treated at all, and only the survival time of the mice was recorded and a survival curve was plotted.
(3) Results of the experiment
As shown in fig. 2, the oncolytic virus, Olaparib, and PD-1 antibody used in combination significantly inhibited the size of the mouse breast cancer orthotopic tumor;
as shown in fig. 3, the use of oncolytic virus, Olaparib in combination with PD-1 antibody significantly reduced the number of lung metastases of breast cancer in mice;
as shown in fig. 4, the use of oncolytic virus, Olaparib in combination with PD-1 antibody significantly increased the number of mouse CD45+ cells;
as shown in FIG. 5, the use of oncolytic virus, Olaparib in combination with PD-1 antibody significantly increased the expression level of mouse CD4+ T cell PD-1;
as shown in FIG. 6, the use of oncolytic virus, Olaparib in combination with PD-1 antibody significantly increased the expression level of PD-1 from mouse CD8+ T cells;
as shown in fig. 7, the use of oncolytic virus, Olaparib in combination with PD-1 antibody significantly prolonged the survival of mice.
In conclusion, the inventor finds that the oncolytic virus oHSV and Olaparib can increase the expression level of PD-1 of CD4+ T cells and CD8+ T cells when used together, so that an immune checkpoint inhibitor, namely a PD-1 antibody is introduced, the curative effect of a treatment scheme in breast cancer is further improved, and the oHSV and Olaparib are used together to reduce the number of lung metastasis foci of the breast cancer in mice;
the oHSV, the Olaparib and the PD-1 antibody are used together, the size of the breast cancer in situ of the mouse and the number of lung metastasis foci are inhibited, and the life cycle of the mouse is obviously prolonged; simultaneously activating the immune system of the organism to increase the number of CD45 positive cells; and up-regulates the expression level of PD-1, play a role in enhancing the curative effect of the immune checkpoint inhibitor; finally, the survival time of the tumor-bearing mice is prolonged. Wherein the using dose of the oHSV is 5x107 PFU/mouse, and the using dose of the Olaparib is 50 mg/kg; the PD-1 antibody was used at a dose of 200. mu.g/mouse.
The foregoing description of specific embodiments of the present invention has been presented. It is to be understood that the present invention is not limited to the specific embodiments described above, and that various changes or modifications may be made by one skilled in the art within the scope of the appended claims without departing from the spirit of the invention. The embodiments and features of the embodiments of the present application may be combined with each other arbitrarily without conflict.
Sequence listing
<110> Shanghai university of transportation
<120> a method for inhibiting tumor metastasis by using oncolytic virus in combination with PARP inhibitor
<130> KAG45783
<160> 6
<170> SIPOSequenceListing 1.0
<210> 1
<211> 777
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 1
tctgcacgca catgcttgcc tgtcaaactc taccaccccg gcacgctctc tgtctccatg 60
gcccgccgcc gccatcgcgg cccccgccgc ccccggccgc ccgggcccac gggcgcggtc 120
ccaaccgcac agtcccaggt aacctccacg cccaactcgg aacccgtggt caggagcgcg 180
cccgcggccg ccccgccgcc gccccccgcc agtgggcccc cgccttcttg ttcgctgctg 240
ctgcgccagt ggctccacgt tcccgagtcc gcgtccgacg acgacgacga cgactggccg 300
gacagccccc cgcccgagcc ggcgccagag gcccggccca ccgccgccgc cccccgcccc 360
cggtccccac cgcccggcgc gggcccgggg ggcggggcta acccctccca ccccccctca 420
cgccccttcc gccttccgcc gcgcctcgcc ctccgcctgc gcgtcaccgc agagcacctg 480
gcgcgcctgc gcctgcgacg cgcgggcggg gagggggcgc cgaagccccc cgcgaccccc 540
gcgacccccg cgacccccac gcgggtgcgc ttctcgcccc acgtccgggt gcgccacctg 600
gtggtctggg cctcggccgc ccgcctggcg cgccgcggct cgtgggcccg cgagcgggcc 660
gaccgggctc ggttccggcg ccgggtggcg gaggccgagg cggtcatcgg gccgtgcctg 720
gggcccgagg cccgtgcccg ggccctggcc cgcggagccg gcccggcgaa ctcagtc 777
<210> 2
<211> 716
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 2
cgttacaccc gaggcggcct gggtcttccg cggagctccc gggagctccg caccaagccg 60
ctctccggag agacgatggc aggagccgcg catatatacg cttggagccg gcccgccccc 120
gaggcgggcc cgccctcgga gggcgggact ggccaatcgg cggccgccag cgcggcgggg 180
cccggccaac cagcgtccgc cgagtcgtcg gggcccggcc cactgggcgg taactcccgc 240
ccagtgggcc gggccgccca cttcccggta tggtaattaa aaacttgcag aggccttgtt 300
ccgcttcccg gtatggtaat tagaaactca ttaatgggcg gccccggccg cccttcccgc 360
ttccggcaat tcccgcggcc cttaatgggc aaccccggta ttccccgcct cccgcgccgc 420
gcgtaaccac tcccctgggg ttccgggtta tgttaattgc ttttttggcg gaacacacgg 480
cccctcgcgc attggcccgc gggtcgctca atgaacccgc attggtcccc tggggttccg 540
ggtatggtaa tgagtttctt cgggaaggcg ggaagccccg gggcaccgac gcaggccaag 600
cccctgttgc gtcggcggga ggggcatgct aatggggttc tttgggggac accgggttgg 660
tcccccaaat cgggggccgg gccgtgcatg ctaatgatat tctttggggg cgccgg 716
<210> 3
<211> 429
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 3
tggctgcaga gcctgctgct cttgggcact gtggcctgca gcatctctgc acccgcccgc 60
tcgcccagcc ccagcacgca gccctgggag catgtgaatg ccatccagga ggcccggcgt 120
ctcctgaacc tgagtagaga cactgctgct gagatgaatg aaacagtaga agtcatctca 180
gaaatgtttg acctccagga gccgacctgc ctacagaccc gcctggagct gtacaagcag 240
ggcctgcggg gcagcctcac caagctcaag ggccccttga ccatgatggc cagccactac 300
aagcagcact gccctccaac cccggaaact tcctgtgcaa cccagattat cacctttgaa 360
agtttcaaag agaacctgaa ggactttctg cttgtcatcc cctttgactg ctgggagcca 420
gtccaggag 429
<210> 4
<211> 1520
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 4
cgttacataa cttacggtaa atggcccgcc tggctgaccg cccaacgacc cccgcccatt 60
gacgtcaata atgacgtatg ttcccatagt aacgccaata gggactttcc attgacgtca 120
atgggtggac tatttacggt aaactgccca cttggcagta catcaagtgt atcatatgcc 180
aagtacgccc cctattgacg tcaatgacgg taaatggccc gcctggcatt atgcccagta 240
catgacctta tgggactttc ctacttggca gtacatctac gtattagtca tcgctattac 300
catggtgatg cggttttggc agtacatcaa tgggcgtgga tagcggtttg actcacgggg 360
atttccaagt ctccacccca ttgacgtcaa tgggagtttg ttttggcacc aaaatcaacg 420
ggactttcca aaatgtcgta acaactccgc cccattgacg caaatgggcg gtaggcgtgt 480
acggtgggag gtctatataa gcagagctcg tttagtgaac cgtcagatcg cctggagacg 540
ccatccacgc tgttttgacc tccatagaag acaccgggac cgatccagcc tccggactct 600
agaggatccg gtactcgagg aactgaaaaa ccagaaagtt aactggtaag tttagtcttt 660
ttgtctttta tttcaggtcc cgatccggtg gtggtgcaaa tcaaagaact gctcctcagt 720
ggatgttgcc tttacttcta ggcctgtacg gaagtgttac ttctgctcta aaagctgcgg 780
aattgtaccc gcgggccacc atggtgagca agggcgagga gctgttcacc ggggtggtgc 840
ccatcctggt cgagctggac ggcgacgtaa acggccacaa gttcagcgtg tccggcgagg 900
gcgagggcga tgccacctac ggcaagctga ccctgaagtt catctgcacc accggcaagc 960
tgcccgtgcc ctggcccacc ctcgtgacca ccctgaccta cggcgtgcag tgcttcagcc 1020
gctaccccga ccacatgaag cagcacgact tcttcaagtc cgccatgccc gaaggctacg 1080
tccaggagcg caccatcttc ttcaaggacg acggcaacta caagacccgc gccgaggtga 1140
agttcgaggg cgacaccctg gtgaaccgca tcgagctgaa gggcatcgac ttcaaggagg 1200
acggcaacat cctggggcac aagctggagt acaactacaa cagccacaac gtctatatca 1260
tggccgacaa gcagaagaac ggcatcaagg tgaacttcaa gatccgccac aacatcgagg 1320
acggcagcgt gcagctcgcc gaccactacc agcagaacac ccccatcggc gacggccccg 1380
tgctgctgcc cgacaaccac tacctgagca cccagtccgc cctgagcaaa gaccccaacg 1440
agaagcgcga tcacatggtc ctgctggagt tcgtgaccgc cgccgggatc actctcggca 1500
tggacgagct gtacaagtaa 1520
<210> 5
<211> 1035
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 5
atgtccaatt tactgaccgt acaccaaaat ttgcctgcat taccggtcga tgcaacgagt 60
gatgaggttc gcaagaacct gatggacatg ttcagggatc gccaggcgtt ttctgagcat 120
acctggaaaa tgcttctgtc cgtttgccgg tcgtgggcgg catggtgcaa gttgaataac 180
cggaaatggt ttcccgcaga acctgaagat gttcgcgatt atcttctata tcttcaggcg 240
cgcggtctgg cagtaaaaac tatccagcaa catttgggcc agctaaacat gcttcatcgt 300
cggtccgggc tgccacgacc aagtgacagc aatgctgttt cactggttat gcggcggatc 360
cgaaaagaaa acgttgatgc cggtgaacgt gcaaaacagg ctctagcgtt cgaacgcact 420
gatttcgacc aggttcgttc actcatggaa aatagcgatc gctgccagga tatacgtaat 480
ctggcatttc tggggattgc ttataacacc ctgttacgta tagccgaaat tgccaggatc 540
agggttaaag atatctcacg tactgacggt gggagaatgt taatccatat tggcagaacg 600
aaaacgctgg ttagcaccgc aggtgtagag aaggcactta gcctgggggt aactaaactg 660
gtcgagcgat ggatttccgt ctctggtgta gctgatgatc cgaataacta cctgttttgc 720
cgggtcagaa aaaatggtgt tgccgcgcca tctgccacca gccagctatc aactcgcgcc 780
ctggaaggga tttttgaagc aactcatcga ttgatttacg gcgctaagga tgactctggt 840
cagagatacc tggcctggtc tggacacagt gcccgtgtcg gagccgcgcg agatatggcc 900
cgcgctggag tttcaatacc ggagatcatg caagctggtg gctggaccaa tgtaaatatt 960
gtcatgaact atatccgtaa cctggatagt gaaacagggg caatggtgcg cctgctggaa 1020
gatggcgatt agtaa 1035
<210> 6
<211> 20
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 6
cggagccggc ccggcgaact 20

Claims (10)

1. An antitumor pharmaceutical composition characterized by: comprises an oncolytic virus and a PARP inhibitor, wherein the oncolytic virus is selected from I type herpes simplex virus, the PARP inhibitor is Olaparib, and the molecular formula of the Olaparib is C24H23FN4O3(ii) a The structural formula of Olaparib is shown as formula I:
Figure FDA0003316740370000011
2. the antitumor pharmaceutical composition as claimed in claim 1, wherein: the oncolytic virus is prepared by inserting miRT124 and hGMCSF into type I herpes simplex virus.
3. The antitumor pharmaceutical composition as claimed in claim 1, wherein: the dosage of the oncolytic virus is 10 multiplied by 107-100×107The dosage of PFU and the Olaparib is 100-1000 mg/kg.
4. The antitumor pharmaceutical composition as claimed in claim 1, wherein: the composition further comprises a PD-1 antibody.
5. The antitumor pharmaceutical composition as claimed in claim 4, wherein: the dosage of the PD-1 antibody is 450 mu g-750 mu g.
6. The antitumor pharmaceutical composition as claimed in claim 1, wherein: the oncolytic virus is prepared by a method comprising the following steps:
s1, amplifying sequences at two ends of HSV-1 and ICP34.5 genes respectively;
s2, connecting sequences at two ends of hGM-CSF, CMV-GFP, 4xmIRT124 and ICP34.5 and plasmid pCMV-GFP by using a homologous recombination method to form a complete donor plasmid which is marked as pULL34.5-hGM-CSF-lox 2272-CMV-eGFP-lox2272-4xmIRT 124;
s3, designing sgRNA aiming at an ICP34.5 region, connecting the sgRNA to a Lenti-CRISPR-V2 vector in an enzyme digestion connection mode, and marking as Lenti-CRISPR-ICP 34.5;
s4, co-transfecting HEK293T cells with donor plasmid pULL34.5-hGM-CSF-lox 2272-CMV-eGFP-lox2272-4xmIRT124 and Lenti-CRISPR-ICP34.5 plasmid targeting ICP34.5, and collecting culture supernatant after the cells are completely diseased; adding the supernatant into Vero cells, observing fluorescent plaques after several hours, and picking the fluorescent plaques; obtaining HSV-1 inserted with hGMCSF, CMV-GFP and 4xmIRT124, and marking as oHSV-GFP;
s5, amplifying a Cre fragment by using a plasmid with the Cre fragment as a template, and connecting the Cre fragment to a pCCL-PGK-eGFP vector by using an enzyme digestion connection method, wherein the pCCL-PGK-eGFP vector is marked as pCCL-PGK-eGFP;
s6, transfecting the pCCL-PGK-Cre plasmid prepared in the step S5 to a HEK293T cell, adding the oHSV-GFP prepared in the step S4 in a dosage of 1.0 MOI, and collecting a culture supernatant after the cell is completely diseased; adding the supernatant into Vero cells, observing non-fluorescent plaques, and selecting the non-fluorescent plaques; HSV-1 with hGMCSF and 4xmIRT124 inserted was obtained and designated oHSV.
7. The antitumor pharmaceutical composition as claimed in claim 6,
step S4 includes the following steps:
co-transfecting HEK293T cells with a donor plasmid pULL34.5-hGM-CSF-lox 2272-CMV-eGFP-lox2272-4xmIRT124 and a Lenti-CRISPR-ICP34.5 plasmid targeting ICP34.5, adding HSV-1 at the dose of MOI (1.0) after 24 hours, and collecting culture supernatant after the cells are completely diseased; adding the supernatant into Vero cells by a 10-fold dilution method, observing fluorescent plaques after 48 hours, and selecting the fluorescent plaques; after multiple rounds of purification, HSV-1 inserted with hGMCSF, CMV-GFP and 4xmIRT124 is obtained and is marked as oHSV-GFP.
Step S6 includes the steps of:
transfecting HEK293T cells with the pCCL-PGK-Cre plasmid prepared in the step S5, adding the oHSV-GFP prepared in the step S4 in a dosage of MOI (1.0) after 24 hours, and collecting culture supernatant after the cells are completely diseased; adding the supernatant into Vero cells, observing no fluorescent plaque after 48 hours, and selecting the non-fluorescent plaque; after multiple rounds of purification by the method, HSV-1 inserted with hGMCSF and 4xmIRT124 is obtained and is marked as oHSV.
8. An anti-tumor medication system, which is characterized in that: the system comprises the anti-neoplastic pharmaceutical composition of any one of claims 1-7.
9. Use of an anti-tumor pharmaceutical composition as defined in any one of claims 1 to 8 in the preparation of an anti-tumor medicament or formulation.
10. Use according to claim 9, characterized in that: the tumor is liver cancer, colorectal cancer, bladder cancer, breast cancer, cervical cancer, prostatic cancer, glioma, melanoma, pancreatic cancer, nasopharyngeal carcinoma, lung cancer or gastric cancer.
CN202111233544.9A 2021-10-22 2021-10-22 Application of oncolytic virus, PARP inhibitor and PD-1 antibody in preparation of antitumor drugs Active CN114010666B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111233544.9A CN114010666B (en) 2021-10-22 2021-10-22 Application of oncolytic virus, PARP inhibitor and PD-1 antibody in preparation of antitumor drugs

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111233544.9A CN114010666B (en) 2021-10-22 2021-10-22 Application of oncolytic virus, PARP inhibitor and PD-1 antibody in preparation of antitumor drugs

Publications (2)

Publication Number Publication Date
CN114010666A true CN114010666A (en) 2022-02-08
CN114010666B CN114010666B (en) 2024-05-07

Family

ID=80057099

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111233544.9A Active CN114010666B (en) 2021-10-22 2021-10-22 Application of oncolytic virus, PARP inhibitor and PD-1 antibody in preparation of antitumor drugs

Country Status (1)

Country Link
CN (1) CN114010666B (en)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140162888A1 (en) * 2010-04-06 2014-06-12 Caris Life Sciences Luxembourg Holdings Circulating biomarkers for disease
US20160250267A1 (en) * 2013-10-28 2016-09-01 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Oncolytic hsv vector
CN109985240A (en) * 2017-12-29 2019-07-09 广州威溶特医药科技有限公司 PARP inhibitor and oncolytic virus are in the application for preparing anti-tumor drug
US20200102361A1 (en) * 2017-05-24 2020-04-02 Universitat Autònoma De Barcelona Viral expression construct comprising a fibroblast growth factor 21 (fgf21) coding sequence
US20200206285A1 (en) * 2017-07-26 2020-07-02 Oncorus, Inc. Oncolytic viral vectors and uses thereof
US20210138008A1 (en) * 2018-04-13 2021-05-13 Beijing Wellgene Company Ltd. Recombinant oncolytic virus composition and use thereof
CN113164589A (en) * 2018-06-29 2021-07-23 维西欧制药公司 Compositions and methods for modulating monocyte and macrophage inflammatory phenotype and immunotherapy uses thereof
CN113164484A (en) * 2018-11-29 2021-07-23 复诺健生物科技加拿大有限公司 HSV vectors with reduced neurotoxicity
US20210322545A1 (en) * 2016-02-24 2021-10-21 Children's Hospital Of Eastern Ontario Research Institute Inc. Smc combination therapy for the treatment of cancer

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140162888A1 (en) * 2010-04-06 2014-06-12 Caris Life Sciences Luxembourg Holdings Circulating biomarkers for disease
US20160250267A1 (en) * 2013-10-28 2016-09-01 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Oncolytic hsv vector
US20210322545A1 (en) * 2016-02-24 2021-10-21 Children's Hospital Of Eastern Ontario Research Institute Inc. Smc combination therapy for the treatment of cancer
US20200102361A1 (en) * 2017-05-24 2020-04-02 Universitat Autònoma De Barcelona Viral expression construct comprising a fibroblast growth factor 21 (fgf21) coding sequence
US20200206285A1 (en) * 2017-07-26 2020-07-02 Oncorus, Inc. Oncolytic viral vectors and uses thereof
CN109985240A (en) * 2017-12-29 2019-07-09 广州威溶特医药科技有限公司 PARP inhibitor and oncolytic virus are in the application for preparing anti-tumor drug
US20210138008A1 (en) * 2018-04-13 2021-05-13 Beijing Wellgene Company Ltd. Recombinant oncolytic virus composition and use thereof
CN113164589A (en) * 2018-06-29 2021-07-23 维西欧制药公司 Compositions and methods for modulating monocyte and macrophage inflammatory phenotype and immunotherapy uses thereof
CN113164484A (en) * 2018-11-29 2021-07-23 复诺健生物科技加拿大有限公司 HSV vectors with reduced neurotoxicity

Also Published As

Publication number Publication date
CN114010666B (en) 2024-05-07

Similar Documents

Publication Publication Date Title
CN107759701B (en) Chimeric antigen receptor, NK cell modified by chimeric antigen receptor, coding DNA, mRNA, expression vector, preparation method and application
Huang et al. A broadly applicable, personalized heat shock protein-mediated oncolytic tumor vaccine
CN108243607A (en) For the genetic engineering of the macrophage of immunotherapy
BRPI0611794A2 (en) methods and compositions for inducing multivalent immune responses against dominant and subdominant epitopes expressed in cancer cells and tumor stroma
AU2020397271B2 (en) Chimeric antigen receptor T cell which can be efficiently prepared and safely used, preparation method therefor and use thereof
CN110295149A (en) A kind of CH-P60-117C plants of 3 type duck hepatitis A virus of mutant strain and construction method
CN112391412A (en) Replication-type oncolytic adenovirus for regulating lipid metabolism and application thereof
KR102601875B1 (en) Immunologically cloaking anti-tumor adenovirus
CN114657210B (en) Nanomaterial based on GSDMD protein N-terminal peptide fragment and application thereof
CN109055375B (en) Method for activating gene expression by CRISPR (clustered regularly interspaced short palindromic repeats) auxiliary trans-enhancer and application of method
KR20190042473A (en) A gene vaccine for preventing and treating severe fever with thrombocytopenia syndrome
CN114350615B (en) STAT2 gene deletion cell strain and preparation method and application thereof
CN109456991B (en) Protocatechuic acid regulated switch system, regulating method and application thereof
CN114010666B (en) Application of oncolytic virus, PARP inhibitor and PD-1 antibody in preparation of antitumor drugs
CN101008009B (en) Cell model for quick screening of histone deacetylase inhibitor
CN109456992B (en) Protocatechuic acid regulated multifunctional gene expression platform and application thereof
CN102061310B (en) Construction and application of human FHL1C eukaryotic expression vector
CN110747232A (en) Long-acting stably expressed baculovirus vector and construction method thereof
CN110295180A (en) 3 type duck hepatitis A virus mutated gene ISA-A117C-C4334A of one kind and construction method
CN103131709B (en) Ribonucleic acid (RNA) interference fragment of zinc finger-x (zfx) gene and application of RNA interference fragment in mouse sex control
CN113980915B (en) Novel CXCL 10-expressing replicative oncolytic adenovirus and application thereof
KR20210040992A (en) Chemokine-reactive activated natural killer cells with secondary homing activation against a validated target
CN114075574A (en) Construct, oncolytic virus and application thereof
TWI720319B (en) Ultrasound-sensing proteins and method for stimulating cells
CA3226725A1 (en) Composition and uses thereof

Legal Events

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