CN113425856A - Pharmaceutical composition containing genetically modified oncolytic virus and application thereof in treating cancer - Google Patents

Pharmaceutical composition containing genetically modified oncolytic virus and application thereof in treating cancer Download PDF

Info

Publication number
CN113425856A
CN113425856A CN202110764637.8A CN202110764637A CN113425856A CN 113425856 A CN113425856 A CN 113425856A CN 202110764637 A CN202110764637 A CN 202110764637A CN 113425856 A CN113425856 A CN 113425856A
Authority
CN
China
Prior art keywords
oncolytic virus
tumor
virus
cells
monoclonal antibody
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
CN202110764637.8A
Other languages
Chinese (zh)
Other versions
CN113425856B (en
Inventor
亓爱杰
李少波
韦素碧
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.)
CARBIOGENE THERAPEUTICS Co.,Ltd.
Original Assignee
Nuosa Union Beijing Biomedical Technology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nuosa Union Beijing Biomedical Technology Co ltd filed Critical Nuosa Union Beijing Biomedical Technology Co ltd
Priority to CN202110764637.8A priority Critical patent/CN113425856B/en
Publication of CN113425856A publication Critical patent/CN113425856A/en
Application granted granted Critical
Publication of CN113425856B publication Critical patent/CN113425856B/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
    • A61K35/761Adenovirus
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K35/768Oncolytic viruses not provided for in groups A61K35/761 - A61K35/766
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K48/0008Medicinal 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 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • 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
    • 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
    • C07K14/54Interleukins [IL]
    • 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
    • C12N2710/10011Adenoviridae
    • C12N2710/10021Viruses as such, e.g. new isolates, mutants or their genomic sequences

Abstract

The present invention relates to a pharmaceutical composition comprising a genetically modified oncolytic virus and its use in the treatment of cancer. The invention prepares an oncolytic virus expressing IL-24, gefitinib and a monoclonal antibody specifically aiming at PD-1. The oncolytic virus, gefitinib and monoclonal antibody can be effectively used for treating nasopharyngeal carcinoma. The combined use of the oncolytic virus, gefitinib and a monoclonal antibody can find that the oncolytic virus has a synergistic treatment effect and has an excellent application prospect.

Description

Pharmaceutical composition containing genetically modified oncolytic virus and application thereof in treating cancer
Technical Field
The present invention relates to the field of biology, and more particularly, to pharmaceutical compositions comprising genetically modified oncolytic viruses and their use in the treatment of cancer.
Background
Oncolytic virus is a natural or genetically modified virus, can selectively infect tumor cells, can replicate in large quantities in the tumor cells and finally lyse the tumor cells, and can release a large quantity of tumor antigens simultaneously to improve the tumor microenvironment.
The oncolytic virus plays an anti-tumor role through direct oncolytic action and immunity activity induction of an organism, and due to the lack of necessary conditions for replication in normal cells, the oncolytic virus can reproduce and proliferate in a tumor cell in a targeted manner, finally, the tumor cell is cracked and killed, and the anti-oncolytic virus has no influence on healthy cells. Therefore, the oncolytic virus has good safety and has the advantage of long-acting expression of anti-tumor exogenous genes. Currently common oncolytic viruses include Ad, herpes simplex virus type 1 (HSV-1), Vesicular Stomatitis Virus (VSV), Newcastle Disease Virus (NDV), measles virus (measles virus), poliovirus (poliovirus), parvovirus (parvovirus), reovirus (reoviruses), VV, and the like.
Research has shown that many factors such as lack of co-stimulatory molecules in tumor cells seriously affect the induction and generation of tumor antigen-specific CTL and the corresponding killing activity. By inserting genes with products expressing tumor killing activity, such as immune activation genes, prodrug invertase genes, anti-tumor angiogenesis genes, apoptosis inducing genes and the like, into the genome of the oncolytic virus, the oncolytic virus can be directly used as a delivery vehicle of a target gene, so that the target gene is continuously expressed in tumor cells along with the replication of the virus, and the killing activity of the oncolytic virus is further enhanced.
The gene for expressing the immunoinflammatory mediator is inserted into the genome of the oncolytic virus, so that the local immunoinflammatory mediator in a tumor microenvironment is abnormally increased, the anti-tumor immune response of an organism is activated, and the anti-tumor effect of the oncolytic virus is enhanced. At present, various immune activation genes are applied to genetic recombination of oncolytic viruses, such as granulocyte-macrophage colony stimulating factor (GM-CSF), interleukins (IL-2, IL-12, IL-15 and IL-18), interferons (IFN-alpha and IFN-beta) and the like. Also, recombinant adenovirus RdB/IL-12/IL-18 capable of simultaneously expressing IL-12 and IL-18 has been designed, IL-12 stimulates the anti-tumor immune response of the organism by activating NK cells and cytotoxic T cells, IL-18 enhances the NK cytotoxic effect and promotes the differentiation of T cells. In mouse melanoma transplantation animal model, obvious increase of IL-12, IL-18, IFN-gamma and GM-CSF concentration was detected in tumor body after RdB/IL-12/IL-18 local injection, and in tumor tissue section, massive NK cell, CIM and CD8T cell infiltration and extensive necrosis of tumor tissue were observed, further demonstrating strong killing activity of IL-12 and IL-18 combined action on tumor cells. The same results were also verified in a mouse colon cancer animal model, and compared with the control group, the gene recombinant measles virus MV-GMCSF expressing GM-CSF can significantly delay the progression of mouse colon cancer and prolong the median survival time of mice.
In recent years, research related to oncolytic viruses has been greatly advanced, and has attracted much attention. However, strictly speaking, the marketed and approved product is only a 2015 FDA approved T-vec (Imlygic) of an Advance company, which is used for local treatment of melanoma patients and is the first drug for obtaining FDA approved oncolytic virus treatment. In 2016, were approved for marketing in Europe and Canada, respectively. In China, Cocury (H101) of three-dimensional organisms is approved by CFDA in 2005, but the clinical curative effect of the product is not internationally recognized until now due to the superposition of various factors such as imperfect domestic clinical test standards, loose examination and approval policies, the war of product patents and the like. In addition, the development of oncolytic virus is currently carried out by a plurality of companies in China, and great progress is made, so that clinical applications of the oncolytic virus are submitted to a drug administration by the plurality of companies in 2020. Meanwhile, tumor immunotherapy gradually inclines to combination therapy, and clinical evidence shows that oncolytic virus can enhance the response rate of tumors to immune therapy methods such as immune regulation checkpoint inhibitors, CAR-T cells and the like, and the oncolytic virus is an ideal object in combination therapy strategies. Making oncolytic viruses a hot direction in tumor immunization studies.
However, currently, there are also few proposals for the use of oncolytic virus products, particularly modified oncolytic virus products, in combination with immune checkpoint inhibitors, which are provided in the home.
Disclosure of Invention
In one aspect, it is an object of the present invention to provide means for improving cancer treatment.
This is achieved according to the invention by the subject-matter defined in the claims.
The present invention provides a pharmaceutical composition comprising (a) an oncolytic virus and (b) a PD-1 inhibitor and (c) gefitinib. The invention also provides the use of the pharmaceutical composition for the treatment of cancer.
The pharmaceutical composition of the present invention may include the oncolytic virus of the present invention, including pharmaceutically acceptable ingredients, buffers, excipients, adjuvants, preservatives, fillers, stabilizers, thickeners and other ingredients commonly used in formulation. In addition, in some cases, in addition to the oncolytic virus of the invention, other therapeutically effective tumor-dissolving viruses and/or therapeutically effective drugs may be included, such as anti-cancer agents, adjunct ingredients (e.g., immune checkpoint inhibitors, such as CTLA-4 blocks, and immune activators, such as GM-CSF).
Preferably, in the pharmaceutical composition, the oncolytic virus and the checkpoint inhibitor are present in effective doses and are combined with a pharmaceutically acceptable carrier.
Further, the oncolytic virus is a transgenic oncolytic virus expressing IL-24.
Further, the transgenic oncolytic virus is pAdxsi adenovirus expressing IL-24.
Further, the oncolytic virus is pAd-HS4-AFP-E1A-IL-24 virus plasmid, which is obtained by a person skilled in the art through a conventional construction method or a whole gene synthesis method, as long as the virus can express IL-24.
Furthermore, the invention also provides a specific PD-1 monoclonal antibody, which is a monoclonal antibody which is obtained by immunizing a mouse by the PD-1 protein and is specifically combined with PD-1 by a hybridoma screening technology. The variable region of the light chain of the monoclonal antibody is shown as SEQ ID NO: 1, and the heavy chain variable region is shown as SEQ ID NO: 2, the monoclonal antibody has a good binding effect with a binding constant of 5.9nM to PD-1 protein, and has good specificity and no reaction with other proteins.
Light chain variable region (SEQ ID NO: 1)
DIVITQSPALMAASPGEKVTITCTCYSDEGYHNCNWYQQKSGISPKPWIYLPEFYQCGVPARFSGSGSGTSYSLTITSMEAEDAATYYCINLILGCERFGAGTKLELK
Heavy chain variable region (SEQ ID NO: 2)
EVQLEESGTELARPGASVKLSCKASGYIFSFWALGWIKQRPGQGLEWIGNFTTQAVERTKHDFQFGKATLTADKSSSTAYMQLSSLASEDSAVYYCAGGAKRCLNWGLGTTLAVSS。
It is noted that in this disclosure, and in the claims and/or paragraphs particularly, the terms "comprising," "including," and the like may have the meaning attributed to it in U.S. patent statutes; for example, they can mean "including", "comprising", "including", and the like; and the terms "consisting essentially of and" consisting essentially of have their definitions in U.S. patent statutes, e.g., they allow for elements not expressly listed but exclude elements found in the prior art or that affect a basic or novel feature of the invention.
In some embodiments, the oncolytic virus is derived from a poxviridae family. In some embodiments, the oncolytic virus is derived from a chordaviridae or arbovirus subfamily.
In some embodiments, the oncolytic virus is from an adenovirus. Or other available oncolytic viruses.
In some embodiments, the cancer is a solid tumor. In some embodiments, the solid tumor is a sarcoma or carcinoma.
In some embodiments, the solid tumor is fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyoma, rhabdomyosarcoma, colon carcinoma, pancreatic carcinoma, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary adenocarcinoma, bladder cancer, medullary carcinoma, bronchial cancer, renal cell carcinoma, liver cancer, bile duct cancer, choriocarcinoma, seminoma, embryonic cancer, Wilm's tumor, cervical cancer, uterine cancer, testicular cancer, lung cancer, small cell lung cancer, bladder cancer, epithelial cancer, glioma, astrocytoma, medulloblastoma, craniopharyngioma, epididymoma, spongioblastoma, acoustic neuroma, oligodendroglioma, glioma, meningioma, melanoma, neuroblastoma or retinoblastoma.
In some embodiments, the oncolytic virus, gefitinib, and the PD-1 antibody are administered together. In some embodiments, the method comprises administering the oncolytic virus and the PD-1 inhibitor for at least 1 day, at least 2 days, at least 3 days, at least 1 week, at least 2 weeks, at least 3 weeks, at least 4 weeks, at least 1 month, at least 3 months, at least 6 months, or at least 1 year. In some embodiments, the method comprises administering to the subject an oncolytic virus, a nuclear mass transport inhibitor, or both, twice weekly, biweekly, every three weeks, every four weeks, monthly, or every two months. In some embodiments, the oncolytic virus is administered locally to the cancer tissue to be treated. In some embodiments, the oncolytic virus is administered by intratumoral injection. In some embodiments, the multiple infection of the tissue by the oncolytic virus (MoI) is from about 0.01 to about 10. In some embodiments, the multiple infection (MOI) of the tissue by the oncolytic virus is from about 0.05 to about 5.
Advantageous effects
The invention prepares an oncolytic virus expressing IL-24, gefitinib and a monoclonal antibody specifically aiming at PD-1. The use of the oncolytic virus together with a monoclonal antibody may be useful for the treatment of nasopharyngeal carcinoma. The combined use of the oncolytic virus and the monoclonal antibody can find that the combined use of the oncolytic virus and the monoclonal antibody has synergistic treatment effect and excellent application prospect.
Drawings
FIG. 1 is a graph showing the effect of the use of the PD-1 antibody in combination with an oncolytic virus
FIG. 2 graph of the effect of the PD-1 antibody in combination with oncolytic virus and gefitinib
Detailed Description
Examples will be given below and further illustrate the present invention, but the examples are merely illustrative of the embodiments of the present invention and do not limit the scope of the present invention.
EXAMPLE 1 preparation of transgenic oncolytic viruses
Plasmids containing HS-4-AFP-E1A-PolyA were purchased from Shanghai three-dimensional Biotechnology, Inc. pShuttle-Basic-EGFP recombinant shuttle vector and pAdxsi recombinant adenovirus backbone vector were purchased from Nossel genome research center, Inc.
According to the construction method in 'research on the effect of oncolytic adenovirus carrying IL-24 gene for inhibiting primary liver cancer lung metastasis, Ph paper of Chack university, 2009', a target gene fragment is obtained by amplifying plasmid HS4-AFP-E1A-PolyA, cloned to pGEM-T Easy vector to obtain HS4-AFP-E1A-PolyA-T cloning vector, IL-24 gene is amplified, connected to pShuttle-Basic-EGFP vector to replace EGFP to obtain pShuttle-Basic-IL-24, then the fragment of HS4-AFP-E1A-PolyA is cut from HS4-AFP-E1A-PolyA-T cloning vector, connected to pShuttle-Basic-IL-24 vector to obtain pShuttle-Basic-IL-24-AFP-E1A, and transferred to pAdxsi adenovirus recombinant skeleton vector, the pAd-HS4-AFP-E1A-IL-24 virus plasmid is obtained, and the construction is correct through enzyme digestion identification.
Packaging and purifying the recombinant adenovirus: lipofectamine 2000 liposomes transfected 293 cells to obtain virus, stored at-80 ℃. (3) Adenovirus amplification and purification: purification by cesium chloride gradient centrifugation and titer determination was performed using TCID 50. The titers of the amplified viruses are all 2.1 multiplied by 10 by the method of TCID5011PFU/ml。
EXAMPLE 2 preparation of PD-1 inhibitor
The immunogen is PD-1 recombinant protein (P1210, Okay organism). Mice were immunized by the general method. Briefly, an appropriate amount of Freund's adjuvant was taken out into a 1.5ml EP tube and mixed well with a shaker. The antigen protein solution was prepared with PBS. Mixing adjuvant and protein antigen solution according to required amount, emulsifying antigen by injector to form stable water-in-oil solution, and injecting into animal. According to the result of serum titer measurement, after the first immunization, 3 times of boosting immunization is usually required to achieve a good immune effect. And (3) selecting immune mice with high serum titer to perform cell fusion after intraperitoneal injection. Prior to cell fusion, mouse myeloma cells (SP2/0-Ag14, ATCC # CRL-1581) were cultured in logarithmic growth phase. After sacrifice of the immunization, the mice were harvested in sterile environment and splenic B lymphocytes and SP2/0 myeloma cells were fused using PEG chemistry. After the fusion, the cells were plated in 96-well cell culture plates, and the growth of viable hybridoma cells was observed under a microscope after 7 to 10 days. Two weeks after cell plating, culture supernatants from each well were collected and hybridoma screened using human PD-1-his protein antigen by ELISA. Briefly, the microplate was coated with 50ul of 2ug/ml of human PD-1-his antigen in PBS overnight at 4 ℃. After 4 additional PBST washes, the plates were blocked for 3 hours at 37 ℃ with 200. mu.l/well of 5% skim milk powder in PBST. The plate was washed again, 50. mu.l/well of hybridoma supernatant was added, incubated at 37 ℃ for 40 minutes, and then the plate was washed 5 times. Diluted horseradish peroxidase-labeled secondary goat-anti-mouse antibody was added at 100. mu.l/well, incubated at 37 ℃ for 40 minutes, and then the plate was washed 5 times and patted dry. The absorbance of each well at 450 nm was measured by adding 100. mu.l/well of TMB chromogenic substrate, developing at room temperature for 20 minutes, and then stopping with a 2M solution of sulfuric acid. And (4) selecting hybridoma hole cells with positive ELISA color development, transferring the hybridoma hole cells into a 24-hole plate, and continuing to culture. And performing a second round of re-screening by an ELISA method, screening out hybridomas which specifically recognize PD-1 antigens and can block the combination of PD-1, and performing subcloning by a limiting dilution method to obtain the target monoclonal cell strain 3F 2.
Through sequence identification, the variable region of the light chain of the monoclonal antibody is shown as SEQ ID NO: 1, and the heavy chain variable region is shown as SEQ ID NO: 1, the monoclonal antibody has a good binding effect with a binding constant of 5.9nM to PD-1 protein, and has good specificity and no reaction with other proteins.
Light chain variable region (SEQ ID NO: 1)
DIVITQSPALMAASPGEKVTITCTCYSDEGYHNCNWYQQKSGISPKPWIYLPEFYQCGVPARFSGSGSGTSYSLTITSMEAEDAATYYCINLILGCERFGAGTKLELK
Heavy chain variable region (SEQ ID NO: 2)
EVQLEESGTELARPGASVKLSCKASGYIFSFWALGWIKQRPGQGLEWIGNFTTQAVERTKHDFQFGKATLTADKSSSTAYMQLSSLASEDSAVYYCAGGAKRCLNWGLGTTLAVSS。
Example 3 Effect of transgenic oncolytic tumor vaccine on CTL cells
The high titer virus prepared in example 1 was selected to infect human nasopharyngeal carcinoma cells CNE2, while the empty virus was transfected as a control. G418 (250. mu.g/ml) was screened for 2 weeks to obtain positive clones. The integration of the IL24 gene into CEN2 cells was confirmed by PCR detection. The double-antibody sandwich ELISA detects IL24 transfection positive cells, and the cell amount detected is 106And (4) respectively. The results are shown in table 1 below.
TABLE 1 secretion of IL-24
Type (B) Amount of secretion
IL24 positive 945.3±23.4pg/24h
As can be seen from the results in Table 1, the secretion of IL24 in the transgenic cells was 945.3. + -. 23.4pg/24h, and the secretion of IL24 in the control cells was low, indicating that the transgene was successful.
Mitomycin C (40 mug.10)6Cells/ml, 37 ℃,1h) treated wild type tumor cells, empty vector and IL24 gene transfected tumor cells were stimulator cells (5X 10)5) Peripheral blood mononuclear cells (PBMC, 10) from healthy donors7) Mixed culture at 37 deg.C and 5% CO2After 1 week of incubation, the reaction cells were collected and restimulated at 2X 106Separately adding stimulating cells (2X 10) to the reaction cells5) Autologous feeder cells (mitomycin C-treated PBMC, 10)5) Culturing for 1 week, collecting the reaction cells, and culturing for 4 hr51The CTL killing activity was detected by Cr release assay. The results are shown in Table 2.
TABLE 2CTL killing Activity results
Type (B) Killing activity (%)
Mitomycin-treated wild-type tumor cells 12.3±1.9
Empty virus tumor cells 12.6±1.8
Tumor cells transformed with IL-24 virus 76.5±2.3
As can be seen from Table 2, the IL-24 gene modified tumor vaccine can induce strong CTL response, and the killing power of the tumor vaccine reaches (76.5 +/-2.3)%, so that the tumor vaccine has a better effect.
Example 4 verification of tumor-inhibiting Effect of oncolytic Virus
Balb-c nude mice were purchased from Shanghai Slek laboratory animal center, female, 4 weeks old, and had a body mass of 15-20 g. Conventionally culturing human nasopharyngeal carcinoma cell CNE2, amplifying, collecting cells in logarithmic growth phase, digesting, and making into single cell suspension. And (5) carrying out ear-cutting marking on the experimental animal, and recording the quality of the experimental animal. 0.3 percent sodium pentobarbital is injected into the abdominal cavity of an anesthesia experimental animal according to the dose of 30-40 mg/kg. 100 μ l of CNE2 cell suspension was injected subcutaneously into bilateral flank of nude mice, and the amount of cells injected into each side of each nude mouse was 1X 106And (4) respectively. Animal grouping and treatment: nude mice were randomly divided into 3 groups: 10 oncolytic virus treated groups and oncolytic virus + PD-1 mab treated groups as well as control groups. After injecting the tumor into the experimental animal for 4 days, measuring the size of the subcutaneous tumor by using a vernier caliper, and when the subcutaneous tumor is ill, measuring the subcutaneous tumor by using the vernier caliperWhen the tumor on the side of the virus group was grown to 5mm (about day 8), the virus was injected intratumorally. Virome treatment the virus of example 1 was injected twice a week at 5X 105Mu.l for 4 weeks, and the control group was injected twice weekly with 100. mu.l of phosphate buffered saline for 4 weeks. Oncolytic Virus + PD-1 monoclonal antibody treatment group twice weekly injections of the virus of example 1 at 5X 105Mu.l 50. mu.l and PD-1 mAb 1mg/kg, injected for 4 weeks. Tumor size measurement: the maximum major diameter (a) and maximum width (b) of the tumor were measured after each inoculation, and the tumor volume V was calculated according to the formula (a × b2/2) to plot the tumor volume growth curve. The results are shown in FIG. 1.
As can be seen from the results in FIG. 1, the addition of oncolytic virus effectively inhibited tumor growth, and in particular, reduced tumor volume after 3 weeks. After the PD-1 antibody is added and the oncolytic virus is used together, the tumor volume can be more obviously shortened, and in the 4 th week, the tumor volume is only 80mm3And the inhibitor has obvious inhibiting effect.
Example 5 verification of tumor-inhibiting Effect of oncolytic Virus in combination with antitumor drug
Balb-c nude mice were purchased from Shanghai Slek laboratory animal center, female, 4 weeks old, and had a body mass of 15-20 g. Conventionally culturing human nasopharyngeal carcinoma cell CNE2, amplifying, collecting cells in logarithmic growth phase, digesting, and making into single cell suspension. And (5) carrying out ear-cutting marking on the experimental animal, and recording the quality of the experimental animal. 0.3 percent sodium pentobarbital is injected into the abdominal cavity of an anesthesia experimental animal according to the dose of 30-40 mg/kg. 100 μ l of CNE2 cell suspension was injected subcutaneously into bilateral flank of nude mice, and the amount of cells injected into each side of each nude mouse was 1X 106And (4) respectively. Animal grouping and treatment: nude mice were randomly divided into 3 groups: 10 oncolytic virus + PD-1 monoclonal antibody treatment groups, oncolytic virus + PD-1 monoclonal antibody + gefitinib treatment groups and control groups. Subcutaneous tumor size was measured with a vernier caliper starting 4d after tumor injection into the experimental animals, and intratumoral virus injection treatment was performed when the tumor on the virus-treated side grew to 5mm (about day 8). The control group was injected twice weekly with 50 μ l of phosphate buffered saline for a total of 4 weeks. Oncolytic virus + PD-1 monoclonal antibody treatment group twice weeklyVirus 5X 10 of example 15Mu.l 50. mu.l and PD-1 mAb 1mg/kg, injected for 4 weeks. Oncolytic virus + PD-1 monoclonal antibody treatment group and oncolytic virus + PD-1 monoclonal antibody + gefitinib treatment group were injected twice weekly with 5X 10 of the virus of example 15Mu.l 50. mu.l and PD-1 mAb 1mg/kg + gefitinib 2mg/kg for a total of 4 weeks. Tumor size measurement: the maximum major diameter (a) and maximum width (b) of the tumor were measured after each inoculation, and the tumor volume V was calculated according to the formula (a × b2/2) to plot the tumor volume growth curve. The results are shown in FIG. 2.
As can be seen from the results in FIG. 2, the addition of oncolytic virus effectively inhibited tumor growth, and in particular, reduced tumor volume after 3 weeks. After the PD-1 antibody is added and the oncolytic virus and gefitinib are used together, the tumor volume can be more remarkably shortened, and in the 4 th week, the tumor volume is only 30mm3About 80mm in comparison with the addition of PD-1 antibody and oncolytic virus3Has obviously improved inhibiting effect.
The invention has been described in detail with reference to specific embodiments and illustrative examples, but the description is not intended to be construed in a limiting sense. Those skilled in the art will appreciate that various equivalent substitutions, modifications or improvements may be made to the technical solution of the present invention and its embodiments without departing from the spirit and scope of the present invention, which fall within the scope of the present invention. The scope of the invention is defined by the appended claims.
Sequence listing
<110> Nosai Union (Beijing) biomedical science and technology Co., Ltd
<120> a pharmaceutical composition comprising a genetically modified oncolytic virus and its use in the treatment of cancer
<160> 2
<170> SIPOSequenceListing 1.0
<210> 1
<211> 108
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 1
Asp Ile Val Ile Thr Gln Ser Pro Ala Leu Met Ala Ala Ser Pro Gly
1 5 10 15
Glu Lys Val Thr Ile Thr Cys Thr Cys Tyr Ser Asp Glu Gly Tyr His
20 25 30
Asn Cys Asn Trp Tyr Gln Gln Lys Ser Gly Ile Ser Pro Lys Pro Trp
35 40 45
Ile Tyr Leu Pro Glu Phe Tyr Gln Cys Gly Val Pro Ala Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Ser Tyr Ser Leu Thr Ile Thr Ser Met Glu
65 70 75 80
Ala Glu Asp Ala Ala Thr Tyr Tyr Cys Ile Asn Leu Ile Leu Gly Cys
85 90 95
Glu Arg Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys
100 105
<210> 2
<211> 116
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 2
Glu Val Gln Leu Glu Glu Ser Gly Thr Glu Leu Ala Arg Pro Gly Ala
1 5 10 15
Ser Val Lys Leu Ser Cys Lys Ala Ser Gly Tyr Ile Phe Ser Phe Trp
20 25 30
Ala Leu Gly Trp Ile Lys Gln Arg Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Asn Phe Thr Thr Gln Ala Val Glu Arg Thr Lys His Asp Phe Gln
50 55 60
Phe Gly Lys Ala Thr Leu Thr Ala Asp Lys Ser Ser Ser Thr Ala Tyr
65 70 75 80
Met Gln Leu Ser Ser Leu Ala Ser Glu Asp Ser Ala Val Tyr Tyr Cys
85 90 95
Ala Gly Gly Ala Lys Arg Cys Leu Asn Trp Gly Leu Gly Thr Thr Leu
100 105 110
Ala Val Ser Ser
115

Claims (2)

1. A pharmaceutical combination for use in the treatment of nasopharyngeal carcinoma, characterized in that said combination consists of oncolytic virus, gefitinib and PD-1 monoclonal antibody; wherein, the oncolytic virus is pAdxsi adenovirus for specifically expressing IL-24; the variable region of the light chain of the monoclonal antibody is shown as SEQ ID NO: 1, and the heavy chain variable region is shown as SEQ ID NO: 2, respectively.
2. Use of a combination of an oncolytic virus, gefitinib and a PD-1 monoclonal antibody for the preparation of a pharmaceutical composition for the treatment of nasopharyngeal carcinoma, wherein the oncolytic virus is a pAdxsi adenovirus specifically expressing IL-24; the variable region of the light chain of the monoclonal antibody is shown as SEQ ID NO: 1, and the heavy chain variable region is shown as SEQ ID NO: 2, respectively.
CN202110764637.8A 2021-07-07 2021-07-07 Pharmaceutical composition containing genetically modified oncolytic virus and application thereof in treating cancer Active CN113425856B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110764637.8A CN113425856B (en) 2021-07-07 2021-07-07 Pharmaceutical composition containing genetically modified oncolytic virus and application thereof in treating cancer

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110764637.8A CN113425856B (en) 2021-07-07 2021-07-07 Pharmaceutical composition containing genetically modified oncolytic virus and application thereof in treating cancer

Publications (2)

Publication Number Publication Date
CN113425856A true CN113425856A (en) 2021-09-24
CN113425856B CN113425856B (en) 2022-01-04

Family

ID=77759296

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110764637.8A Active CN113425856B (en) 2021-07-07 2021-07-07 Pharmaceutical composition containing genetically modified oncolytic virus and application thereof in treating cancer

Country Status (1)

Country Link
CN (1) CN113425856B (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110785180A (en) * 2017-04-21 2020-02-11 贝勒医学院 Oncolytic viral therapy and immunotherapy
US20200289591A1 (en) * 2017-09-26 2020-09-17 Hangzhou Converd Co., Ltd. Isolated Recombinant Oncolytic Poxvirus, Pharmaceutical Composition, and Use Thereof in Treatment of Tumors and/or Cancer
WO2021000813A1 (en) * 2019-06-30 2021-01-07 方树彬 Monoclonal antibody targeting pd-1 and use thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110785180A (en) * 2017-04-21 2020-02-11 贝勒医学院 Oncolytic viral therapy and immunotherapy
US20200289591A1 (en) * 2017-09-26 2020-09-17 Hangzhou Converd Co., Ltd. Isolated Recombinant Oncolytic Poxvirus, Pharmaceutical Composition, and Use Thereof in Treatment of Tumors and/or Cancer
WO2021000813A1 (en) * 2019-06-30 2021-01-07 方树彬 Monoclonal antibody targeting pd-1 and use thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
QTF73967.1: "immunoglobulin light chain variable region, partial [synthetic construct]", 《GENBANK》 *
SINKOVICS J G: "A novel conditionally oncolytic adenovirus for nasopharyngeal cancer gene therapy", 《ACTA MICROBIOLOGIC A HUNGARICA》 *
曹华等: "新型溶肿瘤腺病毒Ad-TD-RFP对人鼻咽癌细胞的治疗作用", 《中华耳鼻咽喉头颈外科杂志》 *

Also Published As

Publication number Publication date
CN113425856B (en) 2022-01-04

Similar Documents

Publication Publication Date Title
JP7295192B2 (en) modified oncolytic virus
CN110128550B (en) Novel replicative oncolytic adenovirus capable of simultaneously blocking immune check points PD-L1 and TIGIT and application
US20240100106A1 (en) Isolated recombinant oncolytic adenoviruses, pharmaceutical compositions, and uses thereof for drugs for treatment of tumors and/or cancers
KR102227180B1 (en) Recombinant herpes simplex virus and uses thereof
CN109745343B (en) Recombinant oncolytic virus compositions and uses thereof
JP7066812B2 (en) Therapeutic compositions and methods of use for treating cancer
US20210252135A1 (en) Treatment using oncolytic virus
JP7248325B2 (en) Isolated Recombinant Oncolytic Vaccinia Viruses, Pharmaceutical Compositions, and Uses Thereof for Medicines for the Treatment of Tumors and/or Cancers
CN112584849A (en) Therapeutic agents comprising nucleic acids and CAR-modified immune cells and uses thereof
JP7132339B2 (en) Pseudorabies virus for treating tumors
CN111606999B (en) Replicative oncolytic adenovirus with functions of activating immune co-stimulatory signaling pathway and blocking immune checkpoint and application thereof
WO2018171103A1 (en) Programmable oncolytic virus vaccine system and application thereof
CN113425856B (en) Pharmaceutical composition containing genetically modified oncolytic virus and application thereof in treating cancer
CN113244411B (en) Method for inducing CTL cell by gene modified oncolytic virus and application of CTL cell in tumor treatment
CN109419818B (en) Echovirus for treating tumors
CN114657150A (en) Recombinant oncolytic adenovirus for improving immunotherapy and application thereof
GB2583296A (en) Dna molecules producing custom designed replicating and non-replicating negative stranded rna viruses and uses there of
CN116590242A (en) Recombinant oncolytic adenovirus and application thereof
CN117025555A (en) Oncolytic adenovirus expressing dual-specificity T cell adapter, construction method and application
CN116981483A (en) Adenovirus encoding IL-15

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
TA01 Transfer of patent application right

Effective date of registration: 20211214

Address after: Room 2006-2008, 20 / F, building 5, no.688, Bin'an Road, Changhe street, Binjiang District, Hangzhou City, Zhejiang Province 310000

Applicant after: CARBIOGENE THERAPEUTICS Co.,Ltd.

Address before: Room 01-12, 1st floor, building 1, No.3, Yongchang North Road, Beijing Economic and Technological Development Zone, Daxing District, Beijing

Applicant before: Nuosa Union (Beijing) Biomedical Technology Co.,Ltd.

TA01 Transfer of patent application right
GR01 Patent grant
GR01 Patent grant