WO2024119387A1 - System for controlling target gene expression in response to hypoxic environment and use thereof - Google Patents
System for controlling target gene expression in response to hypoxic environment and use thereof Download PDFInfo
- Publication number
- WO2024119387A1 WO2024119387A1 PCT/CN2022/137157 CN2022137157W WO2024119387A1 WO 2024119387 A1 WO2024119387 A1 WO 2024119387A1 CN 2022137157 W CN2022137157 W CN 2022137157W WO 2024119387 A1 WO2024119387 A1 WO 2024119387A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- gene
- virus
- gene encoding
- seq
- nucleotide sequence
- 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.)
- Ceased
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
Definitions
- the present invention belongs to the field of biomedicine technology, and specifically relates to a system for controlling the expression of a target gene in response to a hypoxic environment and its use in preparing a drug for treating hypoxic diseases, such as cancer.
- Oncolytic virus-mediated immunotherapy is another important emerging tumor treatment strategy after the three major conventional treatment methods (surgery, radiotherapy and chemotherapy) and immunotherapy.
- Other tumor immunotherapies such as cell immunotherapy, are expensive and difficult to solve the problem of the universality of cell immunotherapy.
- oncolytic viruses have the advantages of high killing efficiency, good targeting, small adverse reactions, diverse tumor killing pathways, low drug resistance and low cost.
- Studies have shown that oncolytic viruses can be applied to different types of tumors or tumors at different stages of progression, and can even improve the overall survival rate of patients with metastatic or incurable cancer.
- oncolytic virus therapy is considered to be one of the important means of saving lives, and oncolytic virus therapy can induce complete regression or remission of tumors.
- oncolytic virus therapy is receiving widespread attention and has a very broad prospect in the treatment of tumors.
- oncolytic virus therapy began to be introduced into cancer treatment strategies.
- oncolytic viruses have evolved from the initial generation of products that were not genetically modified, that is, directly using wild virus strains or attenuated wild virus strains, such as reovirus and Newcastle disease virus, to the research and development stage of genetically modified virus strains.
- the viruses that can proliferate in tumor cells through genetic recombination mainly include herpes simplex virus, adenovirus, measles virus, and vaccinia virus, and have been iteratively upgraded to the third generation of new and powerful oncolytic viruses, that is, the stage of gene insertion and combined therapy enhancement, which is also the main research direction of oncolytic viruses in the current clinical stage.
- the current improved strategy is to modify the oncolytic virus gene or its delivery method to adapt it to the patient's body system and specifically activate the immune system to achieve targeted killing of tumors; or to use oncolytic viruses in combination with immune checkpoint inhibitors such as PD-1 and CTLA-4, using oncolytic viruses to break the tumor tissue microenvironment, so that immune checkpoint inhibitors such as PD-1 and CTLA-4 can play a better role.
- the object of the present invention is to provide a system for controlling the transcription and expression of a target gene in response to a hypoxic environment.
- the system is induced by a hypoxia-sensitive artificial transcription factor to be efficiently expressed in a normoxic environment, and the expression is inhibited in a hypoxic environment, thereby ensuring the hypoxia sensitivity of the system; and the low-level expression of the artificial transcription factor induced in the hypoxic environment releases the inhibition of the target gene, thereby starting the efficient transcription and expression of the target gene, achieving targeted delivery of the target gene in a hypoxic environment, and avoiding serious toxic side effects caused by systemic administration.
- the present invention also provides the application of the hypoxia-sensitive transcription expression regulation system, which can be used to prepare drugs for treating hypoxic diseases such as solid tumors by accessing different target genes.
- the present invention provides a system for controlling the expression of a target gene in response to a hypoxic environment, which comprises a gene encoding a hypoxia sensing unit and a gene encoding a target gene expression unit, which are separated from or fused to each other, wherein the gene encoding the hypoxia sensing unit comprises a gene encoding a hypoxia response element (HRE) and a gene encoding an artificial transcription factor (ATF) connected via a gene encoding a virus transcription promoter (VTP), and the gene encoding the target gene expression unit comprises a gene encoding an artificial transcription factor recognition element (ATFRE) and a target gene (gene of interest, GOI) connected via a gene encoding a virus transcription promoter.
- HRE hypoxia response element
- ATF artificial transcription factor
- VTP virus transcription promoter
- the gene encoding the target gene expression unit comprises a gene encoding an artificial transcription factor recognition element (ATFRE) and a target gene (gene of interest, GO
- the hypoxic environment refers to cells or tissues with an oxygen content of less than 2%.
- Tumor tissues and cells have a great demand for energy substances such as oxygen and glucose.
- hypoxia of tumor cells and tissues occurs, and the oxygen content of cells and tissues is often less than 2%.
- a gene encoding a joint is included between the gene encoding VTP and the gene encoding HRE, and between the gene encoding VTP and the gene encoding ATF.
- the nucleotide sequence of the gene encoding the joint is a random sequence, generally 10-20 bp in size.
- the nucleotide sequence of the gene encoding HRE is as shown in SEQ ID NO: 1, or is a repeated nucleotide sequence of more than 1, preferably 3, and more preferably 5 nucleotide sequences as shown in SEQ ID NO: 1.
- the VTP is selected from one or more of PE/L, truncated PE/L (PE/L-L), further truncated PE/L (PE/L-S), P7.5, truncated P7.5 (P7.5-71), further truncated P7.5 (P7.5-24), LTRminiP or P11.
- the nucleotide sequence of the PE/L is as shown in SEQ ID NO: 2; the nucleotide sequence of the PE/L-L is as shown in SEQ ID NO: 3; the nucleotide sequence of the PE/L-S is as shown in SEQ ID NO: 4; the nucleotide sequence of the P7.5 is as shown in SEQ ID NO: 5; the nucleotide sequence of the P7.5-71 is as shown in SEQ ID NO: 6; the nucleotide sequence of the P7.5-24 is as shown in SEQ ID NO: 7; the nucleotide sequence of the LTRminiP is as shown in SEQ ID NO: 8; the nucleotide sequence of the P11 is as shown in SEQ ID NO: 37.
- the gene encoding ATF is selected from one or more of the CRE gene with a denuclearized localization signal (dNLS), the CRE-VP64 gene with a denuclearized localization signal, the CRE-VP64-ODD gene with a denuclearized localization signal, the CRE-RAP94 gene with a denuclearized localization signal, the CRE-RAP94-ODD gene with a denuclearized localization signal, or the CRE-VP160 gene with a denuclearized localization signal, and is preferably the CRE-VP64 gene with a denuclearized localization signal.
- dNLS denuclearized localization signal
- the CRE-VP64 gene with a denuclearized localization signal the CRE-VP64-ODD gene with a denuclearized localization signal
- the CRE-RAP94 gene with a denuclearized localization signal the CRE-RAP94 gene with a denuclearized local
- the nucleotide sequence of the CRE gene with the denuclearization localization signal is shown in SEQ ID NO: 21 (the corresponding amino acid sequence is shown in SEQ ID NO: 22); the nucleotide sequence of the CRE-VP64 gene with the denuclearization localization signal is shown in SEQ ID NO: 23 (the corresponding amino acid sequence is shown in SEQ ID NO: 24); the nucleotide sequence of the CRE-VP64-ODD gene with the denuclearization localization signal is shown in SEQ ID NO: 25 (the corresponding amino acid sequence is shown in SEQ ID NO: 26
- the nucleotide sequence of the CRE-RAP94 gene with the nuclear localization signal is shown in SEQ ID NO: 27 (the corresponding amino acid sequence is shown in SEQ ID NO: 28); the nucleotide sequence of the CRE-RAP94-ODD gene with the nuclear localization signal is shown in SEQ ID NO: 29 (the corresponding amino acid sequence is shown in
- the gene encoding ATFRE is one or more Loxp genes.
- the nucleotide sequence of the gene encoding ATFRE is a repeated gene sequence of more than 1, preferably 3, more preferably 5, and further preferably 6 Loxp genes, such as the nucleotide sequence shown in SEQ ID NO: 20.
- the GOI is selected from one or more of co-stimulatory molecules, cytokines, negative regulatory molecules, blocking antibodies of signal pathways, chemokines or killer molecules, preferably killer molecules, and more preferably one or more of the encoding genes of BiTE.
- the composition of the hypoxia sensing unit is 5*SEQ ID NO: 1-gene encoding VTP-dNLS-CRE-VP64 gene, and/or the composition of the target gene expression unit is SEQ ID NO: 20-gene encoding VTP-GOI. More preferably, the composition of the hypoxia sensing unit is 5*SEQ ID NO: 1-gene encoding PE/L-dNLS-CRE-VP64 gene, and/or the composition of the target gene expression unit is SEQ ID NO: 20-gene encoding P7.5-GOI.
- composition of the hypoxia sensing unit is 5*SEQ ID NO: 1-gene encoding PE/L-dNLS-CRE-VP64 gene, and/or the composition of the target gene expression unit is SEQ ID NO: 20-gene encoding P11-GOI.
- the system for controlling the expression of a target gene in response to a hypoxic environment comprises:
- the vector is selected from one or more of pUC18, pUC19, pUC57, pcDNA3, pcDNA4, pcDNA5, pcDNA6, pCMV, pEF1, PEGFP, pET, pEasy, pAc5, pAcGP, pAcYCDuet, pBluescript, pBudCE, pCAMBIA, pCold, pCR2, pCR3, pCR4, pDsRED, pGEM, pGL, pIRES, pPIC, pMAL or pVRCSV1.0, etc.
- the present invention provides a recombinant virus, the genome of which comprises the system for controlling the expression of a target gene in response to a hypoxic environment according to the present invention.
- the virus is selected from one or more of vaccinia virus, adenovirus, herpes simplex virus type I (HSV-1), herpes simplex virus type II (HSV-2), vesicular stomatitis virus, echovirus, reovirus, alphavirus, yellow fever virus, coxsackievirus, Newcastle disease virus, measles virus, poliovirus, Zika virus, lymphocytic choriomeningitis virus, M1 virus, Marburg virus, lentivirus or retrovirus.
- HSV-1 herpes simplex virus type I
- HSV-2 herpes simplex virus type II
- vesicular stomatitis virus echovirus
- reovirus alphavirus
- yellow fever virus coxsackievirus
- Newcastle disease virus measles virus
- poliovirus Zika virus
- lymphocytic choriomeningitis virus M1 virus
- Marburg virus Marburg virus
- lentivirus or retrovirus lentivirus
- the virus for treating hypoxic diseases is selected from one or more of vaccinia virus, adenovirus, herpes simplex virus type I (HSV-1), herpes simplex virus type II (HSV-2), vesicular stomatitis virus, echovirus, reovirus, alphavirus, yellow fever virus, coxsackievirus, Newcastle disease virus, measles virus, poliovirus, Zika virus, lymphocytic choriomeningitis virus, M1 virus, Marburg virus, lentivirus or retrovirus.
- HSV-1 herpes simplex virus type I
- HSV-2 herpes simplex virus type II
- vesicular stomatitis virus echovirus
- reovirus alphavirus
- yellow fever virus coxsackievirus
- Newcastle disease virus measles virus
- poliovirus Zika virus
- lymphocytic choriomeningitis virus M1 virus, Marburg virus, lentivirus or retrovirus.
- the present invention provides use of the system for controlling target gene expression in response to a hypoxic environment according to the present invention, the recombinant virus according to the present invention, or the pharmaceutical composition according to the present invention in the preparation of a medicament for treating hypoxic diseases.
- the hypoxic disease is cancer.
- the cancer is leukemia and/or lymphoma.
- the cancer is a solid tumor, such as one or more of neuroblastoma, lung cancer, breast cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumor (GIST), liver cancer, cervical cancer, ovarian cancer, kidney cancer, pancreatic cancer, nasopharyngeal cancer, small intestine cancer, large intestine cancer, colorectal cancer, bladder cancer, bone cancer, prostate cancer, sarcoma, thyroid cancer or brain cancer.
- GIST gastrointestinal stromal tumor
- the drug is an oncolytic virus drug.
- the drug is an injection.
- the present invention provides a method for treating hypoxic diseases, comprising administering to a subject in need thereof a therapeutically effective amount of a system for controlling expression of a target gene in response to a hypoxic environment according to the present invention and a virus for treating a hypoxic disease, a recombinant virus according to the present invention, or a pharmaceutical composition according to the present invention.
- the hypoxic disease is cancer.
- the cancer is leukemia and/or lymphoma.
- the cancer is a solid tumor, such as one or more of neuroblastoma, lung cancer, breast cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumor (GIST), liver cancer, cervical cancer, ovarian cancer, kidney cancer, pancreatic cancer, nasopharyngeal cancer, small intestine cancer, large intestine cancer, colorectal cancer, bladder cancer, bone cancer, prostate cancer, sarcoma, thyroid cancer or brain cancer.
- GIST gastrointestinal stromal tumor
- the administration is by intratumoral injection and/or intravenous administration.
- the present invention provides a combination of molecular elements that can enable viruses to efficiently express exogenous genes under hypoxic conditions, and the expression is better than that under normoxic conditions, and its preparation method and application.
- the regulatory system comprising an artificial transcription factor that senses hypoxia can be applied to the treatment of hypoxic diseases such as solid tumors, especially oncolytic viruses for the treatment of solid tumors.
- the regulatory system comprising an artificial transcription factor that senses hypoxia consists of a hypoxia response element, a hypoxia-sensitive artificial transcription factor, an artificial transcription factor recognition element, and a target gene expression frame.
- the gene in the plasmid shown in Figure 1a was inserted into the vaccinia virus genome, and the cells were infected for 24 hours under normoxic (21% O 2 ) and hypoxic (1% O 2 ) conditions, or the gene in the plasmid shown in Figure 1b was inserted into the vaccinia virus genome to overexpress the dNLS-HIF-1 ⁇ (WT) gene at the same time. It can be observed that compared with normoxic conditions, the expression of luciferase under hypoxic conditions is downregulated (Figure 1c).
- Figure 3a-g shows the map of the regulatory plasmid of the ATFRE-VTP-GOI model.
- Figure 3a shows the map of the plasmid loaded with 6*Loxp-LTRminiP-Luciferase-mCherry at the VGF2 gene site of vaccinia virus;
- Figure 3b shows the map of the plasmid loaded with 6*Loxp-PE/L-S-Luciferase-mCherry at the VGF2 gene site of vaccinia virus;
- Figure 3c shows the map of the plasmid loaded with 6*Loxp-PE/L-L-Luciferase-mCherry at the VGF2 gene site of vaccinia virus;
- Figure 3d shows the map of the plasmid loaded with 6*Loxp-
- Figure 3e shows a map of a plasmid loaded with 6*Loxp-P7.5-24-Luciferase-mCherry at the vaccinia virus VGF2
- Figure 5a-d shows that HRE-VTP-ATF and ATFRE-VTP-GOI can mediate the virus to specifically express the target gene under hypoxic conditions.
- Figure 5a shows the map of the plasmid loaded with 6*Loxp-P7.5- ⁇ CD47- ⁇ CD3-BiTE at the vaccinia virus C9 gene site
- Figures 5b and 5c show the maps of the plasmid integrated with HRE-PE/L-L-dNLS-CRE-RAP94-ODD at the vaccinia virus C16 and VGF1 gene sites, respectively
- Figure 5d shows that 6*Loxp-P7.5- ⁇ CD47- ⁇ CD3-BiTE and HRE-PE/L-L-dNLS-CRE-RAP94-ODD were inserted into the vaccinia virus genome by recombination technology, and SKOV3 tumor cells were infected under normoxic and hypoxic conditions for 24 hours. It can be observed that the amount of killing factor
- Figure 6a-g shows that HRE-VTP-ATF and ATFRE-VTP-GOI can mediate the specific expression of target genes under hypoxic conditions.
- Figure 6a-d shows the quality of vaccinia virus C9 gene locus loaded with 6*Loxp-P7.5- ⁇ CD47- ⁇ CD3-BiTE, 6*Loxp-P11- ⁇ CD47- ⁇ CD3-BiTE, 6*Loxp-P7.5- ⁇ IGF1R- ⁇ CD3-BiTE and 6*Loxp-P11- ⁇ IGF1R- ⁇ CD3-BiTE
- Figure 6e shows a map of the plasmid integrating HRE-PE/L-L-dNLS-CRE-VP64 at the vaccinia virus C16 gene locus;
- Figures 6f and 6g show the use of recombination technology to integrate 6*Loxp-P7.5- ⁇ CD47- ⁇ CD3-BiTE and HRE-PE/L-L
- the nucleotide sequence shown in SEQ ID NO: 11 (the corresponding amino acid sequence is shown in SEQ ID NO: 12) was synthesized by Shanghai Jierui Biotechnology Co., Ltd. and cloned into the pHAGE expression vector to obtain the pHAGE-dNLS-HIF-1 ⁇ (WT)-P2A-eGFP overexpression plasmid, and its plasmid map is shown in Figure 1b.
- the nucleotide sequences shown in SEQ ID NO: 13-19 were synthesized by Shanghai Jierui Bioengineering Co., Ltd. and cloned into the VGF2 gene recombinant plasmid respectively to obtain the 6*Loxp-PE/L-Luciferase-mCherry recombinant plasmid carrying the nucleotide sequence shown in SEQ ID NO: 13; the 6*Loxp-PE/L-L-Luciferase-mCherry recombinant plasmid carrying the nucleotide sequence shown in SEQ ID NO: 14; the 6*Loxp-PE/L-S-Luciferase-mCherry recombinant plasmid carrying the nucleotide sequence shown in SEQ ID NO: 15; and the 6*Loxp-P 7.5-Luciferase-mCherry recombinant plasmid; 6*Loxp-P7.5-71-Luciferase-
- the nucleotide sequences shown in SEQ ID NO: 21, 23, 25, 27, 29 and 31 were synthesized by Shanghai Jierui Biotechnology Co., Ltd. and cloned into the pHAGE expression vector respectively to obtain the pHAGE-dNLS-CRE-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 21; the pHAGE-dNLS-CRE-VP64-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 23; and the pHAGE-dNLS-CRE-VP64-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 25.
- LS-CRE-VP64-ODD-P2A-eGFP overexpression plasmid LS-CRE-VP64-ODD-P2A-eGFP overexpression plasmid; pHAGE-dNLS-CRE-RAP94-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 27; pHAGE-dNLS-CRE-RAP94-ODD-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 29; and pHAGE-dNLS-CRE-VP160-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 31.
- the above overexpression plasmids all use green fluorescence signals as recombination screening signals, and their plasmid maps are shown in Figures 2a-f.
- the nucleotide sequences shown in SEQ ID NO: 1, 3 and 9 were synthesized by Shanghai Jierui Bioengineering Co., Ltd., spliced together by homologous recombination, and cloned into the VGF-2 gene recombinant plasmid to obtain the pSC65-VGF-2-HRE-PE/L-L-Luciferase-mCherry recombinant plasmid carrying the HRE-PE/L-L-Luciferase gene.
- the recombinant plasmid uses a red fluorescent signal as a recombination screening signal, and its plasmid map is shown in Figure 1a.
- the recombinant plasmid uses a blue fluorescent signal as a recombination screening signal.
- the nucleotide sequences shown in SEQ ID NO: 20, 37 and 33 were synthesized, spliced together by homologous recombination, and cloned into the C9 gene recombination plasmid to obtain the pSC65-C9-6*Loxp-P11- ⁇ CD47- ⁇ CD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P11- ⁇ CD47- ⁇ CD3-BiTE gene (see Figure 6b).
- the recombinant plasmid uses a blue fluorescent signal as a recombination screening signal.
- the nucleotide sequences shown in SEQ ID NOs: 20, 5 and 35 (the amino acid sequence corresponding to the nucleotide sequence shown in SEQ ID NO: 35 is shown in SEQ ID NO: 36) were synthesized by Shanghai Jierui Biotechnology Co., Ltd., spliced together by homologous recombination, and cloned into the C9 gene recombination plasmid to obtain the pSC65-C9-6*Loxp-P7.5- ⁇ IGF1R- ⁇ CD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P7.5- ⁇ IGF1R- ⁇ CD3-BiTE gene.
- nucleotide sequences shown in SEQ ID NOs: 20, 37 and 35 were synthesized, spliced together by homologous recombination, and cloned into the C9 gene recombination plasmid to obtain the pSC65-C9-6*Loxp-P11- ⁇ IGF1R- ⁇ CD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P11- ⁇ IGF1R- ⁇ CD3-BiTE gene (see Figure 6d), which used a blue fluorescence signal as a recombination screening signal.
- the nucleotide sequences shown in SEQ ID NO: 1, 3 and 29 were synthesized by Shanghai Jierui Biotechnology Co., Ltd., spliced together by homologous recombination, and cloned into C16 and VGF gene recombinant plasmids, respectively, to obtain pSC65-C16-HRE-PE/L-L-dNLS-CRE-RAP94-ODD-eGFP recombinant plasmid and pSC65-VGF-HRE-PE/L-L-dNLS-CRE-RAP94-ODD-eGFP recombinant plasmid carrying HRE-PE/L-L-dNLS-CRE-RAP94-ODD gene (see Figure 5b and c), and the above recombinant plasmids used green fluorescence signal as the recombination screening signal; Synthesized SEQ ID NO: The nucleotide sequences shown in 1, 3 and 23 were
- TK143 - cells were plated in a 6-well plate, with about 1 ⁇ 106 cells per well. After culturing for about 24 hours, when the cells adhered to the wall and covered the entire bottom surface, the next step was performed.
- Vaccinia virus incubation Infect cells with the wild-type vaccinia virus Tiantan strain at 0.0125/3 PFU (PFU: plaque forming unit, virus titer)/cell, incubate in a 37°C incubator for 1 hour, remove the cells, aspirate the supernatant, rinse with 1 mL PBS, and then add 1 mL DMEM complete medium (DMEM medium + 10% fetal bovine serum (FBS) + 1% penicillin and streptomycin antibiotics (PS)).
- PFU plaque forming unit, virus titer
- Plasmid transfection Transfect TK143- cells with vaccinia gene recombinant plasmid and culture in a 37°C incubator for about 48 hours, the specific time depends on the cell pathological condition.
- TK143- cells Perform small sample amplification of the recombinant vaccinia virus by plating TK143- cells in a six-well plate, with 1 ⁇ 10 6 cells per well. When used, the cells should occupy approximately 100% of the bottom area of the well plate.
- VERO cell plating Take a 10 cm dish, with about 5 ⁇ 10 6 cells in each dish, and ensure that the cell density reaches 100% when inoculating vaccinia virus the next day;
- MOI virus PFU/cell number
- Collect vaccinia virus discard 8 mL of culture medium in the dish, take 2 mL of DMEM maintenance medium to blow off the remaining cells, and collect them in a 15 mL centrifuge tube;
- TK143 cells were plated in a 24-well plate, with approximately 2 ⁇ 10 5 cells per well. The cell density should reach 100% of the bottom area of the 24-well plate when used;
- DMEM dilute the virus solution, starting from 1:100, and make 10-fold dilutions to a final volume of 1100 ⁇ L.
- Virus plaque counting First, observe whether the number of virus plaques decreases tenfold, then count the number of plaques in the two duplicate wells with only single-digit plaques in the inoculated virus, and get the sum of the plaque values in the two wells. Multiply it by the reciprocal value of the dilution corresponding to the well to get the titer of the virus in 1 mL.
- vaccinia virus infection experiments were conducted in TK143- cells under normoxic and hypoxic conditions to test the expression of target genes by vaccinia viruses in which a hypoxia-sensitive transcriptional expression control system was integrated into the viral genome.
- Figure 1a shows the map of the plasmid carrying HRE-PE/L-L-Luciferase-mCherry at the vaccinia virus VGF2 gene site constructed in Example 1.
- the target gene in the plasmid of Figure 1a was inserted into the vaccinia virus genome using the vaccinia virus recombinant plasmid and the method of Example 2, and verified.
- TK143 - cells were plated into 96-well plates at 1 ⁇ 104 cells/well. The next day, the cells were attached to the wall and divided into two groups. One group was transfected with the overexpression plasmid of pHAGE-dNLS-HIF-1 ⁇ (WT)-P2A-eGFP (200ng per well), and the other group was a blank control group (Blank). 24 hours after plasmid transfection, the cells were infected with the recombinant vaccinia virus obtained in step 1, and the multiplicity of infection (MOI) was 0.02. After infection, the cells were placed in hypoxic conditions (1% O2 concentration) and normoxic conditions (21% O2 concentration) for 24 hours. After the culture, the cells were collected and the expression of luciferase was detected.
- hypoxic conditions 1% O2 concentration
- normoxic conditions 21% O2 concentration
- the luciferase activity value of the target cells was detected by a microplate luminescence detector. According to the results shown in Figure 1c, the hypoxia sensing mode HRE-VTP-GOI can significantly inhibit the expression of the target gene under hypoxic conditions.
- the Blank group and the HIF-1 ⁇ overexpression group had 31.8 and 50.4-fold downregulation, respectively, indicating that overexpression of HIF-1 ⁇ protein further increased the inhibition of target gene expression.
- Example 4 Different ATFs in vaccinia virus can inhibit the expression of the target gene in ATFRE-VTP-GOI
- a vaccinia virus infection experiment under hypoxic conditions was conducted in TK143- cells to test the expression of a target gene by a vaccinia virus in which a hypoxia-sensitive transcriptional expression control system was integrated into the viral genome.
- the plasmid constructed in Example 1 with 6*Loxp-LTRminiP-Luciferase-mCherry at the vaccinia virus VGF2 gene site FIG. 3 a
- the plasmid with 6*Loxp-PE/L-S-Luciferase-mCherry at the vaccinia virus VGF2 gene site FIG. 3 b
- the plasmid with 6*Loxp-PE/L-L-Luciferase-mCherry at the vaccinia virus VGF2 gene site FIG.
- FIG. 3 c the plasmid with 6*Loxp-PE/L-Luciferase-mCherry at the vaccinia virus VGF2 gene site
- FIG. 3 e The target genes were inserted into the vaccinia virus genome by using a plasmid containing erase-mCherry ( FIG. 3d ), a plasmid containing 6*Loxp-P7.5-24-Luciferase-mCherry at the vaccinia virus VGF2 gene site ( FIG. 3e ), a plasmid containing 6*Loxp-P7.5-71-Luciferase-mCherry at the vaccinia virus VGF2 gene site ( FIG. 3f ), and a plasmid containing 6*Loxp-P7.5-Luciferase-mCherry at the vaccinia virus VGF2 gene site ( FIG. 3g ) and the method of Example 2, respectively, and verified.
- TK143 - cells were plated into 96-well plates at 1 ⁇ 104 cells/well. After the cells adhered to the wall the next day, the plasmids shown in Figure 2 were transfected (200 ng per well). 24 hours after plasmid transfection, the recombinant vaccinia virus obtained in step 1 was infected, and the virus infection multiplicity (MOI) was 0.02. After infection, the cells were cultured for another 24 hours. After the culture was completed, the cells were collected and the expression of luciferase was detected.
- MOI virus infection multiplicity
- luciferase activity value of the target cells was detected by a microplate luminescence detector. As shown in Figures 4a and 4b, different types of ATFs under the control of different viral promoters could inhibit the expression of the target gene in the ATFRE-VTP-GOI model, and the reduction factor of luciferase expression inhibited by different viral promoters was between 5-10 times.
- Example 5 The hypoxia-sensitive transcriptional expression regulatory system can mediate the specific expression of the target gene by vaccinia virus under hypoxic conditions at different integration sites
- vaccinia virus infection experiments were conducted in TK143- cells under normoxic and hypoxic conditions to test the expression of target genes by vaccinia viruses with hypoxia-sensitive transcriptional expression regulatory systems at different integration sites of the viral genome.
- TK143 - cells were plated into 12-well plates at 2 ⁇ 10 5 cells/well, and infected after the cells adhered to the wall the next day.
- the virus infection multiplicity (MOI) was 0.02, and the cells were infected with vaccinia virus with HRE-PE/LL-dNLS-CRE-RAP94-ODD and 6*Loxp-P7.5- ⁇ CD47- ⁇ CD3-BiTE genes integrated at the C16 and C9 gene loci; and infected with vaccinia virus with HRE-PE/LL-dNLS-CRE-RAP94-ODD and 6*Loxp-P7.5- ⁇ CD47- ⁇ CD3-BiTE genes integrated at the VGF and C9 gene loci.
- the cells were cultured under hypoxic conditions (1% O 2 concentration) and normoxic conditions (21% O 2 concentration) for 24 hours, and the cell supernatant was collected after the culture to detect the expression of killing factors.
- the tumor cell killing efficiency was evaluated by luciferase-based cytotoxicity assay.
- SKOV3-Luc human ovarian cancer cells modified by firefly luciferase gene
- SKOV3-Luc human ovarian cancer cells modified by firefly luciferase gene
- Cell killing rate (%) (luciferase activity value of virus-infected supernatant in control group - luciferase activity value of experimental group) / luciferase activity value of virus-infected supernatant in control group ⁇ 100
- Example 6 In vitro antitumor effect of oncolytic virus controlled by hypoxia-sensitive transcriptional expression regulatory system
- vaccinia virus infection experiments were conducted in TK143- cells under normoxic and hypoxic conditions to test the expression of target genes by viruses with hypoxia-sensitive transcriptional expression regulatory systems.
- the vaccinia virus was recombined using the pSC65-C16-HRE-PE/L-L-dNLS-CRE-VP64-eGFP recombinant plasmid carrying the HRE-PE/L-L-dNLS-CRE-VP64 gene constructed in Example 1 and the pSC65-C9-6*Loxp-P11- ⁇ IGF1R- ⁇ CD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P11- ⁇ IGF1R- ⁇ CD3-BiTE gene and the method of Example 2, and the results were verified.
- the tumor cell killing efficiency was evaluated by luciferase-based cytotoxicity assay.
- SKOV3-Luc human ovarian cancer cells modified by firefly luciferase gene
- SKOV3-Luc human ovarian cancer cells modified by firefly luciferase gene
- Cell killing rate (%) (luciferase activity value of virus-infected supernatant in control group - luciferase activity value of experimental group) / luciferase activity value of virus-infected supernatant in control group ⁇ 100
- Figure 6f shows vaccinia virus with HRE-PE/L-L-dNLS-CRE-VP64 and 6*Loxp-P7.5- ⁇ CD47- ⁇ CD3-BiTE genes integrated into the C16 and C9 gene loci, and vaccinia virus with HRE-PE/L-L-dNLS-CRE-VP64 and 6*Loxp-P7.5- ⁇ CD47- ⁇ CD3-BiTE genes integrated into the C16 and C9 gene loci.
- the tumor killing activity of vaccinia virus carrying 6*Loxp-P11- ⁇ CD47- ⁇ CD3-BiTE gene was low under normoxic conditions, with killing rates of 18.2% and 13.8%, respectively.
- the vaccinia virus with the 1R- ⁇ CD3-BiTE gene and the vaccinia virus with the HRE-PE/L-L-dNLS-CRE-VP64 and 6*Loxp-P11- ⁇ IGF1R- ⁇ CD3-BiTE genes integrated at the C16 and C9 gene loci had low tumor killing activity under normoxic conditions, with killing rates of 5.3% and 6.2%, respectively. However, they could selectively and efficiently kill SKOV3 tumor cells under hypoxic conditions (killing rates were 21.5% and 18.5%, respectively), that is, the killing activity of specific tumor cell killing under hypoxic conditions was 3.1 times and 4.1 times that under normoxic conditions, respectively.
- Example 7 In vivo anti-tumor effect of oncolytic virus controlled by hypoxia-sensitive transcriptional expression control system
- Hair removal can be done using a depilatory cream or an animal shaver to expose the skin at the site of tumor cell inoculation.
- mice The tumor formation and health status of the mice were observed every 2-3 days after cell inoculation. After tumor formation, the baseline tumor volume was measured with a vernier caliper and subsequent experiments were performed.
- vaccinia virus with hypoxia-sensitive regulatory system i.e., vaccinia virus with HRE-PE/LL-dNLS-CRE-VP64 and 6*Loxp-P7.5- ⁇ CD47- ⁇ CD3-BiTE genes integrated into C16 and C9 sites (TTV-P7.5-347-CRE-VP64), vaccinia virus with HRE-PE/LL-dNLS-CRE-VP64 and 6*Loxp-P11- ⁇ CD47- ⁇ CD3-BiTE genes integrated into C16 and C9 sites (TTV-P11-347-CRE-VP64) and control wild-type vaccinia virus were reinfused intratumorally.
- the tumor size was measured every 2-3 days. The long and short diameters of the tumor were measured with a vernier caliper.
- the tumor volume was calculated as follows:
- the results are shown in Figure 7.
- the vaccinia viruses controlled by the hypoxia-sensitive transcriptional expression control system can effectively inhibit the growth of SKOV3 (ovarian cancer) and NCI-H292 (lung cancer).
- the tumor inhibition rates 66 days after virus injection were as high as 100% and 80%, respectively.
- the survival of mice can be significantly prolonged. All the vaccinia virus mice in the wild-type control group died on the 39th day, while the mice in the TTV-P7.5-347-CRE-VP64 and TTV-P11-347-CRE-VP64 groups were still alive at 60 days.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Genetics & Genomics (AREA)
- Engineering & Computer Science (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Public Health (AREA)
- Wood Science & Technology (AREA)
- General Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Pharmacology & Pharmacy (AREA)
- Biomedical Technology (AREA)
- Medicinal Chemistry (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Physics & Mathematics (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Microbiology (AREA)
- Plant Pathology (AREA)
- Epidemiology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
本发明属于生物医药技术领域,具体涉及一种响应缺氧环境控制目的基因表达的系统及其在制备治疗乏氧性疾病,例如癌症的药物中的用途。The present invention belongs to the field of biomedicine technology, and specifically relates to a system for controlling the expression of a target gene in response to a hypoxic environment and its use in preparing a drug for treating hypoxic diseases, such as cancer.
溶瘤病毒介导的免疫疗法是继三大常规治疗方法(手术、放疗和化疗)以及免疫治疗之后的又一重要的新兴肿瘤治疗策略。其他肿瘤免疫疗法,例如细胞免疫治疗的费用高昂,而且难以解决细胞免疫治疗通用性的问题。与之相比,溶瘤病毒具有杀伤效率高、靶向性好、不良反应小、杀伤肿瘤途径多样、耐药性低以及成本低廉等优势。已有研究显示,溶瘤病毒可以应用于不同类型的肿瘤或者不同进展阶段的肿瘤,甚至可以提高转移性或无法治愈的癌症患者的总生存率。对于晚期癌症病人,溶瘤病毒疗法被认为是挽救生命重要的手段之一,而且溶瘤病毒治疗能够诱导肿瘤完全消退或缓解。目前,溶瘤病毒疗法正受到人们的广泛关注,在肿瘤的治疗方面具有十分广阔的前景。Oncolytic virus-mediated immunotherapy is another important emerging tumor treatment strategy after the three major conventional treatment methods (surgery, radiotherapy and chemotherapy) and immunotherapy. Other tumor immunotherapies, such as cell immunotherapy, are expensive and difficult to solve the problem of the universality of cell immunotherapy. In comparison, oncolytic viruses have the advantages of high killing efficiency, good targeting, small adverse reactions, diverse tumor killing pathways, low drug resistance and low cost. Studies have shown that oncolytic viruses can be applied to different types of tumors or tumors at different stages of progression, and can even improve the overall survival rate of patients with metastatic or incurable cancer. For patients with advanced cancer, oncolytic virus therapy is considered to be one of the important means of saving lives, and oncolytic virus therapy can induce complete regression or remission of tumors. At present, oncolytic virus therapy is receiving widespread attention and has a very broad prospect in the treatment of tumors.
上世纪初,溶瘤病毒疗法开始被引入癌症治疗策略中。发展至今,溶瘤病毒已经由最初不经基因修饰的初代产品,即直接利用野生病毒株或者减毒野生病毒株,比如呼肠孤病毒和新城疫病毒等,发展到基因改造的病毒株的研发阶段。经过基因重组从而能在肿瘤细胞内进行增殖的病毒主要有单纯疱疹病毒、腺病毒、麻疹病毒以及牛痘病毒等,而且已经迭代升级到了第三代全新的强效溶瘤病毒,即基因插入及联合治疗增效阶段,这也是目前临床阶段中溶瘤病毒的主要研究方向。At the beginning of the last century, oncolytic virus therapy began to be introduced into cancer treatment strategies. To date, oncolytic viruses have evolved from the initial generation of products that were not genetically modified, that is, directly using wild virus strains or attenuated wild virus strains, such as reovirus and Newcastle disease virus, to the research and development stage of genetically modified virus strains. The viruses that can proliferate in tumor cells through genetic recombination mainly include herpes simplex virus, adenovirus, measles virus, and vaccinia virus, and have been iteratively upgraded to the third generation of new and powerful oncolytic viruses, that is, the stage of gene insertion and combined therapy enhancement, which is also the main research direction of oncolytic viruses in the current clinical stage.
然而,在近百年的发展过程中,由于溶瘤病毒自身的限制、肿瘤组织的异质性和肿瘤细胞的复杂性,虽然已经有多款溶瘤病毒药物上市,但是治疗效果甚微,而且单独给药的效果也难以到达临床预期,因此其应用开始受到质疑,这极大限制了溶瘤病毒在实体瘤中的临床应用。目前改进的策略是改造溶瘤病毒基因或者其运载方式,使其适应患者机体系统并且特异性激活免疫系统,实现对肿瘤的定向杀伤;或者采用溶瘤病毒与PD-1、CTLA-4等免 疫检查点抑制剂的联用,借助溶瘤病毒打破肿瘤组织微环境,使PD-1、CTLA-4等免疫检查点抑制剂能够更好地发挥作用。However, in the development process of nearly 100 years, due to the limitations of oncolytic viruses themselves, the heterogeneity of tumor tissues and the complexity of tumor cells, although a number of oncolytic virus drugs have been launched, the therapeutic effect is minimal, and the effect of single administration is difficult to reach clinical expectations. Therefore, its application has begun to be questioned, which greatly limits the clinical application of oncolytic viruses in solid tumors. The current improved strategy is to modify the oncolytic virus gene or its delivery method to adapt it to the patient's body system and specifically activate the immune system to achieve targeted killing of tumors; or to use oncolytic viruses in combination with immune checkpoint inhibitors such as PD-1 and CTLA-4, using oncolytic viruses to break the tumor tissue microenvironment, so that immune checkpoint inhibitors such as PD-1 and CTLA-4 can play a better role.
溶瘤病毒疗法面临的巨大挑战不仅在于如何找到高敏感性病毒种类和与之相匹配的适应症,另一个挑战还来自于病毒本身。溶瘤病毒作为身体的异源病原体,无论如何削减病毒活性,都不可避免地会引发机体的免疫反应。需要通过一些安全的调控机制,保障溶瘤病毒最大程度上在肿瘤细胞内复制,而在正常细胞内不复制,在提高肿瘤靶向特异性的同时,其安全性也要得到保障。The huge challenge facing oncolytic virus therapy is not only how to find highly sensitive virus types and matching indications, but another challenge comes from the virus itself. As a heterologous pathogen in the body, oncolytic viruses will inevitably trigger the body's immune response no matter how the viral activity is reduced. Some safe regulatory mechanisms are needed to ensure that oncolytic viruses replicate in tumor cells to the greatest extent possible and do not replicate in normal cells. While improving tumor targeting specificity, their safety must also be guaranteed.
实体瘤相对缺氧的肿瘤微环境是基因治疗的理想靶点,可以通过缺氧信号使得溶瘤病毒仅在肿瘤组织中复制进行肿瘤杀伤,从而提升肿瘤治疗的特异性,避免或减少对正常组织的损伤。已有研究表明,在缺氧条件下,细胞核产生的缺氧诱导因子-1(hypoxia-inducible factor-1,HIF-1)表达水平升高,其与靶基因的结合增加,促进靶基因转录,从而引起细胞的一系列针对缺氧的反应。现已发现的HIF-1的靶基因有60多种,它们都具有缺氧反应元件(hypoxia responsive element,HRE)。HRE由HIF1结合点(共有序列为5’-TACGTGCT-3’)和两侧的功能序列构成。HIF1结合点突变导致基因对缺氧敏感的转录反应丧失。因此,可以利用缺氧反应元件作为启动表达的控制元件。The relatively hypoxic tumor microenvironment of solid tumors is an ideal target for gene therapy. Hypoxia signals can be used to allow oncolytic viruses to replicate only in tumor tissues to kill tumors, thereby improving the specificity of tumor therapy and avoiding or reducing damage to normal tissues. Studies have shown that under hypoxic conditions, the expression level of hypoxia-inducible factor-1 (HIF-1) produced in the cell nucleus increases, and its binding to target genes increases, promoting target gene transcription, thereby causing a series of cellular responses to hypoxia. More than 60 target genes of HIF-1 have been discovered, and they all have hypoxia responsive elements (HRE). HRE consists of the HIF1 binding site (the consensus sequence is 5'-TACGTGCT-3') and functional sequences on both sides. Mutations in the HIF1 binding site result in the loss of the gene's transcriptional response to hypoxia. Therefore, the hypoxia response element can be used as a control element to initiate expression.
发明内容Summary of the invention
本发明的目的在于提供一种响应缺氧环境控制目的基因的转录和表达的系统,该系统通过缺氧敏感的人工转录因子在常氧环境下经诱导而高效表达,在缺氧环境下表达被抑制,保证了该系统的缺氧敏感性;而缺氧环境下诱导的人工转录因子低水平表达解除了对目的基因的抑制,从而启动目的基因的高效转录和表达,实现缺氧环境下目的基因的靶向递送,避免系统性给药引起的严重毒副作用。通过该种转录表达调控系统,可以增强药物的安全性,但不影响其有效性。本发明还提供了该缺氧敏感的转录表达调控系统的应用,其可通过接入不同的目的基因来制备治疗如实体瘤等缺氧性疾病的药物。The object of the present invention is to provide a system for controlling the transcription and expression of a target gene in response to a hypoxic environment. The system is induced by a hypoxia-sensitive artificial transcription factor to be efficiently expressed in a normoxic environment, and the expression is inhibited in a hypoxic environment, thereby ensuring the hypoxia sensitivity of the system; and the low-level expression of the artificial transcription factor induced in the hypoxic environment releases the inhibition of the target gene, thereby starting the efficient transcription and expression of the target gene, achieving targeted delivery of the target gene in a hypoxic environment, and avoiding serious toxic side effects caused by systemic administration. Through this transcription expression regulation system, the safety of the drug can be enhanced without affecting its effectiveness. The present invention also provides the application of the hypoxia-sensitive transcription expression regulation system, which can be used to prepare drugs for treating hypoxic diseases such as solid tumors by accessing different target genes.
一方面,本发明提供一种响应缺氧环境控制目的基因表达的系统,其包含彼此分开的或融合的编码缺氧感应单元的基因和编码目的基因表达单 元的基因,其中所述编码缺氧感应单元的基因包含经由编码病毒转录启动子(virus transcription promoter,VTP)的基因连接的编码缺氧反应元件(hypoxia response element,HRE)的基因和编码人工转录因子(artificial transcription factor,ATF)的基因,以及所述编码目的基因表达单元的基因包含经由编码病毒转录启动子的基因连接的编码人工转录因子识别元件(artificial transcription factor recognition element,ATFRE)的基因和目的基因(gene of interest,GOI)。On the one hand, the present invention provides a system for controlling the expression of a target gene in response to a hypoxic environment, which comprises a gene encoding a hypoxia sensing unit and a gene encoding a target gene expression unit, which are separated from or fused to each other, wherein the gene encoding the hypoxia sensing unit comprises a gene encoding a hypoxia response element (HRE) and a gene encoding an artificial transcription factor (ATF) connected via a gene encoding a virus transcription promoter (VTP), and the gene encoding the target gene expression unit comprises a gene encoding an artificial transcription factor recognition element (ATFRE) and a target gene (gene of interest, GOI) connected via a gene encoding a virus transcription promoter.
在本发明中,所述缺氧环境是指氧含量低于2%的细胞或组织。肿瘤组织和细胞对氧和葡萄糖等能量物质的需求量很大,随着肿瘤组织供血不足随之出现肿瘤细胞和组织的缺氧,其细胞和组织氧含量往往低于2%。In the present invention, the hypoxic environment refers to cells or tissues with an oxygen content of less than 2%. Tumor tissues and cells have a great demand for energy substances such as oxygen and glucose. As the blood supply to tumor tissues is insufficient, hypoxia of tumor cells and tissues occurs, and the oxygen content of cells and tissues is often less than 2%.
根据本发明的一些实施方式,所述编码VTP的基因与编码HRE的基因之间、以及所述编码VTP的基因与编码ATF的基因之间包含编码接头的基因。编码接头的基因的核苷酸序列为随机序列,一般为10-20bp大小。According to some embodiments of the present invention, a gene encoding a joint is included between the gene encoding VTP and the gene encoding HRE, and between the gene encoding VTP and the gene encoding ATF. The nucleotide sequence of the gene encoding the joint is a random sequence, generally 10-20 bp in size.
根据本发明的一些实施方式,所述编码VTP的基因与编码ATFRE的基因之间、以及所述编码VTP的基因与GOI之间包含编码接头的基因。编码接头的基因的核苷酸序列为随机序列,一般为10-20bp大小。According to some embodiments of the present invention, a gene encoding a linker is included between the gene encoding VTP and the gene encoding ATFRE, and between the gene encoding VTP and GOI. The nucleotide sequence of the gene encoding the linker is a random sequence, generally 10-20 bp in size.
根据本发明的一些实施方式,所述编码HRE的基因的核苷酸序列如SEQ ID NO:1所示,或者为多于1个、优选3个、更优选5个如SEQ ID NO:1所示的核苷酸序列的重复核苷酸序列。According to some embodiments of the present invention, the nucleotide sequence of the gene encoding HRE is as shown in SEQ ID NO: 1, or is a repeated nucleotide sequence of more than 1, preferably 3, and more preferably 5 nucleotide sequences as shown in SEQ ID NO: 1.
根据本发明的一些实施方式,所述VTP选自PE/L、截短型PE/L(PE/L-L)、再次截短型PE/L(PE/L-S)、P7.5、截短型P7.5(P7.5-71)、再次截短型P7.5(P7.5-24)、LTRminiP或P11中的一种或多种。According to some embodiments of the present invention, the VTP is selected from one or more of PE/L, truncated PE/L (PE/L-L), further truncated PE/L (PE/L-S), P7.5, truncated P7.5 (P7.5-71), further truncated P7.5 (P7.5-24), LTRminiP or P11.
根据本发明的一些具体实施方式,所述PE/L的核苷酸序列如SEQ ID NO:2所示;所述PE/L-L的核苷酸序列如SEQ ID NO:3所示;所述PE/L-S的核苷酸序列如SEQ ID NO:4所示;所述P7.5的核苷酸序列如SEQ ID NO:5所示;所述P7.5-71的核苷酸序列如SEQ ID NO:6所示;所述P7.5-24的核苷酸序列如SEQ ID NO:7所示;所述LTRminiP的核苷酸序列如SEQ ID NO:8所示;所述P11的核苷酸序列如SEQ ID NO:37所示。According to some specific embodiments of the present invention, the nucleotide sequence of the PE/L is as shown in SEQ ID NO: 2; the nucleotide sequence of the PE/L-L is as shown in SEQ ID NO: 3; the nucleotide sequence of the PE/L-S is as shown in SEQ ID NO: 4; the nucleotide sequence of the P7.5 is as shown in SEQ ID NO: 5; the nucleotide sequence of the P7.5-71 is as shown in SEQ ID NO: 6; the nucleotide sequence of the P7.5-24 is as shown in SEQ ID NO: 7; the nucleotide sequence of the LTRminiP is as shown in SEQ ID NO: 8; the nucleotide sequence of the P11 is as shown in SEQ ID NO: 37.
根据本发明的一些实施方式,所述编码ATF的基因选自去核定位信号(dNLS)的CRE基因、去核定位信号的CRE-VP64基因、去核定位信号的CRE-VP64-ODD基因、去核定位信号的CRE-RAP94基因、去核定位信 号的CRE-RAP94-ODD基因或去核定位信号的CRE-VP160基因中的一种或多种,优选为去核定位信号的CRE-VP64基因。According to some embodiments of the present invention, the gene encoding ATF is selected from one or more of the CRE gene with a denuclearized localization signal (dNLS), the CRE-VP64 gene with a denuclearized localization signal, the CRE-VP64-ODD gene with a denuclearized localization signal, the CRE-RAP94 gene with a denuclearized localization signal, the CRE-RAP94-ODD gene with a denuclearized localization signal, or the CRE-VP160 gene with a denuclearized localization signal, and is preferably the CRE-VP64 gene with a denuclearized localization signal.
根据本发明的一些具体实施方式,所述去核定位信号的CRE基因的核苷酸序列如SEQ ID NO:21所示(对应的氨基酸序列如SEQ ID NO:22所示);所述去核定位信号的CRE-VP64基因的核苷酸序列如SEQ ID NO:23所示(对应的氨基酸序列如SEQ ID NO:24所示);所述去核定位信号的CRE-VP64-ODD基因的核苷酸序列如SEQ ID NO:25所示(对应的氨基酸序列如SEQ ID NO:26所示);所述去核定位信号的CRE-RAP94基因的核苷酸序列如SEQ ID NO:27所示(对应的氨基酸序列如SEQ ID NO:28所示);所述去核定位信号的CRE-RAP94-ODD基因的核苷酸序列如SEQ ID NO:29所示(对应的氨基酸序列如SEQ ID NO:30所示);所述去核定位信号的CRE-VP160基因的核苷酸序列如SEQ ID NO:31所示(对应的氨基酸序列如SEQ ID NO:32所示)。According to some specific embodiments of the present invention, the nucleotide sequence of the CRE gene with the denuclearization localization signal is shown in SEQ ID NO: 21 (the corresponding amino acid sequence is shown in SEQ ID NO: 22); the nucleotide sequence of the CRE-VP64 gene with the denuclearization localization signal is shown in SEQ ID NO: 23 (the corresponding amino acid sequence is shown in SEQ ID NO: 24); the nucleotide sequence of the CRE-VP64-ODD gene with the denuclearization localization signal is shown in SEQ ID NO: 25 (the corresponding amino acid sequence is shown in SEQ ID NO: 26 The nucleotide sequence of the CRE-RAP94 gene with the nuclear localization signal is shown in SEQ ID NO: 27 (the corresponding amino acid sequence is shown in SEQ ID NO: 28); the nucleotide sequence of the CRE-RAP94-ODD gene with the nuclear localization signal is shown in SEQ ID NO: 29 (the corresponding amino acid sequence is shown in SEQ ID NO: 30); the nucleotide sequence of the CRE-VP160 gene with the nuclear localization signal is shown in SEQ ID NO: 31 (the corresponding amino acid sequence is shown in SEQ ID NO: 32).
根据本发明的一些实施方式,所述编码ATFRE的基因为一个或多个Loxp基因。According to some embodiments of the present invention, the gene encoding ATFRE is one or more Loxp genes.
根据本发明的一些具体实施方式,所述编码ATFRE的基因的核苷酸序列为多于1个、优选3个、更优选5个、进一步优选6个Loxp基因的重复基因序列,例如SEQ ID NO:20所示的核苷酸序列。According to some specific embodiments of the present invention, the nucleotide sequence of the gene encoding ATFRE is a repeated gene sequence of more than 1, preferably 3, more preferably 5, and further preferably 6 Loxp genes, such as the nucleotide sequence shown in SEQ ID NO: 20.
根据本发明的一些实施方式,所述GOI选自共刺激分子、细胞因子、负调分子及信号通路的阻断抗体、趋化因子或杀伤分子中的一种或多种,优选为杀伤分子,更优选为BiTE的编码基因中的一种或多种。According to some embodiments of the present invention, the GOI is selected from one or more of co-stimulatory molecules, cytokines, negative regulatory molecules, blocking antibodies of signal pathways, chemokines or killer molecules, preferably killer molecules, and more preferably one or more of the encoding genes of BiTE.
根据本发明的一些具体实施方式,所述GOI为αCD47-αCD3-BiTE和/或αIGF1R-αCD3-BiTE。According to some specific embodiments of the present invention, the GOI is αCD47-αCD3-BiTE and/or αIGF1R-αCD3-BiTE.
根据本发明的一些优选实施方式,所述缺氧感应单元的组成为5*SEQ ID NO:1-编码VTP的基因-dNLS-CRE-VP64基因,和/或所述目的基因表达单元的组成为SEQ ID NO:20-编码VTP的基因-GOI。更优选地,所述缺氧感应单元的组成为5*SEQ ID NO:1-编码PE/L的基因-dNLS-CRE-VP64基因,和/或所述目的基因表达单元的组成为SEQ ID NO:20-编码P7.5的基因-GOI。更优选地,所述缺氧感应单元的组成为5*SEQ ID NO:1-编码PE/L的基因-dNLS-CRE-VP64基因,和/或所述目的基因表达单元为SEQ ID NO:20-编码P11的基因-GOI。According to some preferred embodiments of the present invention, the composition of the hypoxia sensing unit is 5*SEQ ID NO: 1-gene encoding VTP-dNLS-CRE-VP64 gene, and/or the composition of the target gene expression unit is SEQ ID NO: 20-gene encoding VTP-GOI. More preferably, the composition of the hypoxia sensing unit is 5*SEQ ID NO: 1-gene encoding PE/L-dNLS-CRE-VP64 gene, and/or the composition of the target gene expression unit is SEQ ID NO: 20-gene encoding P7.5-GOI. More preferably, the composition of the hypoxia sensing unit is 5*SEQ ID NO: 1-gene encoding PE/L-dNLS-CRE-VP64 gene, and/or the composition of the target gene expression unit is SEQ ID NO: 20-gene encoding P11-GOI.
根据本发明的一些实施方式,根据本发明的响应缺氧环境控制目的基因表达的系统包含:According to some embodiments of the present invention, the system for controlling the expression of a target gene in response to a hypoxic environment according to the present invention comprises:
包含所述编码缺氧感应单元的基因的第一载体和包含所述编码目的基因表达单元的基因的第二载体。A first vector comprising the gene encoding the hypoxia sensing unit and a second vector comprising the gene encoding the target gene expression unit.
根据本发明的一些实施方式,根据本发明的响应缺氧环境控制目的基因表达的系统包含一种载体,所述载体包含融合的所述编码缺氧感应单元的基因和所述编码目的基因表达单元的基因。According to some embodiments of the present invention, the system for controlling the expression of a target gene in response to a hypoxic environment comprises a vector comprising a fused gene encoding the hypoxia sensing unit and the gene encoding the target gene expression unit.
根据本发明的一些实施方式,所述载体选自pUC18、pUC19、pUC57、pcDNA3、pcDNA4、pcDNA5、pcDNA6、pCMV、pEF1、PEGFP、pET、pEasy、pAc5、pAcGP、pAcYCDuet、pBluescript、pBudCE、pCAMBIA、pCold、pCR2、pCR3、pCR4、pDsRED、pGEM、pGL、pIRES、pPIC、pMAL或pVRCSV1.0等中的一种或多种。According to some embodiments of the present invention, the vector is selected from one or more of pUC18, pUC19, pUC57, pcDNA3, pcDNA4, pcDNA5, pcDNA6, pCMV, pEF1, PEGFP, pET, pEasy, pAc5, pAcGP, pAcYCDuet, pBluescript, pBudCE, pCAMBIA, pCold, pCR2, pCR3, pCR4, pDsRED, pGEM, pGL, pIRES, pPIC, pMAL or pVRCSV1.0, etc.
另一方面,本发明提供一种重组病毒,其基因组包含根据本发明的响应缺氧环境控制目的基因表达的系统。In another aspect, the present invention provides a recombinant virus, the genome of which comprises the system for controlling the expression of a target gene in response to a hypoxic environment according to the present invention.
根据本发明的一些实施方式,所述病毒选自痘苗病毒、腺病毒、I型单纯疱疹病毒(HSV-1)、II型单纯疱疹病毒(HSV-2)、水疱性口炎病毒、埃可病毒、呼肠孤病毒、甲病毒、黄热病毒、柯萨奇病毒、新城疫病毒、麻疹病毒、脊髓灰质炎病毒、寨卡病毒、淋巴细胞脉络丛脑膜炎病毒、M1病毒、马尔堡病毒、慢病毒或逆转录病毒中的一种或多种。According to some embodiments of the present invention, the virus is selected from one or more of vaccinia virus, adenovirus, herpes simplex virus type I (HSV-1), herpes simplex virus type II (HSV-2), vesicular stomatitis virus, echovirus, reovirus, alphavirus, yellow fever virus, coxsackievirus, Newcastle disease virus, measles virus, poliovirus, Zika virus, lymphocytic choriomeningitis virus, M1 virus, Marburg virus, lentivirus or retrovirus.
再一方面,本发明提供一种用于治疗乏氧性疾病的药物组合物,其包含根据本发明的响应缺氧环境控制目的基因表达的系统和用于治疗乏氧性疾病的病毒、或者根据本发明的重组病毒,以及任选的药学上可接受的辅料。In another aspect, the present invention provides a pharmaceutical composition for treating hypoxic diseases, comprising a system for controlling the expression of a target gene in response to a hypoxic environment according to the present invention and a virus for treating a hypoxic disease, or a recombinant virus according to the present invention, and optional pharmaceutically acceptable excipients.
根据本发明的一些实施方式,所述治疗乏氧性疾病的病毒选自痘苗病毒、腺病毒、I型单纯疱疹病毒(HSV-1)、II型单纯疱疹病毒(HSV-2)、水疱性口炎病毒、埃可病毒、呼肠孤病毒、甲病毒、黄热病毒、柯萨奇病毒、新城疫病毒、麻疹病毒、脊髓灰质炎病毒、寨卡病毒、淋巴细胞脉络丛脑膜炎病毒、M1病毒、马尔堡病毒、慢病毒或逆转录病毒中的一种或多种。According to some embodiments of the present invention, the virus for treating hypoxic diseases is selected from one or more of vaccinia virus, adenovirus, herpes simplex virus type I (HSV-1), herpes simplex virus type II (HSV-2), vesicular stomatitis virus, echovirus, reovirus, alphavirus, yellow fever virus, coxsackievirus, Newcastle disease virus, measles virus, poliovirus, Zika virus, lymphocytic choriomeningitis virus, M1 virus, Marburg virus, lentivirus or retrovirus.
根据本发明的一些实施方式,所述药学上可接受的辅料选自磷酸钠、磷酸氢二钠二水合物、磷酸二氢钠二水合物、氯化钠、山梨糖醇、肌醇、 质量比为0.001%的泊洛沙姆188水溶液、质量比为0.005%的泊洛沙姆188水溶液、三羟甲基氨基甲烷(Tris)、氯化镁或注射用水中的一种或多种。According to some embodiments of the present invention, the pharmaceutically acceptable excipient is selected from one or more of sodium phosphate, disodium hydrogen phosphate dihydrate, sodium dihydrogen phosphate dihydrate, sodium chloride, sorbitol, inositol, a 0.001% by mass aqueous solution of poloxamer 188, a 0.005% by mass aqueous solution of poloxamer 188, tris(hydroxymethyl)aminomethane (Tris), magnesium chloride or water for injection.
根据本发明的一些实施方式,所述药物组合物还包括其他治疗乏氧性疾病的药物活性成分,例如PD-1免疫检查点抑制剂和/或CTLA-4免疫检查点抑制剂。According to some embodiments of the present invention, the pharmaceutical composition further comprises other active pharmaceutical ingredients for treating hypoxic diseases, such as PD-1 immune checkpoint inhibitors and/or CTLA-4 immune checkpoint inhibitors.
另一方面,本发明提供根据本发明的响应缺氧环境控制目的基因表达的系统、根据本发明的重组病毒或根据本发明的药物组合物在制备治疗乏氧性疾病的药物中的用途。In another aspect, the present invention provides use of the system for controlling target gene expression in response to a hypoxic environment according to the present invention, the recombinant virus according to the present invention, or the pharmaceutical composition according to the present invention in the preparation of a medicament for treating hypoxic diseases.
根据本发明的一些实施方式,所述乏氧性疾病为癌症。优选地,所述癌症为白血病和/或淋巴瘤。优选地,所述癌症为实体瘤,例如神经母细胞瘤、肺癌、乳腺癌、食管癌、胃癌、胃肠道间质瘤(GIST)、肝癌、子宫颈癌、卵巢癌、肾癌、胰腺癌、鼻咽癌、小肠癌、大肠癌、结直肠癌、膀胱癌、骨癌、前列腺癌、肉瘤、甲状腺癌或脑癌中的一种或多种。According to some embodiments of the present invention, the hypoxic disease is cancer. Preferably, the cancer is leukemia and/or lymphoma. Preferably, the cancer is a solid tumor, such as one or more of neuroblastoma, lung cancer, breast cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumor (GIST), liver cancer, cervical cancer, ovarian cancer, kidney cancer, pancreatic cancer, nasopharyngeal cancer, small intestine cancer, large intestine cancer, colorectal cancer, bladder cancer, bone cancer, prostate cancer, sarcoma, thyroid cancer or brain cancer.
根据本发明的一些实施方式,所述药物为溶瘤病毒药物。According to some embodiments of the present invention, the drug is an oncolytic virus drug.
根据本发明的一些实施方式,所述药物为注射剂。According to some embodiments of the present invention, the drug is an injection.
相应地,本发明提供一种治疗乏氧性疾病的方法,其包括向有需要的对象给予治疗有效量的根据本发明的响应缺氧环境控制目的基因表达的系统和用于治疗乏氧性疾病的病毒、根据本发明的重组病毒或根据本发明的药物组合物。Accordingly, the present invention provides a method for treating hypoxic diseases, comprising administering to a subject in need thereof a therapeutically effective amount of a system for controlling expression of a target gene in response to a hypoxic environment according to the present invention and a virus for treating a hypoxic disease, a recombinant virus according to the present invention, or a pharmaceutical composition according to the present invention.
根据本发明的一些实施方式,所述乏氧性疾病为癌症。优选地,所述癌症为白血病和/或淋巴瘤。优选地,所述癌症为实体瘤,例如神经母细胞瘤、肺癌、乳腺癌、食管癌、胃癌、胃肠道间质瘤(GIST)、肝癌、子宫颈癌、卵巢癌、肾癌、胰腺癌、鼻咽癌、小肠癌、大肠癌、结直肠癌、膀胱癌、骨癌、前列腺癌、肉瘤、甲状腺癌或脑癌中的一种或多种。According to some embodiments of the present invention, the hypoxic disease is cancer. Preferably, the cancer is leukemia and/or lymphoma. Preferably, the cancer is a solid tumor, such as one or more of neuroblastoma, lung cancer, breast cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumor (GIST), liver cancer, cervical cancer, ovarian cancer, kidney cancer, pancreatic cancer, nasopharyngeal cancer, small intestine cancer, large intestine cancer, colorectal cancer, bladder cancer, bone cancer, prostate cancer, sarcoma, thyroid cancer or brain cancer.
根据本发明的一些实施方式,所述给予采用瘤内注射和/或静脉给药方式。According to some embodiments of the present invention, the administration is by intratumoral injection and/or intravenous administration.
本发明提供一种能够使病毒在缺氧条件下高效表达外源基因,且表达优于常氧条件的分子元件组合及其制备方法与应用。包含感应缺氧环境的人工转录因子的调控系统可应用在实体瘤等缺氧性疾病的治疗,特别是用于实体瘤治疗的溶瘤病毒。包含感应缺氧的人工转录因子的调控系统由缺氧反 应元件、缺氧敏感人工转录因子、人工转录因子识别元件和目的基因表达框组成,该系统具有以下三大优势:1)感应缺氧的人工转录因子可在常氧环境下诱导高效表达,而在缺氧环境下表达抑制,保证了该系统的缺氧敏感性;2)缺氧环境下诱导人工转录因子低水平表达,解除对目的基因的抑制,从而启动目的基因高效转录和表达,从而实现缺氧环境的靶向递送,避免系统性给药引起的严重毒副作用;3)目的基因编码的蛋白并未作任何的缺氧敏感功能域修饰,可以确保目的基因编码的蛋白功能不受干扰,发挥出其最大的活性。The present invention provides a combination of molecular elements that can enable viruses to efficiently express exogenous genes under hypoxic conditions, and the expression is better than that under normoxic conditions, and its preparation method and application. The regulatory system comprising an artificial transcription factor that senses hypoxia can be applied to the treatment of hypoxic diseases such as solid tumors, especially oncolytic viruses for the treatment of solid tumors. The regulatory system comprising an artificial transcription factor that senses hypoxia consists of a hypoxia response element, a hypoxia-sensitive artificial transcription factor, an artificial transcription factor recognition element, and a target gene expression frame. The system has the following three advantages: 1) The artificial transcription factor that senses hypoxia can induce efficient expression in a normoxic environment, and inhibit expression in a hypoxic environment, thereby ensuring the hypoxia sensitivity of the system; 2) Inducing low-level expression of artificial transcription factors in a hypoxic environment, relieving the inhibition of the target gene, thereby starting the efficient transcription and expression of the target gene, thereby achieving targeted delivery in a hypoxic environment, and avoiding serious toxic side effects caused by systemic administration; 3) The protein encoded by the target gene has not been modified by any hypoxia-sensitive functional domain, which can ensure that the protein function encoded by the target gene is not disturbed and exerts its maximum activity.
应理解,在本发明范围内,本发明的上述各项技术特征和在下文(如实施例)中具体描述的各项技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅限制,在此不再一一赘述。It should be understood that within the scope of the present invention, the above-mentioned various technical features of the present invention and the various technical features specifically described below (such as embodiments) can be combined with each other to form a new or preferred technical solution. Due to space limitations, they will not be described one by one here.
附图的简要说明BRIEF DESCRIPTION OF THE DRAWINGS
以下,结合附图来详细说明本发明的实施方案,其中:The embodiments of the present invention are described in detail below with reference to the accompanying drawings, wherein:
图1a-c显示HRE-VTP-GOI模式的缺氧调控作用。图1a为在痘苗病毒的VGF2基因位点装载HRE-PE/L-Luciferase-mCherry的质粒的图谱;图1b为装载dNLS-HIF-1α(WT)-P2A-eGFP的过表达基因载体pHGAE质粒的图谱。利用重组技术,将图1a所示质粒中的基因插入痘苗病毒基因组中,在常氧(21%O 2)和缺氧(1%O 2)条件下感染细胞24h,或者同时将图1b所示质粒中的基因插入痘苗病毒基因组中过表达dNLS-HIF-1α(WT)基因,可观察到相比于常氧条件,缺氧条件下的荧光素酶表达都下调(图1c)。 Figures 1a-c show the hypoxic regulation of the HRE-VTP-GOI model. Figure 1a is a map of the plasmid loaded with HRE-PE/L-Luciferase-mCherry at the VGF2 gene site of vaccinia virus; Figure 1b is a map of the overexpression gene vector pHGAE plasmid loaded with dNLS-HIF-1α (WT)-P2A-eGFP. Using recombinant technology, the gene in the plasmid shown in Figure 1a was inserted into the vaccinia virus genome, and the cells were infected for 24 hours under normoxic (21% O 2 ) and hypoxic (1% O 2 ) conditions, or the gene in the plasmid shown in Figure 1b was inserted into the vaccinia virus genome to overexpress the dNLS-HIF-1α (WT) gene at the same time. It can be observed that compared with normoxic conditions, the expression of luciferase under hypoxic conditions is downregulated (Figure 1c).
图2a-f显示具有不同种ATF基因的HRE-VTP-ATF模式的质粒的图谱。图2a显示过表达去核定位信号的CRE基因的质粒(pHAGE-dNLS-CRE-P2A-eGFP)的图谱;图2b显示过表达去核定位信号的CRE-VP64基因的质粒(pHAGE-dNLS-CRE-VP64-P2A-eGFP)的图谱;图2c显示过表达去核定位信号的CRE-VP64-ODD基因的质粒(pHAGE-dNLS-CRE-VP64-ODD-P2A-eGFP)的图谱;图2d显示过表达去核定位信号的CRE-RAP94基因的质粒(pHAGE-dNLS-CRE-RAP94-P2A-eGFP)的图谱;图2e显示过表达去核定位信号的CRE-RAP94-ODD基因的质粒(pHAGE-dNLS-CRE-RAP94-ODD-P2A-eGFP)的图谱;图2f显示过表达 去核定位信号的CRE-VP160基因的质粒(pHAGE-dNLS-CRE-VP160-P2A-eGFP)的图谱。Figure 2a-f shows maps of plasmids with HRE-VTP-ATF patterns of different ATF genes. Figure 2a shows a map of a plasmid (pHAGE-dNLS-CRE-P2A-eGFP) overexpressing a CRE gene with a denuclearization localization signal; Figure 2b shows a map of a plasmid (pHAGE-dNLS-CRE-VP64-P2A-eGFP) overexpressing a CRE-VP64 gene with a denuclearization localization signal; Figure 2c shows a map of a plasmid (pHAGE-dNLS-CRE-VP64-ODD-P2A-eGFP) overexpressing a CRE-VP64-ODD gene with a denuclearization localization signal; Figure 2d shows a map of a plasmid (pHAGE-dNLS-CRE-RAP94-P2A-eGFP) overexpressing a CRE-RAP94 gene with a denuclearization localization signal; Figure 2e shows a map of a plasmid (pHAGE-dNLS-CRE-RAP94-ODD-P2A-eGFP) overexpressing a CRE-RAP94-ODD gene with a denuclearization localization signal; Figure 2f shows a map of a plasmid (pHAGE-dNLS-CRE-RAP94-ODD-P2A-eGFP) overexpressing a CRE-RAP94-ODD gene with a denuclearization localization signal; Map of the plasmid containing the CRE-VP160 gene without the nuclear localization signal (pHAGE-dNLS-CRE-VP160-P2A-eGFP).
图3a-g显示ATFRE-VTP-GOI模式的调控质粒的图谱。图3a显示在痘苗病毒VGF2基因位点装载6*Loxp-LTRminiP-Luciferase-mCherry的质粒的图谱;图3b显示在痘苗病毒VGF2基因位点装载6*Loxp-PE/L-S-Luciferase-mCherry的质粒的图谱;图3c显示在痘苗病毒VGF2基因位点装载6*Loxp-PE/L-L-Luciferase-mCherry的质粒的图谱;图3d显示在痘苗病毒VGF2基因位点装载6*Loxp-PE/L-Luciferase-mCherry的质粒的图谱;图3e显示在痘苗病毒VGF2基因位点装载6*Loxp-P7.5-24-Luciferase-mCherry的质粒的图谱;图3f显示在痘苗病毒VGF2基因位点装载6*Loxp-P7.5-71-Luciferase-mCherry的质粒的图谱;图3g显示在痘苗病毒VGF2基因位点装载6*Loxp-P7.5-Luciferase-mCherry的质粒的图谱。Figure 3a-g shows the map of the regulatory plasmid of the ATFRE-VTP-GOI model. Figure 3a shows the map of the plasmid loaded with 6*Loxp-LTRminiP-Luciferase-mCherry at the VGF2 gene site of vaccinia virus; Figure 3b shows the map of the plasmid loaded with 6*Loxp-PE/L-S-Luciferase-mCherry at the VGF2 gene site of vaccinia virus; Figure 3c shows the map of the plasmid loaded with 6*Loxp-PE/L-L-Luciferase-mCherry at the VGF2 gene site of vaccinia virus; Figure 3d shows the map of the plasmid loaded with 6*Loxp- Figure 3e shows a map of a plasmid loaded with 6*Loxp-P7.5-24-Luciferase-mCherry at the vaccinia virus VGF2 gene site; Figure 3f shows a map of a plasmid loaded with 6*Loxp-P7.5-71-Luciferase-mCherry at the vaccinia virus VGF2 gene site; Figure 3g shows a map of a plasmid loaded with 6*Loxp-P7.5-Luciferase-mCherry at the vaccinia virus VGF2 gene site.
图4a和b显示利用ATFRE-VTP-GOI模式,加载ATF可抑制目的基因的表达。利用重组技术将图3所显示的质粒中的基因分别插入痘苗病毒基因组中,再将图2所显示的质粒中的ATF基因进行过表达,可观察到荧光素酶表达下调,其中Blank代表没有转染图2所示质粒的空白对照。Figures 4a and b show that loading ATF can inhibit the expression of the target gene using the ATFRE-VTP-GOI model. The genes in the plasmid shown in Figure 3 were inserted into the vaccinia virus genome using recombination technology, and then the ATF gene in the plasmid shown in Figure 2 was overexpressed, and downregulation of luciferase expression was observed, where Blank represents a blank control without transfection of the plasmid shown in Figure 2.
图5a-d显示HRE-VTP-ATF和ATFRE-VTP-GOI可介导病毒在缺氧条件下特异性表达目的基因。其中,图5a显示在痘苗病毒C9基因位点装载6*Loxp-P7.5-αCD47-αCD3-BiTE的质粒的图谱;图5b和5c分别显示在痘苗病毒C16和VGF1基因位点整合HRE-PE/L-L-dNLS-CRE-RAP94-ODD的质粒的图谱;图5d显示利用重组技术,将6*Loxp-P7.5-αCD47-αCD3-BiTE和HRE-PE/L-L-dNLS-CRE-RAP94-ODD插入痘苗病毒基因组中,在常氧和缺氧条件下感染SKOV3肿瘤细胞24h,可观察到在缺氧条件下病毒释放杀伤因子αCD47-αCD3-BiTE的量要显著高于在常氧条件下释放的量,即在缺氧环境下选择性杀伤SKOV3肿瘤细胞。Figure 5a-d shows that HRE-VTP-ATF and ATFRE-VTP-GOI can mediate the virus to specifically express the target gene under hypoxic conditions. Among them, Figure 5a shows the map of the plasmid loaded with 6*Loxp-P7.5-αCD47-αCD3-BiTE at the vaccinia virus C9 gene site; Figures 5b and 5c show the maps of the plasmid integrated with HRE-PE/L-L-dNLS-CRE-RAP94-ODD at the vaccinia virus C16 and VGF1 gene sites, respectively; Figure 5d shows that 6*Loxp-P7.5-αCD47-αCD3-BiTE and HRE-PE/L-L-dNLS-CRE-RAP94-ODD were inserted into the vaccinia virus genome by recombination technology, and SKOV3 tumor cells were infected under normoxic and hypoxic conditions for 24 hours. It can be observed that the amount of killing factor αCD47-αCD3-BiTE released by the virus under hypoxic conditions is significantly higher than the amount released under normoxic conditions, that is, SKOV3 tumor cells are selectively killed under hypoxic environment.
图6a-g显示HRE-VTP-ATF和ATFRE-VTP-GOI可介导病毒在缺氧条件下特异性表达目的基因。其中,图6a-d显示在痘苗病毒C9基因位点装载6*Loxp-P7.5-αCD47-αCD3-BiTE、6*Loxp-P11-αCD47-αCD3-BiTE、6*Loxp-P7.5-αIGF1R-αCD3-BiTE和6*Loxp-P11-αIGF1R-αCD3-BiTE的质 粒的图谱;图6e显示在痘苗病毒C16基因位点整合HRE-PE/L-L-dNLS-CRE-VP64的质粒的图谱;图6f和6g显示利用重组技术,分别将6*Loxp-P7.5-αCD47-αCD3-BiTE和HRE-PE/L-L-dNLS-CRE-VP64、6*Loxp-P11-αCD47-αCD3-BiTE和HRE-PE/L-L-dNLS-CRE-VP64、6*Loxp-P7.5-αIGF1R-αCD3-BiTE和HRE-PE/L-L-dNLS-CRE-VP64、以及6*Loxp-P11-αIGF1R-αCD3-BiTE和HRE-PE/L-L-dNLS-CRE-VP64装入痘苗病毒基因组中,在常氧和缺氧条件下感染SKOV3细胞24h,可观察到在缺氧条件下病毒释放杀伤因子αCD47-αCD3-BiTE或者αIGF1R-αCD3-BiTE的量要显著高于在常氧条件下释放的量,即在缺氧环境下选择性杀伤SKOV3肿瘤细胞。Figure 6a-g shows that HRE-VTP-ATF and ATFRE-VTP-GOI can mediate the specific expression of target genes under hypoxic conditions. Among them, Figure 6a-d shows the quality of vaccinia virus C9 gene locus loaded with 6*Loxp-P7.5-αCD47-αCD3-BiTE, 6*Loxp-P11-αCD47-αCD3-BiTE, 6*Loxp-P7.5-αIGF1R-αCD3-BiTE and 6*Loxp-P11-αIGF1R-αCD3-BiTE Figure 6e shows a map of the plasmid integrating HRE-PE/L-L-dNLS-CRE-VP64 at the vaccinia virus C16 gene locus; Figures 6f and 6g show the use of recombination technology to integrate 6*Loxp-P7.5-αCD47-αCD3-BiTE and HRE-PE/L-L-dNLS-CRE-VP64, 6*Loxp-P11-αCD47-αCD3-BiTE and HRE-PE/L-L-dNLS-CRE-VP64, 6*Loxp-P7.5-αIGF1R-αCD3-BiTE, respectively. E and HRE-PE/L-L-dNLS-CRE-VP64, as well as 6*Loxp-P11-αIGF1R-αCD3-BiTE and HRE-PE/L-L-dNLS-CRE-VP64 were loaded into the vaccinia virus genome, and SKOV3 cells were infected under normoxic and hypoxic conditions for 24 hours. It was observed that the amount of killing factors αCD47-αCD3-BiTE or αIGF1R-αCD3-BiTE released by the virus under hypoxic conditions was significantly higher than that released under normoxic conditions, that is, SKOV3 tumor cells were selectively killed under hypoxic conditions.
图7显示瘤内注射的缺氧敏感转录表达调控系统控制的溶瘤病毒(TTV-P7.5-347-CRE-VP64和TTV-P11-347-CRE-VP64)都可以有效地抑制肿瘤的生长。其中,图7a显示对NCI-H292肿瘤细胞的抑制;图7b显示对SKOV3肿瘤细胞的抑制;TTV:Tiantan vaccinia virus,痘苗病毒天坛株;UTD:Untransduced,即细胞因子诱导培养的,但是未经过转导的T细胞。Figure 7 shows that intratumorally injected hypoxia-sensitive transcriptional expression control system-controlled oncolytic viruses (TTV-P7.5-347-CRE-VP64 and TTV-P11-347-CRE-VP64) can effectively inhibit tumor growth. Among them, Figure 7a shows the inhibition of NCI-H292 tumor cells; Figure 7b shows the inhibition of SKOV3 tumor cells; TTV: Tiantan vaccinia virus, vaccinia virus Tiantan strain; UTD: Untransduced, that is, cytokine-induced cultured but not transduced T cells.
实施发明的最佳方式Best Mode for Carrying Out the Invention
下面结合具体实施例来进一步描述本发明,本发明的优点和特点将会随着这些描述更为清楚。以下实施例仅用于说明本发明,并不用来限制本发明的范围。The present invention will be further described below in conjunction with specific examples, and the advantages and features of the present invention will become clearer with these descriptions. The following examples are only used to illustrate the present invention and are not intended to limit the scope of the present invention.
以下实施例中的实验方法,如无特殊说明,均为本领域的常规实验方法。以下实施例中所使用的实验材料,若无特殊说明,均为常规生化试剂,由销售公司购买所得。其中,DMEM培养基购自Corning公司。胎牛血清购自BI公司。LIPOFECTAMINE 3000转染试剂盒购自Thermo Fisher Scientific公司。基因合成由上海捷瑞生物工程有限公司完成。Stabl3化学感受态细胞购自上海唯地生物技术有限公司。无内毒素质粒小提试剂盒购自OMEGA公司。TK143 -细胞和SKOV3卵巢癌细胞购自美国ATCC。荧光显微镜购自日本尼康。荧光素酶底物购自普洛麦格生物技术有限公司。GloMax96微孔板发光检测仪购自普洛麦格生物技术有限公司。重症联合免疫缺陷小鼠(B-NDG)购自上海南方模式生物科技股份有限公司。 The experimental methods in the following examples, unless otherwise specified, are all conventional experimental methods in the art. The experimental materials used in the following examples, unless otherwise specified, are all conventional biochemical reagents purchased from sales companies. Among them, DMEM culture medium was purchased from Corning. Fetal bovine serum was purchased from BI. LIPOFECTAMINE 3000 transfection kit was purchased from Thermo Fisher Scientific. Gene synthesis was completed by Shanghai Jierui Bioengineering Co., Ltd. Stabl3 chemical competent cells were purchased from Shanghai Weidi Biotechnology Co., Ltd. Endotoxin-free plasmid miniprep kit was purchased from OMEGA. TK143 - cells and SKOV3 ovarian cancer cells were purchased from ATCC, USA. Fluorescence microscope was purchased from Nikon, Japan. Luciferase substrate was purchased from Promega Biotechnology Co., Ltd. GloMax96 microplate luminescence detector was purchased from Promega Biotechnology Co., Ltd. Severe combined immunodeficiency mice (B-NDG) were purchased from Shanghai South Model Organisms Technology Co., Ltd.
实施例1 痘苗病毒重组质粒与人工转录因子过表达质粒的构建Example 1 Construction of vaccinia virus recombinant plasmid and artificial transcription factor overexpression plasmid
由上海捷瑞生物工程有限公司合成SEQ ID NO:11所示的核苷酸序列(对应氨基酸序列如SEQ ID NO:12所示),并克隆至pHAGE表达载体,获得pHAGE-dNLS-HIF-1α(WT)-P2A-eGFP过表达质粒,其质粒图谱如图1b所示。The nucleotide sequence shown in SEQ ID NO: 11 (the corresponding amino acid sequence is shown in SEQ ID NO: 12) was synthesized by Shanghai Jierui Biotechnology Co., Ltd. and cloned into the pHAGE expression vector to obtain the pHAGE-dNLS-HIF-1α (WT)-P2A-eGFP overexpression plasmid, and its plasmid map is shown in Figure 1b.
由上海捷瑞生物工程有限公司合成SEQ ID NO:13-19所示的核苷酸序列,并分别克隆至VGF2基因重组质粒,获得携带SEQ ID NO:13所示核苷酸序列的6*Loxp-PE/L-Luciferase-mCherry重组质粒;携带SEQ ID NO:14所示核苷酸序列的6*Loxp-PE/L-L-Luciferase-mCherry重组质粒;携带SEQ ID NO:15所示核苷酸序列的6*Loxp-PE/L-S-Luciferase-mCherry重组质粒;携带SEQ ID NO:16所示核苷酸序列的6*Loxp-P7.5-Luciferase-mCherry重组质粒;携带SEQ ID NO:17所示核苷酸序列的6*Loxp-P7.5-71-Luciferase-mCherry重组质粒;携带SEQ ID NO:18所示核苷酸序列的6*Loxp-P7.5-24-Luciferase-mCherry重组质粒;和携带SEQ ID NO:19所示核苷酸序列的6*Loxp-LTRminP-Luciferase-mCherry重组质粒(其中Luciferase的核苷酸序列如SEQ ID NO:9所示,其氨基酸序列如SEQ ID NO:10所示)。上述重组质粒均采用红色荧光信号作为重组筛选信号,其质粒图谱如图3a-g所示。The nucleotide sequences shown in SEQ ID NO: 13-19 were synthesized by Shanghai Jierui Bioengineering Co., Ltd. and cloned into the VGF2 gene recombinant plasmid respectively to obtain the 6*Loxp-PE/L-Luciferase-mCherry recombinant plasmid carrying the nucleotide sequence shown in SEQ ID NO: 13; the 6*Loxp-PE/L-L-Luciferase-mCherry recombinant plasmid carrying the nucleotide sequence shown in SEQ ID NO: 14; the 6*Loxp-PE/L-S-Luciferase-mCherry recombinant plasmid carrying the nucleotide sequence shown in SEQ ID NO: 15; and the 6*Loxp-P 7.5-Luciferase-mCherry recombinant plasmid; 6*Loxp-P7.5-71-Luciferase-mCherry recombinant plasmid carrying the nucleotide sequence shown in SEQ ID NO: 17; 6*Loxp-P7.5-24-Luciferase-mCherry recombinant plasmid carrying the nucleotide sequence shown in SEQ ID NO: 18; and 6*Loxp-LTRminP-Luciferase-mCherry recombinant plasmid carrying the nucleotide sequence shown in SEQ ID NO: 19 (wherein the nucleotide sequence of Luciferase is shown in SEQ ID NO: 9, and its amino acid sequence is shown in SEQ ID NO: 10). The above recombinant plasmids all use red fluorescent signals as recombination screening signals, and their plasmid maps are shown in Figures 3a-g.
由上海捷瑞生物工程有限公司合成SEQ ID NO:21、23、25、27、29和31所示的核苷酸序列,并分别克隆至pHAGE表达载体,获得携带SEQ ID NO:21所示核苷酸序列的pHAGE-dNLS-CRE-P2A-eGFP过表达质粒;携带SEQ ID NO:23所示核苷酸序列的pHAGE-dNLS-CRE-VP64-P2A-eGFP过表达质粒;携带SEQ ID NO:25所示核苷酸序列的pHAGE-dNLS-CRE-VP64-ODD-P2A-eGFP过表达质粒;携带SEQ ID NO:27所示核苷酸序列的pHAGE-dNLS-CRE-RAP94-P2A-eGFP过表达质粒;携带SEQ ID NO:29所示核苷酸序列的pHAGE-dNLS-CRE-RAP94-ODD-P2A-eGFP过表达质粒;和携带SEQ ID NO:31所示核苷酸序列的pHAGE-dNLS-CRE-VP160-P2A-eGFP过表达质粒。上述过表达质粒均采用绿色荧光信号作为重组筛选信号,其质粒图谱如图2a-f所示。The nucleotide sequences shown in SEQ ID NO: 21, 23, 25, 27, 29 and 31 were synthesized by Shanghai Jierui Biotechnology Co., Ltd. and cloned into the pHAGE expression vector respectively to obtain the pHAGE-dNLS-CRE-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 21; the pHAGE-dNLS-CRE-VP64-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 23; and the pHAGE-dNLS-CRE-VP64-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 25. LS-CRE-VP64-ODD-P2A-eGFP overexpression plasmid; pHAGE-dNLS-CRE-RAP94-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 27; pHAGE-dNLS-CRE-RAP94-ODD-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 29; and pHAGE-dNLS-CRE-VP160-P2A-eGFP overexpression plasmid carrying the nucleotide sequence shown in SEQ ID NO: 31. The above overexpression plasmids all use green fluorescence signals as recombination screening signals, and their plasmid maps are shown in Figures 2a-f.
由上海捷瑞生物工程有限公司合成SEQ ID NO:1、3和9所示的核苷酸序列,通过同源重组的方法拼接在一起,并分别克隆至VGF-2基因重组质粒,获得携带HRE-PE/L-L-Luciferase基因的pSC65-VGF-2-HRE-PE/L-L-Luciferase-mCherry重组质粒。该重组质粒采用红色荧光信号作为重组筛选信号,其质粒图谱如图1a所示。The nucleotide sequences shown in SEQ ID NO: 1, 3 and 9 were synthesized by Shanghai Jierui Bioengineering Co., Ltd., spliced together by homologous recombination, and cloned into the VGF-2 gene recombinant plasmid to obtain the pSC65-VGF-2-HRE-PE/L-L-Luciferase-mCherry recombinant plasmid carrying the HRE-PE/L-L-Luciferase gene. The recombinant plasmid uses a red fluorescent signal as a recombination screening signal, and its plasmid map is shown in Figure 1a.
由上海捷瑞生物工程有限公司合成SEQ ID NO:20、5和33所示的核苷酸序列(SEQ ID NO:33所示的核苷酸序列对应的氨基酸序列如SEQ ID NO:34所示),通过同源重组的方法拼接在一起,并克隆至C9基因重组质粒,获得携带6*Loxp-P7.5-αCD47-αCD3-BiTE基因的pSC65-C9-6*Loxp-P7.5-αCD47-αCD3-BiTE-BFP重组质粒(见图5a和6a),该重组质粒采用蓝色荧光信号作为重组筛选信号。合成SEQ ID NO:20、37和33所示的核苷酸序列,通过同源重组的方法拼接在一起,并克隆至C9基因重组质粒,获得携带6*Loxp-P11-αCD47-αCD3-BiTE基因的pSC65-C9-6*Loxp-P11-αCD47-αCD3-BiTE-BFP重组质粒(见图6b),该重组质粒采用蓝色荧光信号作为重组筛选信号。The nucleotide sequences shown in SEQ ID NOs: 20, 5 and 33 (the amino acid sequence corresponding to the nucleotide sequence shown in SEQ ID NO: 33 is shown in SEQ ID NO: 34) were synthesized by Shanghai Jierui Bioengineering Co., Ltd., spliced together by homologous recombination, and cloned into the C9 gene recombination plasmid to obtain the pSC65-C9-6*Loxp-P7.5-αCD47-αCD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P7.5-αCD47-αCD3-BiTE gene (see Figures 5a and 6a). The recombinant plasmid uses a blue fluorescent signal as a recombination screening signal. The nucleotide sequences shown in SEQ ID NO: 20, 37 and 33 were synthesized, spliced together by homologous recombination, and cloned into the C9 gene recombination plasmid to obtain the pSC65-C9-6*Loxp-P11-αCD47-αCD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P11-αCD47-αCD3-BiTE gene (see Figure 6b). The recombinant plasmid uses a blue fluorescent signal as a recombination screening signal.
由上海捷瑞生物工程有限公司合成SEQ ID NO:20、5和35所示的核苷酸序列(SEQ ID NO:35所示的核苷酸序列对应的氨基酸序列如SEQ ID NO:36所示),通过同源重组的方法拼接在一起,并克隆至C9基因重组质粒,获得携带6*Loxp-P7.5-αIGF1R-αCD3-BiTE基因的pSC65-C9-6*Loxp-P7.5-αIGF1R-αCD3-BiTE-BFP重组质粒(见图6c),该重组质粒采用蓝色荧光信号作为重组筛选信号;合成SEQ ID NO:20、37和35所示的核苷酸序列,通过同源重组的方法拼接在一起,并克隆至C9基因重组质粒,获得携带6*Loxp-P11-αIGF1R-αCD3-BiTE基因的pSC65-C9-6*Loxp-P11-αIGF1R-αCD3-BiTE-BFP重组质粒(见图6d),该重组质粒采用蓝色荧光信号作为重组筛选信号。The nucleotide sequences shown in SEQ ID NOs: 20, 5 and 35 (the amino acid sequence corresponding to the nucleotide sequence shown in SEQ ID NO: 35 is shown in SEQ ID NO: 36) were synthesized by Shanghai Jierui Biotechnology Co., Ltd., spliced together by homologous recombination, and cloned into the C9 gene recombination plasmid to obtain the pSC65-C9-6*Loxp-P7.5-αIGF1R-αCD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P7.5-αIGF1R-αCD3-BiTE gene. The nucleotide sequences shown in SEQ ID NOs: 20, 37 and 35 were synthesized, spliced together by homologous recombination, and cloned into the C9 gene recombination plasmid to obtain the pSC65-C9-6*Loxp-P11-αIGF1R-αCD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P11-αIGF1R-αCD3-BiTE gene (see Figure 6d), which used a blue fluorescence signal as a recombination screening signal.
由上海捷瑞生物工程有限公司合成SEQ ID NO:1、3和29所示的核苷酸序列,通过同源重组的方法拼接在一起,并分别克隆至C16和VGF基因重组质粒,获得携带HRE-PE/L-L-dNLS-CRE-RAP94-ODD基因的pSC65-C16-HRE-PE/L-L-dNLS-CRE-RAP94-ODD-eGFP重组质粒和pSC65-VGF-HRE-PE/L-L-dNLS-CRE-RAP94-ODD-eGFP重组质粒(见图5b和c),上述重组质粒采用绿色荧光信号作为重组筛选信号;合成SEQ ID NO: 1、3和23所示的核苷酸序列,通过同源重组的方法拼接在一起,并克隆至C16基因重组质粒,获得携带HRE-PE/L-L-dNLS-CRE-VP64基因的pSC65-C16-HRE-PE/L-L-dNLS-CRE-VP64-eGFP重组质粒(见图6e)。The nucleotide sequences shown in SEQ ID NO: 1, 3 and 29 were synthesized by Shanghai Jierui Biotechnology Co., Ltd., spliced together by homologous recombination, and cloned into C16 and VGF gene recombinant plasmids, respectively, to obtain pSC65-C16-HRE-PE/L-L-dNLS-CRE-RAP94-ODD-eGFP recombinant plasmid and pSC65-VGF-HRE-PE/L-L-dNLS-CRE-RAP94-ODD-eGFP recombinant plasmid carrying HRE-PE/L-L-dNLS-CRE-RAP94-ODD gene (see Figure 5b and c), and the above recombinant plasmids used green fluorescence signal as the recombination screening signal; Synthesized SEQ ID NO: The nucleotide sequences shown in 1, 3 and 23 were spliced together by homologous recombination and cloned into the C16 gene recombination plasmid to obtain the pSC65-C16-HRE-PE/L-L-dNLS-CRE-VP64-eGFP recombinant plasmid carrying the HRE-PE/L-L-dNLS-CRE-VP64 gene (see Figure 6e).
实施例2 痘苗病毒的重组、纯化和滴度测定Example 2 Recombination, purification and titer determination of vaccinia virus
1.采用痘苗基因重组质粒进行痘苗病毒的重组1. Recombination of vaccinia virus using vaccinia gene recombinant plasmid
1.1细胞准备:将TK143 -细胞铺在6孔板中,每孔约1×10 6个。培养24小时左右,当细胞贴壁并且铺满整个底面时,进行下一步操作。 1.1 Cell preparation: TK143 - cells were plated in a 6-well plate, with about 1× 106 cells per well. After culturing for about 24 hours, when the cells adhered to the wall and covered the entire bottom surface, the next step was performed.
1.2痘苗病毒孵育:以0.0125/3PFU(PFU:空斑形成单位,病毒液滴度)/细胞用野生型痘苗病毒天坛株感染细胞,在37℃孵箱中孵育1小时后取出,吸掉上清,并用1mL PBS冲洗一遍,再加入1mL DMEM完全培养基(DMEM培养基+10%胎牛血清(FBS)+1%青霉素和链霉素抗生素(PS))。1.2 Vaccinia virus incubation: Infect cells with the wild-type vaccinia virus Tiantan strain at 0.0125/3 PFU (PFU: plaque forming unit, virus titer)/cell, incubate in a 37°C incubator for 1 hour, remove the cells, aspirate the supernatant, rinse with 1 mL PBS, and then add 1 mL DMEM complete medium (DMEM medium + 10% fetal bovine serum (FBS) + 1% penicillin and streptomycin antibiotics (PS)).
1.3质粒转染:用痘苗基因重组质粒转染TK143 -细胞。在37℃孵箱中培养48小时左右,具体时间根据细胞病变情况而定。 1.3 Plasmid transfection: Transfect TK143- cells with vaccinia gene recombinant plasmid and culture in a 37°C incubator for about 48 hours, the specific time depends on the cell pathological condition.
1.4准备病毒铺斑用的2×DMEM维持培养基(DMEM培养基+2%FBS+1%PS),加入2%预热的低熔点琼脂糖。1.4
1.5吸掉6孔板中的上清,将4mL铺斑用的混合物加入6孔板中,每孔300μL。然后小心放入4℃冰箱促进凝固,待低熔点琼脂糖凝固后再转入37℃孵箱中培养。1.5 Aspirate the supernatant in the 6-well plate, add 4 mL of the mixture for plating into the 6-well plate, 300 μL per well. Then carefully place it in a 4°C refrigerator to promote solidification, and transfer it to a 37°C incubator for culture after the low melting point agarose solidifies.
1.6在荧光显微镜下挑取重组的病毒噬斑,加入500μL的DMEM完全培养基。在-80℃下反复冻融三次以上,使病毒尽量多的释放。该纯化过程至少需要进行5次。1.6 Pick the recombinant virus plaques under a fluorescence microscope and add 500 μL of DMEM complete medium. Repeat freezing and thawing at -80°C for more than three times to release as much virus as possible. This purification process needs to be performed at least 5 times.
1.7进行重组痘苗病毒的小样扩增,铺TK143 -细胞于六孔板,每孔1×10 6个细胞,使用时细胞约为孔板底面积的100%。 1.7 Perform small sample amplification of the recombinant vaccinia virus by plating TK143- cells in a six-well plate, with 1×10 6 cells per well. When used, the cells should occupy approximately 100% of the bottom area of the well plate.
1.8在接种病毒前,将孔中的培养基换成2mL DMEM维持培养基。将纯化得到的含有荧光的病毒液反复吹打至散开。每孔加入100μL左右病毒液。在37℃孵箱中孵育48小时左右,根据病毒斑形成情况收样。1.8 Before inoculating the virus, replace the culture medium in the well with 2 mL DMEM maintenance medium. Repeatedly blow the purified virus solution containing fluorescence until it is dispersed. Add about 100 μL of virus solution to each well. Incubate in a 37°C incubator for about 48 hours, and collect samples according to the formation of virus plaques.
1.9收样:将孔里的培养基上清小心吸出1mL。用剩下的1mL培养基将细胞充分吹下,收于EP管中,可以用于后续基因组的提取以及作为毒种进行扩增。1.9 Sample collection: Carefully aspirate 1 mL of the culture medium supernatant in the well. Use the remaining 1 mL of culture medium to fully blow off the cells and collect them in an EP tube, which can be used for subsequent genome extraction and as a virus seed for amplification.
2.重组痘苗病毒的扩增与纯化2. Amplification and Purification of Recombinant Vaccinia Virus
2.1 VERO细胞铺板:取10cm皿,每个皿约5×10 6个细胞,保证第二天接种痘苗病毒时细胞密度达100%为宜; 2.1 VERO cell plating: Take a 10 cm dish, with about 5 × 10 6 cells in each dish, and ensure that the cell density reaches 100% when inoculating vaccinia virus the next day;
2.2病毒接种前,需将DMEM完全培养基换成8mL DMEM维持培养基,将病毒接种到DMEM维持培养基的细胞中,接种量约为0.02MOI(MOI=病毒PFU/细胞数)。继续在37℃、5%CO 2的孵箱中培养48小时左右,根据病毒斑形成情况收样; 2.2 Before virus inoculation, replace DMEM complete medium with 8 mL DMEM maintenance medium, inoculate the virus into the cells in DMEM maintenance medium, and the inoculation amount is about 0.02 MOI (MOI = virus PFU/cell number). Continue to culture in an incubator at 37°C and 5% CO2 for about 48 hours, and collect samples according to the formation of virus plaques;
2.3收痘苗病毒:弃掉皿内8mL培养基,取用2mL DMEM维持培养基将剩余的细胞吹下,收于15mL离心管中;2.3 Collect vaccinia virus: discard 8 mL of culture medium in the dish, take 2 mL of DMEM maintenance medium to blow off the remaining cells, and collect them in a 15 mL centrifuge tube;
2.4冻存24小时后,将收得的病毒液再反复冻融2次,用36%的蔗糖溶液进行密度梯度离心,在16000g、4℃下离心90min,小心倒掉上清,用PBS缓冲液溶解离心管内的病毒沉淀,分装保存于-80℃下,待测定病毒滴度。2.4 After 24 hours of freezing, freeze-thaw the collected virus solution twice, perform density gradient centrifugation with 36% sucrose solution, centrifuge at 16000g and 4°C for 90 minutes, carefully pour out the supernatant, dissolve the virus precipitate in the centrifuge tube with PBS buffer, and store it in aliquots at -80°C until the virus titer is determined.
3.重组痘苗病毒的滴度测定3. Titer determination of recombinant vaccinia virus
3.1 TK143 -细胞的准备:将TK143 -细胞铺在24孔板中,每孔约为2×10 5个细胞,使用时细胞密度达到24孔板底面积的100%; 3.1 Preparation of TK143 cells: TK143 cells were plated in a 24-well plate, with approximately 2 × 10 5 cells per well. The cell density should reach 100% of the bottom area of the 24-well plate when used;
3.2稀释病毒,用DMEM维持培养基稀释痘苗病毒液,从1:100开始,做10倍比稀释,终体积为1100μL;3.2 Dilute the virus. Use DMEM to dilute the vaccinia virus solution, starting from 1:100, and make 10-fold dilutions to a final volume of 1100 μL.
3.3弃掉24孔板中的DMEM完全培养基,取稀释好的病毒液500μL加入孔中,做两个复孔。在37℃、5%CO 2的孵箱中孵育48小时左右,根据病毒噬斑形成情况决定铺斑时间; 3.3 Discard the DMEM complete medium in the 24-well plate, take 500 μL of the diluted virus solution and add it to the wells, and make two duplicate wells. Incubate in an incubator at 37°C and 5% CO 2 for about 48 hours, and determine the plaque spreading time according to the formation of virus plaques;
3.4病毒噬斑计数:首先观察病毒噬斑的数目是否呈十倍比的趋势递减,随后统计种毒的两个复孔中仅有个位数噬斑的复孔中的噬斑数量,得到两个孔中噬斑数值之和,再乘以该孔所对应稀释度的倒数值即为1mL中病毒的滴度。3.4 Virus plaque counting: First, observe whether the number of virus plaques decreases tenfold, then count the number of plaques in the two duplicate wells with only single-digit plaques in the inoculated virus, and get the sum of the plaque values in the two wells. Multiply it by the reciprocal value of the dilution corresponding to the well to get the titer of the virus in 1 mL.
实施例3 痘苗病毒中HRE-VTP-GOI模式的缺氧调控作用Example 3 Hypoxia-regulated effects of the HRE-VTP-GOI model in vaccinia virus
本实施例在TK143 -细胞中进行了常氧和缺氧条件下的痘苗病毒感染实验,测试在病毒基因组整合缺氧敏感转录表达控制系统的痘苗病毒表达目的基因的情况。 In this example, vaccinia virus infection experiments were conducted in TK143- cells under normoxic and hypoxic conditions to test the expression of target genes by vaccinia viruses in which a hypoxia-sensitive transcriptional expression control system was integrated into the viral genome.
1.图1a显示实施例1构建的在痘苗病毒VGF2基因位点装载HRE-PE/L-L-Luciferase-mCherry的质粒的图谱。利用重组技术,采用该痘苗 病毒重组质粒和实施例2的方法将图1a的质粒中的目的基因插入痘苗病毒基因组中,并进行验证。1. Figure 1a shows the map of the plasmid carrying HRE-PE/L-L-Luciferase-mCherry at the vaccinia virus VGF2 gene site constructed in Example 1. Using recombinant technology, the target gene in the plasmid of Figure 1a was inserted into the vaccinia virus genome using the vaccinia virus recombinant plasmid and the method of Example 2, and verified.
2.将TK143
-细胞按1×10
4个细胞/孔铺到96孔板内,待第二天细胞贴壁后分成两组,一组转染pHAGE-dNLS-HIF-1α(WT)-P2A-eGFP的过表达质粒(每孔200ng),另一组为空白对照组(Blank)。在质粒转染24h后,分别感染步骤1获得的重组痘苗病毒,感染复数(MOI)为0.02。感染后,将细胞分别放置于缺氧条件(1%O
2浓度)和常氧条件下(21%O
2浓度)下培养24h,培养结束后收集细胞,检测荧光素酶的表达情况。
2. TK143 - cells were plated into 96-well plates at 1× 104 cells/well. The next day, the cells were attached to the wall and divided into two groups. One group was transfected with the overexpression plasmid of pHAGE-dNLS-HIF-1α(WT)-P2A-eGFP (200ng per well), and the other group was a blank control group (Blank). 24 hours after plasmid transfection, the cells were infected with the recombinant vaccinia virus obtained in
使用 微孔板发光检测仪检测靶细胞的荧光素酶活力值,根据图1c所示的结果,缺氧感应模式HRE-VTP-GOI在缺氧条件下可以显著抑制目的基因表达,Blank组和过表达HIF-1α组分别有31.8和50.4倍的下调,表明过表达HIF-1α蛋白进一步增加了对目的基因表达的抑制。 use The luciferase activity value of the target cells was detected by a microplate luminescence detector. According to the results shown in Figure 1c, the hypoxia sensing mode HRE-VTP-GOI can significantly inhibit the expression of the target gene under hypoxic conditions. The Blank group and the HIF-1α overexpression group had 31.8 and 50.4-fold downregulation, respectively, indicating that overexpression of HIF-1α protein further increased the inhibition of target gene expression.
实施例4 在痘苗病毒中不同种ATF均可抑制ATFRE-VTP-GOI中的目的基因的表达Example 4 Different ATFs in vaccinia virus can inhibit the expression of the target gene in ATFRE-VTP-GOI
本实施例在TK143 -细胞中进行了缺氧条件下的痘苗病毒感染实验,测试在病毒基因组整合缺氧敏感转录表达控制系统的痘苗病毒表达目的基因的情况。 In this example, a vaccinia virus infection experiment under hypoxic conditions was conducted in TK143- cells to test the expression of a target gene by a vaccinia virus in which a hypoxia-sensitive transcriptional expression control system was integrated into the viral genome.
1.利用重组技术,采用实施例1构建的在痘苗病毒VGF2基因位点装载6*Loxp-LTRminiP-Luciferase-mCherry的质粒(图3a)、在痘苗病毒VGF2基因位点装载6*Loxp-PE/L-S-Luciferase-mCherry的质粒(图3b)、在痘苗病毒VGF2基因位点装载6*Loxp-PE/L-L-Luciferase-mCherry的质粒(图3c)、在痘苗病毒VGF2基因位点装载6*Loxp-PE/L-Luciferase-mCherry的质粒(图3d)、在痘苗病毒VGF2基因位点装载6*Loxp-P7.5-24-Luciferase-mCherry的质粒(图3e)、在痘苗病毒VGF2基因位点装载6*Loxp-P7.5-71-Luciferase-mCherry的质粒(图3f)和在痘苗病毒VGF2基因位点装载6*Loxp-P7.5-Luciferase-mCherry的质粒(图3g)和实施例2的方法将目的基因分别插入痘苗病毒基因组中,并进行验证。1. Using recombinant technology, the plasmid constructed in Example 1 with 6*Loxp-LTRminiP-Luciferase-mCherry at the vaccinia virus VGF2 gene site ( FIG. 3 a ), the plasmid with 6*Loxp-PE/L-S-Luciferase-mCherry at the vaccinia virus VGF2 gene site ( FIG. 3 b ), the plasmid with 6*Loxp-PE/L-L-Luciferase-mCherry at the vaccinia virus VGF2 gene site ( FIG. 3 c ), and the plasmid with 6*Loxp-PE/L-Luciferase-mCherry at the vaccinia virus VGF2 gene site ( FIG. 3 e ) were used. The target genes were inserted into the vaccinia virus genome by using a plasmid containing erase-mCherry ( FIG. 3d ), a plasmid containing 6*Loxp-P7.5-24-Luciferase-mCherry at the vaccinia virus VGF2 gene site ( FIG. 3e ), a plasmid containing 6*Loxp-P7.5-71-Luciferase-mCherry at the vaccinia virus VGF2 gene site ( FIG. 3f ), and a plasmid containing 6*Loxp-P7.5-Luciferase-mCherry at the vaccinia virus VGF2 gene site ( FIG. 3g ) and the method of Example 2, respectively, and verified.
2.将TK143
-细胞按1×10
4个细胞/孔铺到96孔板内,待第二天细胞贴壁后,分别转染图2所示质粒(每孔200ng)。在质粒转染24h后,分别感染步骤1获得的重组痘苗病毒,病毒感染复数(MOI)为0.02。感染后,细胞 继续培养24h,培养结束后收集细胞,检测荧光素酶的表达情况。
2. TK143 - cells were plated into 96-well plates at 1× 104 cells/well. After the cells adhered to the wall the next day, the plasmids shown in Figure 2 were transfected (200 ng per well). 24 hours after plasmid transfection, the recombinant vaccinia virus obtained in
3.使用 微孔板发光检测仪检测靶细胞的荧光素酶活力值,根据图4a和4b所示,不同病毒启动子控制下,不同种ATF均可抑制ATFRE-VTP-GOI模式中的目的基因的表达,不同病毒的启动子抑制荧光素酶表达的下降倍数在5-10倍之间。 3. Use The luciferase activity value of the target cells was detected by a microplate luminescence detector. As shown in Figures 4a and 4b, different types of ATFs under the control of different viral promoters could inhibit the expression of the target gene in the ATFRE-VTP-GOI model, and the reduction factor of luciferase expression inhibited by different viral promoters was between 5-10 times.
实施例5 缺氧敏感转录表达调控系统在不同整合位点均可介导痘苗病毒在缺氧条件下特异性表达目的基因Example 5 The hypoxia-sensitive transcriptional expression regulatory system can mediate the specific expression of the target gene by vaccinia virus under hypoxic conditions at different integration sites
本实施例在TK143 -细胞中进行了常氧和缺氧条件下痘苗病毒感染实验,测试在病毒基因组的不同整合位点具有缺氧敏感转录表达调控系统的痘苗病毒表达目的基因的情况。 In this example, vaccinia virus infection experiments were conducted in TK143- cells under normoxic and hypoxic conditions to test the expression of target genes by vaccinia viruses with hypoxia-sensitive transcriptional expression regulatory systems at different integration sites of the viral genome.
1.采用实施例1构建的携带携HRE-PE/L-L-dNLS-CRE-RAP94-ODD基因的pSC65-C16-HRE-PE/L-L-dNLS-CRE-RAP94-ODD-eGFP重组质粒和携带6*Loxp-P7.5-αCD47-αCD3-BiTE基因的pSC65-C9-6*Loxp-P7.5-αCD47-αCD3-BiTE-BFP重组质粒和实施例2的方法重组痘苗病毒,并进行验证。1. The pSC65-C16-HRE-PE/L-L-dNLS-CRE-RAP94-ODD-eGFP recombinant plasmid carrying the HRE-PE/L-L-dNLS-CRE-RAP94-ODD gene and the pSC65-C9-6*Loxp-P7.5-αCD47-αCD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P7.5-αCD47-αCD3-BiTE gene constructed in Example 1 and the method of Example 2 were used to recombinant vaccinia virus and verified.
2.采用实施例1构建的携带携HRE-PE/L-L-dNLS-CRE-RAP94-ODD基因的pSC65-VGF-HRE-PE/L-L-dNLS-CRE-RAP94-ODD-eGFP重组质粒和携带6*Loxp-P7.5-αCD47-αCD3-BiTE基因的pSC65-C9-6*Loxp-P7.5-αCD47-αCD3-BiTE-BFP重组质粒和实施例2的方法重组痘苗病毒,并进行验证。2. The pSC65-VGF-HRE-PE/L-L-dNLS-CRE-RAP94-ODD-eGFP recombinant plasmid carrying the HRE-PE/L-L-dNLS-CRE-RAP94-ODD gene and the pSC65-C9-6*Loxp-P7.5-αCD47-αCD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P7.5-αCD47-αCD3-BiTE gene constructed in Example 1 and the method of Example 2 were used to recombinant vaccinia virus and verified.
3.将TK143 -细胞按2×10 5个细胞/孔铺到12孔板内,待第二天细胞贴壁后进行感染。病毒感染复数(MOI)为0.02,分别感染在C16和C9基因位点整合HRE-PE/L-L-dNLS-CRE-RAP94-ODD和6*Loxp-P7.5-αCD47-αCD3-BiTE基因的痘苗病毒;以及分别感染在VGF和C9基因位点整合HRE-PE/L-L-dNLS-CRE-RAP94-ODD和6*Loxp-P7.5-αCD47-αCD3-BiTE基因的痘苗病毒。感染后,将细胞分别放置于缺氧条件(1%O 2浓度)和常氧条件下(21%O 2浓度)下培养24h,培养结束后收集细胞上清,检测杀伤因子的表达情况。 3. TK143 - cells were plated into 12-well plates at 2×10 5 cells/well, and infected after the cells adhered to the wall the next day. The virus infection multiplicity (MOI) was 0.02, and the cells were infected with vaccinia virus with HRE-PE/LL-dNLS-CRE-RAP94-ODD and 6*Loxp-P7.5-αCD47-αCD3-BiTE genes integrated at the C16 and C9 gene loci; and infected with vaccinia virus with HRE-PE/LL-dNLS-CRE-RAP94-ODD and 6*Loxp-P7.5-αCD47-αCD3-BiTE genes integrated at the VGF and C9 gene loci. After infection, the cells were cultured under hypoxic conditions (1% O 2 concentration) and normoxic conditions (21% O 2 concentration) for 24 hours, and the cell supernatant was collected after the culture to detect the expression of killing factors.
4.肿瘤细胞杀伤效率由基于荧光素酶的细胞杀伤检测方法(Luciferase-based cytotoxicity assay)进行评估。首先,将1×10
4个SKOV3-Luc (萤火虫荧光素酶基因修饰的人卵巢癌细胞)接种于96孔平底黑板上,每孔100μL培养基,置于37℃、5%CO
2细胞培养箱中培养18h。第二天,将步骤3获得的培养上清加到对应的孔中,设置共培养细胞上清(对照组),培养结束后使用
微孔板发光检测仪检测靶细胞的荧光素酶活力值。
4. The tumor cell killing efficiency was evaluated by luciferase-based cytotoxicity assay. First, 1×10 4 SKOV3-Luc (human ovarian cancer cells modified by firefly luciferase gene) were inoculated on a 96-well flat-bottom black plate, 100 μL culture medium per well, and cultured in a 37°C, 5% CO 2 cell culture incubator for 18 hours. On the second day, the culture supernatant obtained in
细胞杀伤率的计算公式如下所示:The calculation formula of cell killing rate is as follows:
细胞杀伤率(%)=(对照组病毒感染上清荧光素酶活力值-实验组荧光素酶活力值)/对照组病毒感染上清荧光素酶活力值×100Cell killing rate (%) = (luciferase activity value of virus-infected supernatant in control group - luciferase activity value of experimental group) / luciferase activity value of virus-infected supernatant in control group × 100
5.结果如图5d所示。在痘苗病毒不同基因位点C16/VGF和C9分别整合HRE-PE/L-L-dNLS-CRE-RAP94-ODD和6*Loxp-P7.5-αCD47-αCD3-BiTE基因的溶瘤病毒在常氧条件下的肿瘤杀伤活性较低,杀伤率分别仅为10.4%和5.04%,而在缺氧环境下则能选择性高效杀伤SKOV3肿瘤细胞(杀伤率分别为17.7%和12%),即缺氧条件下特异性杀伤肿瘤细胞的杀伤活性分别是常氧条件下的1.7倍和2.4倍。5. The results are shown in Figure 5d. The oncolytic virus with HRE-PE/L-L-dNLS-CRE-RAP94-ODD and 6*Loxp-P7.5-αCD47-αCD3-BiTE genes integrated into different gene sites C16/VGF and C9 of vaccinia virus respectively had low tumor killing activity under normoxic conditions, with killing rates of only 10.4% and 5.04%, respectively, but could selectively and efficiently kill SKOV3 tumor cells under hypoxic conditions (killing rates were 17.7% and 12%, respectively), that is, the killing activity of specific tumor cell killing under hypoxic conditions was 1.7 times and 2.4 times that under normoxic conditions, respectively.
实施例6 缺氧敏感转录表达调控系统控制的溶瘤病毒的体外抗肿瘤作用Example 6 In vitro antitumor effect of oncolytic virus controlled by hypoxia-sensitive transcriptional expression regulatory system
本实施例在TK143 -细胞中进行了常氧和缺氧条件下痘苗病毒感染实验,测试具有缺氧敏感转录表达调控系统的病毒表达目的基因的情况。 In this example, vaccinia virus infection experiments were conducted in TK143- cells under normoxic and hypoxic conditions to test the expression of target genes by viruses with hypoxia-sensitive transcriptional expression regulatory systems.
1.采用实施例1构建的携带HRE-PE/L-L-dNLS-CRE-VP64基因的pSC65-C16-HRE-PE/L-L-dNLS-CRE-VP64-eGFP重组质粒和携带6*Loxp-P7.5-αCD47-αCD3-BiTE基因的pSC65-C9-6*Loxp-P7.5-αCD47-αCD3-BiTE-BFP重组质粒和实施例2的方法重组痘苗病毒,并进行验证;采用实施例1构建的携带HRE-PE/L-L-dNLS-CRE-VP64基因的pSC65-C16-HRE-PE/L-L-dNLS-CRE-VP64-eGFP重组质粒和携带核酸序列6*Loxp-P11-αCD47-αCD3-BiTE基因的pSC65-C9-6*Loxp-P11-αCD47-αCD3-BiTE-BFP重组质粒和实施例2的方法重组痘苗病毒,并进行验证。1. The pSC65-C16-HRE-PE/L-L-dNLS-CRE-VP64-eGFP recombinant plasmid carrying the HRE-PE/L-L-dNLS-CRE-VP64 gene and the pSC65-C9-6*Loxp-P7.5-αCD47-αCD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P7.5-αCD47-αCD3-BiTE gene constructed in Example 1 and the method of Example 2 were used to recombinant vaccinia virus and verified; The vaccinia virus was recombined using the pSC65-C16-HRE-PE/L-L-dNLS-CRE-VP64-eGFP recombinant plasmid carrying the HRE-PE/L-L-dNLS-CRE-VP64 gene constructed in Example 1 and the pSC65-C9-6*Loxp-P11-αCD47-αCD3-BiTE-BFP recombinant plasmid carrying the
2.采用实施例1构建的携带HRE-PE/L-L-dNLS-CRE-VP64基因的pSC65-C16-HRE-PE/L-L-dNLS-CRE-VP64-eGFP重组质粒和携带 6*Loxp-P7.5-αIGF1R-αCD3-BiTE基因的pSC65-C9-6*Loxp-P7.5-αIGF1R-αCD3-BiTE-BFP重组质粒和实施例2的方法重组痘苗病毒,并进行验证;采用实施例1构建的携带HRE-PE/L-L-dNLS-CRE-VP64基因的pSC65-C16-HRE-PE/L-L-dNLS-CRE-VP64-eGFP重组质粒和携带6*Loxp-P11-αIGF1R-αCD3-BiTE基因的pSC65-C9-6*Loxp-P11-αIGF1R-αCD3-BiTE-BFP重组质粒和实施例2的方法重组痘苗病毒,并进行验证。2. The pSC65-C16-HRE-PE/L-L-dNLS-CRE-VP64-eGFP recombinant plasmid carrying the HRE-PE/L-L-dNLS-CRE-VP64 gene constructed in Example 1 and the The vaccinia virus was recombined using the pSC65-C9-6*Loxp-P7.5-αIGF1R-αCD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P7.5-αIGF1R-αCD3-BiTE gene and the method of Example 2, and the results were verified. The vaccinia virus was recombined using the pSC65-C16-HRE-PE/L-L-dNLS-CRE-VP64-eGFP recombinant plasmid carrying the HRE-PE/L-L-dNLS-CRE-VP64 gene constructed in Example 1 and the pSC65-C9-6*Loxp-P11-αIGF1R-αCD3-BiTE-BFP recombinant plasmid carrying the 6*Loxp-P11-αIGF1R-αCD3-BiTE gene and the method of Example 2, and the results were verified.
3.将TK143 -细胞按2×10 5个细胞/孔铺到12孔板内,待第二天细胞贴壁后进行感染。病毒感染复数(MOI)为0.02,分别感染在C16和C9基因位点整合HRE-PE/L-L-dNLS-CRE-VP64和6*Loxp-P7.5-αCD47-αCD3-BiTE基因的痘苗病毒、在C16和C9基因位点整合HRE-PE/L-L-dNLS-CRE-VP64和6*Loxp-P11-αCD47-αCD3-BiTE基因的痘苗病毒、在C16和C9基因位点整合HRE-PE/L-L-dNLS-CRE-VP64和6*Loxp-P7.5-αIGF1R-αCD3-BiTE基因的痘苗病毒、以及在C16和C9基因位点整合HRE-PE/L-L-dNLS-CRE-VP64和6*Loxp-P11-αIGF1R-αCD3-BiTE基因的痘苗病毒。感染后,将细胞分别放置于缺氧条件(1%O 2浓度)和常氧条件下(21%O 2浓度)下培养24h,培养结束后收集细胞上清,检测杀伤因子的表达情况。 3. Plate TK143- cells at 2× 105 cells/well into a 12-well plate and infect them after the cells adhere to the plate the next day. The virus infection multiplicity (MOI) was 0.02, and the cells were infected with vaccinia virus with HRE-PE/LL-dNLS-CRE-VP64 and 6*Loxp-P7.5-αCD47-αCD3-BiTE genes integrated at the C16 and C9 gene loci, vaccinia virus with HRE-PE/LL-dNLS-CRE-VP64 and 6*Loxp-P11-αCD47-αCD3-BiTE genes integrated at the C16 and C9 gene loci, vaccinia virus with HRE-PE/LL-dNLS-CRE-VP64 and 6*Loxp-P7.5-αIGF1R-αCD3-BiTE genes integrated at the C16 and C9 gene loci, and vaccinia virus with HRE-PE/LL-dNLS-CRE-VP64 and 6*Loxp-P11-αIGF1R-αCD3-BiTE genes integrated at the C16 and C9 gene loci. After infection, the cells were cultured under hypoxic conditions (1% O 2 concentration) and normoxic conditions (21% O 2 concentration) for 24 h. After the culture, the cell supernatant was collected to detect the expression of the killing factor.
4.肿瘤细胞杀伤效率由基于荧光素酶的细胞杀伤检测方法(Luciferase-based cytotoxicity assay)进行评估。首先,将1×10
4个SKOV3-Luc(萤火虫荧光素酶基因修饰的人卵巢癌细胞)接种于96孔平底黑板上,每孔100μL培养基,置于37℃、5%CO
2细胞培养箱中培养18h。第二天,将步骤3获得的培养上清加到对应的孔中,设置共培养细胞上清(对照组),培养结束后使用
微孔板发光检测仪检测靶细胞的荧光素酶活力值。
4. The tumor cell killing efficiency was evaluated by luciferase-based cytotoxicity assay. First, 1×10 4 SKOV3-Luc (human ovarian cancer cells modified by firefly luciferase gene) were inoculated on a 96-well flat-bottom black plate, 100 μL culture medium per well, and cultured in a 37°C, 5% CO 2 cell culture incubator for 18 hours. On the second day, the culture supernatant obtained in
细胞杀伤率的计算公式如下所示,The calculation formula of cell killing rate is as follows:
细胞杀伤率(%)=(对照组病毒感染上清荧光素酶活力值-实验组荧光素酶活力值)/对照组病毒感染上清荧光素酶活力值×100Cell killing rate (%) = (luciferase activity value of virus-infected supernatant in control group - luciferase activity value of experimental group) / luciferase activity value of virus-infected supernatant in control group × 100
5.结果如图6f和6g所示。其中,图6f显示在C16和C9基因位点整合HRE-PE/L-L-dNLS-CRE-VP64和6*Loxp-P7.5-αCD47-αCD3-BiTE基因的痘苗病毒,以及C16和C9基因位点整合HRE-PE/L-L-dNLS-CRE-VP64和 6*Loxp-P11-αCD47-αCD3-BiTE基因的痘苗病毒在常氧条件下的肿瘤杀伤活性较低,杀伤率分别为18.2%和13.8%,在缺氧环境下则能选择性高效杀伤SKOV3肿瘤细胞(杀伤率分别为30.9%和27.2%),即缺氧条件下特异性杀伤肿瘤细胞的杀伤活性分别是常氧条件下的1.7倍和2.1倍;图6g显示在C16和C9基因位点整合HRE-PE/L-L-dNLS-CRE-VP64和6*Loxp-P7.5-αIGF1R-αCD3-BiTE基因的痘苗病毒,以及在C16和C9基因位点整合HRE-PE/L-L-dNLS-CRE-VP64和6*Loxp-P11-αIGF1R-αCD3-BiTE基因的痘苗病毒在常氧条件下的肿瘤杀伤活性较低,杀伤率分别为5.3%和6.2%,在缺氧环境下则能选择性高效杀伤SKOV3肿瘤细胞(杀伤率分别为21.5%和18.5%),即缺氧条件下特异性杀伤肿瘤细胞的杀伤活性分别是常氧条件下的3.1倍和4.1倍。5. The results are shown in Figures 6f and 6g. Figure 6f shows vaccinia virus with HRE-PE/L-L-dNLS-CRE-VP64 and 6*Loxp-P7.5-αCD47-αCD3-BiTE genes integrated into the C16 and C9 gene loci, and vaccinia virus with HRE-PE/L-L-dNLS-CRE-VP64 and 6*Loxp-P7.5-αCD47-αCD3-BiTE genes integrated into the C16 and C9 gene loci. The tumor killing activity of vaccinia virus carrying 6*Loxp-P11-αCD47-αCD3-BiTE gene was low under normoxic conditions, with killing rates of 18.2% and 13.8%, respectively. However, it could selectively and efficiently kill SKOV3 tumor cells under hypoxia (killing rates of 30.9% and 27.2%, respectively), that is, the killing activity of specific tumor cell killing under hypoxia was 1.7 times and 2.1 times that under normoxic conditions, respectively; Figure 6g shows that HRE-PE/L-L-dNLS-CRE-VP64 and 6*Loxp-P7.5-αIGF were integrated at the C16 and C9 gene loci. The vaccinia virus with the 1R-αCD3-BiTE gene and the vaccinia virus with the HRE-PE/L-L-dNLS-CRE-VP64 and 6*Loxp-P11-αIGF1R-αCD3-BiTE genes integrated at the C16 and C9 gene loci had low tumor killing activity under normoxic conditions, with killing rates of 5.3% and 6.2%, respectively. However, they could selectively and efficiently kill SKOV3 tumor cells under hypoxic conditions (killing rates were 21.5% and 18.5%, respectively), that is, the killing activity of specific tumor cell killing under hypoxic conditions was 3.1 times and 4.1 times that under normoxic conditions, respectively.
实施例7 缺氧敏感转录表达控制系统控制的溶瘤病毒的体内抗肿瘤作用Example 7 In vivo anti-tumor effect of oncolytic virus controlled by hypoxia-sensitive transcriptional expression control system
提前一天对无菌隔离器饲养的B-NDG小鼠进行背部局部脱毛,脱毛可以使用脱毛膏或动物剃毛器,使其暴露出肿瘤细胞接种部位的皮肤即可。One day in advance, perform local hair removal on the back of B-NDG mice housed in a sterile isolator. Hair removal can be done using a depilatory cream or an animal shaver to expose the skin at the site of tumor cell inoculation.
左手固定小鼠,即左手同时抓紧小鼠的头部、颈部和背部皮肤,使其背部朝左翻,充分暴露背部右侧剃毛部位后,右手用酒精棉球对其进行消毒处理。1mL胰岛素注射器吹打混匀预先准备好的SKOV3或NCI-H292肿瘤细胞后,吸取125μL细胞悬液(5×10 6个肿瘤细胞),将针头以针尖与皮肤呈30°-40°角度斜着刺入小鼠皮下,缓慢推注细胞悬液,避免细胞溢漏。待125μL细胞悬液注射完毕,留针2-3秒后迅速拔出,可见注射部位的皮下鼓起一个清晰可见的小包。 Fix the mouse with the left hand, that is, grasp the mouse's head, neck and back skin with the left hand at the same time, turn the mouse's back to the left, fully expose the shaved area on the right side of the back, and disinfect it with an alcohol cotton ball in the right hand. After blowing and mixing the pre-prepared SKOV3 or NCI-H292 tumor cells with a 1mL insulin syringe, draw 125μL of cell suspension (5×10 6 tumor cells), insert the needle obliquely into the mouse's subcutaneous tissue at an angle of 30°-40° between the needle tip and the skin, and slowly push the cell suspension to avoid cell leakage. After the injection of 125μL of cell suspension is completed, leave the needle in for 2-3 seconds and then quickly pull it out. A clearly visible small bag can be seen under the skin at the injection site.
细胞接种后每隔2-3天观察小鼠的成瘤情况和健康状态,成瘤后用游标卡尺测量基线肿瘤体积并进行后续实验。The tumor formation and health status of the mice were observed every 2-3 days after cell inoculation. After tumor formation, the baseline tumor volume was measured with a vernier caliper and subsequent experiments were performed.
瘤内回输5×10 3PFU具有缺氧敏感调控系统的痘苗病毒,即C16和C9位点整合HRE-PE/L-L-dNLS-CRE-VP64和6*Loxp-P7.5-αCD47-αCD3-BiTE基因的痘苗病毒(TTV-P7.5-347-CRE-VP64),以及C16和C9位点整合HRE-PE/L-L-dNLS-CRE-VP64和6*Loxp-P11-αCD47-αCD3-BiTE基因的痘苗病毒(TTV-P11-347-CRE-VP64)和对照野生型痘苗病毒,每隔2-3天测量肿瘤大小,用游标卡尺分别测量肿瘤的长径和短径,肿瘤体积的计算公式如 下: 5×10 3 PFU of vaccinia virus with hypoxia-sensitive regulatory system, i.e., vaccinia virus with HRE-PE/LL-dNLS-CRE-VP64 and 6*Loxp-P7.5-αCD47-αCD3-BiTE genes integrated into C16 and C9 sites (TTV-P7.5-347-CRE-VP64), vaccinia virus with HRE-PE/LL-dNLS-CRE-VP64 and 6*Loxp-P11-αCD47-αCD3-BiTE genes integrated into C16 and C9 sites (TTV-P11-347-CRE-VP64) and control wild-type vaccinia virus were reinfused intratumorally. The tumor size was measured every 2-3 days. The long and short diameters of the tumor were measured with a vernier caliper. The tumor volume was calculated as follows:
体积=(长径×短径 2)/2。 Volume = (longer diameter × shorter diameter2 )/2.
结果如图7所示,缺氧敏感转录表达控制系统控制的痘苗病毒(TTV-P7.5-347-CRE-VP64和TTV-P11-347-CRE-VP64)均可以有效抑制SKOV3(卵巢癌)和NCI-H292(肺癌)的生长,病毒注射66天的肿瘤抑制率分别高达100%和80%,且相比野生型对照组的痘苗病毒,可显著延长小鼠的生存期,野生型对照组痘苗病毒小鼠在39天就全部死亡,而TTV-P7.5-347-CRE-VP64和TTV-P11-347-CRE-VP64组的小鼠在60天仍有存活。The results are shown in Figure 7. The vaccinia viruses controlled by the hypoxia-sensitive transcriptional expression control system (TTV-P7.5-347-CRE-VP64 and TTV-P11-347-CRE-VP64) can effectively inhibit the growth of SKOV3 (ovarian cancer) and NCI-H292 (lung cancer). The
以上所述仅是本发明的优选实施例而已,并非对本发明做任何形式上的限制,虽然本发明已经以优选实施例揭示如上,然而其并非用以限定本发明。任何熟悉本专业的技术人员,在不脱离本发明技术方案的范围内,可利用上述揭示的技术内容作出些许变动或修饰以获得具有等同变化的等效实施例,但凡是未脱离本发明技术方案的内容且依据本发明的技术实质对以上实施例所作的任何修改、等同变化与修饰均仍落入本发明的范围。The above description is only the preferred embodiment of the present invention, and does not limit the present invention in any form. Although the present invention has been disclosed as the preferred embodiment, it is not used to limit the present invention. Any technician familiar with the profession can make some changes or modifications to the technical content disclosed above without departing from the scope of the technical solution of the present invention to obtain an equivalent embodiment with equivalent changes. However, any modification, equivalent change and modification made to the above embodiment without departing from the content of the technical solution of the present invention and based on the technical essence of the present invention still fall within the scope of the present invention.
Claims (12)
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| PCT/CN2022/137157 WO2024119387A1 (en) | 2022-12-07 | 2022-12-07 | System for controlling target gene expression in response to hypoxic environment and use thereof |
Applications Claiming Priority (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| PCT/CN2022/137157 WO2024119387A1 (en) | 2022-12-07 | 2022-12-07 | System for controlling target gene expression in response to hypoxic environment and use thereof |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024119387A1 true WO2024119387A1 (en) | 2024-06-13 |
Family
ID=91378469
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2022/137157 Ceased WO2024119387A1 (en) | 2022-12-07 | 2022-12-07 | System for controlling target gene expression in response to hypoxic environment and use thereof |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024119387A1 (en) |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1319139A (en) * | 1998-09-23 | 2001-10-24 | 牛津生物医学(英国)有限公司 | Polynucleotide constructs and uses thereof |
| CN1720066A (en) * | 2002-10-01 | 2006-01-11 | 杜克大学 | Targeted tumor therapy by use of recombinant adenovirus vectors that selectively replicate in hypoxic regions of tumors |
| CN101200732A (en) * | 2007-07-10 | 2008-06-18 | 西安交通大学 | Hypoxia response element gene therapy plasmid and its construction method |
| CN114249807A (en) * | 2021-08-13 | 2022-03-29 | 上海鑫湾生物科技有限公司 | Hypoxia-triggered artificial transcription factor, transcription control system and application thereof |
-
2022
- 2022-12-07 WO PCT/CN2022/137157 patent/WO2024119387A1/en not_active Ceased
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1319139A (en) * | 1998-09-23 | 2001-10-24 | 牛津生物医学(英国)有限公司 | Polynucleotide constructs and uses thereof |
| CN1720066A (en) * | 2002-10-01 | 2006-01-11 | 杜克大学 | Targeted tumor therapy by use of recombinant adenovirus vectors that selectively replicate in hypoxic regions of tumors |
| CN101200732A (en) * | 2007-07-10 | 2008-06-18 | 西安交通大学 | Hypoxia response element gene therapy plasmid and its construction method |
| CN114249807A (en) * | 2021-08-13 | 2022-03-29 | 上海鑫湾生物科技有限公司 | Hypoxia-triggered artificial transcription factor, transcription control system and application thereof |
Non-Patent Citations (1)
| Title |
|---|
| HARADA HIROSHI, INOUE MASAHIRO, ITASAKA SATOSHI, HIROTA KIICHI, MORINIBU AKIYO, SHINOMIYA KAZUMI, ZENG LIHUA, OU GUANGFEI, ZHU YUX: "Cancer cells that survive radiation therapy acquire HIF-1 activity and translocate towards tumour blood vessels", NATURE COMMUNICATIONS, vol. 3, no. 1, 17 April 2012 (2012-04-17), XP093034919, DOI: 10.1038/ncomms1786 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20240100106A1 (en) | Isolated recombinant oncolytic adenoviruses, pharmaceutical compositions, and uses thereof for drugs for treatment of tumors and/or cancers | |
| CN107349226B (en) | Application of alphavirus in preparing anti-tumor medicine | |
| CN111315873B (en) | Isolated recombinant oncolytic poxviruses, pharmaceutical compositions and their use in medicaments for treating tumors and/or cancer | |
| CN115103912B (en) | A plasmid combination and its use in preparing modified immune cells | |
| CN116966281A (en) | Methods and uses of engineering T cells to load oncolytic viruses | |
| JP5846650B2 (en) | Vector-produced tumor target cells | |
| CN116064670A (en) | System for controlling virus replication in response to anoxic environment and application thereof | |
| WO2024056106A1 (en) | Segmented vesicular stomatitis virus vector, preparation method therefor, and use thereof | |
| WO2024119387A1 (en) | System for controlling target gene expression in response to hypoxic environment and use thereof | |
| Ahmed | Oncolytic virotherapy using neural stem cells as a novel treatment option for glioblastoma multiforme | |
| CN117085049A (en) | Application of oncolytic herpes simplex virus oHSV combined with BET inhibitor JQ1 in the treatment of malignant glioma | |
| Mi et al. | The enhanced efficacy of herpes simplex virus by lentivirus mediated VP22 and cytosine deaminase gene therapy against glioma | |
| CN118186004A (en) | A system for controlling target gene expression in response to hypoxic environment and its use | |
| WO2024082263A1 (en) | System for controlling virus replication in response to hypoxic environment and use thereof | |
| US20240318147A1 (en) | Recombinant oncolytic virus, and construction method therefor and use thereof | |
| CN116286911A (en) | A spliceable system for regulating viral replication in response to anoxic environment and its use | |
| CN120678921A (en) | Application, method and medicine of PPARG gene or/and its expression product in preparing medicine for inhibiting hepatocellular carcinoma | |
| CN111979204B (en) | Oncolytic vaccinia virus carrying sponge lectin gene, construction method and application | |
| CN109718385B (en) | Function and application of Klotho-beta | |
| CN120695178A (en) | Application, method and medicine of MYOD1 gene or/and its expression product in preparing medicine for inhibiting hepatocellular carcinoma | |
| WO2025217985A1 (en) | Use of glucose-1-phosphate in treatment of tumor | |
| CN118994351A (en) | Preparation and application of macrophage fusing plasmodium vivax SRA protein | |
| CN117210409A (en) | Pharmaceutical composition and application thereof | |
| WO2025242232A1 (en) | Modified virus targeting abnormal ets family transcription factor-related tumors | |
| CN116808061A (en) | Application of Anti-miRNA-32-5p in preparation of antitumor preparation |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 22967547 Country of ref document: EP Kind code of ref document: A1 |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 22967547 Country of ref document: EP Kind code of ref document: A1 |