WO2018171103A1 - 可编程的溶瘤病毒疫苗系统及其应用 - Google Patents
可编程的溶瘤病毒疫苗系统及其应用 Download PDFInfo
- Publication number
- WO2018171103A1 WO2018171103A1 PCT/CN2017/096043 CN2017096043W WO2018171103A1 WO 2018171103 A1 WO2018171103 A1 WO 2018171103A1 CN 2017096043 W CN2017096043 W CN 2017096043W WO 2018171103 A1 WO2018171103 A1 WO 2018171103A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- nucleic acid
- acid molecule
- protein
- promoter
- expression
- Prior art date
Links
- 244000309459 oncolytic virus Species 0.000 title description 21
- 229960005486 vaccine Drugs 0.000 title description 3
- 150000007523 nucleic acids Chemical class 0.000 claims abstract description 308
- 102000039446 nucleic acids Human genes 0.000 claims abstract description 284
- 108020004707 nucleic acids Proteins 0.000 claims abstract description 284
- 230000014509 gene expression Effects 0.000 claims abstract description 267
- 108091006104 gene-regulatory proteins Proteins 0.000 claims abstract description 120
- 102000034356 gene-regulatory proteins Human genes 0.000 claims abstract description 120
- 230000001105 regulatory effect Effects 0.000 claims abstract description 71
- 108091006106 transcriptional activators Proteins 0.000 claims abstract description 28
- 230000027455 binding Effects 0.000 claims abstract description 24
- 108090000623 proteins and genes Proteins 0.000 claims description 305
- 210000004027 cell Anatomy 0.000 claims description 281
- 241000701161 unidentified adenovirus Species 0.000 claims description 145
- 102000004169 proteins and genes Human genes 0.000 claims description 134
- 206010028980 Neoplasm Diseases 0.000 claims description 128
- 239000002679 microRNA Substances 0.000 claims description 123
- 108700011259 MicroRNAs Proteins 0.000 claims description 111
- 241000700605 Viruses Species 0.000 claims description 83
- 239000013598 vector Substances 0.000 claims description 73
- 210000004881 tumor cell Anatomy 0.000 claims description 71
- 238000000034 method Methods 0.000 claims description 55
- 239000013604 expression vector Substances 0.000 claims description 49
- 230000002401 inhibitory effect Effects 0.000 claims description 42
- 101150024821 tetO gene Proteins 0.000 claims description 35
- 201000007270 liver cancer Diseases 0.000 claims description 34
- 208000014018 liver neoplasm Diseases 0.000 claims description 32
- 108010002350 Interleukin-2 Proteins 0.000 claims description 28
- 102100023635 Alpha-fetoprotein Human genes 0.000 claims description 26
- 238000004806 packaging method and process Methods 0.000 claims description 26
- 108091028043 Nucleic acid sequence Proteins 0.000 claims description 24
- 201000011510 cancer Diseases 0.000 claims description 23
- 239000012642 immune effector Substances 0.000 claims description 23
- 229940121354 immunomodulator Drugs 0.000 claims description 23
- 239000012636 effector Substances 0.000 claims description 22
- 230000029812 viral genome replication Effects 0.000 claims description 22
- 102000013462 Interleukin-12 Human genes 0.000 claims description 20
- 108010065805 Interleukin-12 Proteins 0.000 claims description 20
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 claims description 18
- 239000013612 plasmid Substances 0.000 claims description 17
- 101150075174 E1B gene Proteins 0.000 claims description 15
- 102000003812 Interleukin-15 Human genes 0.000 claims description 14
- 108090000172 Interleukin-15 Proteins 0.000 claims description 14
- 102000037865 fusion proteins Human genes 0.000 claims description 13
- 108020001507 fusion proteins Proteins 0.000 claims description 13
- 239000002773 nucleotide Substances 0.000 claims description 13
- 108010026331 alpha-Fetoproteins Proteins 0.000 claims description 12
- 239000003814 drug Substances 0.000 claims description 12
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 12
- 230000010076 replication Effects 0.000 claims description 12
- 239000011701 zinc Substances 0.000 claims description 12
- 125000003729 nucleotide group Chemical group 0.000 claims description 11
- 241001430294 unidentified retrovirus Species 0.000 claims description 11
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 claims description 10
- 229910052725 zinc Inorganic materials 0.000 claims description 10
- 101150029662 E1 gene Proteins 0.000 claims description 8
- 229940079593 drug Drugs 0.000 claims description 8
- 108091062762 miR-21 stem-loop Proteins 0.000 claims description 8
- 108091041631 miR-21-1 stem-loop Proteins 0.000 claims description 8
- 108091044442 miR-21-2 stem-loop Proteins 0.000 claims description 8
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 claims description 7
- 241000700618 Vaccinia virus Species 0.000 claims description 7
- 101710185494 Zinc finger protein Proteins 0.000 claims description 7
- 102100023597 Zinc finger protein 816 Human genes 0.000 claims description 7
- 230000002159 abnormal effect Effects 0.000 claims description 7
- 230000009368 gene silencing by RNA Effects 0.000 claims description 7
- 206010006187 Breast cancer Diseases 0.000 claims description 6
- 208000026310 Breast neoplasm Diseases 0.000 claims description 6
- 206010009944 Colon cancer Diseases 0.000 claims description 6
- 208000001333 Colorectal Neoplasms Diseases 0.000 claims description 6
- 101150091887 Ctla4 gene Proteins 0.000 claims description 6
- 101150005585 E3 gene Proteins 0.000 claims description 6
- 101150066038 E4 gene Proteins 0.000 claims description 6
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 6
- 101710150114 Protein rep Proteins 0.000 claims description 6
- 101710152114 Replication protein Proteins 0.000 claims description 6
- 239000002502 liposome Substances 0.000 claims description 6
- 201000005202 lung cancer Diseases 0.000 claims description 6
- 208000020816 lung neoplasm Diseases 0.000 claims description 6
- 201000001441 melanoma Diseases 0.000 claims description 6
- 108091025686 miR-199a stem-loop Proteins 0.000 claims description 6
- 101150061166 tetR gene Proteins 0.000 claims description 6
- 206010060862 Prostate cancer Diseases 0.000 claims description 5
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims description 5
- 230000003042 antagnostic effect Effects 0.000 claims description 5
- 108091023663 let-7 stem-loop Proteins 0.000 claims description 5
- 108091063478 let-7-1 stem-loop Proteins 0.000 claims description 5
- 108091049777 let-7-2 stem-loop Proteins 0.000 claims description 5
- -1 miniCMV Proteins 0.000 claims description 5
- 238000002360 preparation method Methods 0.000 claims description 5
- 241001529453 unidentified herpesvirus Species 0.000 claims description 5
- 102100025475 Carcinoembryonic antigen-related cell adhesion molecule 5 Human genes 0.000 claims description 4
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 claims description 4
- 108091027766 Mir-143 Proteins 0.000 claims description 4
- 101710089372 Programmed cell death protein 1 Proteins 0.000 claims description 4
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 claims description 4
- 102000003425 Tyrosinase Human genes 0.000 claims description 4
- 108060008724 Tyrosinase Proteins 0.000 claims description 4
- 108010022366 Carcinoembryonic Antigen Proteins 0.000 claims description 3
- 102000019034 Chemokines Human genes 0.000 claims description 3
- 108010012236 Chemokines Proteins 0.000 claims description 3
- 108010089335 Cholecystokinin A Receptor Proteins 0.000 claims description 3
- 101150082674 E2 gene Proteins 0.000 claims description 3
- 101000655352 Homo sapiens Telomerase reverse transcriptase Proteins 0.000 claims description 3
- 108091026807 MiR-214 Proteins 0.000 claims description 3
- 108091028695 MiR-224 Proteins 0.000 claims description 3
- 108091028684 Mir-145 Proteins 0.000 claims description 3
- 108091028049 Mir-221 microRNA Proteins 0.000 claims description 3
- 108091062140 Mir-223 Proteins 0.000 claims description 3
- 108091061960 Naked DNA Proteins 0.000 claims description 3
- 102000004316 Oxidoreductases Human genes 0.000 claims description 3
- 108090000854 Oxidoreductases Proteins 0.000 claims description 3
- 108010072866 Prostate-Specific Antigen Proteins 0.000 claims description 3
- 108010002687 Survivin Proteins 0.000 claims description 3
- 230000002759 chromosomal effect Effects 0.000 claims description 3
- 108091064282 miR-125 stem-loop Proteins 0.000 claims description 3
- 108091037066 miR-125-1 stem-loop Proteins 0.000 claims description 3
- 108091062107 miR-125-2 stem-loop Proteins 0.000 claims description 3
- 108091079767 miR-125-3 stem-loop Proteins 0.000 claims description 3
- 108091032320 miR-146 stem-loop Proteins 0.000 claims description 3
- 108091024530 miR-146a stem-loop Proteins 0.000 claims description 3
- 108091049730 miR-1792 stem-loop Proteins 0.000 claims description 3
- 108091027698 miR-18-1 stem-loop Proteins 0.000 claims description 3
- 108091090961 miR-18-2 stem-loop Proteins 0.000 claims description 3
- 108091074450 miR-200c stem-loop Proteins 0.000 claims description 3
- 108091061917 miR-221 stem-loop Proteins 0.000 claims description 3
- 108091063489 miR-221-1 stem-loop Proteins 0.000 claims description 3
- 108091055391 miR-221-2 stem-loop Proteins 0.000 claims description 3
- 108091031076 miR-221-3 stem-loop Proteins 0.000 claims description 3
- 108091023525 miR-95 stem-loop Proteins 0.000 claims description 3
- 239000002086 nanomaterial Substances 0.000 claims description 3
- 150000003180 prostaglandins Chemical class 0.000 claims description 3
- 230000002441 reversible effect Effects 0.000 claims description 3
- 108010057856 Adenovirus E2 Proteins Proteins 0.000 claims description 2
- 239000013603 viral vector Substances 0.000 claims description 2
- 108010074708 B7-H1 Antigen Proteins 0.000 claims 2
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 claims 2
- 101000773083 Homo sapiens 5,6-dihydroxyindole-2-carboxylic acid oxidase Proteins 0.000 claims 1
- 102000007066 Prostate-Specific Antigen Human genes 0.000 claims 1
- 210000004907 gland Anatomy 0.000 claims 1
- 102000046634 human TYRP1 Human genes 0.000 claims 1
- 108091057442 miR-25b stem-loop Proteins 0.000 claims 1
- 239000002574 poison Substances 0.000 claims 1
- 231100000614 poison Toxicity 0.000 claims 1
- 230000000174 oncolytic effect Effects 0.000 description 63
- 210000001744 T-lymphocyte Anatomy 0.000 description 35
- 230000000694 effects Effects 0.000 description 33
- 238000011282 treatment Methods 0.000 description 33
- 239000004098 Tetracycline Substances 0.000 description 31
- 229930101283 tetracycline Natural products 0.000 description 31
- 229960002180 tetracycline Drugs 0.000 description 31
- 235000019364 tetracycline Nutrition 0.000 description 31
- 150000003522 tetracyclines Chemical class 0.000 description 29
- 102000004127 Cytokines Human genes 0.000 description 28
- 108090000695 Cytokines Proteins 0.000 description 28
- 102000000588 Interleukin-2 Human genes 0.000 description 26
- 230000004913 activation Effects 0.000 description 26
- 230000006870 function Effects 0.000 description 25
- 241000699670 Mus sp. Species 0.000 description 23
- 108020004414 DNA Proteins 0.000 description 22
- 230000033228 biological regulation Effects 0.000 description 22
- 230000005764 inhibitory process Effects 0.000 description 21
- 230000001629 suppression Effects 0.000 description 21
- 238000002474 experimental method Methods 0.000 description 20
- 230000001404 mediated effect Effects 0.000 description 19
- 241000699666 Mus <mouse, genus> Species 0.000 description 18
- 230000028993 immune response Effects 0.000 description 18
- 101100519207 Mus musculus Pdcd1 gene Proteins 0.000 description 17
- 230000002062 proliferating effect Effects 0.000 description 17
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 16
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 16
- 208000015181 infectious disease Diseases 0.000 description 16
- 108020004999 messenger RNA Proteins 0.000 description 16
- 239000000047 product Substances 0.000 description 16
- 210000001519 tissue Anatomy 0.000 description 16
- 239000006228 supernatant Substances 0.000 description 14
- 101000600434 Homo sapiens Putative uncharacterized protein encoded by MIR7-3HG Proteins 0.000 description 13
- 108010074328 Interferon-gamma Proteins 0.000 description 13
- 102100037401 Putative uncharacterized protein encoded by MIR7-3HG Human genes 0.000 description 13
- 230000001413 cellular effect Effects 0.000 description 13
- 230000000295 complement effect Effects 0.000 description 13
- 108091070501 miRNA Proteins 0.000 description 13
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 12
- 229940045513 CTLA4 antagonist Drugs 0.000 description 12
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 12
- 210000000822 natural killer cell Anatomy 0.000 description 12
- 239000000090 biomarker Substances 0.000 description 11
- 239000012634 fragment Substances 0.000 description 11
- 108091008146 restriction endonucleases Proteins 0.000 description 11
- 230000035897 transcription Effects 0.000 description 11
- 238000013518 transcription Methods 0.000 description 11
- 108010001515 Galectin 4 Proteins 0.000 description 10
- 102100039556 Galectin-4 Human genes 0.000 description 10
- 102000013529 alpha-Fetoproteins Human genes 0.000 description 10
- 238000006243 chemical reaction Methods 0.000 description 10
- 238000001514 detection method Methods 0.000 description 10
- 230000009977 dual effect Effects 0.000 description 10
- 230000004927 fusion Effects 0.000 description 10
- 210000000987 immune system Anatomy 0.000 description 10
- 230000001939 inductive effect Effects 0.000 description 10
- 230000001225 therapeutic effect Effects 0.000 description 10
- 230000004663 cell proliferation Effects 0.000 description 9
- 238000010276 construction Methods 0.000 description 9
- 230000012010 growth Effects 0.000 description 9
- 230000004044 response Effects 0.000 description 9
- 230000002103 transcriptional effect Effects 0.000 description 9
- 230000003612 virological effect Effects 0.000 description 9
- 102000052510 DNA-Binding Proteins Human genes 0.000 description 8
- 101001068133 Homo sapiens Hepatitis A virus cellular receptor 2 Proteins 0.000 description 8
- 102000008070 Interferon-gamma Human genes 0.000 description 8
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 8
- 108091027981 Response element Proteins 0.000 description 8
- AIYUHDOJVYHVIT-UHFFFAOYSA-M caesium chloride Chemical compound [Cl-].[Cs+] AIYUHDOJVYHVIT-UHFFFAOYSA-M 0.000 description 8
- 238000003776 cleavage reaction Methods 0.000 description 8
- 238000010586 diagram Methods 0.000 description 8
- 238000005516 engineering process Methods 0.000 description 8
- 230000002068 genetic effect Effects 0.000 description 8
- 230000001965 increasing effect Effects 0.000 description 8
- 239000003112 inhibitor Substances 0.000 description 8
- 239000008101 lactose Substances 0.000 description 8
- 210000000265 leukocyte Anatomy 0.000 description 8
- 210000002540 macrophage Anatomy 0.000 description 8
- 230000007017 scission Effects 0.000 description 8
- 230000009885 systemic effect Effects 0.000 description 8
- 238000001890 transfection Methods 0.000 description 8
- 238000012546 transfer Methods 0.000 description 8
- 230000004568 DNA-binding Effects 0.000 description 7
- 102000016928 DNA-directed DNA polymerase Human genes 0.000 description 7
- 230000009471 action Effects 0.000 description 7
- 210000000349 chromosome Anatomy 0.000 description 7
- 238000013461 design Methods 0.000 description 7
- 230000002147 killing effect Effects 0.000 description 7
- 239000002609 medium Substances 0.000 description 7
- 238000011160 research Methods 0.000 description 7
- 230000004614 tumor growth Effects 0.000 description 7
- 102100025248 C-X-C motif chemokine 10 Human genes 0.000 description 6
- 108010014303 DNA-directed DNA polymerase Proteins 0.000 description 6
- 101710199711 Early E1A protein Proteins 0.000 description 6
- 241000196324 Embryophyta Species 0.000 description 6
- 108090000144 Human Proteins Proteins 0.000 description 6
- 102000003839 Human Proteins Human genes 0.000 description 6
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 6
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 6
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 6
- 108020005202 Viral DNA Proteins 0.000 description 6
- 210000001015 abdomen Anatomy 0.000 description 6
- 239000012190 activator Substances 0.000 description 6
- 230000001772 anti-angiogenic effect Effects 0.000 description 6
- 230000008901 benefit Effects 0.000 description 6
- 230000000875 corresponding effect Effects 0.000 description 6
- 230000001419 dependent effect Effects 0.000 description 6
- 239000003623 enhancer Substances 0.000 description 6
- 229940044627 gamma-interferon Drugs 0.000 description 6
- 238000001415 gene therapy Methods 0.000 description 6
- 210000002865 immune cell Anatomy 0.000 description 6
- 210000005229 liver cell Anatomy 0.000 description 6
- 239000003550 marker Substances 0.000 description 6
- 230000007246 mechanism Effects 0.000 description 6
- 210000000440 neutrophil Anatomy 0.000 description 6
- 108700020911 DNA-Binding Proteins Proteins 0.000 description 5
- 241000282412 Homo Species 0.000 description 5
- 102100037850 Interferon gamma Human genes 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 238000004458 analytical method Methods 0.000 description 5
- 239000000427 antigen Substances 0.000 description 5
- 108091007433 antigens Proteins 0.000 description 5
- 102000036639 antigens Human genes 0.000 description 5
- 210000003494 hepatocyte Anatomy 0.000 description 5
- 238000000338 in vitro Methods 0.000 description 5
- 238000002347 injection Methods 0.000 description 5
- 239000007924 injection Substances 0.000 description 5
- 239000003446 ligand Substances 0.000 description 5
- 210000004698 lymphocyte Anatomy 0.000 description 5
- 210000004962 mammalian cell Anatomy 0.000 description 5
- 238000004519 manufacturing process Methods 0.000 description 5
- 239000000463 material Substances 0.000 description 5
- 239000000203 mixture Substances 0.000 description 5
- 230000035755 proliferation Effects 0.000 description 5
- 238000004088 simulation Methods 0.000 description 5
- 108700014590 single-stranded DNA binding proteins Proteins 0.000 description 5
- 239000000243 solution Substances 0.000 description 5
- 238000004114 suspension culture Methods 0.000 description 5
- 230000014616 translation Effects 0.000 description 5
- 238000013519 translation Methods 0.000 description 5
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 5
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 4
- 101710098275 C-X-C motif chemokine 10 Proteins 0.000 description 4
- 108091033409 CRISPR Proteins 0.000 description 4
- 102100034458 Hepatitis A virus cellular receptor 2 Human genes 0.000 description 4
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 4
- 102000014961 Protein Precursors Human genes 0.000 description 4
- 108020004459 Small interfering RNA Proteins 0.000 description 4
- 230000006044 T cell activation Effects 0.000 description 4
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 4
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 4
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 4
- 230000006907 apoptotic process Effects 0.000 description 4
- 210000003719 b-lymphocyte Anatomy 0.000 description 4
- 230000004071 biological effect Effects 0.000 description 4
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 4
- 238000000502 dialysis Methods 0.000 description 4
- 230000004069 differentiation Effects 0.000 description 4
- 238000000684 flow cytometry Methods 0.000 description 4
- 230000004048 modification Effects 0.000 description 4
- 238000012986 modification Methods 0.000 description 4
- 238000010172 mouse model Methods 0.000 description 4
- 230000007886 mutagenicity Effects 0.000 description 4
- 231100000299 mutagenicity Toxicity 0.000 description 4
- 210000004882 non-tumor cell Anatomy 0.000 description 4
- 210000004940 nucleus Anatomy 0.000 description 4
- 238000011580 nude mouse model Methods 0.000 description 4
- 230000007918 pathogenicity Effects 0.000 description 4
- 108091007428 primary miRNA Proteins 0.000 description 4
- 230000008569 process Effects 0.000 description 4
- 238000012545 processing Methods 0.000 description 4
- 230000004853 protein function Effects 0.000 description 4
- 102000005962 receptors Human genes 0.000 description 4
- 108020003175 receptors Proteins 0.000 description 4
- 230000004083 survival effect Effects 0.000 description 4
- 239000000725 suspension Substances 0.000 description 4
- 230000008685 targeting Effects 0.000 description 4
- 102000053602 DNA Human genes 0.000 description 3
- 101710096438 DNA-binding protein Proteins 0.000 description 3
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- 108060002716 Exonuclease Proteins 0.000 description 3
- 108700039691 Genetic Promoter Regions Proteins 0.000 description 3
- 102000003886 Glycoproteins Human genes 0.000 description 3
- 108090000288 Glycoproteins Proteins 0.000 description 3
- 108020005004 Guide RNA Proteins 0.000 description 3
- 101100012844 Homo sapiens AFP gene Proteins 0.000 description 3
- 101710192606 Latent membrane protein 2 Proteins 0.000 description 3
- 206010025323 Lymphomas Diseases 0.000 description 3
- 241000699660 Mus musculus Species 0.000 description 3
- 101710163270 Nuclease Proteins 0.000 description 3
- 108010016790 RNA-Induced Silencing Complex Proteins 0.000 description 3
- 102000000574 RNA-Induced Silencing Complex Human genes 0.000 description 3
- 238000011529 RT qPCR Methods 0.000 description 3
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 3
- 235000014680 Saccharomyces cerevisiae Nutrition 0.000 description 3
- 241000700584 Simplexvirus Species 0.000 description 3
- 101710109576 Terminal protein Proteins 0.000 description 3
- 108091023040 Transcription factor Proteins 0.000 description 3
- 102000040945 Transcription factor Human genes 0.000 description 3
- 239000011324 bead Substances 0.000 description 3
- 230000002457 bidirectional effect Effects 0.000 description 3
- 239000000969 carrier Substances 0.000 description 3
- 230000024245 cell differentiation Effects 0.000 description 3
- 230000019522 cellular metabolic process Effects 0.000 description 3
- 238000005119 centrifugation Methods 0.000 description 3
- 230000009089 cytolysis Effects 0.000 description 3
- 102000013165 exonuclease Human genes 0.000 description 3
- 239000000710 homodimer Substances 0.000 description 3
- 230000001900 immune effect Effects 0.000 description 3
- 229940088592 immunologic factor Drugs 0.000 description 3
- 230000001976 improved effect Effects 0.000 description 3
- 230000008595 infiltration Effects 0.000 description 3
- 238000001764 infiltration Methods 0.000 description 3
- 230000003834 intracellular effect Effects 0.000 description 3
- 230000000670 limiting effect Effects 0.000 description 3
- 238000011068 loading method Methods 0.000 description 3
- 230000003472 neutralizing effect Effects 0.000 description 3
- 108700025694 p53 Genes Proteins 0.000 description 3
- 230000001737 promoting effect Effects 0.000 description 3
- 238000003753 real-time PCR Methods 0.000 description 3
- 230000006798 recombination Effects 0.000 description 3
- 238000005215 recombination Methods 0.000 description 3
- 238000007920 subcutaneous administration Methods 0.000 description 3
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 description 3
- 238000011144 upstream manufacturing Methods 0.000 description 3
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 2
- 229940123373 Adenovirus E1A gene Drugs 0.000 description 2
- 229920000936 Agarose Polymers 0.000 description 2
- 241000219194 Arabidopsis Species 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 208000023275 Autoimmune disease Diseases 0.000 description 2
- 108091032955 Bacterial small RNA Proteins 0.000 description 2
- 108700031361 Brachyury Proteins 0.000 description 2
- 101150091609 CD274 gene Proteins 0.000 description 2
- 101100506090 Caenorhabditis elegans hil-2 gene Proteins 0.000 description 2
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 2
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 2
- 230000004543 DNA replication Effects 0.000 description 2
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 2
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 2
- 108091005941 EBFP Proteins 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 206010017533 Fungal infection Diseases 0.000 description 2
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 2
- 108010068250 Herpes Simplex Virus Protein Vmw65 Proteins 0.000 description 2
- 101000858088 Homo sapiens C-X-C motif chemokine 10 Proteins 0.000 description 2
- 101100407305 Homo sapiens CD274 gene Proteins 0.000 description 2
- 101001120822 Homo sapiens Putative microRNA 17 host gene protein Proteins 0.000 description 2
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 2
- 241000598171 Human adenovirus sp. Species 0.000 description 2
- 241000700588 Human alphaherpesvirus 1 Species 0.000 description 2
- 108060003951 Immunoglobulin Proteins 0.000 description 2
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 2
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 2
- 102000010789 Interleukin-2 Receptors Human genes 0.000 description 2
- 108010038453 Interleukin-2 Receptors Proteins 0.000 description 2
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 2
- 108090000364 Ligases Proteins 0.000 description 2
- 102000003960 Ligases Human genes 0.000 description 2
- 101100407308 Mus musculus Pdcd1lg2 gene Proteins 0.000 description 2
- 208000031888 Mycoses Diseases 0.000 description 2
- 230000006051 NK cell activation Effects 0.000 description 2
- 241000244206 Nematoda Species 0.000 description 2
- 102000043276 Oncogene Human genes 0.000 description 2
- 108700020796 Oncogene Proteins 0.000 description 2
- 108700030875 Programmed Cell Death 1 Ligand 2 Proteins 0.000 description 2
- 102100024213 Programmed cell death 1 ligand 2 Human genes 0.000 description 2
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 2
- 102100038280 Prostaglandin G/H synthase 2 Human genes 0.000 description 2
- 102100038358 Prostate-specific antigen Human genes 0.000 description 2
- 108010078762 Protein Precursors Proteins 0.000 description 2
- 102000002727 Protein Tyrosine Phosphatase Human genes 0.000 description 2
- 102100026055 Putative microRNA 17 host gene protein Human genes 0.000 description 2
- 108091034057 RNA (poly(A)) Proteins 0.000 description 2
- 230000004570 RNA-binding Effects 0.000 description 2
- IWUCXVSUMQZMFG-AFCXAGJDSA-N Ribavirin Chemical compound N1=C(C(=O)N)N=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 IWUCXVSUMQZMFG-AFCXAGJDSA-N 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 230000006052 T cell proliferation Effects 0.000 description 2
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 2
- 102000043124 TIM family Human genes 0.000 description 2
- 108091054435 TIM family Proteins 0.000 description 2
- 210000000447 Th1 cell Anatomy 0.000 description 2
- 210000000068 Th17 cell Anatomy 0.000 description 2
- 230000003213 activating effect Effects 0.000 description 2
- 150000001413 amino acids Chemical class 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 238000000137 annealing Methods 0.000 description 2
- 239000002246 antineoplastic agent Substances 0.000 description 2
- 229940041181 antineoplastic drug Drugs 0.000 description 2
- 230000001640 apoptogenic effect Effects 0.000 description 2
- 208000006673 asthma Diseases 0.000 description 2
- 230000003305 autocrine Effects 0.000 description 2
- 210000003651 basophil Anatomy 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 210000004204 blood vessel Anatomy 0.000 description 2
- 108091092328 cellular RNA Proteins 0.000 description 2
- 230000007969 cellular immunity Effects 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 238000010367 cloning Methods 0.000 description 2
- 230000004186 co-expression Effects 0.000 description 2
- 230000002596 correlated effect Effects 0.000 description 2
- 230000001472 cytotoxic effect Effects 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 238000012217 deletion Methods 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 210000004443 dendritic cell Anatomy 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 201000010099 disease Diseases 0.000 description 2
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 2
- 230000003828 downregulation Effects 0.000 description 2
- 210000003979 eosinophil Anatomy 0.000 description 2
- 210000001339 epidermal cell Anatomy 0.000 description 2
- 210000002919 epithelial cell Anatomy 0.000 description 2
- 210000002950 fibroblast Anatomy 0.000 description 2
- 238000011331 genomic analysis Methods 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 2
- 239000008187 granular material Substances 0.000 description 2
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 2
- 231100000844 hepatocellular carcinoma Toxicity 0.000 description 2
- 210000005260 human cell Anatomy 0.000 description 2
- 210000004408 hybridoma Anatomy 0.000 description 2
- 230000005965 immune activity Effects 0.000 description 2
- 102000018358 immunoglobulin Human genes 0.000 description 2
- 230000016784 immunoglobulin production Effects 0.000 description 2
- 230000001506 immunosuppresive effect Effects 0.000 description 2
- 230000000415 inactivating effect Effects 0.000 description 2
- 230000002779 inactivation Effects 0.000 description 2
- 230000006698 induction Effects 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 229960003130 interferon gamma Drugs 0.000 description 2
- 229940117681 interleukin-12 Drugs 0.000 description 2
- 101150044508 key gene Proteins 0.000 description 2
- 108091053735 lin-4 stem-loop Proteins 0.000 description 2
- 108091032363 lin-4-1 stem-loop Proteins 0.000 description 2
- 108091028008 lin-4-2 stem-loop Proteins 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 210000000207 lymphocyte subset Anatomy 0.000 description 2
- 210000003810 lymphokine-activated killer cell Anatomy 0.000 description 2
- 239000006166 lysate Substances 0.000 description 2
- 239000012139 lysis buffer Substances 0.000 description 2
- 230000036210 malignancy Effects 0.000 description 2
- 108091091360 miR-125b stem-loop Proteins 0.000 description 2
- 108091007426 microRNA precursor Proteins 0.000 description 2
- 210000001616 monocyte Anatomy 0.000 description 2
- 230000007896 negative regulation of T cell activation Effects 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 230000010355 oscillation Effects 0.000 description 2
- 230000002018 overexpression Effects 0.000 description 2
- 230000003076 paracrine Effects 0.000 description 2
- 230000001717 pathogenic effect Effects 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 239000013600 plasmid vector Substances 0.000 description 2
- 229920000642 polymer Polymers 0.000 description 2
- 230000029279 positive regulation of transcription, DNA-dependent Effects 0.000 description 2
- 230000001323 posttranslational effect Effects 0.000 description 2
- 108020000494 protein-tyrosine phosphatase Proteins 0.000 description 2
- 230000007115 recruitment Effects 0.000 description 2
- 230000014493 regulation of gene expression Effects 0.000 description 2
- 210000003289 regulatory T cell Anatomy 0.000 description 2
- 238000010839 reverse transcription Methods 0.000 description 2
- 229960000329 ribavirin Drugs 0.000 description 2
- HZCAHMRRMINHDJ-DBRKOABJSA-N ribavirin Natural products O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1N=CN=C1 HZCAHMRRMINHDJ-DBRKOABJSA-N 0.000 description 2
- 102000012498 secondary active transmembrane transporter activity proteins Human genes 0.000 description 2
- 108040003878 secondary active transmembrane transporter activity proteins Proteins 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 230000011664 signaling Effects 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 210000004989 spleen cell Anatomy 0.000 description 2
- 210000000130 stem cell Anatomy 0.000 description 2
- 230000000638 stimulation Effects 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 230000036964 tight binding Effects 0.000 description 2
- 231100000331 toxic Toxicity 0.000 description 2
- 230000002588 toxic effect Effects 0.000 description 2
- 230000037426 transcriptional repression Effects 0.000 description 2
- 230000009261 transgenic effect Effects 0.000 description 2
- 230000001052 transient effect Effects 0.000 description 2
- 230000009385 viral infection Effects 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- 238000001262 western blot Methods 0.000 description 2
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- 102100030310 5,6-dihydroxyindole-2-carboxylic acid oxidase Human genes 0.000 description 1
- 241000711404 Avian avulavirus 1 Species 0.000 description 1
- 208000003174 Brain Neoplasms Diseases 0.000 description 1
- 108010076491 BsaI endonuclease Proteins 0.000 description 1
- 238000011740 C57BL/6 mouse Methods 0.000 description 1
- 238000011746 C57BL/6J (JAX™ mouse strain) Methods 0.000 description 1
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 1
- 101710132601 Capsid protein Proteins 0.000 description 1
- 102100034927 Cholecystokinin receptor type A Human genes 0.000 description 1
- 101710094648 Coat protein Proteins 0.000 description 1
- 108010037462 Cyclooxygenase 2 Proteins 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- 238000007702 DNA assembly Methods 0.000 description 1
- 102000005768 DNA-Activated Protein Kinase Human genes 0.000 description 1
- 108010006124 DNA-Activated Protein Kinase Proteins 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 238000008157 ELISA kit Methods 0.000 description 1
- 241000701959 Escherichia virus Lambda Species 0.000 description 1
- 108700039887 Essential Genes Proteins 0.000 description 1
- 108700024394 Exon Proteins 0.000 description 1
- 101150066002 GFP gene Proteins 0.000 description 1
- 206010064571 Gene mutation Diseases 0.000 description 1
- 102100021181 Golgi phosphoprotein 3 Human genes 0.000 description 1
- 101000721661 Homo sapiens Cellular tumor antigen p53 Proteins 0.000 description 1
- 101000946804 Homo sapiens Cholecystokinin receptor type A Proteins 0.000 description 1
- 101000725401 Homo sapiens Cytochrome c oxidase subunit 2 Proteins 0.000 description 1
- 101000608765 Homo sapiens Galectin-4 Proteins 0.000 description 1
- 101000605127 Homo sapiens Prostaglandin G/H synthase 2 Proteins 0.000 description 1
- 101710125418 Major capsid protein Proteins 0.000 description 1
- 102100025169 Max-binding protein MNT Human genes 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 206010027480 Metastatic malignant melanoma Diseases 0.000 description 1
- 108091007780 MiR-122 Proteins 0.000 description 1
- 108091033773 MiR-155 Proteins 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 206010028851 Necrosis Diseases 0.000 description 1
- 101710141454 Nucleoprotein Proteins 0.000 description 1
- 101710160107 Outer membrane protein A Proteins 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 101710083689 Probable capsid protein Proteins 0.000 description 1
- 108091007744 Programmed cell death receptors Proteins 0.000 description 1
- 241001112090 Pseudovirus Species 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- 238000002123 RNA extraction Methods 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 108700008625 Reporter Genes Proteins 0.000 description 1
- 208000007660 Residual Neoplasm Diseases 0.000 description 1
- 108020004682 Single-Stranded DNA Proteins 0.000 description 1
- 108091027967 Small hairpin RNA Proteins 0.000 description 1
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 1
- 101100038645 Streptomyces griseus rppA gene Proteins 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 108700025695 Suppressor Genes Proteins 0.000 description 1
- 108700026226 TATA Box Proteins 0.000 description 1
- 108010017842 Telomerase Proteins 0.000 description 1
- 108091036066 Three prime untranslated region Proteins 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 108700025716 Tumor Suppressor Genes Proteins 0.000 description 1
- 102000044209 Tumor Suppressor Genes Human genes 0.000 description 1
- 108010064978 Type II Site-Specific Deoxyribonucleases Proteins 0.000 description 1
- 108090000848 Ubiquitin Proteins 0.000 description 1
- 102000044159 Ubiquitin Human genes 0.000 description 1
- 108020000999 Viral RNA Proteins 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 240000008042 Zea mays Species 0.000 description 1
- 235000016383 Zea mays subsp huehuetenangensis Nutrition 0.000 description 1
- 235000002017 Zea mays subsp mays Nutrition 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 101150055123 afp gene Proteins 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- 230000005975 antitumor immune response Effects 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 230000001363 autoimmune Effects 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 108700041737 bcl-2 Genes Proteins 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 230000008827 biological function Effects 0.000 description 1
- 230000005540 biological transmission Effects 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- 230000003157 cancerolytic effect Effects 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000022534 cell killing Effects 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 230000006037 cell lysis Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000010307 cell transformation Effects 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 238000012411 cloning technique Methods 0.000 description 1
- 238000000749 co-immunoprecipitation Methods 0.000 description 1
- 238000012761 co-transfection Methods 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 238000005520 cutting process Methods 0.000 description 1
- 230000007402 cytotoxic response Effects 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 239000003480 eluent Substances 0.000 description 1
- 230000013020 embryo development Effects 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 230000007613 environmental effect Effects 0.000 description 1
- 238000001976 enzyme digestion Methods 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 230000007849 functional defect Effects 0.000 description 1
- 150000002256 galaktoses Chemical class 0.000 description 1
- 230000030279 gene silencing Effects 0.000 description 1
- 238000003144 genetic modification method Methods 0.000 description 1
- 201000010536 head and neck cancer Diseases 0.000 description 1
- 208000014829 head and neck neoplasm Diseases 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 238000007490 hematoxylin and eosin (H&E) staining Methods 0.000 description 1
- 238000000265 homogenisation Methods 0.000 description 1
- 230000006658 host protein synthesis Effects 0.000 description 1
- 102000007579 human kallikrein-related peptidase 3 Human genes 0.000 description 1
- 108010071652 human kallikrein-related peptidase 3 Proteins 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 239000000367 immunologic factor Substances 0.000 description 1
- 238000001114 immunoprecipitation Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 238000011081 inoculation Methods 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- PGHMRUGBZOYCAA-UHFFFAOYSA-N ionomycin Natural products O1C(CC(O)C(C)C(O)C(C)C=CCC(C)CC(C)C(O)=CC(=O)C(C)CC(C)CC(CCC(O)=O)C)CCC1(C)C1OC(C)(C(C)O)CC1 PGHMRUGBZOYCAA-UHFFFAOYSA-N 0.000 description 1
- PGHMRUGBZOYCAA-ADZNBVRBSA-N ionomycin Chemical compound O1[C@H](C[C@H](O)[C@H](C)[C@H](O)[C@H](C)/C=C/C[C@@H](C)C[C@@H](C)C(/O)=C/C(=O)[C@@H](C)C[C@@H](C)C[C@@H](CCC(O)=O)C)CC[C@@]1(C)[C@@H]1O[C@](C)([C@@H](C)O)CC1 PGHMRUGBZOYCAA-ADZNBVRBSA-N 0.000 description 1
- 238000005304 joining Methods 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 239000012160 loading buffer Substances 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 235000009973 maize Nutrition 0.000 description 1
- 108010082117 matrigel Proteins 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000008384 membrane barrier Effects 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 230000001394 metastastic effect Effects 0.000 description 1
- 208000021039 metastatic melanoma Diseases 0.000 description 1
- 206010061289 metastatic neoplasm Diseases 0.000 description 1
- 108091051828 miR-122 stem-loop Proteins 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 239000010447 natron Substances 0.000 description 1
- 230000017074 necrotic cell death Effects 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 230000003121 nonmonotonic effect Effects 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 238000012858 packaging process Methods 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 238000009521 phase II clinical trial Methods 0.000 description 1
- 238000009522 phase III clinical trial Methods 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 230000001124 posttranscriptional effect Effects 0.000 description 1
- 239000002244 precipitate Substances 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 239000003531 protein hydrolysate Substances 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 238000004445 quantitative analysis Methods 0.000 description 1
- 230000008439 repair process Effects 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- 231100000241 scar Toxicity 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 239000012679 serum free medium Substances 0.000 description 1
- 208000037968 sinus cancer Diseases 0.000 description 1
- 239000004055 small Interfering RNA Substances 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 230000009870 specific binding Effects 0.000 description 1
- 108010068698 spleen exonuclease Proteins 0.000 description 1
- 210000004988 splenocyte Anatomy 0.000 description 1
- 230000010473 stable expression Effects 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 239000011550 stock solution Substances 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 230000009469 supplementation Effects 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 108091006107 transcriptional repressors Proteins 0.000 description 1
- 239000012096 transfection reagent Substances 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 230000001960 triggered effect Effects 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 239000000107 tumor biomarker Substances 0.000 description 1
- 230000005740 tumor formation Effects 0.000 description 1
- 230000005909 tumor killing Effects 0.000 description 1
- 231100000588 tumorigenic Toxicity 0.000 description 1
- 230000000381 tumorigenic effect Effects 0.000 description 1
- 108010014402 tyrosinase-related protein-1 Proteins 0.000 description 1
- 238000002255 vaccination Methods 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 238000012795 verification Methods 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 238000003466 welding Methods 0.000 description 1
Images
Classifications
-
- 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
- C12N15/86—Viral vectors
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/76—Viruses; Subviral particles; Bacteriophages
- A61K35/761—Adenovirus
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/76—Viruses; Subviral particles; Bacteriophages
- A61K35/768—Oncolytic viruses not provided for in groups A61K35/761 - A61K35/766
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/0005—Vertebrate antigens
- A61K39/0011—Cancer antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
-
- 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
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/53—Colony-stimulating factor [CSF]
- C07K14/535—Granulocyte CSF; Granulocyte-macrophage CSF
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/54—Interleukins [IL]
- C07K14/5434—IL-12
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/54—Interleukins [IL]
- C07K14/5443—IL-15
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/54—Interleukins [IL]
- C07K14/55—IL-2
-
- 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
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- 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
- C12N7/00—Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/525—Virus
- A61K2039/5256—Virus expressing foreign proteins
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/57—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
- A61K2039/575—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
-
- 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
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/14—Type of nucleic acid interfering N.A.
-
- 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
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/10011—Adenoviridae
- C12N2710/10034—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
-
- 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
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/10011—Adenoviridae
- C12N2710/10041—Use of virus, viral particle or viral elements as a vector
- C12N2710/10043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- 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
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/10011—Adenoviridae
- C12N2710/10311—Mastadenovirus, e.g. human or simian adenoviruses
- C12N2710/10334—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
-
- 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
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/10011—Adenoviridae
- C12N2710/10311—Mastadenovirus, e.g. human or simian adenoviruses
- C12N2710/10341—Use of virus, viral particle or viral elements as a vector
- C12N2710/10343—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
Definitions
- the present invention relates to the field of biomedicine, in particular, the present invention relates to an expression system and its use, and more particularly, to the use of an expression system, a recombinant virus, a recombinant cell, and an expression system, a recombinant virus, and a recombinant cell in the preparation of a medicament, Further, the present invention also relates to a method of expressing a protein of interest using an expression system.
- Oncolytic virus refers to a type of virus that has the ability to replicate and package to achieve tumor killing. At present, most studies have modified the expression of some of the less virulent strains in nature, allowing them to express their packaging in tumor cells and then achieve oncolysis. There are two main principles for using tumorigenic viruses to recognize tumor cells. First, they selectively infect tumor cells by inactivating or defective tumor suppressor genes in target cells. Second, they selectively use tumor-specific promoters to regulate viruses. The expression of key genes allows the oncolytic virus to replicate in large amounts in tumor cells and express toxic proteins to destroy tumor cells, and/or simultaneously secrete cytokines to stimulate the autoimmune system to attack tumor cells.
- oncolytic virus cannot replicate in normal organism cells without killing effect, so the oncolytic virus has higher anti-tumor effect and lower side effects.
- oncolytic virus treatment has caused widespread concern, and relevant research has made great progress.
- adenovirus, herpes simplex virus-1 (HSV-1), Newcastle disease virus, etc. have been transformed into oncolytic viruses.
- HSV-1 herpes simplex virus-1
- Newcastle disease virus etc.
- oncolytic adenovirus products (Gendicine and oncorine) have been used in clinical treatment in China, mainly for the treatment of head and neck cancer, sinus cancer. The principle of Gendicine and oncorine is similar.
- OncoVEX GM-CSF The genetically engineered herpes simplex virus OncoVEX GM-CSF, developed by biotechnology company BioVex, was approved by the FDA in October 2015 to become the first oncolytic virus product to be marketed in the US and Europe.
- OncoVex selectively kills tumor cells while expressing an immune response that secretes GM-CSF to initiate the body's immune system to kill residual tumor cells and their metastatic sites.
- a phase II trial of metastatic melanoma published by BioVex in 2009 showed that 26% of 50 patients responded to treatment and 8 patients returned to full health. The company was acquired by Agmen for $1 billion in 2011 for advancement.
- Adl ⁇ Ad52 human adenoviruses
- Ad2A and E1B which are involved in cell transformation
- E2 region encodes DNA binding.
- the protein is involved in viral replication; the E3 region encodes a glycoprotein that appears on the surface of the host cell; the E4 region is located at the right end of the Ad2 genome and is regulated by the DNA binding protein encoded by the E2 region; the fifth transcription unit synthesizes the Ad2 protein during the viral infection.
- the cellular transcription factor first binds to the enhancer upstream of the E1A region, expressing the E1A protein, which regulates cellular metabolism and makes the viral DNA more easily replicated in the cell.
- the E1A protein also activates promoters of other early genes (E1B, E2A, E2B, E3, and E4), where E2B drives three additional early gene transcriptional terminal protein precursors (pTPs) involved in viral replication.
- ssDBP single-stranded DNA binding proteins
- DNA pol DNA polymerase
- E1A expression to regulate the replication of packaging of adenovirus in targeted cells
- removal of some viral packaging non-essential genes to reduce virus-to-non- The toxicity of the target cells, while increasing the packaging capacity of the virus, such as E3, E4, etc.
- replacing the coat protein of the adenovirus changing the targeting of the virus to specific cells and tissues.
- the present invention aims to solve at least one of the related technical problems to some extent.
- the inventors designed and constructed a gene line that is suitable for adenovirus regulation and can respond to different microenvironments.
- this genetic line the inventors utilized multiple levels of biomarkers. Specifically, first, a specific promoter is used as a master switch for regulating gene lines, that is, a specific promoter is used to regulate the expression of the total activator, and the expression of the adenovirus E1A gene is further regulated. Second, the target sequence of the microRNA is used to respond to the microRNA expression characteristics of different microenvironments as a secondary switch for regulating gene lines.
- the closed two-way suppression switch can respond to changes in the microenvironment more efficiently, while effectively avoiding Give way.
- the E1B gene was removed, and the ability of E1B to recognize the complementary deletion of P53 gene further improved the ability of adenovirus to distinguish tumor cells from normal cells.
- the removal of the E3 gene reduces the toxic effects of oncolytic adenovirus on normal cells and increases the packaging capacity of adenoviral vectors.
- the genome length of type 2 and type 5 human adenoviruses is 36K. It is very difficult and time consuming to reconstruct adenovirus using traditional single plasmid construction methods (such as restriction enzyme ligation).
- the inventors designed a rapid construction method for the adenovirus constructed by the gene line involved in the present invention: the first step is to construct a first-level component library, and the desired components are constructed on the corresponding plasmids.
- the primary component library includes three libraries: a suppressor element A library mainly expressing a suppressor element on one side of the gene line, an E1A gene of an adenovirus, and an effector gene (such as an immune factor or a killer gene) co-expressed with an adenovirus; and a suppressor element B
- the library mainly expresses the suppressor gene on the other side of the gene line, regulates the effective flipping of the gene line, and enhances the safety of the switch; the specific promoter library regulates the total switch of the downstream gene line through the tissue or tumor-specific promoter.
- the third step is to integrate the gene line into the modified adenoviral vector by Gateway (removing the E1 gene to facilitate manual control of the adenovirus; removing part of the E3 sequence to expand the packaging capacity).
- oncolytic adenoviruses can carry and simultaneously express multiple genes.
- the inventors simultaneously expressed single or multiple cellular immune-related genes: such as IL-2, GM-CSF, and anti- PD-1 scFv, anti-PD-L1 scFv, and fusion proteins between these factors and the like.
- immune-related genes such as IL-2, GM-CSF, and anti- PD-1 scFv, anti-PD-L1 scFv, and fusion proteins between these factors and the like.
- oncolytic viruses Due to the carrying of immune-related genes, oncolytic viruses cause systemic immune responses while attacking tumor cells, but there are also risks and hidden dangers that cause immune overreaction. Therefore, what kind of immune response gene is carried, and the corresponding treatment of how much dose of virus, as well as the mode of administration of the virus treatment, strongly influence the therapeutic effect of the oncolytic virus.
- the inventors attempted to model the process of infecting adenovirus-infected target cells by means of bioinformatics, and studied the characteristics of oncolytic adenovirus killing tumor cells, and improved the treatment of oncolytic adenovirus as much as possible. effect.
- the invention proposes an expression system.
- the expression system comprises: a first nucleic acid molecule comprising a cell-specific promoter; a second nucleic acid molecule, the second nucleic acid molecule and the first nucleic acid molecule Operably linked, the second nucleic acid molecule encoding a transcriptional activator; a third nucleic acid molecule comprising a first recognition sequence of the transcriptional activator; a fourth nucleic acid molecule, the fourth nucleic acid molecule
- the third nucleic acid molecule is operably linked, the fourth nucleic acid molecule comprising a first promoter and a first regulatory element; a fifth nucleic acid molecule, the fifth nucleic acid molecule being operably linked to a fourth nucleic acid molecule,
- the fifth nucleic acid molecule encodes a first regulatory protein; a sixth nucleic acid molecule comprising a second recognition sequence of the transcriptional activator; a seventh nucleic acid molecule
- the expression system may further comprise at least one of the following additional technical features:
- the cell-specific promoter is a tumor cell-specific promoter selected from the group consisting of an alpha-fetoprotein-specific promoter, a Survivin gene promoter, and a human telomerase.
- Reverse transcriptase gene promoter cholecystokinin A receptor gene promoter, carcinoembryonic antigen promoter, proto-oncogene human epidermal growth factor receptor 2 promoter, prostaglandin internal oxidase reductase 2 promoter, chemokine At least one of receptor-4, E2F-1 gene promoter, mucin promoter, prostate specific antigen, human tyrosinase-related protein 1, and tyrosinase promoter.
- the expression system of the examples of the present application can be initiated in the microenvironment of specific tumor cells, and the specificity of the expression system for gene expression regulation is further enhanced.
- the transcriptional activator is at least one selected from the group consisting of Gal4VP16, Gal4VP64, Gal4esn, dCas9-VP16, dCas9-VP64, dCas9-VPR, dCas9-VTR, and rtTA.
- the first recognition sequence and the second recognition sequence are each independently selected from at least one of a target sequence of 5 ⁇ UAS, 7 ⁇ tetO and dCas9.
- the first promoter and the second promoter are each independently selected from a miniCMV, a TATA box.
- the first regulatory protein and the second regulatory protein are each independently selected from at least one of Lacl, tetR, zinc finger (zinc finger), KRAB, tetR-KRAB, dCas9-KRAB.
- the first regulatory element and the second regulatory element are each independently selected from at least one of a tetO, a LacO, a zinc finger target site, and a target sequence of dCas9.
- the first regulatory protein is LacI
- the second regulatory element comprises a plurality of repeating LacO sequences, at least one of the plurality of repeated LacO sequences being set at the second promoter Downstream.
- the LacO sequence can be specifically bound, thereby inhibiting the function of the second promoter.
- the LacI/LacO inhibition system of the embodiment of the present invention it has been experimentally shown that the system can effectively inhibit the expression of genes downstream of the promoter.
- the second regulatory protein is tetR-KRAB
- the first regulatory element comprises a plurality of repeated tetO sequences, at least one of the plurality of repeated tetO sequences being set at the first initiation Downstream of the child.
- At least one of the fifth nucleic acid molecule and the ninth nucleic acid molecule further comprises a sequence encoding a protein of interest.
- the fifth nucleic acid molecule comprises a sequence encoding the protein of interest, and the protein of interest comprises at least one selected from the group consisting of a viral replication packaging protein and an immune effector.
- the viral replication packaging protein, immune effector may be present in the form of a fusion protein.
- the viral replication packaging protein can effectively ensure the survival and replication of the expression system vector in the host; the expression of the immune effector can effectively activate the body's immune system, thereby promoting immune killing of specific cells such as tumor cells.
- the virus replication packaging-related protein comprises at least one selected from the group consisting of an adenovirus E1 gene, an E1A gene, an E1B gene, an E2 gene, and an E4 gene.
- the immune effector comprises a sequence selected from the group consisting of an inhibitory sequence that antagonizes the PD-1 gene, an inhibitory sequence that antagonizes the PD-L1 gene, an inhibitory sequence that antagonizes the CTLA4 gene, an inhibitory sequence that antagonizes the Tim-3 gene, and GM.
- - at least one of CSF, IL-2, IL-12, IL-15.
- the above immune effector factors may be present in the form of a fusion protein.
- the target protein and the first regulatory protein are expressed in the form of a fusion protein, and the target protein and the first regulatory protein are linked by a cleavable linker peptide.
- the target protein and the first regulatory protein are regulated and expressed under the same promoter, and are expressed after being cleaved at the linker peptide, and the target protein is separated from the first regulatory protein, and the target protein and the first regulatory protein function independently of each other.
- the ninth nucleic acid molecule and the tenth nucleic acid molecule independently inhibit expression of the first regulatory protein or the second regulatory protein, respectively, by RNA interference.
- the microRNA is a specific microRNA expressed by different cellular microenvironments
- the ninth or tenth nucleic acid molecule is a specific target sequence of the microRNA, and then the microRNA target expressed in the specific microenvironment can be realized by RNA interference (RNAi)
- RNAi RNA interference
- the ninth nucleic acid molecule comprises a nucleic acid sequence specifically recognized by a first microRNA, the tenth nucleic acid molecule comprising a nucleic acid sequence specifically recognized by a second microRNA, the first microRNA being normal A cell-specific microRNA, the second microRNA being an abnormal cell-specific microRNA.
- the first regulatory protein is expressed in abnormal cells, and is not expressed or underexpressed in normal cells, while the second regulatory protein is expressed in normal cells and is not expressed or expressed in abnormal cells.
- the first microRNA comprises at least one selected from the group consisting of miR199a, miR95, miR125, miR125b, Let-7, miR143, miR145 and miR200C.
- the above microRNA is highly expressed in normal liver cells.
- the second microRNA comprises at least one selected from the group consisting of miR21, miR223, miR224, miR221, miR18, miR214, miR146a and miR1792.
- the second microRNA is a microRNA specifically expressed by liver cancer cells (HepG2, Huh7, PLC). Furthermore, the first regulatory protein is expressed in liver cancer cells, and is not expressed or expressed in normal cells, while the second regulatory protein is expressed in normal cells and is not expressed or expressed in liver cancer cells.
- the first nucleic acid molecule and the second nucleic acid molecule are supported on a first expression vector
- the selected ninth nucleic acid molecule is supported on a second expression vector
- the sixth nucleic acid molecule, the seventh nucleic acid molecule, the eighth nucleic acid molecule, and optionally the tenth nucleic acid molecule are loaded in a third On the expression vector.
- the first, second and third expression vectors serve as a loading vector for the expression system to effect regulation of the specific expression of the gene of interest in a suitable microenvironment, such as a cell.
- the selection of the expression vector is not particularly limited as long as the function of the expression system in a suitable microenvironment can be achieved.
- the first expression vector, the second expression vector and the third expression vector are each independently selected from at least one of the following:
- Plasmids, viruses, stable cell lines, and other material carriers such as nanomaterials, liposomes, molecularly coupled vectors, naked DNA, chromosomal vectors, polymers.
- the virus comprises at least one selected from the group consisting of an adenovirus, a vaccinia virus, a herpes virus, and a retrovirus.
- the first expression vector, the second expression vector and the third expression vector are constructed to be loaded on the same vector.
- the order of joining the first expression vector, the second expression vector, and the third expression vector is not particularly limited as long as it does not affect the realization of the biological function of the system.
- loading on the same expression vector can effectively solve the problem that the co-transfection efficiency of a plurality of large fragment vectors is extremely low.
- the same vector is an adenoviral vector.
- adenovirus As a gene therapy vector, adenovirus has a wide range of main genes, low pathogenicity to humans, infection and expression of genes in proliferating and non-proliferating cells, high titer, homology with human genes, no insertional mutagenicity, and can be cultured in suspension. The advantage of amplifying in the liquid and simultaneously expressing multiple genes.
- the first expression vector comprises BsaI, AFP III, Gal4VP16 and BsaI (BsaI-AFP III-Gal4VP16-BsaI) from the 5' end to the 3' end.
- the first expression vector carries a nucleic acid having the nucleotide sequence set forth in SEQ ID NO: 1.
- the second expression vector comprises: BsaI, 5 ⁇ UAS, tetO, miniCMV, tetO, E1A, 2A, immune effector (Effector), LacI, microRNA199a from 5' to 3'.
- BsaI BsaI-5 ⁇ UAS-tetO-miniCMV-tetO-E1A-2A-Effector-LacI-miR199a target site-BsaI).
- the effector specifically refers to IL-2, hGM-CSF, mGM-CSF, anti-PD-1 scFv, anti-PD-L1 scFv, IL-2 fusion anti-PD-1 scfv, hGM-CSF fusion anti- PD-1scfv, mGM-CSF fusion anti-PD-1scfv, IL-2 fusion anti-PD-L1scfv, hGM-CSF fusion anti-PD-L1scfv, mGM-CSF fusion anti-PD-L1scfv.
- the second expression vector carries a nucleic acid having the nucleotide sequence shown in any one of SEQ ID NOS: 2 to 7.
- SEQ ID NO: 2 is the sequence of BsaI-5 ⁇ UAS-tetO-miniCMV-tetO-E1A-2A-EBFP-2A-LacI-miR199a target site-BsaI;
- SEQ ID NO: 3 is the sequence of BsaI-5 ⁇ UAS-tetO-miniCMV-tetO-E1A-2A-hIL-2-2A-LacI-miR199a target site-BsaI;
- SEQ ID NO: 4 is the sequence of BsaI-5 ⁇ UAS-tetO-miniCMV-tetO-E1A-2A-hGM-CSF-2A-LacI-miR199a target site-BsaI;
- SEQ ID NO: 5 is the sequence of BsaI-5 ⁇ UAS-tetO-miniCMV-tetO-E1A-2A-mGM-CSF-2A-LacI-miR199a target site-BsaI;
- SEQ ID NO: 6 is the sequence of BsaI-5 ⁇ UAS-tetO-miniCMV-tetO-E1A-2A-anti-PD-1scFv-2A-LacI-miR199a target site-BsaI;
- SEQ ID NO: 7 is the sequence of BsaI-5 x UAS-tetO-miniCMV-tetO-E1A-2A-anti-PD-L1scFv-2A-LacI-miR199a target site-BsaI.
- the third expression vector comprises BsaI, 5 ⁇ UAS, LacO, miniCMV, LacO, tetR-KRAB, microRNA21 specific target site and BsaI from 5' to 3'.
- the third expression vector carries a nucleic acid having the nucleotide sequence set forth in SEQ ID NO: 8.
- the adenovirus comprises: from 5' to 3', inverted terminal repeat ITR, package signal, AFPIII, Gal4VP16, 5 x UAS ,tetO,miniCMV,tetO,E1A,2A,Effector,2A,LacI,miR199a target site,5 ⁇ UAS,LacO,miniCMV,LacO,tetR-KRAB,miR21target site,adenovirus E2 gene region,E3 gene region (removed Region 28922-30801), E4 gene region, second inverted terminal repeat (ITR).
- the inventors injected the above adenovirus into a tumor mouse model to significantly inhibit the growth of mouse tumors.
- the above adenovirus can be used as a safe and effective oncolytic virus vaccine to achieve safe and effective specific killing of related tumors.
- the adenoviral vector carries a nucleic acid having the nucleotide sequence shown in SEQ ID NOS: 9 to 14.
- SEQ ID NO: 9 is the sequence of the EBFP-bearing nucleic acid carried by the adenoviral vector
- SEQ ID NO: 10 is the sequence of a nucleic acid carrying hIL-2 carried by an adenoviral vector
- SEQ ID NO: 11 is the sequence of a nucleic acid carrying hGMCSF carried by an adenoviral vector
- SEQ ID NO: 12 is the sequence of a nucleic acid carrying mGMCSF carried by an adenoviral vector
- SEQ ID NO: 13 is the sequence of a nucleic acid carrying an anti-PD-1 scFv carried by an adenoviral vector
- SEQ ID NO: 14 is the sequence of a nucleic acid carrying an anti-PD-L1 scFv carried by an adenoviral vector.
- the adenovirus is obtained by removing the E1 gene and a part of the E3 gene associated with adenoviral replication packaging, and the E1A gene is constructed by a stepwise Golden Gate method.
- the logic is finally inserted into the adenoviral vector by way of Gateway or Gibson.
- the invention proposes a recombinant virus.
- the recombinant virus comprises: a first nucleic acid molecule, the first nucleic acid molecule comprising a tumor cell-specific promoter, and the tumor cell-specific promoter is an alpha-fetoprotein-specific promoter; a nucleic acid molecule operably linked to the first nucleic acid molecule, the second nucleic acid molecule encoding a transcriptional activator, the transcriptional activator is Gal4VP16; a third nucleic acid molecule, the third The nucleic acid molecule comprises a first recognition sequence of the transcriptional activator, the first recognition sequence being 5 x UAS; a fourth nucleic acid molecule operably linked to the third nucleic acid molecule, The fourth nucleic acid molecule comprises a first promoter and a first regulatory element, the first promoter is a miniCMV, and the first regulatory element comprises a plurality of repeating tet
- the first regulatory protein LacI and the target protein are specifically expressed in tumor cells
- the second regulatory protein tetR-KRAB is specifically not expressed or underexpressed in tumor cells, and thus the inhibition mechanism of the first promoter miniCMV mediated by tetR-KRAB is released, the first regulatory protein LacI, the target protein in the first promoter miniCMV Under the control of efficacious expression, the LacI-mediated inhibition mechanism effectively inhibited the function of the second promoter miniCMV, and the expression of tetR-KRAB was further inhibited.
- expression of the protein in the tumor cell can be more specifically expressed (such as the target protein Lacl) or not expressed (such as tetR-KRAB), and the expression efficiency or non-expression efficiency in the tumor cell can be achieved. And high specificity.
- the above recombinant virus may further include at least one of the following additional technical features:
- the recombinant virus is at least one selected from the group consisting of a retrovirus, an adenovirus, a herpes virus, and a vaccinia virus.
- the recombinant virus is an adenovirus.
- adenovirus has a wide host range as a gene therapy vector, low pathogenicity to humans, infection and expression of genes in proliferating and non-proliferating cells, high titer, homology to human genes, and no insertional mutagenicity. The advantage of being able to amplify in suspension culture and simultaneously express multiple genes.
- the immune effector comprises a restriction sequence selected from the group consisting of an antagonistic PD-1 gene, an inhibitory sequence which antagonizes the PD-L1 gene, an inhibitory sequence which antagonizes the CTLA4 gene, an inhibitory sequence which antagonizes the Tim-3 gene, and IL. - 2, at least one of IL-15, IL-12, GM-CSF.
- the immune effector may be in the form of a fusion protein.
- the invention proposes a recombinant cell.
- the recombinant cell comprises an expression system as described above.
- the recombinant cell according to the embodiment of the invention can effectively activate the systemic immune response of the human body, and attack the xenogeneic cells, such as tumor cells, with high safety and specificity.
- the above recombinant cell may further comprise at least one of the following additional technical features:
- At least a portion of the expression system is integrated into the genome of the recombinant cell.
- the expression system replicates as the recombinant cell genome replicates, and the expression system continues to be effective in regulating the expression of the protein of interest.
- the invention provides the use of the expression system described above, the recombinant virus described above, and the recombinant cells described above for the preparation of a medicament for the treatment of cancer.
- the expression system described in the present application can achieve specific expression of a target protein in tumor cells, and the drug according to the embodiment of the present application is more effective, specific, and safer for treating cancer.
- the above use may further include at least one of the following additional technical features:
- the cancer comprises liver cancer, lung cancer, colorectal cancer, melanoma, breast cancer or prostate cancer.
- the inventors have found that the therapeutic effect of the drug according to the embodiment of the present invention on liver cancer, lung cancer, colorectal cancer, melanoma, breast cancer or prostate cancer is more remarkable.
- the invention proposes a method of expressing a protein of interest using an expression system, which is the expression system described above.
- the method comprises: (1) causing the fifth nucleic acid molecule to comprise a nucleic acid sequence encoding the protein of interest; (2) inhibiting the tenth nucleic acid molecule from the second regulatory protein Expression to express the protein of interest.
- the second regulatory protein-mediated inhibition mechanism of the first promoter is released, and the target protein is co-operated by the cell-specific promoter and the first promoter. Highly expressed in specific cells.
- the above method may further include at least one of the following additional technical features:
- the expression is carried out in a cell.
- the cells can provide a microenvironment for the expression of the protein of interest, and the expression of the protein of interest in the cell is more efficient.
- the tenth nucleic acid molecule comprises a nucleic acid sequence specifically recognized by a second microRNA, and in step (2) comprises contacting the second microRNA with the tenth nucleic acid molecule.
- the invention proposes a method of expressing a protein of interest using an expression system, which is the expression system described above.
- the method comprises: (1) causing the eighth nucleic acid molecule to comprise a nucleic acid sequence encoding a first protein of interest, the fifth nucleic acid molecule comprising a nucleic acid sequence encoding a second protein of interest; 2) expressing the first protein of interest or the second protein of interest by one of: inhibiting expression of the first regulatory protein by the ninth nucleic acid molecule to express the first protein of interest; A tenth nucleic acid molecule inhibits expression of the second regulatory protein to express the second protein of interest.
- the second regulatory protein-mediated inhibition of the first promoter is released, and the second protein of interest is co-operating with the cell-specific promoter and the first promoter.
- the first protein of interest is efficiently activated in a specific cell by a combination of a cell-specific promoter and a second promoter. expression.
- fusion protein described herein refers to a protein that is co-transcribed under the control of the same promoter, including a fusion protein that is not linked between proteins, or a fusion protein linked by other linker peptides (such as GGGS or 2A sequence). .
- Figure 1 is a schematic diagram of the original structure of the adenovirus of the Gateway system
- FIG. 1 Schematic diagram of the original structure of the Adenovirus of the Golden Gate system
- Figure 4 is a diagram showing the detection of the hAFP gene in the cell line Chang by the method of real-time quantitative PCR according to an embodiment of the present invention. Expression levels in HepG2, Huh7, Hep3B, PLC and Hepa1-6;
- Figure 5 is a promoter for engineering a human AFP gene according to an embodiment of the present invention.
- FIG. 6 is a schematic transmission system of the modified AFP promoter to Chang and HepG2 according to an embodiment of the present invention
- FIG. 7 is a schematic diagram of a microRNA detection system in accordance with an embodiment of the present invention.
- Figure 8 is a diagram showing significant differential expression of microRNA markers in different cell lines in accordance with an embodiment of the present invention.
- FIG. 9 is a schematic diagram of a double suppression switch based on the LacI and tetR-KRAB genes according to an embodiment of the present invention.
- FIG. 10 is a diagram showing a dual suppression switch in response to a synthetic shRNA signal to achieve efficient flipping in accordance with an embodiment of the present invention
- 11 is a dual suppression switch in response to various activation factors in accordance with an embodiment of the present invention.
- CGGA Cluster Golden-Gate and Gateway/Gibson Assemble method
- Figure 13 is a second step of a CGGA (Cascade Golden-Gate and Gateway/Gibson Assemble method) according to an embodiment of the present invention
- CGGA Cluster Golden-Gate and Gateway/Gibson Assemble method
- Figure 15 is a diagram showing the effect of detecting a virus at a cellular level according to an embodiment of the present invention.
- Figure 16 is a graph showing in vitro detection of cytokine activity (plasmid vector expressing cytokine) according to an embodiment of the present invention.
- Figure 17 is a graph showing in vitro detection of cytokine activity (viral vector expressing cytokine) according to an embodiment of the present invention.
- Figure 18 is a nude mouse model for treating HepG2, Huh7 and Hepa1-6 using oncolytic adenovirus according to an embodiment of the present invention
- 19 is a C57 mouse model for treating Hepa1-6 using an oncolytic adenovirus according to an embodiment of the present invention.
- Figure 20 is a graph showing the expression of T cell gamma interferon and Ki67 markers for oncolytic virus treatment for tumor infiltration according to an embodiment of the present invention
- Figure 21 is a challenge experiment of mouse liver cancer cell Hepa1-6 for re-inoculation of mice after treatment according to an embodiment of the present invention
- Figure 22 is a tumor-virus-immune system model in accordance with an embodiment of the present invention.
- Figure 23 is a graph showing the rate of virus replication in a tumor-virus system simulating a minimum tumor volume under different initial tumor titers and initial tumor sizes under different initial tumor sizes, initial viral titers, and viral replication rates, in accordance with an embodiment of the present invention.
- the tumor-virus system changes with time to the final state, the minimum value of the tumor volume changes;
- Figure 24 is a graph showing the final state tumor volume under the influence of the tumor-virus-immunological system simulation immune cell on the different inhibitory effects of tumor and virus according to an embodiment of the present invention.
- the invention proposes an expression system.
- the expression system comprises: a first nucleic acid molecule comprising a cell-specific promoter; a second nucleic acid molecule, the second nucleic acid molecule and the first nucleic acid molecule Operably linked, the second nucleic acid molecule encoding a transcriptional activator; a third nucleic acid molecule comprising a first recognition sequence of the transcriptional activator; a fourth nucleic acid molecule, the fourth nucleic acid molecule
- the third nucleic acid molecule is operably linked, the fourth nucleic acid molecule comprising a first promoter and a first regulatory element; a fifth nucleic acid molecule, the fifth nucleic acid molecule being operably linked to a fourth nucleic acid molecule,
- the fifth nucleic acid molecule encodes a first regulatory protein; a sixth nucleic acid molecule comprising a second recognition sequence of the transcriptional activator; a seventh nucleic acid molecule
- the cell-specific promoter is a tumor cell-specific promoter selected from the group consisting of an alpha-fetoprotein-specific promoter (AFP) and a Survivin gene promoter (SUR).
- Human telomerase reverse transcriptase (hTERT) gene promoter cholecystokinin A receptor gene promoter (CCKAR promoter), carcinoembryonic antigen promoter (CEA promoter), proto-oncogene human epidermal growth factor receptor 2 Human epidermal growth factor receptor-2 (HER2), prostaglandin oxidase reductase 2 promoter (Cyclooxygenase 2, COX2), chemokine receptor-4 (CXCR4), E2F-1 gene Promoter (E2F-1promoter), mucin promoter (Mucin1MUC1), prostate specific antigen (Prostate-specific antigen PSA), human tyrosine related protein 1 (TRP1), tyrosinase promoter (ty)
- the transcriptional activator is at least one selected from the group consisting of Gal4VP16, Gal4esn, Gal4VP64, dCas9-VP16, dCas9-VP64, dCas9-VPR, dCas9-VTR, and rtTA.
- Yeast Gal4-UAS base Since the expression system is one of the most well understood eukaryotic transcriptional regulatory systems, the Gal4 gene encodes a transcriptional activator in yeast (Saccharomyces cerevisiae).
- Gal4 recognizes a 17 bp long sequence of the upstream activation sequence (UAS) of the gene promoter: 5'-CGGRNNRCYNYNYNCNCCG-3' (R stands for ⁇ , Y stands for pyrimidine, and N stands for any deoxynucleotide). Gal4 acts with Gal80, which binds to galactose metabolites. Gal4 has two domains: a DNA binding domain (N-terminal domain) and an activation domain (C-terminal domain). It has a zinc cluster domain, specifically a Zn(2)-Cys(6) dual core cluster domain. The form in which it functions is often a homodimer.
- UAS upstream activation sequence
- Gal4 acts with Gal80, which binds to galactose metabolites.
- Gal4 has two domains: a DNA binding domain (N-terminal domain) and an activation domain (C-terminal domain). It has a zinc cluster domain, specifically a Zn(2)-Cys(6) dual core cluster domain.
- the core sequence that binds to the GAL4 protein in the UAS sequence is a four-copy tandem repeat, each copy containing 17 base pairs, and the sequence 5 '-CGGAGTACTGTCGTGGG-3'.
- Gal4 is present in mammalian cells and activates the target gene only when UAS is recognized, and initiates downstream gene transcription with good specificity and inducibility.
- the protein activation domain of Gal4 can be replaced by the activation domain of herpes simplex virus VP16 protein or 4 copies of VP16, VP64.
- the fusion transcription factor thus formed increases the transcriptional activation activity of Gal4 protein while maintaining the binding specificity of Gal4 protein.
- the tetracycline-inducing system is a bacterial-derived transcriptional activation system for eukaryotic cells.
- the tetracycline response element contains 7 repeats of the 19-nucleotide tetracycline operon sequence 5'-TCCCTATCAGTGATAGAGA-3', which can be recognized by tetracycline-inducible proteins.
- the tetracycline-inducing system includes a tetracycline-inducing inhibition system and a tetracycline-inducing activation system.
- the tetracycline-inducing inhibition system comprises a tetracycline-responsive element and a tetracycline repressor tetR.
- the tetracycline-inducible activation system comprises a tetracycline responsive element and a reverse tetracycline-controlled trans a ctivator rtTA.
- rtTA When tetracycline is absent, rtTA cannot bind to the TRE element and cannot activate downstream gene expression; when tetracycline is present, rtTA binds to TRE in tetracycline to activate downstream gene expression.
- dCas9 nuclease deactivated Cas9
- dCas9 is a mutant form of Cas9 that recognizes the target sequence under gRNA-mediated, but dCas9 has no cleavage activity on the target sequence, and only specifically recognizes the activity of binding DNA.
- dCas9 can be expressed in fusion with various activation elements, it can effectively activate the expression of the target gene.
- the first recognition sequence and the second recognition sequence are each independently selected from at least one of a target sequence of 5 ⁇ UAS, 7 ⁇ tetO and dCas9.
- the 5 ⁇ UAS is the response element of the 5 copies of the Gal4VP16 regulatory system.
- the core sequence that binds to the Gal4 protein in the UAS sequence is a four-copy tandem repeat, each copy containing 17 base pairs and the sequence is 5 '-CGGAGTACTGTCGTGGG-3'.
- 7 ⁇ tetO is a 7-copy TRE that is the recognition sequence for the response element of the tetracycline regulatory system.
- the tetracycline response element contains 7 repeats of the 19-nucleotide tetracycline operon sequence 5'-TCCCTATCAGTGATAGAGA-3', which can be recognized by tetracycline-inducible proteins.
- the first promoter and the second promoter are miniCMV.
- 5 ⁇ UAS-miniCMV is a synthetic inducible promoter.
- 5 ⁇ UAS is the recognition sequence of Gal4 protein
- miniCMV is a partial sequence of the promoter of Cytomegalovirus.
- TRE is also an inducible promoter.
- 7 ⁇ tetO-miniCMV, tetO is the recognition sequence of the response element of the tetracycline system.
- the first regulatory protein and the second regulatory protein are each independently selected from at least one of Lacl, tetR, zinc finger, KRAB, dCas9-KRAB.
- Lactose repressor LacI
- Lactose operon Lactoseoperon LacO
- the base is closely linked to the base sequence of the LacO minor groove. This tight binding prevents the high affinity binding of RNA polymerase from entering the extended state in the promoter region, thereby preventing mRNA transcription and expression of downstream genes.
- the tetracycline repressor tetR TetR contains a conserved helix-turn-helix DNA binding region (forming a homodimer) that binds to the tetracycline operon tetO and inhibits the transcriptional expression of downstream genes.
- Zinc finger zinc finger protein consists of a ring containing about 30 amino acids and a Zn 2+ coordinated to 4 Cys or 2 Cys and 2 His on the ring, forming a structure like a finger.
- Zinc finger proteins are a class that play an important role in gene regulation. According to their conserved domains, zinc finger proteins can be mainly classified into C2H2, C4 and C6.
- Zinc refers to the regulation of gene expression, cell differentiation and embryonic development at the transcriptional and translational levels by specific binding to the target molecule DNA, RNA, DNA-RNA, and to its own or other zinc finger proteins.
- the Krüppel associated box (KRAB) domain is a class of transcriptional repressor regions present in nearly 400 zinc finger proteins in humans. KRAB generally consists of 75 amino acid residues and the smallest inhibitory effective region consists of 45 amino acids. It functions by means of an amphipathic helical connection between proteins. It can be divided into two regions, A and B, which are encoded by different exons, wherein A box plays a central role in transcriptional repression, and B box enhances the transcriptional repression function of A box.
- dCas9 nuclease deactivated Cas9
- dCas9 is a mutant form of Cas9 that recognizes the target sequence under gRNA-mediated, but dCas9 has no cleavage activity on the target sequence, and only specifically recognizes the activity of binding DNA.
- dCas9 and the suppressor region are expressed, the expression of the target gene can be effectively inhibited.
- the first regulatory element and the second regulatory element are each independently selected from at least one of a tetO, a LacO, a zinc finger target site, and a target sequence of dCas9.
- the tetO (tetracycline operon tetO) tetracycline operon can be recognized by tetracycline inhibitors to inhibit the expression of downstream genes.
- the LacO (Lactose operon LacO) lactose operon can be recognized by the lactose repressor (Lactose repressor LacI) to inhibit the expression of downstream genes.
- the zinc finger target site can be recognized by zinc finger proteins to regulate the expression of downstream genes.
- the dCas9 target sequence can be recognized by dCas9-KRAB with the aid of a complementary gRNA and inhibits the expression of downstream genes.
- the first regulatory protein is LacI
- the second regulatory element comprises a plurality of repeating LacO sequences, at least one of the plurality of repeated LacO sequences being set at the second promoter Downstream.
- LacI table Upon arrival, the LacO sequence can be specifically bound, thereby inhibiting the function of the second promoter.
- the Lactose repressor LacI recognizes the specific sequence by the helix-turn-helical element of the DNA binding domain and the large groove of the lactose operon (Lactoseoperon LacO). Binding, while the symmetry-dependent ⁇ -helix residue is closely linked to the base sequence of the LacO minor groove.
- the inventors constructed LacO on both sides of the miniCMV sequence of the 5 ⁇ UAS-miniCMV promoter, and it was experimentally shown that the system can effectively inhibit the expression of genes downstream of the promoter. However, the inventors do not preclude the use of other suppression systems at this location.
- the second regulatory protein is tetR-KRAB
- the first regulatory element comprises a plurality of repeated tetO sequences, at least one of the plurality of repeated tetO sequences being set at the first Downstream of the promoter.
- the inhibition system selected in the embodiment of the present invention is the tetR-KRAB/tetO system
- the tetracycline repressor tetR TetR contains a conserved helix-turn-helix DNA binding region (forming a homodimer), which binds to the tetracycline operon. (tetracycline operon tetO) inhibits transcriptional expression of downstream genes.
- the inventors constructed tetO on both sides of the miniCMV sequence of the 5 ⁇ UAS-miniCMV promoter, and it was shown by experiments that the system can effectively inhibit the expression of genes downstream of the promoter.
- the switching system composed of the first regulatory protein and the second regulatory protein can effectively input signals correspondingly and effectively distinguish different cell lines.
- the inventors here do not preclude the use of other suppression systems.
- At least one of the fifth nucleic acid molecule and the ninth nucleic acid molecule further comprises a sequence encoding a protein of interest.
- the fifth nucleic acid molecule comprises a sequence encoding the protein of interest, and the protein of interest comprises at least one selected from the group consisting of a viral replication packaging protein and an immune effector.
- the viral replication packaging protein can effectively ensure the survival and replication of the expression system vector in the host; the expression of the immune effector can effectively activate the body's immune system, thereby promoting immune killing of specific cells such as tumor cells.
- the virus replication packaging-related protein comprises at least one selected from the group consisting of an adenovirus E1 gene, an E1A gene, an E1B gene, an E2 gene, and an E4 gene.
- adenoviral genes are mainly divided into early expression genes (E1 ⁇ 4) and late expression genes (L1 ⁇ 5). After the adenoviral genome enters the nucleus, the cellular transcription factor first binds to the enhancer upstream of the E1A region, expressing the E1A protein, which regulates cellular metabolism and makes the viral DNA more easily replicated in the cell.
- the E1A protein also activates promoters of other early genes (E1B, E2A, E2B, E3, and E4), where E2B drives three additional early gene transcriptional terminal protein precursors (pTPs) involved in viral replication.
- E1B Single-stranded DNA binding proteins
- DNA pol DNA polymerase
- the E1 region gene expression products are divided into E1A and E1B.
- E1A is mainly composed of two components, 289R (or 13S) and 243R (or 12S).
- E1A protein The main function of the E1A protein is to regulate cellular metabolism, making cells more susceptible to viral replication.
- E1B19K is homologous to the expression product of the Bcl-2 gene, and can prevent apoptosis or necrosis of cells by inactivating and clearing Bax family members.
- the E1B55K gene product can down-regulate the transcription level of the p53 gene, and other adenoviral genes (such as E4 or f6) are involved in this process.
- the E1B55K gene product is also involved in viral replication, transcription of viral late mRNA, and viral RNA transport.
- the E2 region gene expression products are divided into E2A and E2B.
- E2A is DNA Binding Protein (DBP); two main products of E2B are terminal protein precursor (pTP) and viral DNA polymerase (pol).
- the three proteins interact with at least three intracellular factors to initiate adenoviral DNA replication and transcription and translation of the viral late genes.
- the gene product of the E4 region is commonly referred to as orf 1 to 6/7 and is mainly involved in the metabolism of viral messenger RNA. There is also the function of promoting viral DNA replication and shutting down host protein synthesis. Studies have found that some E4 products can bind to DNA-activated protein kinases to prevent viral DNA from concatenating. Since this kinase activates the p53 gene, it is believed that some E4 region gene products can inhibit apoptosis. Many E1B and E4 gene products are involved in antagonizing E1A protein function. For example, E4 inhibits E1A activation of the E2F promoter.
- the immune effector comprises a sequence selected from the group consisting of an inhibitory sequence that antagonizes the PD-1 gene, an inhibitory sequence that antagonizes the PD-L1 gene, an inhibitory sequence that antagonizes the CTLA4 gene, an inhibitory sequence that antagonizes the Tim-3 gene, and GM.
- - at least one of CSF, IL-2, IL-12, IL-15 or a fusion expression form of these factors.
- PD-1 (programmed death 1) programmed death receptor 1 is an important immunosuppressive molecule.
- PD-1 is an important inhibitory molecule on the surface of T cells, and its intracellular domain contains an immunoreceptor tyrosine inhibitory motif ITIM and an immunoreceptor tyrosine transfer motif (ITSM).
- ITSM mediates the recruitment of protein tyrosine phosphatase family phosphatase and the inhibition of T cell activation signals.
- Its ligands are PD-L1 and PD-L2, which play a major role in suppressing T cells in the tumor microenvironment of the immune system. The important role of activation.
- Programmed cell death 1 ligand also known as cluster of differentiation 274 (CD274) or B7 homolog (B7homolog 1, B7-H1), is human A protein in the body that is encoded by the CD274 gene.
- PD-L1 is inducible in hematopoietic cells in a variety of tumor cells and tumor cell microenvironments. Its expression level is positively correlated with the malignancy of some tumors.
- PD-1 antibody and PD-L1 antibody can block the binding of PD-1 to PD-L1, and tissue tumor cells escape from the PD-1/PD-L1 pathway.
- CTLA-4 Cytotoxic T lymphocyte associate protein-4, which is a type of co-stimulatory molecule expressed on the surface of T cells. Similar to the function of CD28, CTLA-4 is capable of specifically binding to CD80/CD86 on the surface of APC to activate downstream signals during T cell activation.
- Treg regulatory T cells
- Activation is accompanied by severe autoimmune diseases.
- CTLA-4 is also expressed in conT cells, and its role is to inhibit the signaling of T cell activation.
- T cell immunoglobulin and mucin-domain containing molecule (TIM) gene family was discovered by Mclntire in 2001 when searching for mouse asthma susceptibility genes and was identified by genomic analysis and cloning. A new family of genes identified that contain immunoglobulin V regions and mucin regions. Tim-3 is an important member of the TIM family and expresses a negative regulatory molecule on the surface of activated Th1 cells. Expression of TIM3 has also been found in CD8 + T cells, Th17 cells, Treg, NK cells, and other lymphocyte subpopulations.
- GM-CSF granulocyte-macrophage Colony Stimulating Factor
- GM-CSF granulocyte-macrophage colony-stimulating factor
- T cells T cells
- NK cells NK cells
- GM-CSF stimulates stem cells to produce granules.
- Cells neutralils, eosinophils, and basophils and monocytes.
- GM-CSF also affects mature cells in the immune system, such as inhibiting the transfer of neutrophils and altering receptor expression on the cell surface. This factor also inhibits fungal infections by activating macrophages.
- IL-2 Interleukin 2
- Interleukin 2 mediates cytokines between white blood cells and white blood cells and between white blood cells and other cells.
- IL-2 is mainly produced by activated T cells, and acts on local target cells in an autocrine and paracrine manner. It is a major cytokine involved in the immune response and has obvious immune effects. It can promote T cell proliferation and produce cytokines, promote B cell proliferation and secretion of Ig, activate macrophages, and enhance NK cell activation and proliferation.
- IL-2 also has a negative regulation effect, which can induce apoptosis of Ag-activated T cells, limit the intensity of immune response, and avoid obvious immune damage.
- Interleukin-12 is a kind of leukocyte-mediated by dendritic cells, macrophages, neutrophils, and human B lymphoblasts (nc-37) in response to antigen stimulation.
- Prime. IL-12 called T cell stimulating factor, participates in the differentiation of naive cells into Th1 and stimulates the production of interferon gamma (IFN- ⁇ ) and tumor necrosis factor alpha (TNF- ⁇ ).
- IFN- ⁇ interferon gamma
- TNF- ⁇ tumor necrosis factor alpha
- IL-12 plays an important role in regulating the activity of natural killer cells and T lymphocytes.
- IL-12 mediates enhanced cytotoxic activity of NK cells and CD8+ cytotoxic T lymphocytes.
- IL-12 also has anti-angiogenic activity, which means it blocks the formation of new blood vessels.
- IP-10 protein 10
- CXCL10 protein 10
- IL-12 has been tested as a possible anti-cancer drug.
- IL-15 is a recently discovered factor that can be produced by a variety of cells such as activated monocytes-macrophages, epidermal cells, and fibroblasts. The molecular structure of IL-15 is much similar to that of IL-2, so that the ⁇ -chain and the ⁇ -chain of the IL-2 receptor can be utilized to bind to target cells to exert a biological activity similar to IL-2.
- IL-15 can induce B cell proliferation and differentiation, and is the only cytokine that can partially replace IL-2 to induce initial antibody production; IL-15 can stimulate T cell and NK cell proliferation, induce LAK cell activity, and can also interact with IL-12. Synergistically stimulate NK cells to produce IFN- ⁇ .
- the target protein and the first regulatory protein are co-expressed under the control of the same promoter, and the cleavable linker peptide is passed between the target protein and the first regulatory protein. connected.
- the target protein and the first regulatory protein are regulated and expressed under the same promoter, and the expression is cleaved at the linker peptide, and the target protein is The first regulatory protein is separated, and the target protein and the first regulatory protein function independently of each other.
- the ninth nucleic acid molecule and the tenth nucleic acid molecule independently inhibit expression of the first regulatory protein or the second regulatory protein, respectively, by the ninth and tenth nucleic acid molecules
- the expression of the first and second regulatory proteins is inhibited by RNA interference, wherein the microRNA is a specific microRNA expressed by different cellular microenvironments, and the ninth or tenth nucleic acid molecule is a specific target sequence of the microRNA, thereby further interfering with RNA interference
- the specific effect of the microRNA expressed in the specific microenvironment on the target sequence can be achieved, thereby achieving specific regulation of the expression of the first regulatory protein or the second regulatory protein.
- MicroRNAs are a class of endogenous small RNAs of about 20-24 nucleotides in length, and several miRNAs can regulate the same gene. The expression of a gene can be finely regulated by a combination of several miRNAs. MicroRNA exists in many forms, the most primitive is pri-miRNA, which is about 300-1000 bases in length; after a single processing, pri-miRNA becomes a pre-miRNA, a microRNA precursor, with a length of about 70-90 bases. The pre-miRNA is digested with Dicer to form a mature miRNA of about 20 to 24 nt in length. The RNA-induced silencing complex (RISC) inhibits the expression of target genes.
- RISC RNA-induced silencing complex
- microRNA-RISC effect of microRNA-RISC on target gene mRNA has always been largely dependent on its degree of complementation with the target gene transcript sequence, in three ways. The first is to cleave the mRNA molecule of the target gene - the miRNA is fully complementary to the target gene, and its mode of action and function are very similar to those of the siRNA, and finally the target mRNA is cleaved. In plants, most of the miRNAs in this way, after the target gene mRNA is broken, the poly(A)-free molecule has multiple U's at the 3' end and is rapidly degraded, and the poly(A)-containing molecule can be stably present. For a while (eg Arabidopsis miR-171).
- miRNAs There is currently one miRNA in the plant that is fully complementary to the three potential target genes, although it is unclear whether these genes are targets of miRNAs. This is the first time that miRNAs are fully complementary to their potential targets, suggesting that miRNAs May contain a similar mode of action as siRNA. The second is to inhibit the translation of the target gene, which does not completely complement the target gene, thereby suppressing translation without affecting the stability of the mRNA. This miRNA is the most widely discovered species (such as nematode lin-4). A very small number of miRNAs in plants inhibit the target gene in this way. The third is binding inhibition - having the above two modes of action: direct binding to cleavage of mRNA when complementary to the target gene; and regulation of gene expression when not intimately bound to the target gene.
- the ninth nucleic acid molecule comprises a nucleic acid sequence specifically recognized by a first microRNA, the tenth nucleic acid molecule comprising a nucleic acid sequence specifically recognizing a second microRNA, the first microRNA being a normal cell Specific microRNAs, the second microRNA is an abnormal cell-specific microRNA.
- the first regulatory protein is expressed in abnormal cells, and is not expressed or underexpressed in normal cells, while the second regulatory protein is expressed in normal cells and is not expressed or expressed in abnormal cells.
- the first microRNA comprises at least one selected from the group consisting of miR199a, miR95, miR125, miRamir25b, Let-7, miR143, miR145, and miR200C.
- the above microRNA is expressed in normal liver cells.
- the second microRNA comprises at least one selected from the group consisting of miR21, miR223, miR224, miR221, miR18, miR214, miR146a, and miR1792 expressed in hepatoma cells (HepG2, Huh7, PLC) .
- the second microRNA is a microRNA specifically expressed by a liver cancer cell.
- the first regulatory protein is expressed in liver cancer cells, and is not expressed or expressed in normal cells, while the second regulatory protein is expressed in normal cells and is not expressed or expressed in liver cancer cells.
- the first nucleic acid molecule and the second nucleic acid molecule are supported on a first expression vector
- the selected ninth nucleic acid molecule is supported on a second expression vector
- the sixth nucleic acid molecule, the seventh nucleic acid molecule, the eighth nucleic acid molecule, and optionally the tenth nucleic acid molecule are loaded in a third On the expression vector.
- the first, second and third expression vectors serve as a loading vector for the expression system to effect regulation of the specific expression of the gene of interest in a suitable microenvironment, such as a cell.
- the selection of the expression vector is not particularly limited as long as the function of the expression system in a suitable microenvironment can be achieved.
- the first expression vector, the second expression vector and the third expression vector are each independently selected from at least one of the following:
- the liposome carrier is a liposome-DNA complex formed by packaging DNA with an artificial bilayer phospholipid.
- the liposome carrier is non-toxic and non-antigenic, and the target gene and the liposome are encapsulated to be protected from nuclease degradation, and the capacity is large, and it can be used alone or in combination with other carriers, and the target gene can be introduced into a specific site by intravenous injection. Disadvantages: short expression time, can not pass the cell membrane barrier.
- Molecularly coupled carrier The molecularly coupled carrier consists of three parts: DNA, DNA binding factor and ligand.
- the virus comprises at least one selected from the group consisting of an adenovirus, a vaccinia virus, and a retrovirus.
- the first expression vector, the second expression vector and the third expression vector are the same vector.
- the inventors load the components in the expression system of the present application on the same expression vector by Cascade Golden-Gate Gibson/Gateway Assemble Method technology, and the plurality of large fragments can be effectively solved by being loaded on the same expression vector.
- the carrier has a very low transfection efficiency.
- the same vector is an adenovirus.
- Adenovirus as a gene therapy vector has the following advantages: 1) a wide range of hosts and low pathogenicity to humans.
- Adenoviral vector systems are widely used for the expression of human and non-human proteins.
- Adenoviruses can infect a range of mammalian cells and are therefore useful for expression of recombinant proteins in most mammalian cells and tissues. It is particularly important to note that adenoviruses are epithelial, whereas most human tumors are derived from epithelial cells.
- the replication genes and pathogenic genes of adenoviruses are quite clear and have been prevalent in the human population (70-80% of adults have neutralizing antibodies to adenovirus).
- Retroviruses can only infect proliferating cells, so DNA transfection cannot be performed in non-proliferating cells, but cells must be maintained in a continuous culture.
- Adenovirus can infect almost all cell types, except for some anti-adenovirus-infected lymphoma cells.
- Adenovirus is the best system for studying gene expression in primary non-proliferating cells, which allows direct comparison between the results obtained in transformed cells and primary cells.
- It can effectively proliferate and has a high titer.
- the adenovirus system produces 10 10 to 10 11 VP/mlL and is concentrated up to 10 13 VP/mL, making it ideal for gene therapy.
- 4) is homologous to human genes.
- Adenoviral vector systems generally use human viruses as vectors and human cells as hosts, thus providing an ideal environment for accurate post-translational processing and proper folding of human proteins. Most human proteins achieve high levels of expression and are fully functional.
- 5) Not integrated into the chromosome, no insertional mutagenicity. Retroviruses can be randomly integrated into the host chromosome, resulting in gene inactivation or activation of oncogenes. The adenovirus does not integrate into the chromosome in almost all known cells except the egg cell, and thus does not interfere with other host genes.
- Integrating a single copy of the virus in an egg cell is a better system for producing transgenic animals with specific characteristics.
- the inventors injected the above adenovirus into a tumor mouse model to significantly inhibit the growth of mouse tumors.
- the above adenovirus can be used as a safe and effective oncolytic virus vaccine to achieve safe and effective specific killing of related tumors.
- the adenovirus is obtained by removing the E1 gene and a part of the E3 gene associated with adenoviral replication packaging, and the E1A gene is constructed by a stepwise Golden Gate method.
- the gene line is finally inserted into the adenoviral vector by way of Gateway or Gibson.
- the inventors inserted a gene line recognizing cancer cells into an adenovirus vector in three steps.
- a marker element that distinguishes normal cells from cancer cells such as a tumor-specific promoter, a suppressor element related to gene line construction, and a recognition sequence of a microRNA that distinguishes cancer cells, is constructed on a primary vector plasmid to constitute a primary component library.
- Esp3I recognition sites are designed at both ends of the component for constructing multiple primary components onto the secondary expression system by means of Golden Gate.
- the expression system library is constructed, and multiple first-level components are selected from the first-level library to assemble them into three expression systems by means of Golden Gate, including a tumor-specific promoter system, which is mediated by the first regulatory element.
- the inhibition system mediated by the inhibition system and the second regulatory element.
- the recognition site of BsaI endonuclease was designed at both ends of each expression system.
- the gene lines are assembled, and the three expression systems are randomly assembled by the Golden Gate method to form a complete gene line. Both ends of the gene line were designed with Gibson homologous sequences or Gateway recognition sites for further experiments.
- the gene line was constructed by Gibson or Gateway to an adenoviral vector in which E1 and part E3 have been removed.
- the above-described manner of obtaining an adenovirus enables rapid modification of a complex, large-fragment oncolytic adenoviral vector.
- the invention proposes a recombinant virus.
- the recombinant virus comprises: a first nucleic acid molecule, the first nucleic acid molecule comprising a tumor cell-specific promoter, and the tumor cell-specific promoter is an alpha-fetoprotein-specific promoter; a nucleic acid molecule operably linked to the first nucleic acid molecule, the second nucleic acid molecule encoding a transcriptional activator, the transcriptional activator is Gal4VP16; a third nucleic acid molecule, the third The nucleic acid molecule comprises a first recognition sequence of the transcriptional activator, the first recognition sequence being 5 x UAS; a fourth nucleic acid molecule operably linked to the third nucleic acid molecule, The fourth nucleic acid molecule comprises a first promoter and a first regulatory element, the first promoter is a miniCMV, and the first regulatory element comprises a plurality of repeating tet
- the first regulatory protein LacI and the target protein are specifically expressed in tumor cells
- the second regulatory protein tetR-KRAB is specifically not expressed or underexpressed in tumor cells, and thus the inhibition mechanism of the first promoter miniCMV mediated by tetR-KRAB is released, the first regulatory protein LacI, the target protein in the first promoter miniCMV start up Under the regulation of effective expression, the LacI-mediated inhibition mechanism effectively inhibited the function of the second promoter miniCMV, and the expression of tetR-KRAB was further inhibited.
- the recombinant virus according to an embodiment of the present invention, more specific expression of the protein in the tumor cell (such as the target protein E1A, the first regulatory protein LacI) or no expression (such as the second regulatory protein tetR-KRAB) can be achieved.
- the expression or non-expression efficiency and specificity in tumor cells are higher.
- the recombinant virus is at least one selected from the group consisting of a retrovirus, an adenovirus, a herpes virus, and a vaccinia virus.
- a retrovirus an adenovirus
- a herpes virus a vaccinia virus.
- vaccinia virus a retroviral vector
- Characteristics of adenovirus vector wide range of host; adenoviral protein expression is not necessary for host proliferation; high virus titer can be obtained; recombinant is very stable; no tumor is caused; high safety; no capsule It is not easily inactivated by complement and can be directly applied in the body; it is not integrated into the chromosome.
- Characteristics of herpes simplex virus vector high titer; large capacity; both proliferating cells and non-proliferating cells can be infected; unintegrated, but long-term existence and stable expression.
- the recombinant virus is an adenovirus.
- adenovirus has many advantages as a gene therapy vector: 1. The host has a wide range and is low in pathogenicity to humans. Adenoviral vector systems are widely used for the expression of human and non-human proteins. Adenoviruses can infect a range of mammalian cells and are therefore useful for expression of recombinant proteins in most mammalian cells and tissues. It is particularly important to note that adenoviruses are epithelial, whereas most human tumors are derived from epithelial cells.
- adenovirus In addition, the replication genes and pathogenic genes of adenoviruses are quite clear and have been prevalent in the human population (70-80% of adults have neutralizing antibodies to adenovirus). Human infection with wild-type adenovirus produces only mild self-limiting symptoms, and ribavirin is effective. 2. Infect and express genes in proliferating and non-proliferating cells. Retroviruses can only infect proliferating cells, so DNA transfection cannot be performed in non-proliferating cells, but cells must be maintained in a continuous culture. Adenovirus can infect almost all cell types, except for some anti-adenovirus-infected lymphoma cells.
- Adenovirus is the best system for studying gene expression in primary non-proliferating cells, which allows direct comparison between the results obtained in transformed cells and primary cells. 3. It can effectively proliferate and has a high titer. The adenovirus system produces 10 10 to 10 11 VP/mL and is concentrated up to 10 13 VP/mlL, making it ideal for gene therapy. 4. Is homologous to human genes. Adenoviral vector systems generally use human viruses as vectors and human cells as hosts, thus providing an ideal environment for accurate post-translational processing and proper folding of human proteins. Most human proteins achieve high levels of expression and are fully functional. 5. Not integrated into the chromosome, no insertional mutagenicity.
- Retroviruses can be randomly integrated into the host chromosome, resulting in gene inactivation or activation of oncogenes.
- the adenovirus does not integrate into the chromosome in almost all known cells except the egg cell, and thus does not interfere with other host genes. Integrating a single copy of the virus in an egg cell is a better system for producing transgenic animals with specific characteristics.
- 6. Can be expanded in suspension culture. 293 cells can be adapted to suspension culture, and this adjustment allows the virus to be expanded in large quantities. A large number of facts have shown that suspension 293 cells can express recombinant proteins in a 1-20 L bioreactor. 7. Can express multiple genes at the same time. This is the first expression system that can be used to design multiple genes in the same cell line or tissue.
- the simplest method is to insert a double expression cassette containing two genes into an adenovirus transfer vector, or co-transfect a target cell strain with a different recombinant virus to express a protein separately. Determination of the MOI ratio of different recombinant viruses can correctly estimate the relative co-expression of each recombinant protein.
- the immune effector comprises a restriction sequence selected from the group consisting of an antagonistic PD-1 gene, an inhibitory sequence which antagonizes the PD-L1 gene, an inhibitory sequence which antagonizes the CTLA4 gene, an inhibitory sequence which antagonizes the Tim-3 gene, and IL. - 2, at least one of IL-12, IL-15, GM-CSF or a fusion expression form of these factors.
- PD-1 programmeed death 1 is a major immunosuppressive molecule.
- PD-1 is an important inhibitory molecule on the surface of T cells, and its intracellular domain contains an immunoreceptor tyrosine inhibitory motif ITIM and an immunoreceptor tyrosine transfer motif (ITSM).
- ITSM mediates the recruitment of protein tyrosine phosphatase family phosphatase and the inhibition of T cell activation signals.
- Its ligands are PD-L1 and PD-L2, which play a major role in suppressing T cells in the tumor microenvironment of the immune system. The important role of activation.
- Programmed cell death 1 ligand also known as cluster of differentiation 274 (CD274) or B7 homolog (B7homolog 1, B7-H1), is human A protein in the body that is encoded by the CD274 gene.
- PD-L1 is inducible in hematopoietic cells in a variety of tumor cells and tumor cell microenvironments. Its expression level is positively correlated with the malignancy of some tumors.
- PD-1 antibody and PD-l1 antibody can block the binding of PD-1 to PD-L1, and tissue tumor cells escape from the PD-1/PD-L1 pathway.
- CTLA-4 Cytotoxic T lymphocyte associate protein-4, which is a type of co-stimulatory molecule expressed on the surface of T cells. Similar to the function of CD28, CTLA-4 is capable of specifically binding to CD80/CD86 on the surface of APC to activate downstream signals during T cell activation.
- Treg regulatory T cells
- Activation is accompanied by severe autoimmune diseases.
- CTLA-4 is also expressed in conT cells, and its role is to inhibit the signaling of T cell activation.
- T cell immunoglobulin and mucin-domain containing molecule (TIM) gene family was discovered by Mclntire in 2001 when searching for mouse asthma susceptibility genes and was identified by genomic analysis and cloning. A new family of genes identified that contain immunoglobulin V regions and mucin regions.
- TIM3 is an important member of the TIM family and expresses a negative regulatory molecule on the surface of activated Th1 cells. Expression of TIM3 has also been found in CD8 + T cells, Th17 cells, Treg, NK cells, and other lymphocyte subpopulations.
- GM-CSF granulocyte-macrophage Colony Stimulating Factor
- GM-CSF granulocyte-macrophage colony-stimulating factor
- T cells T cells
- NK cells NK cells
- GM-CSF stimulates stem cells to produce granules.
- Cells neutralils, eosinophils, and basophils and monocytes.
- GM-CSF also affects mature cells in the immune system, such as inhibiting the transfer of neutrophils and altering receptor expression on the cell surface. This factor also inhibits fungal infections by activating macrophages.
- IL-2 Interleukin 2
- Interleukin 2 mediates cytokines between white blood cells and white blood cells and between white blood cells and other cells.
- IL-2 is mainly produced by activated T cells, and acts on local target cells in an autocrine and paracrine manner. It is a major cytokine involved in the immune response and has obvious immune effects. It can promote T cell proliferation and produce cytokines, promote B cell proliferation and secretion of Ig, activate macrophages, and enhance NK cell activation and proliferation.
- IL-2 also has a negative regulation effect, which can induce apoptosis of Ag-activated T cells, limit the intensity of immune response, and avoid obvious immune damage.
- Interleukin-12 is a kind of leukocyte-mediated by dendritic cells, macrophages, neutrophils, and human B lymphoblasts (nc-37) in response to antigen stimulation.
- Prime. IL-12 called T cell stimulating factor, participates in the differentiation of naive cells into Th1 and stimulates the production of interferon gamma (IFN- ⁇ ) and tumor necrosis factor alpha (TNF- ⁇ ).
- IFN- ⁇ interferon gamma
- TNF- ⁇ tumor necrosis factor alpha
- IL-12 plays an important role in regulating the activity of natural killer cells and T lymphocytes.
- IL-12 mediates enhanced cytotoxic activity of NK cells and CD8+ cytotoxic T lymphocytes.
- IL-12 also has anti-angiogenic activity, which means it blocks the formation of new blood vessels.
- IP-10 protein 10
- CXCL10 protein 10
- IL-12 has been tested as a possible anti-cancer drug.
- IL-15 is a recently discovered factor that can be produced by a variety of cells such as activated monocytes-macrophages, epidermal cells, and fibroblasts. The molecular structure of IL-15 is much similar to that of IL-2, so that the ⁇ -chain and the ⁇ -chain of the IL-2 receptor can be utilized to bind to target cells to exert a biological activity similar to IL-2.
- IL-15 can induce B cell proliferation and differentiation, and is the only cytokine that can partially replace IL-2 to induce initial antibody production; IL-15 can stimulate T cell and NK cell proliferation, induce LAK cell activity, and can also interact with IL-12. Synergistically stimulate NK cells to produce IFN- ⁇ .
- the invention proposes a recombinant cell.
- the recombinant cell comprises an expression system as described above.
- the recombinant cell according to the embodiment of the invention can effectively activate the systemic immune response of the human body, and attack the xenogeneic cells, such as tumor cells, with high safety and specificity.
- At least a portion of the expression system is integrated into the genome of the recombinant cell.
- the expression system replicates as the recombinant cell genome replicates, and the expression system continues to be effective in regulating the expression of the protein of interest.
- the present invention provides the use of the aforementioned expression system, the recombinant virus described above, and the aforementioned recombinant cells for the preparation of a medicament for the treatment of cancer.
- the expression system described in the present application can achieve specific expression of a target protein in tumor cells, and the drug according to the embodiment of the present application is more effective, specific, and safer for treating cancer.
- the inventors used the concept of synthetic biology to design a gene line that responds to multiple targets to regulate the expression of the key gene E1A associated with adenoviral transcription.
- a related cytokine or antibody gene that activates a systemic immune response is co-expressed.
- the gene lines designed by the inventors can regulate the packaging of adenoviruses at multiple levels.
- the inventors used a tumor-specific promoter to regulate
- the microRNA target sequence inside the gene line can distinguish tumor cells from non-tumor cells in response to the expression of microRNAs in different cell lines.
- the inventor designed the gene line to adopt a double suppression system that is very stable and can efficiently respond to external input signals such as cell-specific promoters and microRNA signals, and the gene line can further expand the difference of input signals and be more efficient. Differentiate between tumor cells and non-tumor cells.
- the system removes the E1B gene of the adenovirus, and the E1B gene can interact with the P53 gene of the normal cell to ensure the smooth proliferation of the adenovirus in the cell.
- the adenovirus could not proliferate in normal cells expressing P53.
- the expression of the P53 gene is deleted in many tumor cells, and the adenovirus that does not express E1B can still be efficiently expanded. Therefore, the adenoviral vector of the present embodiment is more specific and safer than the traditional oncolytic adenovirus by multi-angle and multi-level regulation.
- this embodiment also utilizes this adenovirus as a vector to carry a variety of cytokines and antibody genes. At the same time as the oncolytic effect of adenovirus, these co-expressed factors are used to activate the systemic immune response, thereby further improving the effect of oncolytic adenovirus.
- the present patent uses synthetic biology to construct a gene line to regulate the specific expression packaging of oncolytic adenovirus in hepatocellular carcinoma.
- the adenovirus designed and constructed by the inventors is also loaded with cytokines that can activate the immune response of the body. By expressing these cytokines, the body can stimulate the immune response that inhibits tumor growth to achieve the purpose of treating tumors.
- the system designed by this patent has great innovation and technical advantages:
- the gene line designed in this patent comprehensively utilizes multi-level and multi-level biomarkers to distinguish normal cells from cancer cells, and uses double-inhibited closed-loop gene lines to regulate the replication of adenovirus in different cells. Through this genetic line, it is more sensitive to respond to different cell line microenvironments while reducing the effects of gene expression noise.
- This patent utilizes the gene line to carry a variety of cytokines and/or antibody sequences to achieve an effective immune response in the cells of interest.
- the biomarkers involved in the genetic lines are modularized, and each module is built into a library containing biomarkers for different diseases. Establish a genetic line for specific conditions by quickly building up relevant modules.
- the cancer comprises liver cancer, lung cancer, colorectal cancer, melanoma, breast cancer or prostate cancer.
- the inventors have found that the therapeutic effect of the drug according to the embodiment of the present invention on liver cancer, lung cancer, colorectal cancer, melanoma, breast cancer or prostate cancer is more remarkable.
- microRNAs in different tumor cell lines is shown in Table 1.
- the upward arrow indicates microRNAs that are highly expressed in cancer cells compared to normal cells
- the downward arrow indicates microRNAs that are underexpressed in cancer cells compared to normal cells.
- the invention proposes a method of expressing a protein of interest using an expression system, which is the expression system described above.
- the method comprises: (1) causing the fifth nucleic acid molecule to comprise a nucleic acid sequence encoding the protein of interest; (2) inhibiting the tenth nucleic acid molecule from the second regulatory protein Expression to express the protein of interest.
- the second regulatory protein-mediated inhibition mechanism of the first promoter is released, and the target protein is co-operated by the cell-specific promoter and the first promoter. Highly expressed in specific cells.
- the expression is carried out in a cell.
- the cells can provide a microenvironment for the expression of the protein of interest, and the expression of the protein of interest in the cell is more efficient.
- the tenth nucleic acid molecule comprises a nucleic acid sequence specific for the second microRNA, and in step (2) comprises contacting the second microRNA with the tenth nucleic acid molecule.
- MicroRNAs are a class of endogenous small RNAs of about 20-24 nucleotides in length. Multiple miRNAs can also regulate the same gene, and the expression of a single gene can be finely regulated by a combination of several miRNAs. MicroRNA exists in many forms, the most primitive is pri-miRNA, which is about 300-1000 bases in length; after a single processing, pri-miRNA becomes a pre-miRNA, a microRNA precursor, with a length of about 70-90 bases.
- the pre-miRNA is digested with Dicer to form a mature miRNA of about 20 to 24 nt in length.
- MicroRNA-mediated silencing complex RISC inhibits the expression of target genes.
- the effect of microRNA-RISC on target gene mRNA has always been largely dependent on its degree of complementation with the target gene transcript sequence, in three ways. The first is to cleave the mRNA molecule of the target gene - the miRNA is fully complementary to the target gene, and its mode of action and function are very similar to those of the siRNA, and finally the target mRNA is cleaved.
- the poly(A)-free molecule In plants, most of the miRNAs in this way, after the target gene mRNA is broken, the poly(A)-free molecule has multiple U's at the 3' end and is rapidly degraded, and the poly(A)-containing molecule can be stably present.
- miRNAs may contain a similar mode of action as siRNA. The second is to inhibit the translation of the target gene, which does not completely complement the target gene, thereby suppressing translation without affecting the stability of the mRNA.
- This miRNA is the most widely discovered species (such as nematode lin-4). A very small number of miRNAs in plants inhibit the target gene in this way.
- the third is binding inhibition - with the above two modes of action: when it is complementary to the target gene, it directly targets the cleavage of the mRNA; when it is not complete with the target gene When fully integrated, it plays a role in regulating gene expression.
- the invention further provides a method of expressing a protein of interest using an expression system, which is the expression system described above.
- the method comprises: (1) causing the eighth nucleic acid molecule to comprise a nucleic acid sequence encoding a first protein of interest, the fifth nucleic acid molecule comprising a nucleic acid sequence encoding a second protein of interest; 2) expressing the first protein of interest or the second protein of interest by one of: inhibiting expression of the first regulatory protein by the ninth nucleic acid molecule to express the first protein of interest; A tenth nucleic acid molecule inhibits expression of the second regulatory protein to express the second protein of interest.
- the second regulatory protein-mediated inhibition of the first promoter is released, and the second protein of interest is co-operating with the cell-specific promoter and the first promoter.
- the first protein of interest is efficiently activated in a specific cell by a combination of a cell-specific promoter and a second promoter. expression.
- the HEK293 (293-H) cell line was purchased from Invitrogen, the human hepatoma cell line HepG2, the normal liver cell Chang was purchased from ATCC, the human liver cancer cell Huh7 was purchased from the North Natron, and Hepa1-6 was purchased from the ATCC.
- the cells were cultured in Dulbecco's Modified Eagle's Medium, 4.5 g/L glucose, 0.045 unit/mL penicillin and 0.045 g/mL streptomycin, and 10% FBS (purchased from Invitrogen) at 37 °C. , humidity 100%, CO 2 concentration 5%.
- a 24-well plate (Falcon) was taken, and 0.5 mL of a high glucose DMEM medium containing about 7.5 ⁇ 10 4 HEK293 cells was added to each well for 24 hours. Prior to transfection, change the medium and add new DMEM complete medium. Transfection experiments were performed using Attractene Transfection Reagent (Qiagen) or Lipofectamine LTX (Life Technologies). When Attractene was used, the DNA was mixed by volume, and 1.5 ⁇ L of Attractene was added, and allowed to stand at room temperature for 15 minutes, and then added to the cells.
- pDT7004 pUBI-linker-NOS contains a maize ubiquitin promoter (UBI), downstream of the NOS terminator, and no coding protein sequence between UBI and NOS. This application uses pDT7004 to separate plasmid DNA from different experimental groups. After transfection, culture was carried out for 48 hours for flow cytometry analysis or cytokine detection related experiments.
- AFP promoter truncation experiment-related plasmid construction AFP enhancer sequence and AFP promoter sequence were directly PCR-derived from the genome of liver cancer cell line by designing PCR primers. Point mutations in the AFP promoter sequence were obtained by overlay PCR. The AFP enhancer and AFP promoter sequences were constructed in the entry clone sequence, and the recognition sequences of LR enzyme were designed at both ends. The specific promoter was constructed on the same vector as the activation gene Gal4VP16 by LR. The targeting sequence of the microRNA was inserted into the CMV-EYFP vector (XbaI/SalI) by primer annealing. The system level component library is built by Golden Gate.
- Type IIs restriction enzymes are a special class of restriction enzymes, and the most commonly used type II restriction enzymes in molecular biology (recognition sites are palindromes).
- the symmetric sequence, the cleavage site overlaps with the recognition sequence, is a non-palindromic symmetrical sequence, and the cleavage site is located outside the recognition site. Therefore, when the two fragments digested by the type IIs restriction endonuclease are ligated, the original recognition site will no longer exist, and will not be cleaved again in the subsequent restriction enzyme ligation reaction. The effect of welding death.
- the Golden Gate cloning technique utilizes this property of the type IIs restriction enzyme to artificially design different sequences of cleavage sites outside the recognition sequences of these enzymes, and then use the same restriction enzyme to generate different viscous ends, thereby
- the ability to assemble multiple fragments overcomes the deficiencies of traditional multi-fragment assembly requiring the simultaneous use of multiple different restriction enzymes. Compared with the traditional enzyme-cutting technology, the biggest feature of this technology is that it does not introduce any extra "scar", thus achieving the seamless connection of DNA fragments.
- the type IIs restriction endonuclease used in the first-stage element library in this example is Esp3I.
- the system's secondary logic line library was also constructed by Golden Gate, using the Type IIs restriction endonuclease as BsaI.
- Logic ViraPower TM Adenoviral Gateway inserted into the adenoviral vector sequence in TM Expression Kit by Gateway embodiment; inserted into the Adeno-X Adenoviral System adenoviral vector sequences 3 clones described by Gibson.
- the Gateway technology is based on the well-studied ⁇ phage site-specific recombination system (attB ⁇ attP ⁇ attL ⁇ attR) that has been studied. The two reactions of BP and LR constitute the Gateway technology.
- the BP reaction uses an attB DNA fragment or a recombination reaction between an expression clone and an attP donor vector to create an entry clone.
- the LR reaction is a recombination reaction between an attL entry clone and an attR destination vector.
- the LR reaction is used to transfer the sequence of interest to one or more destination vectors in a parallel reaction.
- the inventors employed an LR reaction.
- Gibson assembly technology also known as "Gibson thermostatic one-step assembly method” is a DNA assembly method created by Gibson et al. at the J. CraigVenter Institute in the United States, using the synergistic effect of T5 exonuclease, DNA polymerase and ligase.
- a plurality of DNA fragments with overlapping sequences are assembled in vitro.
- the T5 exonuclease has 5' ⁇ 3' exonuclease activity, and can cleave a DNA fragment having an overlapping region from the 5' end to generate a 3' overhanging end, and then the overlapping sequence of the single-stranded DNA is specific at 50 °C. Annealing (at this time T5 exonuclease is gradually inactivated), and finally DNA polymerase and Taq ligase repair the double-stranded DNA, thereby forming a complete DNA molecule for seamless splicing.
- the quantitative PCR primers were qAFP for: CAAAGCTGAAAATGCAGTTGAATG (SEQ ID NO: 15); qAFP rev: TTCCCCATCCTGCAGACAATCC (SEQ ID NO: 16); GAPDH for: AGAAGGCTGGGGCTCATTTG (SEQ ID NO: 17); GAPDH rev: AGGGGCCATCCACAGTCTTC (SEQ ID NO: 18) ). Fluorescent dye for PowerUp TM Green Master Mix (A25741 Thermo Fisher).
- the high-purity endotoxin-free recombinant adenovirus plasmid was extracted: PacI was linearized and transfected into HEK293 cells, and transfected overnight to complete medium. The first generation was cultured for about one week. The medium was added according to the cell growth state, and then the cells were collected and 1 mL. The culture solution was placed in a 15 mL centrifuge tube, frozen and thawed three times at 37 ° C in liquid nitrogen, and centrifuged at 2000 rpm for 5 minutes. The supernatant was taken as the original solution of the virus solution.
- a large number of virus amplifications (10 15 cm plates) were performed after repeated amplification of the virus for three consecutive generations, and then the virus was purified by CsCl density gradient centrifugation-dialysis.
- the CsCl gradient was prepared by adding 2 mL of a CsCl solution having a density of 1.4 g/mL, and then slowly adding 3 mL of a CsCl solution having a density of 1.3 g/mL, and then adding 5 mL of the virus suspension. Centrifuge at 20000 rpm for 4 hours at 4 °C.
- Viral bands with densities between 1.3 g/mL and 1.4 g/mL were collected into dialysis bags (the dialysis bags were boiled with 10 mM EDTA-Na 2 for 10 minutes before use).
- dialysis buffer 50 g sucrose, 10 mL 1 M Tris-HCl, pH 8.0, 2 mL 1 M MgCl 2 to 1 L
- the mixture was dialyzed overnight at 4 ° C, and the dialysate was changed twice.
- the virus was collected and the virus titer was determined.
- the virus titer was detected by using a viral DNA quantitative method after infecting cells. About 2 x 10 6 HeLa cells/well were seeded onto 6-well tissue culture plates.
- the cell coverage reached 90-100%, and 5 ⁇ L of the virus stock solution was diluted with 500 ⁇ L of serum-free medium to infect the cells and set up two duplicate wells. Incubate at 37 ° C for 3 to 6 hours. The medium was removed and washed twice with 1 mL PBS/well. The cells were lysed by the addition of 500 ⁇ L of freshly prepared NP-40 lysis buffer (0.65% NP-40 substitute (Calbiochem, Billerica, MA, USA), 150 mM NaCl, 10 mM Tris, pH 8.0).
- Standard curve was prepared: The PCR L2 sequence of interest was constructed on a T vector, and the plasmid was diluted to 1 ⁇ 10 9 to 1 ⁇ 10 2 copy / ⁇ L as a standard sequence.
- the number of cells was measured by MTS method: 20 ⁇ L of MTS/PMS mixture (Promega) was added to each well of the cells to be cultured in a 96-well plate, and after mixing, the culture was continued at 37 ° C for 1 to 4 hours. Shake the plate for 10 seconds after color development and mix the colors. The absorbance (OD value) at a wavelength of 490 nm was measured using a microplate reader. A standard curve is drawn to detect the number of cells in the target cell.
- the biological activity of cytokines can be detected by proliferation of factor-dependent cells.
- the biological activities of IL-2, mGM-CSF, and hGM-CSF were detected using CTLL-2, FDC-P1, and TF-1 cell lines, respectively.
- the cells to be tested for factor-dependent were collected, and 1 ⁇ 10 4 cells to be tested were added to a 96-well plate, and then a sample containing the cytokine to be tested was added, and cultured at 37° C. for 24 hours or 48 hours, and cell proliferation was detected using the MTS method. Dose-proliferation standard curve was drawn and the activity level of the corresponding cytokine in the sample was calculated by comparison.
- Co-immunoprecipitation The cells to be tested are collected, and the cells are lysed using an appropriate amount of protein lysate (Biyuntian Co., Ltd.), and the cell debris is removed by centrifugation. A small amount of lysate was taken for Western blot analysis. The remaining lysate was added to 10-30 ⁇ L of protein A/G-agarose beads (Thermo Fisher Scientific) coupled with the corresponding tag antibody, and added to the cell lysate, and slowly shaken at 4 °C. overnight. After incubation, the supernatant was removed by centrifugation.
- protein lysate Boyuntian Co., Ltd.
- the experimental animals were purchased from Huakang Kang or Weitong Lihua Company, and the tumor model was 6-week old male or female mice, which were kept in the environment of SPF.
- the human tumor model was constructed using nude mice. 1 ⁇ 10 7 HepG2 human hepatoma cells mixed with Matrigel (BD) were injected subcutaneously, inoculated into the right abdomen of nude mice, and the size of the subcutaneous tumor mass was measured by vernier calipers every three days.
- the murine tumor model was constructed using C57BL/6J wild type mice, and 1 ⁇ 10 6 Hepa 1-6 mouse liver cancer cells were subcutaneously inoculated into the right abdomen of the mice, and the size of the subcutaneous tumor pieces was measured by vernier calipers every three days.
- oncolytic viruses are a very effective means of treating cancer, and oncolytic adenovirus has carried out many clinical studies.
- the oncolytic adenovirus has a relatively high proportion of leakage, so how to improve the safety of oncolytic adenovirus in the clinical stage is an important issue.
- the regulation of most oncolytic adenoviruses relies on a single biomarker, such as a tumor-specific promoter that regulates the expression of the key gene E1 of adenoviral replication.
- a single biomarker has a relatively high probability of leakage.
- the inventors designed a dual suppression switch system to respond to a variety of biomarkers, and at various levels regulate the expression of the adenovirus E1A gene, thereby improving the recognition specificity of the oncolytic adenovirus and thereby enhancing safety.
- the dual suppression switch system has the following features:
- the tumor specific promoter (pC) regulates the total activator of the dual suppression switch system.
- the activator acts on the activator response promoter (pAct) to regulate the expression of downstream genes.
- One side of the double suppression system expresses the E1A gene associated with adenovirus packaging, the effector (Effector) and the inhibitor a (Rep-a, repressor a), and the targeting of microRNA a (miRNA a) highly expressed in non-tumor cells. sequence.
- the promoter on this side has a recognition sequence (Tb, target of repressor b) expressed on the other side and a targeting sequence in response to the highly expressed microRNA b (miRNA b) in the tumor cells.
- the other side of the double suppression system expresses the repressor b (Rep-b, repressor b).
- the promoter on this side has a recognition sequence (Ta, target of repressor a) on the other side.
- pC initiates the expression of the whole system. Because of the high expression of miRNA b and the low expression of miRNA a in tumor cells, the inhibitor b is degraded by RNA interference, which inhibits the inhibitory effect of suppressor b on the other side. .
- the high expression of the inhibitor a can further inhibit the expression of the inhibitor b, and the double suppression switch system can be rapidly and efficiently flipped to the state of E1A expression, and the expression of the packaging and effector factors of the adenovirus is initiated.
- the double-inhibitor switch enters non-tumor cells, the combination of micro-RNA markers and logic lines through tumor-specific promoters, microRNA markers and logic lines cannot express E1A and effector factors, effectively closing the packaging process of adenovirus, ensuring The specificity and safety of this logic. Therefore, such switches can distinguish between target and non-target cell types at the transcriptional and post-transcriptional levels in response to a variety of input signals.
- the system designed by the inventor can distinguish the target cells from multiple levels and levels, and can effectively improve the safety of the adenovirus.
- AFP liver cancer-specific alpha-fetoprotein
- Alpha-fetoprotein ( ⁇ FP or AFP) is mainly synthesized in fetal liver. There is no AFP expression in normal adults, but when hepatocytes become cancerous, AFP is re-expressed, and AFP is in serum as the disease progresses. The amount will increase dramatically.
- alpha-fetoprotein is a specific clinical indicator for the diagnosis of primary liver cancer. Therefore, in the present example, the inventors selected the alpha-fetoprotein promoter as a promoter marker for distinguishing between liver cancer cells and non-hepatoma cells.
- RNA of Chang, HepG2, Huh7, PLC, Hep3B and Hepa1-6 cells was extracted as shown in FIG. The expression level of the AFP gene in these cells was examined by quantitative PCR.
- Chang is a normal human liver cell
- HepG2, Huh7, PLC, and Hep3B are human liver cancer cells.
- Hepa1-6 is a liver cancer cell of a mouse.
- the human AFP gene is highly expressed in the HepG2 and Huh7 cell lines and is low in the PLC and Hep3B cell lines. A slight expression was also detected in the mouse hepatoma cell Hepa1-6.
- the currently widely used AFP promoter consists of an enhancer and a promoter.
- the enhancer contains the sequences DA (domain A) and DB (domain B) of the two-stage activation promoter.
- the inventors mutated the G at position -119 in the promoter region to A.
- the inventors performed different truncation experiments on the enhancer region.
- AFPI contains an enhancer sequence of 1.8K of AFP (sequence is shown as SEQ ID NO: 21); AFPII contains activation region A (DA), activation region B (DB) and two activation regions.
- Inter-sequence (sequence as shown in SEQ ID NO: 22); AFPIII contains only activation region A (DA) and activation region B (DB) (sequence as shown in SEQ ID NO: 23); AFPIV contains only activation region A (DA) (sequence as shown in SEQ ID NO: 24); AFPV contains only activation region B (DB) (sequence as shown in SEQ ID NO: 25).
- the expression efficiency and specificity of the AFP promoter were detected by transient transfer of these five AFP promoter reporter system plasmids into normal hepatocytes Chang and hepatoma cells HepG2.
- the five AFP promoters have high expression levels in liver cancer cell lines, even exceeding the commonly expressed CMV promoter. From the specificity analysis, in addition to the AFPII reporter gene in Chang, there was no obvious leakage of other promoters. Considering the length of the inserted sequence, the efficiency and specificity of the promoter. In the subsequent experiments, the inventors selected the AFP III promoter to regulate the inventors' dual suppression switch.
- microRNA microRNA
- microRNA microRNA
- Table 1 there are many highly expressed microRNAs in different cancer cells.
- the inventors used miR199a as a microRNA marker to characterize the high expression of normal hepatocytes in normal hepatocytes, and accordingly the inventors used miR21 and miR122 as microRNA markers to characterize the high expression of hepatoma cells in hepatoma cells. Things.
- the inventor designed and constructed a microRNA fluorescent plasmid reporting system, as shown in Figure 7.
- the inventors inserted the target sequence of the microRNA into the 3' untranslated region of the frequently expressed EYFP and the other frequently expressed EBFP as a control fluorescence.
- a total of different cell lines were transferred to detect the expression of the target microRNA in the target cell line.
- the results of the experiment are shown in Fig. 8.
- miR199a is highly expressed in normal hepatocytes; while the expression level of miR21 in hepatoma cell lines is significantly increased, which can be distinguished as normal in the double suppression switch system designed by the inventors.
- An effective microRNA marker for cells and liver cancer cells is highly expressed in normal hepatocytes.
- Double suppression switch can effectively respond to microRNA input signals and achieve stable function in different cell lines.
- the inventors Since the packaging capacity of the oncolytic adenovirus is limited, only 8K foreign genes can be packaged at most, so the inventors have selected short and effective suppressing elements as much as possible in the construction of this embodiment.
- the inventors selected LacI and tetR-KRAB. As shown in Figure 9, the inventors constructed two logical lines: whether the EYFP gene is co-expressed on the tetR-KRAB side. Switch I (SI) does not express EYFP, and Switch II (SII) expresses EYFP. The inventors transiently transferred the two switches into HEK293, and simultaneously shRNA-FF4 and shRNA-FF5. As shown in Figure 10, the inventors' two switches can be efficiently flipped under different input signals.
- SI expressed higher levels on the LacI side, but the level of leakage on the tetR-KRAB side was also higher than that of SII.
- the inventors chose the SI system as the backbone to construct the oncolytic adenovirus carrying the different effectors of the inventors.
- the inventors examined the control effects of different activators on the bidirectional suppression switch. As shown in Figure 11, the inventors examined the regulation efficiency of Gal4VP16, Gal4esn, dCas9-VP64 and rtTA. The experimental results show that the gene line can respond effectively to different activators, so the gene line has good stability.
- a marker element that distinguishes a normal cell from a cancer cell such as a tumor-specific promoter, a regulatory element related to a logical line, and a recognition sequence that distinguishes a microRNA of a cancer cell, is constructed on a primary vector plasmid.
- a primary vector plasmid Form a library of primary components.
- the recognition sites of EI a restriction enzyme with different recognition sites and cleavage sites such as Esp3I and BsaI
- EI a restriction enzyme with different recognition sites and cleavage sites such as Esp3I and BsaI
- the inventors designed an in vitro cell killing assay for oncolytic adenovirus. Different cell lines are infected with different infection numbers. Chang is a normal liver cell, and Huh7 and HepG2 are human liver cancer cell lines. The viability of the cells was measured by the MTS method after the virus culture for six days. As shown in Fig. 15, it was found that when the multiplicity of infection was 100, 10, 1, the liver cancer cells showed significant cytotoxicity and died a lot, and there was a slight cell death at a multiplicity of infection of 0.1.
- the multiplicity of infection when the multiplicity of infection is 10 or less, the growth state of Chang cells is normal and there is no death. At a multiplicity of infection of 100, the cells have a slight cytotoxic response, while the cell volume becomes larger and the cells grow slower. This data indicates that the adenovirus packaged by the inventors can effectively distinguish between liver cancer cells and the growth and proliferation of normal liver cells within a specific multiplicity of infection.
- the adenoviral vectors currently used clinically are human Ad2 or Ad5 type adenoviruses, these viruses are widely present in nature, and neutralizing antibodies that antagonize these adenoviruses already exist in the human body. Therefore, only relying on the oncolytic effect of adenovirus to treat tumors, clinical data show that the treatment effect is not good. Therefore, in the present embodiment, the inventors used the oncolytic adenovirus as a vector to carry the effector gene to the target site of the tumor to cause a systemic immune response. As shown in Figure 16, the inventors first constructed these effectors on a plasmid vector that was frequently expressed, and detected the activity of these effectors by means of a transient.
- the inventors constructed these effector genes into an oncolytic adenovirus vector, and packaged and purified the oncolytic adenovirus carrying these effectors. After infecting the hepatoma cell HepG2, the inventors extracted the supernatant to detect the activity of the immune factor in the supernatant.
- Fig. 17a 12-well plates were inoculated with 3 ⁇ 10 6 HepG2 cells, and Synp-EBFP and synOV-mGM-SCF were used to infect HepG2 with a multiplicity of infection of 10, respectively, at 1, 2, 3 The supernatant was collected for 4 days, and the content of GM-CSF in the supernatant was detected by an ELISA kit.
- Figure 17b12 Orifice plate was inoculated with 3 ⁇ 10 6 HepG2 cells, and HepG2 was infected with synOV-EBFP and synOV-hIL-2 at a multiplicity of infection of 10, respectively.
- the supernatant was collected on 1, 2, 3, and 4 days, respectively, and the supernatant was added to IL.
- the content of IL-2 in the supernatant was detected by MTS.
- FIG. 17c 12-well plates were seeded 3 ⁇ 10 6 HepG2 cells, respectively synOV-EBFP, synOV-anti- PD-1scFv and synOV-anti-PD-L1scFv infected at a multiplicity of infection of 10 HepG2.
- the supernatants were collected on 1, 2, 3, and 4 days, respectively.
- Mouse spleen cells were isolated, T cells were activated with 2 ug/mL anti-mouse CD3 antibody, and the supernatant collected above was added to the spleen cell system at a dilution ratio of 1:10.
- the IFN- ⁇ production in splenocytes was measured by ELISA after 48 hours.
- HepG2 and Huh7 are human hepatocarcinoma cells.
- Hepa1-6 is a mouse hepatoma cell. After the tumor volume reaches 100 mm 3 , it is randomly divided into two groups. The treatment group is intratumorally injected with 1 ⁇ 10 9 oncolytic adenovirus, and injected with PBS. Control group.
- the tumor in the treatment group continued to grow, and the oncolytic virus treatment group could control the growth of the tumor to some extent, but did not effectively control the tumor growth, probably because Hepa1-6 grew faster than HepG2 and Huh7, and at Hepa1-6.
- the expression level of AFP is relatively low. Therefore, the inventors continued to treat the Hepa1-6 tumor model by using the oncolytic adenovirus carrying the immune effector factor in subsequent experiments, and achieved very effective results.
- the therapeutic effect of the inventor's oncolytic adenovirus on a C57 mouse model with an immune system was further examined.
- 1 ⁇ 10 6 Hepa1-6 mouse hepatoma cells were subcutaneously inoculated into the right abdomen of wild-type C57BL/6J mice. After the tumor volume reached 100 mm 3 , each group was intratumorally injected with 1 ⁇ 10.
- 9 Express different cytokine oncolytic adenoviruses, or PBS as a negative control. The volume of the tumor after treatment was observed and the survival time of the tumor-bearing mice was analyzed by Kaplan-Meier method.
- mice with intact immune system oncolytic adenovirus can still inhibit tumor growth and significantly prolong the survival time of tumor-bearing mice.
- One month after the oncolytic adenovirus treatment 1 ⁇ 10 6 Hepa 1-6 mouse liver cancer cells were inoculated again in the left abdomen of the surviving mice, and it was found that the same tumor cells were inoculated in the same part of the same mouse as the primary vaccination. Department, can not grow into a tumor. This proves that in mice treated with oncolytic virus, the body produces specific immunity against tumor cells, which can resist the same tumor cells from growing again in the body.
- Tumors of tumor-bearing mice were taken, and the lymphocyte infiltration of tumor tissues was detected by HE staining after fixation and sectioning. It was found that the lymphocyte infiltration of tumor tissues of mice treated with oncolytic adenovirus was significantly increased compared with the PBS control group, demonstrating that treatment with oncolytic adenovirus recruited more lymphocytes in tumor tissues.
- the infiltrating lymphocytes in the tumor were isolated and purified, and the phenotype of mouse tumor infiltrating T cells was detected by flow cytometry. It was found that the proportion of Ki-67 + T cells infiltrated by tumors in mice treated with oncolytic virus was higher.
- Tumor-infiltrating lymphocytes were added to RPMI 1640 medium containing PMA (20 ng/mL) and Ionomycin (1 ⁇ g/ml L), and cultured at 37 ° C for 4 hours in the presence of Brevedlin A. After cell fixation staining, the expression of ⁇ -interferon in CD4 + and CD8 + T cells was detected by flow cytometry. It was found that in the tumor of the oncolytic adenovirus-treated group, the expression of ⁇ -interferon was higher in T cells, which changed the immune microenvironment inside the tumor and promoted the clearance of tumor cells, as shown in Fig. 20 (a, b). Shown.
- Oncolytic adenovirus treatment causes a systemic anti-tumor immune response
- Mouse Hepa-1-6 tumors were inoculated subcutaneously on the left and right sides of the abdomen of C57BL/6 mice. After tumor formation, the cytokine hIL-2, mGM-CSF, anti-PD- were injected into the right tumor. 1 scFv of oncolytic virus, PBS control and AdGFP control, continued measurement of tumor growth. After 14 days of treatment, tumors on both sides of tumor-bearing mice were taken. Lymphocytes infiltrated in the tumor were isolated and purified, and the phenotype of tumor infiltrating T cells in mice was detected by flow cytometry.
- oncolytic adenovirus The therapeutic situation of oncolytic adenovirus is abstracted into a tumor-virus-immune system, as shown in Figure 22, containing uninfected tumor cells, infected tumor cells, free virus, immune cells activated by oncolytic adenovirus antigens.
- Five components of immune cells activated by tumor antigens are represented by S, I, V, Z V and Z T , respectively.
- the model assumes that the growth of uninfected tumor cells conforms to the generalized logistic growth model, where ⁇ is the growth rate coefficient, K is the environmental capacity, and ⁇ is the nonlinear coefficient.
- Uninfected tumor cells can be infected by free virus and transformed into infected tumor cells with an invasive rate of ⁇ .
- the infected tumor cells will lyse and release more virus with a lysis rate coefficient of ⁇ and a free virus release rate coefficient of ⁇ .
- the free virus naturally decays in the environment with a decay rate coefficient of ⁇ .
- Z V is increased by the activation of oncolytic adenovirus and tumor cell surface antigen infected by virus.
- the rate coefficients are c V1 and c V2 , respectively, and Z V can inhibit the infected tumor cells.
- the rate coefficient is p V .
- Z T is increased by the activation of antigen released by tumor cell lysis, and the rate coefficient is c T , and Z T can inhibit the infected and uninfected tumor cells with a rate coefficient of p T .
- Z V and Z T are naturally attenuated in the environment with a rate coefficient of ⁇ .
- the simulation plot shows the minimum tumor volume during the period when the tumor-virus system changes to the final state over time when the virus replication rate is lower and higher under different initial virus titers and initial tumor size conditions. Changes. It can be seen from the simulation results that increasing the initial titer of the virus helps to control the tumor size.
- the appropriate initial dose can be selected in conjunction with the initial tumor size and dose limit to allow tumor clearance or supplementation with other therapies at the predicted time points, as shown in FIG.
- the tumor size in the final state of the system during the simulation time Under the influence of the different inhibitory effects of immune cells on tumors and viruses, the tumor size in the final state of the system during the simulation time. It can be seen from the simulation results that the system exhibits several dynamic behaviors that tend to completely eliminate tumors, tumor growth to saturation, tumor volume stability, and tumor volume oscillation. In general, the stronger the inhibitory effect of immune cells on tumors, the more the system tends to clear the tumor, and the stronger the inhibitory effect of immune cells on the virus, the more the system tends to lose control of the tumor, and for the tumor to stabilize or oscillate, the system is about two The trend of immunization showed some non-monotonic changes, and the results are shown in Figure 24.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Zoology (AREA)
- Microbiology (AREA)
- Virology (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Wood Science & Technology (AREA)
- Biomedical Technology (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Biotechnology (AREA)
- Mycology (AREA)
- Epidemiology (AREA)
- Biophysics (AREA)
- Immunology (AREA)
- Oncology (AREA)
- Gastroenterology & Hepatology (AREA)
- Toxicology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Physics & Mathematics (AREA)
- Plant Pathology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Peptides Or Proteins (AREA)
Abstract
Description
肝细胞癌 | miR-21↑,18↑,224↑,199↓,195↓,200↓,125↓ |
肺癌 | let-7↓,miR-17-92↑ |
乳腺癌 | miR125b↓,145↓,21↓,155↓ |
颅脑肿瘤 | miR-21↑,221↑,181↓, |
结直肠癌 | miR143↓,145↓ |
淋巴癌 | miR155↑,miR-17-92↑ |
黑色素瘤 | miR21↑,221↑,214↑146a↑137↓200c↓let-7↓ |
Claims (40)
- 一种表达系统,其特征在于,包括:第一核酸分子,所述第一核酸分子含有细胞特异性启动子;第二核酸分子,所述第二核酸分子与所述第一核酸分子可操作地连接,所述第二核酸分子编码转录激活因子;第三核酸分子,所述第三核酸分子含有所述转录激活因子的第一识别序列;第四核酸分子,所述第四核酸分子与所述第三核酸分子可操作地连接,所述第四核酸分子含有第一启动子和第一调控元件;第五核酸分子,所述第五核酸分子与第四核酸分子可操作地连接,所述第五核酸分子编码第一调控蛋白;第六核酸分子,所述第六核酸分子含有所述转录激活因子的第二识别序列;第七核酸分子,所述第七核酸分子与所述第六核酸分子可操作地连接,所述第七核酸分子含有第二启动子和第二调控元件;第八核酸分子,所述第八核酸分子与第七核酸分子可操作地连接,并且所述第八核酸分子编码第二调控蛋白;以及选自下列的至少之一:第九核酸分子,所述第九核酸分子与所述第五核酸分子可操作地连接,所述第九核酸分子被配置为条件性抑制所述第一调控蛋白的表达;第十核酸分子,所述第十核酸分子与所述第八核酸分子可操作地连接,所述第十核酸分子被配置为条件性抑制所述第二调控蛋白的表达,其中,所述第一调控元件适于通过结合所述第二调控蛋白抑制所述第一启动子的功能,所述第二调控元件适于通过结合所述第一调控蛋白抑制所述第二启动子的功能。
- 根据权利要求1所述的表达系统,其特征在于,所述细胞特异性启动子为肿瘤细胞特异性启动子,所述肿瘤细胞特异性启动子为选自甲胎蛋白特异性启动子,Survivin基因启动子,人端粒酶逆转录酶基因启动子,胆囊收缩素A受体基因启动子,癌胚抗原启动子,原癌基因人类表皮生长因子受体2启动子,前列腺素内氧化酶还原酶2启动子,趋化因子受体-4,E2F-1基因启动子,黏蛋白启动子,前列腺特异抗原,人酪氨酸酶相关蛋白1,酪氨酸酶启动子至少之一。
- 根据权利要求1所述的表达系统,其特征在于,所述转录激活因子为选自Gal4VP16、Gal4vp64、dCas9-VPR、dCas9-VP64、dCas9-VP16、dCas9-VTR以及rtTA至少之一。
- 根据权利要求1所述的表达系统,其特征在于,所述第一识别序列与所述第二识别序列分别独立地选自5×UAS,7×tetO以及dCas9的靶标序列至少之一。
- 根据权利要求1所述的表达系统,其特征在于,所述第一启动子与所述第二启动子分别独立地选自miniCMV、TATAbox。
- 根据权利要求1所述的表达系统,其特征在于,所述第一调控蛋白和第二调控蛋白分别独立地选自Lacl、tetR、zinc finger、KRAB、tetR-KRAB、dCas9-KRAB至少之一。
- 根据权利要求6所述的表达系统,其特征在于,所述第一调控元件和所述第二调控元件分别独立地选自tetO、LacO、锌指蛋白靶标序列、dCas9的靶标序列至少之一。
- 根据权利要求7所述的表达系统,其特征在于,所述第一调控蛋白是LacI,所述第二调控元件包括多个重复的LacO序列,所述多个重复的LacO序列的至少之一设置在所述第二启动子的下游。
- 根据权利要求6所述的表达系统,其特征在于,所述第二调控蛋白是tetR-KRAB所述第一调控元件包括多个重复的tetO序列,所述多个重复的tetO序列的至少之一设置在所 述第一启动子的下游。
- 根据权利要求1所述的表达系统,其特征在于,所述第五核酸分子和第九核酸分子的至少之一进一步包括编码目的蛋白的序列。
- 根据权利要求10所述的表达系统,其特征在于,所述第五核酸分子包括编码所述目的蛋白的序列,并且所述目的蛋白包括选自病毒复制包装蛋白、免疫效应因子的至少之一。
- 根据权利要求的11所述的表达系统,其特征在于,所述病毒复制包装相关蛋白包括选自腺病毒E1基因、E1A基因、E1B基因、E2基因和E4基因至少之一。
- 根据权利要求11所述的表达系统,其特征在于,所述免疫效应因子包括选自拮抗PD-1基因的抑制序列、拮抗PD-L1基因的抑制序列、拮抗CTLA4基因的抑制序列、拮抗Tim-3基因的抑制序列、GM-CSF、IL-2、IL-12、IL-15至少之一。
- 根据权利要求10所述的表达系统,其特征在于,所述目标蛋白与所述第一调控蛋白是以融合蛋白的形式表达的,并且所述目标蛋白与所述第一调控蛋白之间通过可切割的连接肽连接的。
- 根据权利要求1所述的表达系统,其特征在于,所述第九核酸分子与所述第十核酸分子分别独立地通过下列方式抑制所述第一调控蛋白或所述第二调控蛋白的表达:借助RNA干扰。
- 根据权利要求15所述的表达系统,其特征在于,所述第九核酸分子含有被第一microRNA特异性识别的核酸序列,所述第十核酸分子含有被第二microRNA特异性识别的核酸序列,所述第一microRNA为正常细胞特异性microRNA,所述第二microRNA为异常细胞特异性microRNA。
- 根据权利要求16所述的表达系统,其特征在于,所述第一microRNA包括选自下列的至少之一:miR199a、miR95、miR125、miR25b、Let-7、miR143、miR145以及miR200C。
- 根据权利要求16所述的表达系统,其特征在于,所述第二microRNA包括选自下列的至少之一:miR21、miR223、miR224、miR221、miR18、miR214、miR146a以及miR1792。
- 根据权利要求1所述的表达系统,其特征在于,所述第一核酸分子与所述第二核酸分子负载在第一表达载体上,所述第三核酸分子、所述第四核酸分子、所述第五核酸分子以及可选的所述第九核酸分子负载在第二表达载体上,所述第六核酸分子、所述第七核酸分子、所述第八核酸分子以及可选的所述第十核酸分子负载在第三表达载体上。
- 根据权利要求19所述的表达系统,其特征在于,所述第一表达载体、第二表达载体和第三表达载体分别独立地选自下列的至少之一:质粒、病毒、纳米材料、脂质体、分子耦联载体、裸露DNA、染色体载体、多聚物。
- 根据权利要求20所述的表达系统,其特征在于,所述病毒包括选自腺病毒、牛痘病毒、疱疹病毒、逆转录病毒的至少之一。
- 根据权利要求19所述的表达系统,其特征在于,所述第一表达载体、第二表达载体和第三表达载体负载在同一个载体上。
- 根据权利要求22所述的表达系统,其特征在于,所述同一个载体为腺病毒载体。
- 根据权利要求19所述的表达系统,其特征在于,所述第一表达载体包括:从5’端到3’端依次为BsaI,AFP III,Gal4VP16以及BsaI,任选地,第一表达载体携带具有SEQ ID NO:1所示核苷酸序列的核酸。
- 根据权利要求19所述的表达系统,其特征在于,所述第二表达载体包括:从5’端到3’端依次为BsaI,5×UAS,tetO,miniCMV,tetO,E1A,2A,免疫效应因子,LacI,microRNA199a特异识别序列以及BsaI,任选地,第二表达载体携带具有SEQ ID NO:2~7任一项所示核苷酸序列的核酸。
- 根据权利要求19所述的表达系统,其特征在于,所述第三表达载体包括:从5’端到3’端依次为BsaI,5×UAS,LacO,miniCMV,LacO,tetR-KRAB,microRNA21特异识别序列以及BsaI,任选地,所述第三表达载体携带具有SEQ ID NO:8所示核苷酸序列的核酸。
- 根据权利要求23所述的表达系统,其特征在于,所述腺病毒包括:从5’端到3’端依次为第一反向末端重复序列,包装信号,AFPIII,Gal4VP16,5×UAS,tetO,miniCMV,tetO,E1A,2A,免疫效应因子,2A,LacI,microRNA199a特异性识别序列,5×UAS,LacO,miniCMV,LacO,tetR-KRAB,microRNA21特异性识别序列,腺病毒E2基因区,E3基因区,E4基因区以及第二反向末端重复序列,任选地,所述腺病毒载体携带具有SEQ ID NO:9~14所示核苷酸序列的核酸。
- 根据权利要求27所述的表达系统,其特征在于,所述腺病毒是通过如下方式获得的:所述的腺病毒载体去除了与腺病毒复制包装相关的E1基因和部分E3基因,E1A基因通过逐级Golden Gate的方法构建到基因线路中,最终通过Gateway或Gibson的方式将基因线路插入到腺病毒载体中。
- 一种重组病毒,其特征在于,包括:第一核酸分子,所述第一核酸分子含有肿瘤细胞特异性启动子,所述肿瘤细胞特异性启动子为甲胎蛋白特异性启动子;第二核酸分子,所述第二核酸分子与所述第一核酸分子可操作地连接,所述第二核酸分子编码转录激活因子,所述转录激活因子为Gal4VP16;第三核酸分子,所述第三核酸分子含有所述转录激活因子的第一识别序列,所述第一识别序列为5×UAS;第四核酸分子,所述第四核酸分子与所述第三核酸分子可操作地连接,所述第四核酸分子含有第一启动子和第一调控元件,所述第一启动子为miniCMV,所述第一调控元件包括多个重复的tetO序列,所述多个重复的tetO序列的至少之一设置在所述第一启动子的下游;第五核酸分子,所述第五核酸分子与第四核酸分子可操作地连接,所述第五核酸分子编码第一调控蛋白,所述第一调控蛋白为LacI;所述第五核酸分子进一步包括编码所述目的蛋白的序列,并且所述目的蛋白包括病毒复制蛋白、免疫效应因子,所述免疫效应因子是以单独或融合蛋白的形式表达的,并且所述毒复制蛋白和所述效应因子之间通过可切割的连接肽连接的,所述目标蛋白与所述第一调控蛋白是以融合蛋白的形式表达的,并且所述目标蛋白与所述第一调控蛋白之间通过可切割的连接肽连接的;第六核酸分子,所述第六核酸分子含有所述转录激活因子的第二识别序列,所述第二识别序列为5×UAS;第七核酸分子,所述第七核酸分子与所述第六核酸分子可操作地连接,所述第七核酸分子含有第二启动子和第二调控元件,所述第二启动子为miniCMV,所述第二调控元件包括多个重复的LacO序列,所述多个重复的LacO序列的至少之一设置在所述第二启动子的下游;第八核酸分子,所述第八核酸分子与第七核酸分子可操作地连接,并且所述第八核酸分子编码第二调控蛋白,所述第二调控蛋白为tetR-KRAB;第九核酸分子,所述第九核酸分子与所述第五核酸分子可操作地连接,所述第九核酸分子被配置为条件性抑制所述第一调控蛋白的表达,所述第九核酸分子含有被第一microRNA特异性识别的核酸序列,所述第一microRNA为正常细胞特异性microRNA;以及第十核酸分子,所述第十核酸分子与所述第八核酸分子可操作地连接,所述第十核酸 分子被配置为条件性抑制所述第二调控蛋白的表达,所述第十核酸分子含有被第二microRNA特异性识别的核酸序列,所述第二microRNA为肿瘤细胞特异性microRNA,其中,所述第一调控元件适于通过结合所述第二调控蛋白抑制所述第一启动子的功能,所述第二调控元件适于通过结合所述第一调控蛋白抑制所述第二启动子的功能。
- 根据权利要求29所述的重组病毒,其特征在于,所述重组病毒为选自腺病毒、牛痘病毒、逆转录病毒、疱疹病毒的至少之一。
- 根据权利要求29所述的重组病毒,其特征在于,所述重组病毒为腺病毒。
- 根据权利要求29所述的重组病毒,其特征在于,所述免疫效应因子包括选自拮抗PD-1基因的抑制序列、拮抗PD-L1基因的抑制序列、拮抗CTLA4基因的抑制序列、拮抗Tim-3基因的抑制序列、IL-2、IL-12、IL-15、GM-CSF至少之一。
- 一种重组细胞,其特征在于,含有权利要求1~28所述的表达系统。
- 根据权利要求33所述的重组细胞,其特征在于,所述表达系统的至少一部分整合于所述重组细胞的基因组中。
- 权利要求1~28任一项所述的表达系统、权利要求29~32任一项所述的重组病毒、权利要求33~34任一项所述的重组细胞在制备药物中的用途,所述药物用于治疗癌症。
- 根据权利要求35所述的用途,其特征在于,所述癌症包括肝癌、肺癌、结直肠癌,黑色素瘤、乳腺癌或前列腺癌。
- 一种利用表达系统表达目的蛋白的方法,所述表达系统为权利要求1~28任一项所述的表达系统,其特征在于,所述方法包括:(1)使所述第五核酸分子包含编码所述目的蛋白的核酸序列;(2)使所述第十核酸分子抑制所述第二调控蛋白的表达,以便表达所述目的蛋白。
- 根据权利要求37所述的方法,其特征在于,所述表达是在细胞中进行的。
- 根据权利要求37所述的方法,其特征在于,所述第十核酸分子含有被第二microRNA特异性识别的核酸序列,在步骤(2)中包括使所述第二microRNA与所述第十核酸分子接触。
- 一种利用表达系统表达目的蛋白的方法,所述表达系统为权利要求1~28任一项所述的表达系统,其特征在于,所述方法包括:(1)使所述第八核酸分子包含编码第一目的蛋白的核酸序列,使所述第五核酸分子包含编码第二目的蛋白的核酸序列;(2)通过下列之一表达所述第一目的蛋白或所述第二目的蛋白:使所述第九核酸分子抑制所述第一调控蛋白的表达,以便表达所述第一目的蛋白;使所述第十核酸分子抑制所述第二调控蛋白的表达,以便表达所述第二目的蛋白。
Priority Applications (6)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US16/470,520 US20200017881A1 (en) | 2017-03-24 | 2017-08-04 | Programmable Oncolytic Virus Vaccine System and Method |
CN201780002478.XA CN108064305B (zh) | 2017-03-24 | 2017-08-04 | 可编程的溶瘤病毒疫苗系统及其应用 |
KR1020197031190A KR102409077B1 (ko) | 2017-03-24 | 2017-08-04 | 프로그램 가능한 항암 바이러스 백신 시스템 및 이의 적용 |
AU2017405929A AU2017405929B2 (en) | 2017-03-24 | 2017-08-04 | Programmable oncolytic virus vaccine system and application thereof |
JP2020500941A JP6961788B2 (ja) | 2017-03-24 | 2017-08-04 | プログラム可能な腫瘍溶解性ウイルスワクチン系及びその適用 |
EP17901776.9A EP3604548A4 (en) | 2017-03-24 | 2017-08-04 | VACCINE SYSTEM WITH PROGRAMMABLE ONCOLYTIC VIRUS AND USE OF IT |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201710184736.2 | 2017-03-24 | ||
CN201710184736 | 2017-03-24 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2018171103A1 true WO2018171103A1 (zh) | 2018-09-27 |
Family
ID=63584602
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2017/096043 WO2018171103A1 (zh) | 2017-03-24 | 2017-08-04 | 可编程的溶瘤病毒疫苗系统及其应用 |
Country Status (7)
Country | Link |
---|---|
US (1) | US20200017881A1 (zh) |
EP (1) | EP3604548A4 (zh) |
JP (1) | JP6961788B2 (zh) |
KR (1) | KR102409077B1 (zh) |
AU (1) | AU2017405929B2 (zh) |
HK (1) | HK1253377A1 (zh) |
WO (1) | WO2018171103A1 (zh) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2020191293A1 (en) * | 2019-03-20 | 2020-09-24 | Javelin Oncology, Inc. | Anti-adam12 antibodies and chimeric antigen receptors, and compositions and methods comprising |
Families Citing this family (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN109402178B (zh) * | 2018-11-16 | 2021-08-03 | 佛山科学技术学院 | 一种小鼠精原干细胞高效重编程的方法及应用 |
EP4176066A1 (en) * | 2020-07-06 | 2023-05-10 | Salk Institute for Biological Studies | Recombinant adenovirus genome having a synthetic transcriptional unit and two step transcriptional regulation and amplification |
WO2022158067A1 (ja) * | 2021-01-20 | 2022-07-28 | 国立大学法人 鹿児島大学 | レプリコンdna、クローニングベクター、クローニングベクターの製造方法、スクリーニングキット及びスクリーニング方法 |
EP4423267A1 (en) * | 2021-10-27 | 2024-09-04 | The Board of Trustees of the Leland Stanford Junior University | Recombinant viral genomes and related compositions and methods |
CN114958782B (zh) * | 2022-03-16 | 2023-03-24 | 中国农业科学院兰州兽医研究所 | 一种iptg诱导性缺失d1133l基因的非洲猪瘟病毒减毒株及其应用 |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN104178506A (zh) * | 2014-04-30 | 2014-12-03 | 清华大学 | Taler蛋白通过空间位阻发挥转录抑制作用及其应用 |
CN104611365A (zh) * | 2014-07-17 | 2015-05-13 | 清华大学 | 利用tale转录抑制子在哺乳动物细胞中模块化构建合成基因线路 |
CN104630267A (zh) * | 2014-07-17 | 2015-05-20 | 清华大学 | 利用tale转录抑制子在哺乳动物细胞中模块化构建合成基因线路的试剂盒 |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20050136035A1 (en) * | 2003-06-03 | 2005-06-23 | Derek Ko | Cell specific replication-competent viral vectors comprising a self processing peptide cleavage site |
US9624476B2 (en) * | 2011-08-23 | 2017-04-18 | National Institute Of Biomedical Innovation | Conditionally replicating adenovirus |
CN106011104B (zh) * | 2015-05-21 | 2019-09-27 | 清华大学 | 利用拆分Cas系统进行基因编辑和表达调控方法 |
-
2017
- 2017-08-04 WO PCT/CN2017/096043 patent/WO2018171103A1/zh unknown
- 2017-08-04 US US16/470,520 patent/US20200017881A1/en not_active Abandoned
- 2017-08-04 JP JP2020500941A patent/JP6961788B2/ja active Active
- 2017-08-04 AU AU2017405929A patent/AU2017405929B2/en active Active
- 2017-08-04 EP EP17901776.9A patent/EP3604548A4/en active Pending
- 2017-08-04 KR KR1020197031190A patent/KR102409077B1/ko active IP Right Grant
-
2018
- 2018-10-03 HK HK18112674.8A patent/HK1253377A1/zh unknown
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN104178506A (zh) * | 2014-04-30 | 2014-12-03 | 清华大学 | Taler蛋白通过空间位阻发挥转录抑制作用及其应用 |
CN104611365A (zh) * | 2014-07-17 | 2015-05-13 | 清华大学 | 利用tale转录抑制子在哺乳动物细胞中模块化构建合成基因线路 |
CN104630267A (zh) * | 2014-07-17 | 2015-05-20 | 清华大学 | 利用tale转录抑制子在哺乳动物细胞中模块化构建合成基因线路的试剂盒 |
Non-Patent Citations (5)
Title |
---|
MA, D.C. ET AL.: "Integration and Exchange of Split dCas9 Domains for Transcriptional Controls in Mammalian Cells", NATURE COMMUNICATIONS, vol. 7, 3 October 2016 (2016-10-03), pages 1 - 7, XP055538578 * |
POULIOT, F. ET AL.: "A Molecular Imaging System Based on Both Transcriptional and Genomic Amplification to Detect Prostate Cancer Cells In Vivo", THE AMERICAN SOCIETY OJ GENE & CELL THERAPY, vol. 21, no. 3, 31 March 2013 (2013-03-31), pages 554 - 560, XP055538580 * |
See also references of EP3604548A4 |
VILABOA, N. ET AL.: "Gene Switches for Deliberate Regulation of Transgene Expression: Recent Advances in System Development and Uses", GENETIC SYNDROMES & GENE THERAPY, vol. 2, no. 3, 11 November 2011 (2011-11-11), pages 1 - 23, XP055117017 * |
XIE, Z. ET AL.: "Multi-Input RNAi-Based Logic Circuit for Identification of Specific Cance Cells", SCIENCE, vol. 333, 2 September 2011 (2011-09-02), pages 1307 - 1311, XP055012892 * |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2020191293A1 (en) * | 2019-03-20 | 2020-09-24 | Javelin Oncology, Inc. | Anti-adam12 antibodies and chimeric antigen receptors, and compositions and methods comprising |
Also Published As
Publication number | Publication date |
---|---|
AU2017405929A1 (en) | 2019-10-17 |
KR20190131533A (ko) | 2019-11-26 |
EP3604548A4 (en) | 2021-01-13 |
JP6961788B2 (ja) | 2021-11-05 |
EP3604548A1 (en) | 2020-02-05 |
JP2020511165A (ja) | 2020-04-16 |
US20200017881A1 (en) | 2020-01-16 |
AU2017405929B2 (en) | 2021-05-20 |
KR102409077B1 (ko) | 2022-06-15 |
HK1253377A1 (zh) | 2019-06-14 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2018171103A1 (zh) | 可编程的溶瘤病毒疫苗系统及其应用 | |
CN109576231B (zh) | 分离的重组溶瘤腺病毒、药物组合物及其在治疗肿瘤和/或癌症的药物中的用途 | |
JP7546925B2 (ja) | 核酸とcar修飾免疫細胞とを含む治療薬およびその使用 | |
CN108064305B (zh) | 可编程的溶瘤病毒疫苗系统及其应用 | |
JP2013500339A (ja) | 癌を治療するための送達ツールとしての脂肪由来間質細胞(asc) | |
Brücher et al. | iMATCH: an integrated modular assembly system for therapeutic combination high-capacity adenovirus gene therapy | |
CN110997902A (zh) | Suv39h1缺陷的免疫细胞 | |
CN111743923A (zh) | 包含分离的重组溶瘤腺病毒和免疫细胞的治疗剂及其应用 | |
Liu et al. | Inhibitory effect of Survivin promoter-regulated oncolytic adenovirus carrying P53 gene against gallbladder cancer | |
Yoon et al. | CRISPR-Cas12a with an oAd induces precise and cancer-specific genomic reprogramming of EGFR and efficient tumor regression | |
JP2014523236A (ja) | 腫瘍溶解性強化B型ヒトアデノウイルスAd11突然変異体の構築とその応用 | |
WO2022012531A1 (zh) | 一种经修饰的免疫细胞的制备方法 | |
WO2021218802A1 (zh) | 可受微小rna调控的分离的重组溶瘤痘病毒及其应用 | |
JP6483019B2 (ja) | 新規アデノウイルス及びその増殖促進方法 | |
CA2542335A1 (en) | Cancer gene therapeutic drug | |
Davola et al. | Genetic modification of oncolytic viruses to enhance antitumor immunity | |
US20240041960A1 (en) | Anti-cancer-associated non-tumor cell agent comprising virus | |
CN117866906B (zh) | Foxr1抑制剂在制备用于治疗肿瘤的药物中的应用 | |
US20240307539A1 (en) | Egfr-targeting chimeric antigen receptor | |
US20220152134A1 (en) | Oncolytic adenoviral vector expressing a member of the b7 family of costimulatory ligands and ada | |
US20220154218A1 (en) | Oncolytic adenoviral vector expressing peptidylarginine deiminase and tissue inhibitor of metalloproteinase | |
Kirchhammer | Tumor-targeted immunotherapy using an engineered adenoviral vector platform | |
CN117120062A (zh) | 用于发现cd8 t细胞中治疗靶标的体内crispr筛选系统 | |
BR112016008973B1 (pt) | Adenovírus oncolítico munido de genes heterólogos |
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: 17901776 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2020500941 Country of ref document: JP Kind code of ref document: A |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
ENP | Entry into the national phase |
Ref document number: 2017405929 Country of ref document: AU Date of ref document: 20170804 Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 20197031190 Country of ref document: KR Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 2017901776 Country of ref document: EP Effective date: 20191024 |