CN105254767A - Protein D and HER2 fusion protein and preparing method and application thereof - Google Patents
Protein D and HER2 fusion protein and preparing method and application thereof Download PDFInfo
- Publication number
- CN105254767A CN105254767A CN201510789201.9A CN201510789201A CN105254767A CN 105254767 A CN105254767 A CN 105254767A CN 201510789201 A CN201510789201 A CN 201510789201A CN 105254767 A CN105254767 A CN 105254767A
- Authority
- CN
- China
- Prior art keywords
- protein
- fusion protein
- her2
- tumor
- seq
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 108020001507 fusion proteins Proteins 0.000 title claims abstract description 70
- 102000037865 fusion proteins Human genes 0.000 title claims abstract description 69
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 title claims abstract description 54
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 title claims abstract description 47
- 102100037840 Dehydrogenase/reductase SDR family member 2, mitochondrial Human genes 0.000 title claims abstract description 16
- 101710188053 Protein D Proteins 0.000 title claims abstract description 16
- 101710132893 Resolvase Proteins 0.000 title claims abstract description 16
- 238000000034 method Methods 0.000 title claims abstract description 10
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 65
- 102000004169 proteins and genes Human genes 0.000 claims abstract description 59
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 46
- 239000000427 antigen Substances 0.000 claims abstract description 43
- 102000036639 antigens Human genes 0.000 claims abstract description 43
- 108091007433 antigens Proteins 0.000 claims abstract description 43
- 239000002671 adjuvant Substances 0.000 claims abstract description 25
- 241000606768 Haemophilus influenzae Species 0.000 claims abstract description 22
- 229940047650 haemophilus influenzae Drugs 0.000 claims abstract description 22
- 208000026310 Breast neoplasm Diseases 0.000 claims abstract description 14
- 125000003275 alpha amino acid group Chemical group 0.000 claims abstract 7
- 210000004027 cell Anatomy 0.000 claims description 33
- 229960005486 vaccine Drugs 0.000 claims description 24
- 241000894006 Bacteria Species 0.000 claims description 22
- 210000003000 inclusion body Anatomy 0.000 claims description 18
- 150000007523 nucleic acids Chemical group 0.000 claims description 18
- 229940021747 therapeutic vaccine Drugs 0.000 claims description 18
- 108091028043 Nucleic acid sequence Proteins 0.000 claims description 17
- 239000013604 expression vector Substances 0.000 claims description 16
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 claims description 15
- 206010006187 Breast cancer Diseases 0.000 claims description 12
- 239000002773 nucleotide Substances 0.000 claims description 12
- 125000003729 nucleotide group Chemical group 0.000 claims description 12
- 239000012634 fragment Substances 0.000 claims description 10
- 102100029727 Enteropeptidase Human genes 0.000 claims description 8
- 108010013369 Enteropeptidase Proteins 0.000 claims description 8
- 241000588724 Escherichia coli Species 0.000 claims description 7
- 102000051957 human ERBB2 Human genes 0.000 claims description 7
- 238000002347 injection Methods 0.000 claims description 7
- 239000007924 injection Substances 0.000 claims description 7
- 108020004705 Codon Proteins 0.000 claims description 6
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 claims description 6
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 claims description 6
- 108010033276 Peptide Fragments Proteins 0.000 claims description 6
- 102000007079 Peptide Fragments Human genes 0.000 claims description 6
- 238000003776 cleavage reaction Methods 0.000 claims description 6
- 238000000746 purification Methods 0.000 claims description 6
- 230000007017 scission Effects 0.000 claims description 6
- 102000004190 Enzymes Human genes 0.000 claims description 5
- 108090000790 Enzymes Proteins 0.000 claims description 5
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 claims description 4
- BPHPUYQFMNQIOC-NXRLNHOXSA-N isopropyl beta-D-thiogalactopyranoside Chemical compound CC(C)S[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O BPHPUYQFMNQIOC-NXRLNHOXSA-N 0.000 claims description 4
- 239000013522 chelant Substances 0.000 claims description 3
- 239000013598 vector Substances 0.000 claims description 3
- 241000233866 Fungi Species 0.000 claims description 2
- 210000004899 c-terminal region Anatomy 0.000 claims description 2
- 241000699670 Mus sp. Species 0.000 abstract description 10
- 230000035755 proliferation Effects 0.000 abstract description 7
- 208000017891 HER2 positive breast carcinoma Diseases 0.000 abstract description 3
- 235000018102 proteins Nutrition 0.000 description 45
- 150000001413 amino acids Chemical class 0.000 description 17
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 16
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 15
- 230000014509 gene expression Effects 0.000 description 12
- 238000010171 animal model Methods 0.000 description 11
- 241000699666 Mus <mouse, genus> Species 0.000 description 10
- 235000001014 amino acid Nutrition 0.000 description 10
- 239000000872 buffer Substances 0.000 description 9
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 8
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 8
- 230000000694 effects Effects 0.000 description 8
- 230000006058 immune tolerance Effects 0.000 description 8
- 230000005847 immunogenicity Effects 0.000 description 8
- 239000011780 sodium chloride Substances 0.000 description 8
- 239000001963 growth medium Substances 0.000 description 7
- 230000028993 immune response Effects 0.000 description 7
- 210000002540 macrophage Anatomy 0.000 description 7
- 239000002609 medium Substances 0.000 description 7
- 239000006228 supernatant Substances 0.000 description 7
- 238000011282 treatment Methods 0.000 description 7
- 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 6
- 101150029707 ERBB2 gene Proteins 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 238000004153 renaturation Methods 0.000 description 6
- 239000011534 wash buffer Substances 0.000 description 6
- 238000005406 washing Methods 0.000 description 6
- XSQUKJJJFZCRTK-UHFFFAOYSA-N Urea Chemical compound NC(N)=O XSQUKJJJFZCRTK-UHFFFAOYSA-N 0.000 description 5
- 239000004202 carbamide Substances 0.000 description 5
- 238000010276 construction Methods 0.000 description 5
- 238000010790 dilution Methods 0.000 description 5
- 239000012895 dilution Substances 0.000 description 5
- 239000003814 drug Substances 0.000 description 5
- 150000004676 glycans Chemical class 0.000 description 5
- 238000000338 in vitro Methods 0.000 description 5
- 239000000203 mixture Substances 0.000 description 5
- PHEDXBVPIONUQT-RGYGYFBISA-N phorbol 13-acetate 12-myristate Chemical compound C([C@]1(O)C(=O)C(C)=C[C@H]1[C@@]1(O)[C@H](C)[C@H]2OC(=O)CCCCCCCCCCCCC)C(CO)=C[C@H]1[C@H]1[C@]2(OC(C)=O)C1(C)C PHEDXBVPIONUQT-RGYGYFBISA-N 0.000 description 5
- 229920001282 polysaccharide Polymers 0.000 description 5
- 239000005017 polysaccharide Substances 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- 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 4
- 108010060123 Conjugate Vaccines Proteins 0.000 description 4
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 4
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 4
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 4
- 230000001580 bacterial effect Effects 0.000 description 4
- 238000004113 cell culture Methods 0.000 description 4
- 238000002512 chemotherapy Methods 0.000 description 4
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- 230000018109 developmental process Effects 0.000 description 4
- 238000011081 inoculation Methods 0.000 description 4
- 230000009465 prokaryotic expression Effects 0.000 description 4
- 238000001959 radiotherapy Methods 0.000 description 4
- 210000004881 tumor cell Anatomy 0.000 description 4
- 102000014914 Carrier Proteins Human genes 0.000 description 3
- 108010078791 Carrier Proteins Proteins 0.000 description 3
- 206010027476 Metastases Diseases 0.000 description 3
- 241001052560 Thallis Species 0.000 description 3
- 230000027455 binding Effects 0.000 description 3
- 201000011510 cancer Diseases 0.000 description 3
- 238000005119 centrifugation Methods 0.000 description 3
- 229940031670 conjugate vaccine Drugs 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 230000004069 differentiation Effects 0.000 description 3
- 229940079593 drug Drugs 0.000 description 3
- 238000000855 fermentation Methods 0.000 description 3
- 230000001024 immunotherapeutic effect Effects 0.000 description 3
- 230000001939 inductive effect Effects 0.000 description 3
- 230000002401 inhibitory effect Effects 0.000 description 3
- 230000005764 inhibitory process Effects 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 238000003760 magnetic stirring Methods 0.000 description 3
- 230000002018 overexpression Effects 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 229940023143 protein vaccine Drugs 0.000 description 3
- 238000012216 screening Methods 0.000 description 3
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 3
- 230000004083 survival effect Effects 0.000 description 3
- 239000000725 suspension Substances 0.000 description 3
- 230000004614 tumor growth Effects 0.000 description 3
- NHBKXEKEPDILRR-UHFFFAOYSA-N 2,3-bis(butanoylsulfanyl)propyl butanoate Chemical compound CCCC(=O)OCC(SC(=O)CCC)CSC(=O)CCC NHBKXEKEPDILRR-UHFFFAOYSA-N 0.000 description 2
- VHUUQVKOLVNVRT-UHFFFAOYSA-N Ammonium hydroxide Chemical compound [NH4+].[OH-] VHUUQVKOLVNVRT-UHFFFAOYSA-N 0.000 description 2
- 102000019260 B-Cell Antigen Receptors Human genes 0.000 description 2
- 108010012919 B-Cell Antigen Receptors Proteins 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 229920001213 Polysorbate 20 Polymers 0.000 description 2
- 238000012300 Sequence Analysis Methods 0.000 description 2
- 241000193998 Streptococcus pneumoniae Species 0.000 description 2
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 2
- 108010092262 T-Cell Antigen Receptors Proteins 0.000 description 2
- 238000009098 adjuvant therapy Methods 0.000 description 2
- 235000011114 ammonium hydroxide Nutrition 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 2
- 230000001363 autoimmune Effects 0.000 description 2
- 230000009286 beneficial effect Effects 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 239000000969 carrier Substances 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 238000012258 culturing Methods 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 2
- 229940088598 enzyme Drugs 0.000 description 2
- 230000004151 fermentation Effects 0.000 description 2
- 238000009472 formulation Methods 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 210000000987 immune system Anatomy 0.000 description 2
- 230000036039 immunity Effects 0.000 description 2
- 238000002649 immunization Methods 0.000 description 2
- 230000003053 immunization Effects 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 208000022760 infectious otitis media Diseases 0.000 description 2
- 230000003834 intracellular effect Effects 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 230000009401 metastasis Effects 0.000 description 2
- 229910052760 oxygen Inorganic materials 0.000 description 2
- 239000001301 oxygen Substances 0.000 description 2
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 2
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 2
- 239000002244 precipitate Substances 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 108091008146 restriction endonucleases Proteins 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- 230000000638 stimulation Effects 0.000 description 2
- 238000003756 stirring Methods 0.000 description 2
- 229940031000 streptococcus pneumoniae Drugs 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 230000009466 transformation Effects 0.000 description 2
- 241001430294 unidentified retrovirus Species 0.000 description 2
- UAIUNKRWKOVEES-UHFFFAOYSA-N 3,3',5,5'-tetramethylbenzidine Chemical compound CC1=C(N)C(C)=CC(C=2C=C(C)C(N)=C(C)C=2)=C1 UAIUNKRWKOVEES-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
- 238000011725 BALB/c mouse Methods 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- 101100282617 Bovine herpesvirus 1.1 (strain Cooper) gC gene Proteins 0.000 description 1
- 206010006458 Bronchitis chronic Diseases 0.000 description 1
- 102000000905 Cadherin Human genes 0.000 description 1
- 108050007957 Cadherin Proteins 0.000 description 1
- 101100314454 Caenorhabditis elegans tra-1 gene Proteins 0.000 description 1
- UXVMQQNJUSDDNG-UHFFFAOYSA-L Calcium chloride Chemical compound [Cl-].[Cl-].[Ca+2] UXVMQQNJUSDDNG-UHFFFAOYSA-L 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 102000029816 Collagenase Human genes 0.000 description 1
- 108060005980 Collagenase Proteins 0.000 description 1
- 238000012286 ELISA Assay Methods 0.000 description 1
- 208000000461 Esophageal Neoplasms Diseases 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 101150054472 HER2 gene Proteins 0.000 description 1
- 102000004895 Lipoproteins Human genes 0.000 description 1
- 108090001030 Lipoproteins Proteins 0.000 description 1
- 201000009906 Meningitis Diseases 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 102000005717 Myeloma Proteins Human genes 0.000 description 1
- 108010045503 Myeloma Proteins Proteins 0.000 description 1
- 206010030155 Oesophageal carcinoma Diseases 0.000 description 1
- 108700026244 Open Reading Frames Proteins 0.000 description 1
- 208000005141 Otitis Diseases 0.000 description 1
- 101710116435 Outer membrane protein Proteins 0.000 description 1
- 206010033128 Ovarian cancer Diseases 0.000 description 1
- 206010061535 Ovarian neoplasm Diseases 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 206010035664 Pneumonia Diseases 0.000 description 1
- 108010039918 Polylysine Proteins 0.000 description 1
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 1
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 1
- 208000006265 Renal cell carcinoma Diseases 0.000 description 1
- 206010057190 Respiratory tract infections Diseases 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 208000004337 Salivary Gland Neoplasms Diseases 0.000 description 1
- 206010061934 Salivary gland cancer Diseases 0.000 description 1
- 210000001744 T-lymphocyte Anatomy 0.000 description 1
- 102000001742 Tumor Suppressor Proteins Human genes 0.000 description 1
- 108010040002 Tumor Suppressor Proteins Proteins 0.000 description 1
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 238000004873 anchoring Methods 0.000 description 1
- 230000005809 anti-tumor immunity Effects 0.000 description 1
- 230000030741 antigen processing and presentation Effects 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- 230000007503 antigenic stimulation Effects 0.000 description 1
- 230000005975 antitumor immune response Effects 0.000 description 1
- 210000003719 b-lymphocyte Anatomy 0.000 description 1
- 230000007924 bacterial virulence factor Effects 0.000 description 1
- 229960000074 biopharmaceutical Drugs 0.000 description 1
- 229960002685 biotin Drugs 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 206010006451 bronchitis Diseases 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- 235000011148 calcium chloride Nutrition 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000012292 cell migration Effects 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 230000007969 cellular immunity Effects 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 208000007451 chronic bronchitis Diseases 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 229960002424 collagenase Drugs 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 239000003398 denaturant Substances 0.000 description 1
- -1 diacylglycerol phosphate Chemical class 0.000 description 1
- 230000003205 diastolic effect Effects 0.000 description 1
- 238000007865 diluting Methods 0.000 description 1
- 238000007599 discharging Methods 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 238000004090 dissolution Methods 0.000 description 1
- 238000004043 dyeing Methods 0.000 description 1
- 208000019258 ear infection Diseases 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 230000013020 embryo development Effects 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 238000011067 equilibration Methods 0.000 description 1
- 239000006167 equilibration buffer Substances 0.000 description 1
- 108700020302 erbB-2 Genes Proteins 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 229940042385 glatiramer Drugs 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 230000008629 immune suppression Effects 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 208000030776 invasive breast carcinoma Diseases 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 230000002147 killing effect Effects 0.000 description 1
- 108020001756 ligand binding domains Proteins 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000001394 metastastic effect Effects 0.000 description 1
- 206010061289 metastatic neoplasm Diseases 0.000 description 1
- 239000010413 mother solution Substances 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- PXHVJJICTQNCMI-UHFFFAOYSA-N nickel Substances [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 1
- LGQLOGILCSXPEA-UHFFFAOYSA-L nickel sulfate Chemical compound [Ni+2].[O-]S([O-])(=O)=O LGQLOGILCSXPEA-UHFFFAOYSA-L 0.000 description 1
- 229910000363 nickel(II) sulfate Inorganic materials 0.000 description 1
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 102000039446 nucleic acids Human genes 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 230000010355 oscillation Effects 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000007918 pathogenicity Effects 0.000 description 1
- 230000002572 peristaltic effect Effects 0.000 description 1
- 230000003285 pharmacodynamic effect Effects 0.000 description 1
- 239000002644 phorbol ester Substances 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- YHHSONZFOIEMCP-UHFFFAOYSA-O phosphocholine Chemical compound C[N+](C)(C)CCOP(O)(O)=O YHHSONZFOIEMCP-UHFFFAOYSA-O 0.000 description 1
- 229950004354 phosphorylcholine Drugs 0.000 description 1
- 229920000656 polylysine Polymers 0.000 description 1
- 229940031937 polysaccharide vaccine Drugs 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 230000001376 precipitating effect Effects 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- 230000012743 protein tagging Effects 0.000 description 1
- 238000005086 pumping Methods 0.000 description 1
- 238000003259 recombinant expression Methods 0.000 description 1
- 208000015347 renal cell adenocarcinoma Diseases 0.000 description 1
- 238000012827 research and development Methods 0.000 description 1
- 210000002345 respiratory system Anatomy 0.000 description 1
- 208000020029 respiratory tract infectious disease Diseases 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 239000012679 serum free medium Substances 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 238000005063 solubilization Methods 0.000 description 1
- 230000007928 solubilization Effects 0.000 description 1
- 230000009870 specific binding Effects 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 238000003151 transfection method Methods 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 230000001131 transforming effect Effects 0.000 description 1
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 description 1
- 201000005112 urinary bladder cancer Diseases 0.000 description 1
- 229940125575 vaccine candidate Drugs 0.000 description 1
- 238000009423 ventilation Methods 0.000 description 1
- 230000001018 virulence Effects 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
Landscapes
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
Abstract
The invention provides fusion protein. The fusion protein is formed by connecting adjuvant protein with the end N or the end C of a tumor-associated antigen through a linker or a direct connecting mode. The adjuvant protein contains Haemophilus influenzae (Hi) protein D or a segment of the Haemophilus influenzae (Hi) protein D; the tumor-associated antigen contains HER2 protein with the amino acid sequence being SEQ ID NO:3 or a segment of the HER2 protein. The invention further provides a preparing method for the fusion protein and application thereof. The prepared recombined protein can restrain proliferation of HER2 positive breast cancer cells in a Blab/c mice body, or delay the increase speed of breast tumors.
Description
Technical Field
The invention relates to the fields of biology and medicine, in particular to a fusion protein capable of preparing HER2/neu antigen positive tumor vaccine, and specifically relates to a fusion protein comprising Haemophilus influenzae (Hi) protein D (protein D, PD) and HER2/neu tumor antigen.
Background
For the treatment of tumors, there are currently surgical therapies, chemotherapies, radiation therapies, and the like. Surgical treatment is of no therapeutic interest for patients with multiple tissue metastases. Chemotherapy and radiation therapy can cause damage to normal cells due to poor specificity. Therefore, clinically, a drug with stronger specificity is needed, which specifically kills tumor cells without damaging normal cells. Tumor immunotherapy is a method of activating Cytotoxic T Lymphocytes (CTL) against tumor antigens in vivo, and killing tumor cells expressing the corresponding tumor antigens by the activated CTL, thereby having specificity. Tumor specific antigens or related antigens are key to the development of biologics with specific CTL activation.
HER2 is a tumor-associated antigen, whose coding gene is located at position 21 of the 17 th chromosome pair (17q21) and encodes a transmembrane receptor-like phosphorylated glycoprotein with a molecular weight of 185kD, and is also referred to as P185. P185 consists of 1255 amino acids, whose extracellular domain is a ligand binding domain consisting of 632 amino acids, this region being rich in cysteine; the transmembrane region is a strong hydrophobic region consisting of 22 amino acids, and serves as a membrane anchoring region to position the whole protein molecule on a cell membrane; the intracellular segment is made up of 580 amino acids and contains the tyrosine kinase activity domain (CoussensL, 1985).
In general, the HER2 gene is mainly expressed during human embryonic development and is involved in the growth and development of various tissues and organs. After human adult life, only in certain tissues, such as: low level expression is present in tissues such as kidney, liver, breast tissue, ovary, pancreas, stomach, etc. In human tumors, HER2/neu molecular expression is altered due to overexpression of the gene product, with HER2 amplification in 20% to 30% of invasive breast cancer patients. Furthermore, overexpression is found in ovarian cancer (17-37%), non-small cell lung cancer (27-56%), interstitial cancer (37%), bladder cancer (36%), esophageal cancer (60-73%), salivary gland cancer (32-62%), primary renal cell cancer (10%), pancreatic cancer (> 50%) (SlamonDJ, 1989; SchneiderPM, 1989; WeinerDB, 1990; YokotaJ, 1988; HouL, 1992; Scheurled, 2000; Seliger B, 2000; Lipponen P, 1994; TagliabueE, 2000). It can be seen that overexpression of HER2 plays a positive role in tumor transformation, and is one of the etiological factors of tumors. In addition, the high expression of HER2 protein, which occurs in the early stages of breast cancer development, also increases the metastatic capacity of tumor cells, and the high expression of HER2 by driving multiple metastasis-related mechanisms including an increase: cell migration, in vitro invasiveness, collagenase type IV activity, influence the synthesis of certain adhesion molecules such as epithelial cadherins, etc. to promote metastasis (Blume-JensenP, 2001). Therefore, HER2 is one of the important targets for tumor therapy vaccines.
Epitopes (also known as epitopes) of antigens are the fundamental units for specific binding of T cell antigen receptors (TCRs) and B cell antigen receptors (BCRs) and antibodies. Epitopes can be divided into T cell epitopes and B cell epitopes by differentiation of binding to TCR and BCR. Because the epitope vaccine lacks three-dimensional structure, has small molecules, poor immunogenicity and short half-life, the epitope vaccine cannot independently induce immune response, namely has no immunogenicity, but can obtain immunogenicity and induce immune response after being crosslinked or combined with carriers such as macromolecular protein or non-antigenic polylysine and the like.
The haemophilus influenzae PD protein is a highly conserved surface lipoprotein that plays an important role in the process of Hi causing respiratory tract infections as a characteristic virulence determinant of Hi. The haemophilus influenzae PD protein was first confirmed in studies of Hi binding to human IgD myeloma protein 4490. Sequence analysis of the coding region DNA of the Haemophilus influenzae PD protein showed that the mature protein of the Haemophilus influenzae PD protein is encoded by 1092bp open reading frame, consists of 346 amino acids and has a relative molecular mass of 42000. Studies have shown that there are only small variations in PD protein among each Hi strain. Sequence analysis at the nucleic acid and amino acid levels showed that each strain had a high degree of conservation of PD and that the gene substitutions were evenly distributed throughout the gene. Analysis of Hi isolated from different stages of infection in patients with chronic bronchitis revealed only minor mutations in the gene for the Haemophilus influenzae PD protein of each strain. Western blot analysis using 3 different murine anti-PD monoclonal antibodies revealed that the PD protein was present in all 127 strains of Hi, with a number of PD molecules per bacterial strain of about 2800. These studies all confirm that the haemophilus influenzae PD protein is an outer membrane protein located on the surface of the bacterial body, has high antigen conservation, and is a potential vaccine candidate protein.
The haemophilus influenzae PD protein is a bacterial virulence factor with diacylglycerol phosphate enzyme activity that causes the release of phosphorylcholine from host epithelial cells. The streptococcus pneumoniae polysaccharide conjugate vaccine taking the PD protein as a carrier protein can prevent the streptococcus pneumoniae otitis. PD protein is by far the first non-typeable haemophilus influenzae antigen capable of inducing a protective response in the human body and is also the most potent candidate carrier protein for polysaccharide conjugate vaccines.
The haemophilus influenzae PD protein is one of novel conjugate vaccine protein carriers, a ten-valent pneumonia conjugate vaccine Synflorix developed by glatiramer in 2009 is marketed in europe, and this vaccine links pneumococcal capsular polysaccharides of three serotypes 1, 5 and 7F to haemophilus influenzae PD protein, and clinical data indicate its good immunogenicity and safety.
Two of the major causative bacteria of acute otitis media disease are pneumococci and acapsular haemophilus influenzae, both of which are considered to be lower respiratory tract infectionsThe main factor of dyeing. Although vaccines of pneumococcal capsular polysaccharides have been used for many years, protection against children is poor. Later, CRM for the heptavalent pneumococcal polysaccharide vaccine197The combined vaccine as a carrier can play a role in protecting children and preventing invasive diseases and acute otitis media caused by pneumococcus. And the disadvantage is CRM197This carrier is also used in conjunction with Hib and meningitis vaccines, and therefore the same carrier may have a negative impact on vaccine immunity. Based on the reasons and the advantages of the haemophilus influenzae PD protein, such as membrane localization, sequence conservation, wide distribution, pathogenicity and good preclinical experimental results, the haemophilus influenzae PD protein becomes a carrier protein of the pneumococcal decavalent polysaccharide vaccine and can prevent the double infection of pneumococcus and non-capsular Hib. Meanwhile, the early work in the laboratory proves that the Hi protein is used as a carrier adjuvant protein, and the exogenous epitope carried by the Hi protein can induce animals to generate good immune response after the addition of or without the addition of an adjuvant.
Disclosure of Invention
The invention aims to provide a fusion protein which can be used for preparing a HER2 antigen positive tumor therapeutic vaccine.
The primary aim of tumor therapeutic vaccine design is to break the immune tolerance of the organism caused by tumor cells, and due to the influence of the immune tolerance of the organism, the immune system of the organism cannot recognize non-self components, and the immune system cannot recognize tumor-related antigens, so that the immune suppression is finally caused. Research reports show that the HER2 protein with the immune complete sequence cannot break immune tolerance and stimulate immune response, and the applicant finds that the HER2 protein with the complete sequence contains epitopes causing the immune tolerance in the research, so that in the design of a tumor therapeutic vaccine, a intercepted partial sequence is selected to replace a full-length sequence to be more beneficial to break the immune tolerance, such as an extracellular region, a transmembrane region, an intracellular region or a plurality of Tcell epitopes and Bcell epitopes of HER 2.
In order to achieve the purpose, the invention provides the following technical scheme:
a fusion protein formed by connecting the adjuvant protein with the N terminal or the C terminal of a tumor-associated antigen through a Linker (Linker) or a direct connection mode; wherein,
the adjuvant protein comprises haemophilus influenzae (haemophilus influenzae, Hi) protein D or a fragment of the haemophilus influenzae protein D;
the tumor-associated antigen comprises an amino acid sequence of SEQ ID NO: 3 or a fragment thereof;
preferably, the tumor-associated antigen is formed by combining an extracellular region fragment of a human HER2 antigen and a peptide fragment of a HER2 epitope, and the amino acid sequence of the extracellular region fragment of the HER2 antigen is represented by SEQ ID NO: 4, the amino acid sequence of the HER2 epitope peptide fragment is SEQ ID NO: 5.
preferably, the amino acid sequence of the linker is-GGGGSGGGGSGGGGS-; preferably, the adjuvant protein is linked to the N-terminus of the tumor associated antigen by direct linkage.
In one embodiment according to the invention, the amino acid sequence of the fusion protein is seq id no: 6.
the present invention also provides an expression vector constructed by linking a nucleotide sequence encoding the fusion protein according to claim 1 or 2 to a commercialized vector selected from one or more of pET-28a-c (+), pET29a, pET-30a-c (+), pET39b (+), pET-40b (+), pET-41a (+) and pET-43.1a (+) and optionally a sequence encoding a cleavage recognition site sequence and/or a sequence encoding a protein tag; preferably, the nucleotide sequence is seq id no: 7.
in one embodiment according to the present invention, the expression vector further comprises a coding nucleic acid sequence corresponding to the sequential arrangement of the restriction enzyme recognition site, the histidine tag and the enterokinase site at the 5' end of the nucleotide sequence, preferably, the sequential arrangement of the restriction enzyme recognition site, the histidine tag and the enterokinase site corresponds to the coding nucleic acid sequence of seq id no: 8.
in another embodiment according to the present invention, the expression vector further comprises a stop codon sequence and a cleavage recognition site sequence located at the 3' end of the nucleotide sequence; preferably, the stop codon sequence and the enzyme cutting recognition site sequence are SEQ ID NO: 9; more preferably, the nucleotide sequence encoding the fusion protein is seq id no: 10.
the invention also provides a host cell containing the expression vector, wherein the host cell is a bacterium or a fungus; preferably, the host cell is escherichia coli; more preferably, the E.coli is selected from BL21(DE3), BL21(DE3) PlysS or TB 1.
The invention further provides a preparation method of the fusion protein, which comprises the steps of inducing the host cell to express the fusion protein through IPTG (Merk, USA), collecting inclusion bodies, extracting the fusion protein in the inclusion bodies, and purifying to obtain the fusion protein;
preferably, the purification is achieved by:
the fusion protein was purified by passing through a chelate column.
In one embodiment of the present invention, the purification step further comprises:
the fusion protein is cut by enterokinase, then the cut fusion protein is loaded on the cut target protein and loaded on Ni2+ -ChelatingSepharose Fastflow, and then the fusion protein is eluted by imidazole with the concentration of 50 mM.
The fusion protein provided by the invention can be used for preparing a HER2 antigen positive tumor vaccine, and the vaccine comprises the fusion protein; preferably, the vaccine is a HER2 antigen positive tumor therapeutic vaccine; preferably, the antigen positive tumor is a breast tumor.
Further, the vaccine also comprises an adjuvant, wherein the adjuvant is selected from one or more of GM-CSF and GM-CSF + CpG; preferably, the vaccine is in the form of injection.
The preparation method and the application of the protein-HER 2 fusion protein therapeutic vaccine comprise the following steps:
taking the construction of a recombinant pET-30a (+) prokaryotic expression vector as an example: firstly, constructing a pET-30a (+) prokaryotic expression vector; ② a recombinant prokaryotic expression vector containing a nucleic acid sequence of PD protein 19-129aa encoding Hi, a nucleic acid sequence of HER2 protein 41-560aa, a nucleic acid sequence of HER2 protein 774-790aa and a nucleic acid sequence of Linker; providing an Escherichia coli for expression, transforming a recombinant prokaryotic expression vector containing a PD protein 19-129aa nucleic acid sequence encoding Hi, a nucleic acid sequence of HER2 protein 41-560aa, a nucleic acid sequence of HER2 protein 774-790aa and a nucleic acid sequence of Linker into the Escherichia coli for expression, and stably expressing a protein D and HER2 fusion protein after transformation; and providing a method for purifying the protein and HER2 fusion protein.
The percentage contents in the product and the method are mass volume percentage contents.
The invention relates to a therapeutic vaccine of protein and HER2 fusion protein. The vaccine comprises 19-129aa of PD protein, 41-560aa of HER2 protein and 774-790aa of HER2 protein, and can stimulate good immune response of organisms and break immune tolerance. The protein fusion protein of ProteinD and HER2 prepared by the invention can stimulate macrophages in vitro to secrete and express TNF-alpha in large quantity. Meanwhile, the protein D and HER2 fusion protein prepared by the invention is subjected to a pharmacodynamic experiment, and the fusion protein therapeutic vaccine formed by immunity injection is added or not added with an adjuvant, so that the proliferation of 4T1-HER2 cells in a breast cancer animal model of a Balb/c mouse can be inhibited within 50 days, the tumor growth inhibition rate reaches over 90 percent, and the cellular immunity activated by the protein D and HER2 fusion protein therapeutic vaccine is obviously higher than that of a PBS control group. Therefore, the developed therapeutic vaccine of the protein D and HER2 fusion protein has good immunotherapeutic effect on inhibiting the proliferation of 4T1-HER2 cells in Balb/c mouse breast cancer animal models under the condition of containing or not containing adjuvants. Wherein the 4T1-HER2 cell is constructed by transfecting cDNA of human HER2 with a mouse breast cancer 4T1 cell strain through retrovirus; wherein the 4T1-HER2 breast cancer animal model is an animal model established subcutaneously by injecting 4T1-HER2 into female BALB/C mice.
The invention has the beneficial effects that:
by screening the antigen epitope and carrying out tandem expression, the HER2 protein immune tolerance epitope is avoided, the self immune tolerance of a tumor patient is broken, and the anti-tumor immunity of an organism is activated.
The recombinant protein prepared by coupling expression of the antigen epitope and the PD protein and using the high immunogenicity of the PD protein as an adjuvant protein can improve the immunogenicity of the recombinant protein, promote antigen presentation and enhance the anti-tumor immune response of an organism.
The recombinant protein prepared by the invention can inhibit the proliferation of HER2 positive breast cancer cells in a Blab/c mouse body, or delay the growth rate of breast tumors.
The recombinant protein prepared by the invention has great application prospect in prevention and treatment of recurrence after human HER2 positive breast cancer operation and adjuvant therapy of chemotherapy and radiotherapy treatment, and can stimulate immune response of organisms and kill cancer cells by utilizing an autoimmune system.
The recombinant protein prepared by the invention has great application prospect in prevention and treatment of relapse after operation of human HER2 positive tumor, adjuvant therapy of chemotherapy and radiotherapy treatment, and can stimulate immune response of organisms and kill cancer cells by utilizing an autoimmune system.
The therapeutic vaccine of the protein and HER2 fusion protein prepared by the invention does not contain an adjuvant, and also can contain one or more of the following adjuvants: GM-CSF; ② CpG; ③ GM-CSF + CpG, etc.
The protein and HER2 fusion protein therapeutic vaccine prepared by the invention can be prepared into various clinically applicable formulations, including but not limited to injection formulations.
Drawings
FIG. 1 depicts the construction of pET30-a (+) -PD-HER2 expression vector;
FIG. 2 shows SDS-PAGE electrophoresis of induced expression of pET30-a (+) -PD-HER2/BL21(DE3) engineering bacteria;
FIG. 3PD-HER2 fusion protein vaccine in vitro cell viability curve;
FIG. 4 experimental tumor growth curves in PD-HER2 fusion protein vaccine mice;
FIG. 5PD-HER2 fusion protein vaccine in vivo experimental survival curves in mice.
Detailed Description
In order to make the objects, technical solutions and advantages of the present invention more apparent, the present invention is further described in detail below with reference to the accompanying drawings and embodiments. It should be understood that the description of the specific embodiments is intended to illustrate and not to limit the invention.
Example 1Construction of recombinant PD-HER2 fusion protein therapeutic vaccine engineering bacteria
Construction of pET30-a (+) -PD-HER2/BL21(DE3) engineering bacteria
The Hi-derived PD protein (GenBank accession number: CAA84716) was selected to have a total length of 364 amino acids, as shown in SEQ ID NO: 1 is shown. Amino acids 19-127 of the PD protein are selected as adjuvant proteins, such as SEQ ID NO: 2, respectively. Tumor associated antigen human HER2(GenBank accession No.: AAA75493.1) has a full length of 1255 amino acids as shown in seq id no: 3, respectively. The HER2 antigen extracellular region fragment (41-560aa) and the HER2 epitope peptide fragment (774-788aa) are selected as vaccine molecule antigens. The amino acid sequence of the HER2 antigen extracellular region fragment (41-560aa) is shown as SEQ ID NO: 4, the HER2 epitope peptide fragment (774-788aa) is shown as SEQ ID NO: 5, respectively.
The recombinant PD-HER2 fusion protein therapeutic vaccine molecule is that amino acids 19-127 of PD protein are connected to the N end of HER2 antigen extracellular region segment (41-560aa) through Linker sequence-GGGGSGGGGSGGS-, and then HER2 epitope peptide segment (774-788aa) is directly connected to the C end of HER2 antigen extracellular region segment (41-560 aa). The designed PD-HER2 fusion protein has an amino acid sequence shown as SEQ ID NO: 6, and the corresponding coding nucleic acid sequence is shown as SEQ ID NO: shown at 7.
For convenient purification, a nucleic acid sequence corresponding to 6 × His tag and enterokinase site (HHHHHHDDDDK) and an NdeI recognition site, such as SEQ ID NO: 8, are sequentially added at the 5' end of the PD-HER2 fusion protein nucleic acid sequence (SEQ ID NO: 7)CATATGCACCACCACCACCACCACGACGACGACGATAAA, wherein the underline indicates the NdeI recognition site, and CACCACCACCACCACCACGACGACGACGATAAA-HHHHHHDDDDK corresponds to the coding nucleic acid sequence. Meanwhile, two stop codon sequences and a NotI recognition sequence are sequentially added at the 3' end of the PD-HER2 fusion protein nucleic acid sequence (SEQ ID NO: 7), such as SEQ ID NO: 9TAATGAGCGGCCGC, the two stop codon sequences underlined, GCGGCCGC is the NotI recognition sequence. The designed PD-HER2 fusion protein nucleotide sequence is entrusted to Nanjing Kingsry Biotech Co., Ltd for whole gene synthesis (the nucleotide sequence is SEQ ID NO: 10). The expression vector pET30-a (+) -PD-HER2 was constructed by subcloning NdeI/NotI sites (TAKARA, Dalian) into an expression vector selected from the commercial vector pET-30a-c (+) (Merk, USA), and the construction scheme is shown in FIG. 1.
The constructed expression vector is transferred into an escherichia coli expression host bacterium BL21(DE3) by a chemical transfection method, and a recombinant expression engineering bacterium pET30-a (+) -PD-HER2/BL21(DE3) is obtained by screening LB + kan solid plates.
2. Induced screening of engineering bacteria
The above pET30-a (+) -PD-HER2/BL21(DE3) engineered bacteria were randomly picked up and inoculated into 10ml LB medium (100ug/ml kan), cultured in 100ml Erlenmeyer flask, and shake-cultured overnight at 37 ℃ and 220 rpm. The next day, the mother solution of overnight culture was transferred to 40ml of LB liquid medium (100ug/ml kan) at a ratio of 1%, and cultured in a 250ml Erlenmeyer flask at 37 ℃ and 220rpm2.5h to OD600The value is about 0.6-1.0. Add 1mMIPTG to the final concentration and induce 4h at 30 ℃.
SDS-PAGE, the results are shown in FIG. 2, lanes 1-3: pET30-a (+) -PD-HER2/BL21(DE3) No. 1-3 engineering bacteria induces 4h total protein; comparison: total protein was not induced; lane M: protein molecular weight Marker; lanes S1, S2: 1. breaking the bacteria of the engineering bacteria No. 2 and supernatant; lanes P1, P2: 1. and (3) breaking the bacteria of the engineering bacteria No. 2 and precipitating. Compared with a control, the specific expression of 3 engineering bacteria at the theoretical molecular weight of 74kD is realized. 2 strains of the induced expression thalli are subjected to bacterium breaking analysis, and all the strains are expressed by inclusion bodies.
Example 2Fermentation of recombinant PD-HER2 fusion protein engineering bacteria
The pET30-a (+) -PD-HER2/BL21(DE3) engineered bacterium constructed in the above example 1 was streaked on LB plate (kan100mg/L), and cultured in a 37 ℃ incubator (Boxun, Shanghai) for about 16-18 hours until a single colony grew. A single colony of the engineering bacteria is selected and inoculated into 20ml LB culture medium (kan100mg/L), and cultured for 8h at 37 ℃ and 230 rpm. 0.1% was transferred to 250ml LB (kan100mg/L), 1L Erlenmeyer flask, cultured at 37 ℃ and 230rpm for 13 hours. Culturing 4 bottles in parallel, preparing 1000ml of bacterial liquid, inoculating 5% of the bacterial liquid into a tank-loading culture medium of a fermentation tank NLF-2220L, adjusting the pH to 7.0 by using ammonia water before inoculation, and controlling the temperature to be 36 ℃ in the fermentation process. The pH value and dissolved oxygen of the culture medium are controlled by feeding ammonia water and increasing the stirring speed and ventilation, and the dissolved oxygen is more than 30 percent. Carbon source depletion, OD in the culture Medium after about 5h600After reaching 20, the culture was continued by starting feeding the medium (40% glycerol + 20% yeast powder) at a rate of 240mL/h/20L of the medium, OD60035, feeding the feed culture medium at the speed of 60mL/h/20L of culture medium to start induction, feeding isopropyl- β -D-thiogalactoside (IPTG, Merk) as an inducer, keeping the feeding speed at a final concentration of 1 mM., maintaining DO above 30%, inducing for 4h, and discharging.
The thalli is collected by centrifugation, and suspended in a bacteria breaking buffer (20mmTris-HCl, 5mM EDTA, PH8.0), and the ratio of the thalli to the bacteria breaking buffer is as follows: 1g of cells: 10ml of buffer. After the suspension was sufficiently suspended, the suspension was crushed by a high-pressure homogenizer (model AH-PILOT manufactured by ATSENGINEERINGINC) under a pressure of 80MPa, and the cells were disrupted by circulation three times, and the inclusion bodies and the disrupted cells were separated by a high-speed refrigerated centrifuge (Hitachi, CR21 GIII). The centrifugation condition is 8000g at 10 ℃, and the centrifugation is carried out for 15 min. The precipitate was collected and the supernatant discarded.
Example 3 isolation and purification of recombinant PD-HER2 fusion protein
1. Inclusion body washing
(1) First washing
The precipitate collected in example 2 was taken and contained as Inclusion Bodies (IB) as a main component. Washed with wash buffer (20mM Tris-HCl, 2M urea (guggai, shanghai), 1% TritonX-100(sigma, USA), 5mM EDTA (guggai, shanghai), 0.2M NaCl (guggai, shanghai), pH 8.0). IB/washing buffer (1 g/20 ml), magnetic stirring at room temperature for 30min, and centrifuging at 8000g for 10min to remove supernatant.
(2) Second washing
The inclusion bodies collected after the first washing were precipitated and washed with a washing buffer (20mM Tris-HCl, 2M urea, 5mM EDTA, 0.2M NaCl, pH 8.0). IB/washing buffer (1 g/20 ml), magnetic stirring at room temperature for 30min, and centrifuging at 8000g for 10min to remove supernatant.
(3) Third washing
The inclusion bodies collected after the third washing were precipitated and washed with a washing buffer (20mM Tris-HCl, 5mM EDTA, 0.2M NaCl, pH 8.0). IB/washing buffer (1 g/20 ml), magnetic stirring at room temperature for 30min, and centrifuging at 8000g for 10min to remove supernatant.
2. Inclusion body solubilization and chromatographic renaturation
(1) Dissolution of IB
Dissolving buffer solution: 20mM Tris-HClpH8.0+8M Urea (national medicine, Shanghai) +5mM DTT (sigma, USA). IB (g): (1 g): (ml): (1 g): (10 ml), and the mixture was dissolved at room temperature for 6 hours with stirring by a magnetic stirrer.
(2) Renaturation
100ml of the inclusion body dissolved sample is taken, added into 900ml of dilution renaturation buffer (1M urea, 5% glycerol, 0.1M NaCl, 50M Tris-HCl, pH8.0) at the flow rate of 0.2ml/min for dilution renaturation, the buffer is stirred at a low rotation speed (60rpm/min) by a magnetic stirrer, the sample is stirred for about 4 hours after the addition is finished, then the mixture is placed into a chromatography cabinet at 4 ℃ for overnight renaturation (12 hours), and the concentration of a denaturant in the sample after dilution renaturation is between 1.5 and 2M.
(3) Purification by chelate chromatography
Column specification and treatment: Φ 3.5cm × H10cm (chelating fillers 80ml, GE, USA); the column was buffered with 0.1M nickel sulfate 4 column volumes and then the dH2O test water was used to wash the column 10 column volumes.
And (3) chelating chromatographic equilibrium: equilibration buffer (2.0M urea, 0.1M NaCl, 50mM Tris-HCl, pH8.0), flow rate 20ml/min, equilibration time 30 min.
Pumping into the chromatographic column by a peristaltic pump at a flow rate of 20 ml/min. After the sample injection is finished, the balance is continued for 20 min. Eluted with buffer (20mMPB, 0.2M NaCl, 5% glycerol, pH8.0) at a flow rate of 20 ml/min. Then eluted with a buffer (20mM PB, 0.2M NaCl +,% glycerol, 40mM imidazole, pH8.0) at a flow rate of 20 ml/min. Then eluted with a buffer (20mM PB, 0.2M NaCl, 5% glycerol, 200mM imidazole, pH8.0) at a flow rate of 20ml/min, and the desired peak was collected.
The collected fusion protein of interest was desalted and digested with 0.5U enterokinase (sigma, USA, 50mM Tris-HCl, 2mM CaCl2, 0.1% Tween-20, pH8.0) per 1mg of fusion protein at 6-8 ℃ for about 17 hours.
The enzyme-digested target protein is loaded on Ni2+-ChelatingSepharose Fastflow (GE, USA), and the target protein and fusion tag were simultaneously hung on the column. However, the binding force of the target protein hanging column is low, and the target protein is eluted by imidazole (Shanghai Biotech, batch number: B421BA0025) with the concentration of 50mM, and the peak of the eluted protein is collected. Then performing CMFF fine purificationObtaining the high-purity protein. The molecular weight of the product is about 72kD as detected by SDS-PAGE, and the purity is more than 95%.
Example 4 in vitro Activity assay of recombinant PD-HER2 fusion protein therapeutic vaccines
PMA stimulation THP-1 differentiation macrophage detection recombinant protein immunogenicity
1. The method uses macrophage differentiated from THP-1 cell (ATCC, USA) to measure the activity of recombinant PD-HER2 fusion protein, and can directly and truly reflect the immunogenicity of the recombinant protein in an in vitro environment.
2. Experimental methods
The activity of the recombinant PD-HER2 fusion protein is determined by adding phorbol ester (PMA, sigma) to stimulate THP-1 cells to differentiate into macrophages, utilizing the capability of the macrophages to take up and present antigens and detecting the content of TNF-alpha generated by the macrophages after adding the antigens through a TNF-alpha detection kit (MABTECH, the code of 3510-1H-6).
2.1PMA stimulation of THP-1 differentiation
2.1.1 harvesting THP-1 cells, centrifuging (300g, 5 min) to collect the cells, and discarding the supernatant;
2.1.2 resuspend cells in 1640 medium with 10% FBS and mix well;
2.1.3 under the mirror for cell count 1, and using the culture medium to adjust the cell density to 7X 105/ml;
2.1.4 setting the concentration of PMA stimulant at 100 ng/ml;
2.1.5 adding the uniformly mixed THP-1 cell sap containing PMA into a 24-hole cell culture plate at a volume of 500 mul/well; placing the cell culture plate at 37 ℃ and a 5% CO2 cell culture box for culturing for 37 h;
2.2 antigenic stimulation of macrophages
2.2.1 serum-free treatment for 24h, medium in the 24-well plate was aspirated and washed once with serum-free 1640 medium, followed by addition of 500. mu.l serum-free medium per well for further 24 h.
2.2.2 Add 500. mu.l of diluted antigen to the corresponding wells in groups as above, continue incubation for 24h, collect the supernatant and use the kit to detect TNF-. alpha.levels.
2.2.3 TNF-alpha ELISA assay
The coating was taken 28. mu.l and PBS was added to 7 ml.100. mu.l/well overnight at 4 ℃.
The plates were washed 2 times with PBS.
Blocking was performed with PBS37 containing 0.1% BSA for 1 hour.
Wash plate 5 times, PBS + 0.05% tween 20;
adding 1 μ g/ml standard, adding PBS containing 0.1% BSA, diluting to 10ng/ml, and making 7 2-fold dilutions; culture supernatants were aspirated from the cell culture plates at 100. mu.l/well. At 37 ℃ for 2 hours.
The plate was washed 5 times.
Adding mAbTNF 5-biotin: mu.l of PBS containing 0.1% BSA was taken up to 7ml.100 μ l/well. The reaction was carried out at 37 ℃ for 1 hour.
The plate was washed 5 times.
streptavidin-HRP 7. mu.l was added to PBS containing 0.1% BSA to 7ml, 100. mu.l/well. The reaction was carried out at 37 ℃ for 1 hour.
The plate was washed 5 times.
Color development: each well was filled with 100. mu.l of TMB substrate solution and acted upon at 26 ℃.
650nm dynamic plate reader (detection every 1min, moderate oscillation 3sec)
Calculation of the diastolic EC with Graphprism5.0 software50And half-effect dilution factor, calculated as:
3. results of the experiment
The PD-HER2 fusion protein prepared by the invention has biological activitySexual EC508 μ g/ml, see FIG. 3.
EXAMPLE 5 recombinant PD-HER2 fusion protein therapeutic vaccine in mice tumor suppressor Activity assay
1. The method uses Balb/c mouse 4T1-HER2 transfer animal model to determine the tumor inhibition activity of the PD-HER2 fusion protein prepared by the invention. Wherein the 4T1-HER2 cell is constructed by transfecting cDNA carrying human HER2 with a retrovirus into a mouse breast cancer 4T1 cell line (ATCC, USA); wherein the 4T1-HER2 breast cancer animal model is an animal model established subcutaneously by injecting 4T1-HER2 into female BALB/C mice.
2. Experimental methods
2.14T1-HER2Balb/c mouse animal model establishment
40 BALB/c mice were inoculated subcutaneously in situ with 1 × 105Cell/tumor 4T1-HER2 suspension.
2.2 recombinant PD-HER2 fusion protein immunized animals
At the same time of tumor inoculation, the mice are divided into 4 groups and are treated by the drug, 10 mice are respectively treated by 2-3 times of immunization injection, and the interval between the two immunizations is 1 week, namely, the drug is respectively administered on 0, 7 and 14 days of tumor inoculation. The grouping is as follows:
control group: injecting PBS buffer;
② vaccine group: the dosage of the vaccine is 100 mu g/mouse;
③ adjuvant group: injection of an adjuvant;
vaccine + adjuvant group: the vaccine is mixed with 100 mug of adjuvant, shaken up and injected.
2.3 Experimental period
The experimental period was 50 natural days from the day of tumor inoculation. During the experiment, tumor volume and body weight of mice were measured 2 times per week, and tumor rate and tumor volume dynamic records were made, along with survival rate.
2.4 results of the experiment
The recombinant protein and Her2 fusion protein can inhibit the proliferation of 4T1-HER2 cells in a breast cancer animal model of Balb/c mice within 50 days, and the tumor growth inhibition rate reaches over 90 percent, as shown in figure 4. Therefore, the research and development of the therapeutic vaccine of the protein D and Her2 fusion protein has good immunotherapeutic effect on inhibiting the proliferation of 4T1-Her2 cells in a Balb/c mouse breast cancer animal model under the condition of containing or not containing an adjuvant, and can improve the survival rate of the Balb/c mouse breast cancer animal model, and the reference of figure 5 shows that the therapeutic vaccine of the protein D and Her2 fusion protein has good immunotherapeutic effect on inhibiting the proliferation of 4T1-Her2 cells in the.
Although the present invention has been described to a certain extent, it is apparent that appropriate changes in the respective conditions may be made without departing from the spirit and scope of the present invention. It is to be understood that the invention is not limited to the described embodiments, but is to be accorded the scope consistent with the claims, including equivalents of each element described.
Claims (10)
1. A fusion protein, which is formed by connecting the adjuvant protein with the N-terminal or C-terminal of a tumor-associated antigen through a Linker (Linker) or a direct connection manner; wherein,
the adjuvant protein comprises haemophilus influenzae (haemophilus influenzae, Hi) protein D or a fragment of the haemophilus influenzae protein D;
the tumor-associated antigen comprises an amino acid sequence of SEQ ID NO: 3 or a fragment thereof, HER2 protein; preferably, the tumor-associated antigen is formed by combining an extracellular region fragment of a human HER2 antigen and a peptide fragment of a HER2 epitope, and the amino acid sequence of the extracellular region fragment of the HER2 antigen is represented by SEQ ID NO: 4, the amino acid sequence of the HER2 epitope peptide fragment is SEQ ID NO: 5;
preferably, the amino acid sequence of the adjuvant protein is seq id no: 2;
preferably, the amino acid sequence of the linker is-GGGGSGGGGSGGGGS-; preferably, the adjuvant protein is linked to the N-terminus of the tumor associated antigen by direct linkage.
2. The fusion protein of claim 1, wherein the amino acid sequence of the fusion protein is seq id no: 6.
3. an expression vector constructed by linking a nucleotide sequence encoding the fusion protein of claim 1 or 2 to a commercial vector selected from one or more of pET-28a-c (+), pET29a, pET-30a-c (+), pET39b (+), pET-40b (+), pET-41a (+) and pET-43.1a (+) and optionally adding a cleavage recognition site sequence and/or a sequence encoding a protein tag; preferably, the nucleotide sequence is seq id no: 7.
4. the expression vector of claim 3, further comprising a coding nucleic acid sequence corresponding to the sequential arrangement of the enzyme cleavage recognition site, the histidine tag and the enterokinase site at the 5' end of the nucleotide sequence, preferably the sequential arrangement of the enzyme cleavage recognition site, the histidine tag and the enterokinase site corresponds to the coding nucleic acid sequence of SEQ ID NO: 8.
5. the expression vector of claim 3 or 4, further comprising a stop codon sequence and a cleavage recognition site sequence located 3' of the nucleotide sequence; preferably, the stop codon sequence and the enzyme cutting recognition site sequence are SEQ ID NO: 9; more preferably, the nucleotide sequence encoding the fusion protein is seq id no: 12.
6. a host cell comprising the expression vector of any one of claims 3 to 5, wherein the host cell is a bacterium or a fungus; preferably, the host cell is escherichia coli; more preferably, the E.coli is selected from BL21(DE3), BL21(DE3) PlysS or TB 1.
7. A method for preparing the fusion protein of claim 1 or 2, wherein the host cell of claim 6 is induced by IPTG to express the fusion protein, the inclusion bodies are collected, the fusion protein in the inclusion bodies is extracted and purified to obtain the fusion protein;
preferably, the purification is achieved by:
the fusion protein was purified by passing through a chelate column.
8. The method of claim 7, wherein the purifying step further comprises:
the fusion protein is cut by enterokinase, then the cut fusion protein is loaded on the cut target protein and loaded on Ni2+ -ChelatingSepharose Fastflow, and then the fusion protein is eluted by imidazole with the concentration of 50 mM.
9. A HER2 antigen positive tumor vaccine comprising the fusion protein of claim 1 or 2; preferably, the vaccine is a HER2 antigen positive tumor therapeutic vaccine; preferably, the antigen positive tumor is a breast tumor.
10. The vaccine of claim 9, further comprising an adjuvant selected from the group consisting of GM-CSF, CpG, and GM-CSF + CpG; preferably, the vaccine is in the form of injection.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201510789201.9A CN105254767A (en) | 2015-11-17 | 2015-11-17 | Protein D and HER2 fusion protein and preparing method and application thereof |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201510789201.9A CN105254767A (en) | 2015-11-17 | 2015-11-17 | Protein D and HER2 fusion protein and preparing method and application thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
CN105254767A true CN105254767A (en) | 2016-01-20 |
Family
ID=55094742
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN201510789201.9A Pending CN105254767A (en) | 2015-11-17 | 2015-11-17 | Protein D and HER2 fusion protein and preparing method and application thereof |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN105254767A (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2017214833A1 (en) * | 2016-06-14 | 2017-12-21 | 石庆学 | Lentiviral vector for specifically promoting high expression of erbb2 gene, and applications thereof |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN101668770A (en) * | 2007-01-15 | 2010-03-10 | 葛兰素史密丝克莱恩生物有限公司 | Vaccine |
CN102392041A (en) * | 2011-12-08 | 2012-03-28 | 重庆科润生物医药研发有限公司 | Preparation method of recombinant human corticotropin releasing factor |
-
2015
- 2015-11-17 CN CN201510789201.9A patent/CN105254767A/en active Pending
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN101668770A (en) * | 2007-01-15 | 2010-03-10 | 葛兰素史密丝克莱恩生物有限公司 | Vaccine |
CN102392041A (en) * | 2011-12-08 | 2012-03-28 | 重庆科润生物医药研发有限公司 | Preparation method of recombinant human corticotropin releasing factor |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2017214833A1 (en) * | 2016-06-14 | 2017-12-21 | 石庆学 | Lentiviral vector for specifically promoting high expression of erbb2 gene, and applications thereof |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN110835371A (en) | anti-CCR 8 monoclonal antibody and application thereof | |
PT2155243E (en) | Compositions and methods comprising klk3, psca, or folh1 antigen | |
CA2674458A1 (en) | Fusion protein | |
WO2021190551A1 (en) | Chimeric antigen receptor that binds to cd19 and use thereof | |
CN108066755B (en) | Genetic engineering subunit vaccine for resisting sheep echinococcosis infection and preparation method and application thereof | |
CN106350527B (en) | One kind can be in the highly expressed diphtheria toxin mutation of bacterium coli solubility | |
US8685670B2 (en) | Isolated chimeric proteins of modified lumazine synthase | |
CN102633867A (en) | Antigenically-changed enterotoxin C2 mutant, coding gene thereof, preparation thereof and application thereof | |
BR112021004241A2 (en) | improved process for preparing recombinant lectin protein | |
Yi et al. | Improved efficacy of DNA vaccination against breast cancer by boosting with the repeat beta-hCG C-terminal peptide carried by mycobacterial heat-shock protein HSP65 | |
CN103421731B (en) | A kind of haemophilus parasuis attenuation salmonella seedling | |
Zhang et al. | Soluble production and function of vascular endothelial growth factor/basic fibroblast growth factor complex peptide | |
CN103360497A (en) | Novel antitumor fusion protein vaccine, and preparation method and application thereof | |
CN105254767A (en) | Protein D and HER2 fusion protein and preparing method and application thereof | |
CN110167576A (en) | The synthesis for targeting the optimization of fibroblast activation protein shares immunogenic composition | |
KR20200046059A (en) | Immunogenic peptides for group A streptococcus | |
CN102153644B (en) | Antigenic peptide for dimerization of epidermal growth factor receptor | |
CN111410693B (en) | CDK5 resistant nano antibody and application thereof | |
CN104558148A (en) | Ciliary neurotrophic factor mutant, and modified mutant and application thereof | |
CN111118063B (en) | FAP alpha and survivin-based DNA and application thereof in preparation of tumor vaccine | |
CN111349645B (en) | Method for improving safety of non-integrated attenuated listeria vaccine | |
CN108671228A (en) | antigen and antigen combination | |
CA3184665A1 (en) | Amuc-1100 polypeptide variants for effecting immune signalling and/or affecting intestinal barrier function and/or modulating metabolic status | |
RU2590701C2 (en) | Antigenic composition and therapeutic application thereof for preventing and treating oncological diseases, recombinant plasmid dna, providing synthesis of hybrid protein, as well as method of producing protein | |
CN106085932A (en) | A kind of structure of the genetic engineering bacterium of VEGF Yu GnRH fusion protein of recombinating |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
C06 | Publication | ||
PB01 | Publication | ||
C10 | Entry into substantive examination | ||
SE01 | Entry into force of request for substantive examination | ||
WD01 | Invention patent application deemed withdrawn after publication | ||
WD01 | Invention patent application deemed withdrawn after publication |
Application publication date: 20160120 |