CN115260304A - Lipid-based protein degradation tool, application and preparation method thereof - Google Patents
Lipid-based protein degradation tool, application and preparation method thereof Download PDFInfo
- Publication number
- CN115260304A CN115260304A CN202210911853.5A CN202210911853A CN115260304A CN 115260304 A CN115260304 A CN 115260304A CN 202210911853 A CN202210911853 A CN 202210911853A CN 115260304 A CN115260304 A CN 115260304A
- Authority
- CN
- China
- Prior art keywords
- lipid
- protein degradation
- hybrid
- based protein
- recognition group
- 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
- 150000002632 lipids Chemical class 0.000 title claims abstract description 259
- 230000017854 proteolysis Effects 0.000 title claims abstract description 96
- 238000002360 preparation method Methods 0.000 title claims abstract description 32
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 89
- 102000004169 proteins and genes Human genes 0.000 claims abstract description 88
- 239000000126 substance Substances 0.000 claims abstract description 66
- 239000002502 liposome Substances 0.000 claims abstract description 28
- 239000003814 drug Substances 0.000 claims abstract description 25
- 229920001184 polypeptide Polymers 0.000 claims abstract description 13
- 102000004196 processed proteins & peptides Human genes 0.000 claims abstract description 13
- 108090000765 processed proteins & peptides Proteins 0.000 claims abstract description 13
- 230000027455 binding Effects 0.000 claims abstract description 9
- 239000002105 nanoparticle Substances 0.000 claims description 58
- 229920000642 polymer Polymers 0.000 claims description 40
- 238000005859 coupling reaction Methods 0.000 claims description 37
- 238000010168 coupling process Methods 0.000 claims description 36
- 230000008878 coupling Effects 0.000 claims description 34
- 239000002245 particle Substances 0.000 claims description 29
- 201000010099 disease Diseases 0.000 claims description 22
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 22
- 238000000034 method Methods 0.000 claims description 21
- 230000002209 hydrophobic effect Effects 0.000 claims description 18
- 210000001808 exosome Anatomy 0.000 claims description 17
- 238000009825 accumulation Methods 0.000 claims description 13
- 230000002159 abnormal effect Effects 0.000 claims description 12
- 210000000170 cell membrane Anatomy 0.000 claims description 11
- 238000011282 treatment Methods 0.000 claims description 11
- 229920001477 hydrophilic polymer Polymers 0.000 claims description 8
- 230000002797 proteolythic effect Effects 0.000 claims description 8
- 150000003384 small molecules Chemical class 0.000 claims description 8
- 206010028980 Neoplasm Diseases 0.000 claims description 7
- 210000004369 blood Anatomy 0.000 claims description 6
- 239000008280 blood Substances 0.000 claims description 6
- 229920001600 hydrophobic polymer Polymers 0.000 claims description 6
- 208000026278 immune system disease Diseases 0.000 claims description 6
- 208000030159 metabolic disease Diseases 0.000 claims description 6
- 239000002114 nanocomposite Substances 0.000 claims description 6
- 230000004770 neurodegeneration Effects 0.000 claims description 6
- 208000015122 neurodegenerative disease Diseases 0.000 claims description 6
- 108091023037 Aptamer Proteins 0.000 claims description 5
- 230000001717 pathogenic effect Effects 0.000 claims description 5
- 206010061218 Inflammation Diseases 0.000 claims description 4
- 208000015181 infectious disease Diseases 0.000 claims description 4
- 230000004054 inflammatory process Effects 0.000 claims description 4
- 244000052769 pathogen Species 0.000 claims description 4
- 229960005486 vaccine Drugs 0.000 claims description 4
- 238000012360 testing method Methods 0.000 claims description 3
- 230000006916 protein interaction Effects 0.000 claims description 2
- 230000006806 disease prevention Effects 0.000 claims 1
- 125000003473 lipid group Chemical group 0.000 claims 1
- 230000015556 catabolic process Effects 0.000 abstract description 34
- 238000006731 degradation reaction Methods 0.000 abstract description 34
- 229940079593 drug Drugs 0.000 abstract description 22
- 230000015572 biosynthetic process Effects 0.000 abstract description 11
- 238000003786 synthesis reaction Methods 0.000 abstract description 11
- 150000001875 compounds Chemical class 0.000 abstract description 6
- 238000011160 research Methods 0.000 abstract description 6
- 238000011161 development Methods 0.000 abstract description 5
- 238000002648 combination therapy Methods 0.000 abstract description 3
- 230000006870 function Effects 0.000 abstract description 3
- 108091008104 nucleic acid aptamers Proteins 0.000 abstract description 2
- 235000018102 proteins Nutrition 0.000 description 67
- 239000000243 solution Substances 0.000 description 43
- 229920001223 polyethylene glycol Polymers 0.000 description 37
- 239000008055 phosphate buffer solution Substances 0.000 description 35
- 102000052116 epidermal growth factor receptor activity proteins Human genes 0.000 description 31
- 108700015053 epidermal growth factor receptor activity proteins Proteins 0.000 description 31
- 210000004027 cell Anatomy 0.000 description 29
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 29
- 239000012528 membrane Substances 0.000 description 29
- PZNPLUBHRSSFHT-RRHRGVEJSA-N 1-hexadecanoyl-2-octadecanoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCCCC(=O)O[C@@H](COP([O-])(=O)OCC[N+](C)(C)C)COC(=O)CCCCCCCCCCCCCCC PZNPLUBHRSSFHT-RRHRGVEJSA-N 0.000 description 27
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 24
- 238000001962 electrophoresis Methods 0.000 description 24
- 239000002202 Polyethylene glycol Substances 0.000 description 23
- 230000000694 effects Effects 0.000 description 18
- -1 cationic lipid Chemical class 0.000 description 17
- 239000000203 mixture Substances 0.000 description 15
- 238000000108 ultra-filtration Methods 0.000 description 14
- NQTADLQHYWFPDB-UHFFFAOYSA-N N-Hydroxysuccinimide Chemical compound ON1C(=O)CCC1=O NQTADLQHYWFPDB-UHFFFAOYSA-N 0.000 description 13
- 108010090849 Amyloid beta-Peptides Proteins 0.000 description 12
- 102000013455 Amyloid beta-Peptides Human genes 0.000 description 12
- 235000012000 cholesterol Nutrition 0.000 description 12
- 238000010586 diagram Methods 0.000 description 12
- NRJAVPSFFCBXDT-HUESYALOSA-N 1,2-distearoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCCCCCCCCCCC NRJAVPSFFCBXDT-HUESYALOSA-N 0.000 description 11
- 206010006187 Breast cancer Diseases 0.000 description 11
- 208000026310 Breast neoplasm Diseases 0.000 description 11
- 210000004881 tumor cell Anatomy 0.000 description 11
- 238000001514 detection method Methods 0.000 description 10
- 238000012377 drug delivery Methods 0.000 description 10
- 238000002296 dynamic light scattering Methods 0.000 description 9
- 125000005647 linker group Chemical group 0.000 description 9
- 239000000463 material Substances 0.000 description 9
- 108020004707 nucleic acids Proteins 0.000 description 9
- 102000039446 nucleic acids Human genes 0.000 description 9
- 150000007523 nucleic acids Chemical class 0.000 description 9
- KSXTUUUQYQYKCR-LQDDAWAPSA-M 2,3-bis[[(z)-octadec-9-enoyl]oxy]propyl-trimethylazanium;chloride Chemical compound [Cl-].CCCCCCCC\C=C/CCCCCCCC(=O)OCC(C[N+](C)(C)C)OC(=O)CCCCCCC\C=C/CCCCCCCC KSXTUUUQYQYKCR-LQDDAWAPSA-M 0.000 description 8
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 8
- 238000002474 experimental method Methods 0.000 description 8
- 239000010410 layer Substances 0.000 description 8
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 description 8
- AHJRHEGDXFFMBM-UHFFFAOYSA-N palbociclib Chemical compound N1=C2N(C3CCCC3)C(=O)C(C(=O)C)=C(C)C2=CN=C1NC(N=C1)=CC=C1N1CCNCC1 AHJRHEGDXFFMBM-UHFFFAOYSA-N 0.000 description 8
- 229960004390 palbociclib Drugs 0.000 description 8
- 238000000746 purification Methods 0.000 description 8
- 239000011541 reaction mixture Substances 0.000 description 8
- 229920002307 Dextran Polymers 0.000 description 7
- 239000004698 Polyethylene Substances 0.000 description 7
- 238000013461 design Methods 0.000 description 7
- 239000010408 film Substances 0.000 description 7
- 229920000573 polyethylene Polymers 0.000 description 7
- LVNGJLRDBYCPGB-UHFFFAOYSA-N 1,2-distearoylphosphatidylethanolamine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(COP([O-])(=O)OCC[NH3+])OC(=O)CCCCCCCCCCCCCCCCC LVNGJLRDBYCPGB-UHFFFAOYSA-N 0.000 description 6
- 238000002835 absorbance Methods 0.000 description 6
- 230000021615 conjugation Effects 0.000 description 6
- 238000001727 in vivo Methods 0.000 description 6
- QZIQJVCYUQZDIR-UHFFFAOYSA-N mechlorethamine hydrochloride Chemical compound Cl.ClCCN(C)CCCl QZIQJVCYUQZDIR-UHFFFAOYSA-N 0.000 description 6
- 239000000872 buffer Substances 0.000 description 5
- 125000002091 cationic group Chemical group 0.000 description 5
- JEJLGIQLPYYGEE-UHFFFAOYSA-N glycerol dipalmitate Natural products CCCCCCCCCCCCCCCC(=O)OCC(CO)OC(=O)CCCCCCCCCCCCCCC JEJLGIQLPYYGEE-UHFFFAOYSA-N 0.000 description 5
- 230000003834 intracellular effect Effects 0.000 description 5
- 108700021021 mRNA Vaccine Proteins 0.000 description 5
- 229940126582 mRNA vaccine Drugs 0.000 description 5
- 229940125645 monoclonal antibody drug Drugs 0.000 description 5
- 150000003839 salts Chemical class 0.000 description 5
- 239000000523 sample Substances 0.000 description 5
- 238000000527 sonication Methods 0.000 description 5
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 4
- 238000005481 NMR spectroscopy Methods 0.000 description 4
- 239000004696 Poly ether ether ketone Substances 0.000 description 4
- 239000004372 Polyvinyl alcohol Substances 0.000 description 4
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 4
- 108020004459 Small interfering RNA Proteins 0.000 description 4
- 238000004113 cell culture Methods 0.000 description 4
- 229960005395 cetuximab Drugs 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 229920001577 copolymer Polymers 0.000 description 4
- 230000018109 developmental process Effects 0.000 description 4
- 238000010790 dilution Methods 0.000 description 4
- 239000012895 dilution Substances 0.000 description 4
- 238000003306 harvesting Methods 0.000 description 4
- 238000012986 modification Methods 0.000 description 4
- 230000004048 modification Effects 0.000 description 4
- 229910052757 nitrogen Inorganic materials 0.000 description 4
- 229920003229 poly(methyl methacrylate) Polymers 0.000 description 4
- 229920001610 polycaprolactone Polymers 0.000 description 4
- 239000004632 polycaprolactone Substances 0.000 description 4
- 229920002530 polyetherether ketone Polymers 0.000 description 4
- 239000004926 polymethyl methacrylate Substances 0.000 description 4
- 229920002451 polyvinyl alcohol Polymers 0.000 description 4
- 239000004055 small Interfering RNA Substances 0.000 description 4
- JEJLGIQLPYYGEE-XIFFEERXSA-N 1,2-dipalmitoyl-sn-glycerol Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](CO)OC(=O)CCCCCCCCCCCCCCC JEJLGIQLPYYGEE-XIFFEERXSA-N 0.000 description 3
- 102000013701 Cyclin-Dependent Kinase 4 Human genes 0.000 description 3
- 108010025464 Cyclin-Dependent Kinase 4 Proteins 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 108050001049 Extracellular proteins Proteins 0.000 description 3
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 3
- 238000010521 absorption reaction Methods 0.000 description 3
- 125000003277 amino group Chemical group 0.000 description 3
- 108010064539 amyloid beta-protein (1-42) Proteins 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- 238000004624 confocal microscopy Methods 0.000 description 3
- 238000001704 evaporation Methods 0.000 description 3
- 230000008020 evaporation Effects 0.000 description 3
- 238000001125 extrusion Methods 0.000 description 3
- 238000004108 freeze drying Methods 0.000 description 3
- 230000036571 hydration Effects 0.000 description 3
- 238000006703 hydration reaction Methods 0.000 description 3
- 229910052739 hydrogen Inorganic materials 0.000 description 3
- 239000001257 hydrogen Substances 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 210000003712 lysosome Anatomy 0.000 description 3
- 230000001868 lysosomic effect Effects 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- 229950010203 nimotuzumab Drugs 0.000 description 3
- WTJKGGKOPKCXLL-RRHRGVEJSA-N phosphatidylcholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCC=CCCCCCCCC WTJKGGKOPKCXLL-RRHRGVEJSA-N 0.000 description 3
- 229920001308 poly(aminoacid) Polymers 0.000 description 3
- 229920001606 poly(lactic acid-co-glycolic acid) Polymers 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 238000001338 self-assembly Methods 0.000 description 3
- 229940126586 small molecule drug Drugs 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 238000009210 therapy by ultrasound Methods 0.000 description 3
- 230000001988 toxicity Effects 0.000 description 3
- 231100000419 toxicity Toxicity 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 3
- OKLASJZQBDJAPH-UHFFFAOYSA-N (2-dodecanoyloxy-3-phosphonooxypropyl) dodecanoate Chemical compound CCCCCCCCCCCC(=O)OCC(COP(O)(O)=O)OC(=O)CCCCCCCCCCC OKLASJZQBDJAPH-UHFFFAOYSA-N 0.000 description 2
- CITHEXJVPOWHKC-UUWRZZSWSA-N 1,2-di-O-myristoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCCCCCCC CITHEXJVPOWHKC-UUWRZZSWSA-N 0.000 description 2
- KILNVBDSWZSGLL-KXQOOQHDSA-N 1,2-dihexadecanoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCCCCCCCCC KILNVBDSWZSGLL-KXQOOQHDSA-N 0.000 description 2
- TZCPCKNHXULUIY-RGULYWFUSA-N 1,2-distearoyl-sn-glycero-3-phosphoserine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP(O)(=O)OC[C@H](N)C(O)=O)OC(=O)CCCCCCCCCCCCCCCCC TZCPCKNHXULUIY-RGULYWFUSA-N 0.000 description 2
- JFBCSFJKETUREV-LJAQVGFWSA-N 1,2-ditetradecanoyl-sn-glycerol Chemical compound CCCCCCCCCCCCCC(=O)OC[C@H](CO)OC(=O)CCCCCCCCCCCCC JFBCSFJKETUREV-LJAQVGFWSA-N 0.000 description 2
- RKDVKSZUMVYZHH-UHFFFAOYSA-N 1,4-dioxane-2,5-dione Chemical compound O=C1COC(=O)CO1 RKDVKSZUMVYZHH-UHFFFAOYSA-N 0.000 description 2
- JLPULHDHAOZNQI-ZTIMHPMXSA-N 1-hexadecanoyl-2-(9Z,12Z-octadecadienoyl)-sn-glycero-3-phosphocholine Chemical class CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCC\C=C/C\C=C/CCCCC JLPULHDHAOZNQI-ZTIMHPMXSA-N 0.000 description 2
- KYHYBIADZPAZKE-LQDDAWAPSA-M 2,3-bis[[(z)-octadec-9-enoyl]oxy]propyl-trimethylazanium;bromide Chemical compound [Br-].CCCCCCCC\C=C/CCCCCCCC(=O)OCC(C[N+](C)(C)C)OC(=O)CCCCCCC\C=C/CCCCCCCC KYHYBIADZPAZKE-LQDDAWAPSA-M 0.000 description 2
- 208000024827 Alzheimer disease Diseases 0.000 description 2
- 102000005427 Asialoglycoprotein Receptor Human genes 0.000 description 2
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 2
- 229940083347 Cyclin-dependent kinase 4 inhibitor Drugs 0.000 description 2
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 2
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 2
- 229910000640 Fe alloy Inorganic materials 0.000 description 2
- ZWZWYGMENQVNFU-UHFFFAOYSA-N Glycerophosphorylserin Natural products OC(=O)C(N)COP(O)(=O)OCC(O)CO ZWZWYGMENQVNFU-UHFFFAOYSA-N 0.000 description 2
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 2
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 2
- 229920000805 Polyaspartic acid Polymers 0.000 description 2
- 229920002873 Polyethylenimine Polymers 0.000 description 2
- 108010020346 Polyglutamic Acid Proteins 0.000 description 2
- 108010039918 Polylysine Proteins 0.000 description 2
- 229930182558 Sterol Natural products 0.000 description 2
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 2
- 102000003970 Vinculin Human genes 0.000 description 2
- 108090000384 Vinculin Proteins 0.000 description 2
- ATBOMIWRCZXYSZ-XZBBILGWSA-N [1-[2,3-dihydroxypropoxy(hydroxy)phosphoryl]oxy-3-hexadecanoyloxypropan-2-yl] (9e,12e)-octadeca-9,12-dienoate Chemical compound CCCCCCCCCCCCCCCC(=O)OCC(COP(O)(=O)OCC(O)CO)OC(=O)CCCCCCC\C=C\C\C=C\CCCCC ATBOMIWRCZXYSZ-XZBBILGWSA-N 0.000 description 2
- AWUCVROLDVIAJX-UHFFFAOYSA-N alpha-glycerophosphate Natural products OCC(O)COP(O)(O)=O AWUCVROLDVIAJX-UHFFFAOYSA-N 0.000 description 2
- 108010006523 asialoglycoprotein receptor Proteins 0.000 description 2
- TZCXTZWJZNENPQ-UHFFFAOYSA-L barium sulfate Chemical compound [Ba+2].[O-]S([O-])(=O)=O TZCXTZWJZNENPQ-UHFFFAOYSA-L 0.000 description 2
- 230000004888 barrier function Effects 0.000 description 2
- 239000011324 bead Substances 0.000 description 2
- 239000006143 cell culture medium Substances 0.000 description 2
- 239000011248 coating agent Substances 0.000 description 2
- 238000000576 coating method Methods 0.000 description 2
- 239000002131 composite material Substances 0.000 description 2
- 239000012141 concentrate Substances 0.000 description 2
- 150000001982 diacylglycerols Chemical class 0.000 description 2
- 235000014113 dietary fatty acids Nutrition 0.000 description 2
- 229960003724 dimyristoylphosphatidylcholine Drugs 0.000 description 2
- 239000006185 dispersion Substances 0.000 description 2
- 210000003617 erythrocyte membrane Anatomy 0.000 description 2
- 239000000194 fatty acid Substances 0.000 description 2
- 229930195729 fatty acid Natural products 0.000 description 2
- 150000004665 fatty acids Chemical class 0.000 description 2
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 2
- 150000002313 glycerolipids Chemical class 0.000 description 2
- 150000002327 glycerophospholipids Chemical class 0.000 description 2
- KWIUHFFTVRNATP-UHFFFAOYSA-N glycine betaine Chemical compound C[N+](C)(C)CC([O-])=O KWIUHFFTVRNATP-UHFFFAOYSA-N 0.000 description 2
- 229940099578 hydrogenated soybean lecithin Drugs 0.000 description 2
- 239000005457 ice water Substances 0.000 description 2
- 238000003384 imaging method Methods 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- JJTUDXZGHPGLLC-UHFFFAOYSA-N lactide Chemical compound CC1OC(=O)C(C)OC1=O JJTUDXZGHPGLLC-UHFFFAOYSA-N 0.000 description 2
- 238000011068 loading method Methods 0.000 description 2
- 230000002132 lysosomal effect Effects 0.000 description 2
- 239000003550 marker Substances 0.000 description 2
- 230000000877 morphologic effect Effects 0.000 description 2
- 239000007908 nanoemulsion Substances 0.000 description 2
- 210000004498 neuroglial cell Anatomy 0.000 description 2
- 230000001575 pathological effect Effects 0.000 description 2
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 2
- 150000003905 phosphatidylinositols Chemical class 0.000 description 2
- YHHSONZFOIEMCP-UHFFFAOYSA-O phosphocholine Chemical compound C[N+](C)(C)CCOP(O)(O)=O YHHSONZFOIEMCP-UHFFFAOYSA-O 0.000 description 2
- 150000003904 phospholipids Chemical class 0.000 description 2
- 229920001562 poly(N-(2-hydroxypropyl)methacrylamide) Polymers 0.000 description 2
- 229920002401 polyacrylamide Polymers 0.000 description 2
- 108010064470 polyaspartate Proteins 0.000 description 2
- 239000004417 polycarbonate Substances 0.000 description 2
- 229920000515 polycarbonate Polymers 0.000 description 2
- 229920002643 polyglutamic acid Polymers 0.000 description 2
- 229920002338 polyhydroxyethylmethacrylate Polymers 0.000 description 2
- 239000004626 polylactic acid Substances 0.000 description 2
- 229920000656 polylysine Polymers 0.000 description 2
- 239000004814 polyurethane Substances 0.000 description 2
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 2
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 2
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 2
- 239000002243 precursor Substances 0.000 description 2
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 2
- 229940121649 protein inhibitor Drugs 0.000 description 2
- 239000012268 protein inhibitor Substances 0.000 description 2
- 238000011002 quantification Methods 0.000 description 2
- 230000009870 specific binding Effects 0.000 description 2
- 150000003408 sphingolipids Chemical class 0.000 description 2
- 235000003702 sterols Nutrition 0.000 description 2
- 238000003756 stirring Methods 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 229940126622 therapeutic monoclonal antibody Drugs 0.000 description 2
- 125000003396 thiol group Chemical group [H]S* 0.000 description 2
- 210000003934 vacuole Anatomy 0.000 description 2
- 239000013598 vector Substances 0.000 description 2
- 238000005406 washing Methods 0.000 description 2
- PAPBSGBWRJIAAV-UHFFFAOYSA-N ε-Caprolactone Chemical compound O=C1CCCCCO1 PAPBSGBWRJIAAV-UHFFFAOYSA-N 0.000 description 2
- KIUKXJAPPMFGSW-DNGZLQJQSA-N (2S,3S,4S,5R,6R)-6-[(2S,3R,4R,5S,6R)-3-Acetamido-2-[(2S,3S,4R,5R,6R)-6-[(2R,3R,4R,5S,6R)-3-acetamido-2,5-dihydroxy-6-(hydroxymethyl)oxan-4-yl]oxy-2-carboxy-4,5-dihydroxyoxan-3-yl]oxy-5-hydroxy-6-(hydroxymethyl)oxan-4-yl]oxy-3,4,5-trihydroxyoxane-2-carboxylic acid Chemical compound CC(=O)N[C@H]1[C@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](O[C@H]3[C@@H]([C@@H](O)[C@H](O)[C@H](O3)C(O)=O)O)[C@H](O)[C@@H](CO)O2)NC(C)=O)[C@@H](C(O)=O)O1 KIUKXJAPPMFGSW-DNGZLQJQSA-N 0.000 description 1
- HMLSBRLVTDLLOI-UHFFFAOYSA-N 1-(dimethylamino)ethyl 2-methylprop-2-enoate Chemical compound CN(C)C(C)OC(=O)C(C)=C HMLSBRLVTDLLOI-UHFFFAOYSA-N 0.000 description 1
- PKJBWOWQJHHAHG-UHFFFAOYSA-N 1-bromo-4-phenylbenzene Chemical compound C1=CC(Br)=CC=C1C1=CC=CC=C1 PKJBWOWQJHHAHG-UHFFFAOYSA-N 0.000 description 1
- OVRJLLUJKRFEBG-UUWRZZSWSA-N 1-palmitoyl-2-lauroyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCCCCC OVRJLLUJKRFEBG-UUWRZZSWSA-N 0.000 description 1
- VHSHLMUCYSAUQU-UHFFFAOYSA-N 2-hydroxypropyl methacrylate Chemical compound CC(O)COC(=O)C(C)=C VHSHLMUCYSAUQU-UHFFFAOYSA-N 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- 102100022749 Aminopeptidase N Human genes 0.000 description 1
- 108090000975 Angiotensin-converting enzyme 2 Proteins 0.000 description 1
- 102100035765 Angiotensin-converting enzyme 2 Human genes 0.000 description 1
- 238000009020 BCA Protein Assay Kit Methods 0.000 description 1
- 108091005625 BRD4 Proteins 0.000 description 1
- 108010017384 Blood Proteins Proteins 0.000 description 1
- 102000004506 Blood Proteins Human genes 0.000 description 1
- 102100029895 Bromodomain-containing protein 4 Human genes 0.000 description 1
- YDNKGFDKKRUKPY-JHOUSYSJSA-N C16 ceramide Natural products CCCCCCCCCCCCCCCC(=O)N[C@@H](CO)[C@H](O)C=CCCCCCCCCCCCCC YDNKGFDKKRUKPY-JHOUSYSJSA-N 0.000 description 1
- 208000025721 COVID-19 Diseases 0.000 description 1
- 238000010354 CRISPR gene editing Methods 0.000 description 1
- 101000715943 Caenorhabditis elegans Cyclin-dependent kinase 4 homolog Proteins 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 229920001661 Chitosan Polymers 0.000 description 1
- 102000003903 Cyclin-dependent kinases Human genes 0.000 description 1
- 108090000266 Cyclin-dependent kinases Proteins 0.000 description 1
- 229920000858 Cyclodextrin Polymers 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 101100266802 Escherichia coli (strain K12) ychF gene Proteins 0.000 description 1
- 239000001116 FEMA 4028 Substances 0.000 description 1
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 1
- JZNWSCPGTDBMEW-UHFFFAOYSA-N Glycerophosphorylethanolamin Natural products NCCOP(O)(=O)OCC(O)CO JZNWSCPGTDBMEW-UHFFFAOYSA-N 0.000 description 1
- 229930186217 Glycolipid Natural products 0.000 description 1
- 101000757160 Homo sapiens Aminopeptidase N Proteins 0.000 description 1
- 101001105486 Homo sapiens Proteasome subunit alpha type-7 Proteins 0.000 description 1
- 108010031792 IGF Type 2 Receptor Proteins 0.000 description 1
- 239000000232 Lipid Bilayer Substances 0.000 description 1
- PEEHTFAAVSWFBL-UHFFFAOYSA-N Maleimide Chemical compound O=C1NC(=O)C=C1 PEEHTFAAVSWFBL-UHFFFAOYSA-N 0.000 description 1
- 102000019218 Mannose-6-phosphate receptors Human genes 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- CERQOIWHTDAKMF-UHFFFAOYSA-M Methacrylate Chemical compound CC(=C)C([O-])=O CERQOIWHTDAKMF-UHFFFAOYSA-M 0.000 description 1
- 102000007474 Multiprotein Complexes Human genes 0.000 description 1
- 108010085220 Multiprotein Complexes Proteins 0.000 description 1
- CRJGESKKUOMBCT-VQTJNVASSA-N N-acetylsphinganine Chemical compound CCCCCCCCCCCCCCC[C@@H](O)[C@H](CO)NC(C)=O CRJGESKKUOMBCT-VQTJNVASSA-N 0.000 description 1
- 208000036110 Neuroinflammatory disease Diseases 0.000 description 1
- 108700001237 Nucleic Acid-Based Vaccines Proteins 0.000 description 1
- 206010057249 Phagocytosis Diseases 0.000 description 1
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- 229920000375 Poly(ethylene glycol)-block-poly(ε−caprolactone) methyl ether Polymers 0.000 description 1
- 102100021201 Proteasome subunit alpha type-7 Human genes 0.000 description 1
- 101100167281 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) CIN4 gene Proteins 0.000 description 1
- 101100016060 Schizosaccharomyces pombe (strain 972 / ATCC 24843) gtp1 gene Proteins 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 229920002125 Sokalan® Polymers 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- GWEVSGVZZGPLCZ-UHFFFAOYSA-N Titan oxide Chemical compound O=[Ti]=O GWEVSGVZZGPLCZ-UHFFFAOYSA-N 0.000 description 1
- GBXZONVFWYCRPT-KVTDHHQDSA-N [(2s,3s,4r,5r)-3,4,5,6-tetrahydroxy-1-oxohexan-2-yl] dihydrogen phosphate Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](C=O)OP(O)(O)=O GBXZONVFWYCRPT-KVTDHHQDSA-N 0.000 description 1
- NRLNQCOGCKAESA-KWXKLSQISA-N [(6z,9z,28z,31z)-heptatriaconta-6,9,28,31-tetraen-19-yl] 4-(dimethylamino)butanoate Chemical compound CCCCC\C=C/C\C=C/CCCCCCCCC(OC(=O)CCCN(C)C)CCCCCCCC\C=C/C\C=C/CCCCC NRLNQCOGCKAESA-KWXKLSQISA-N 0.000 description 1
- 229950008995 aducanumab Drugs 0.000 description 1
- CYUOWZRAOZFACA-UHFFFAOYSA-N aluminum iron Chemical compound [Al].[Fe] CYUOWZRAOZFACA-UHFFFAOYSA-N 0.000 description 1
- 235000001014 amino acid Nutrition 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 150000003863 ammonium salts Chemical group 0.000 description 1
- 230000008485 antagonism Effects 0.000 description 1
- 229940075444 barium iodide Drugs 0.000 description 1
- 229910001638 barium iodide Inorganic materials 0.000 description 1
- WHGYBXFWUBPSRW-FOUAGVGXSA-N beta-cyclodextrin Chemical compound OC[C@H]([C@H]([C@@H]([C@H]1O)O)O[C@H]2O[C@@H]([C@@H](O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O3)[C@H](O)[C@H]2O)CO)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@@H]3O[C@@H]1CO WHGYBXFWUBPSRW-FOUAGVGXSA-N 0.000 description 1
- 235000011175 beta-cyclodextrine Nutrition 0.000 description 1
- 229960004853 betadex Drugs 0.000 description 1
- 229960003237 betaine Drugs 0.000 description 1
- 230000008499 blood brain barrier function Effects 0.000 description 1
- 210000001218 blood-brain barrier Anatomy 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 239000011852 carbon nanoparticle Substances 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 238000006555 catalytic reaction Methods 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 229940106189 ceramide Drugs 0.000 description 1
- ZVEQCJWYRWKARO-UHFFFAOYSA-N ceramide Natural products CCCCCCCCCCCCCCC(O)C(=O)NC(CO)C(O)C=CCCC=C(C)CCCCCCCCC ZVEQCJWYRWKARO-UHFFFAOYSA-N 0.000 description 1
- 229940045110 chitosan Drugs 0.000 description 1
- 239000000306 component Substances 0.000 description 1
- 239000002872 contrast media Substances 0.000 description 1
- PMHQVHHXPFUNSP-UHFFFAOYSA-M copper(1+);methylsulfanylmethane;bromide Chemical compound Br[Cu].CSC PMHQVHHXPFUNSP-UHFFFAOYSA-M 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
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 230000007547 defect Effects 0.000 description 1
- 239000000412 dendrimer Substances 0.000 description 1
- 229920000736 dendritic polymer Polymers 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 229960002086 dextran Drugs 0.000 description 1
- 238000000502 dialysis Methods 0.000 description 1
- 238000007865 diluting Methods 0.000 description 1
- SNKAWJBJQDLSFF-YEUCEMRASA-O dioleoyl phosphatidylcholine Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC(COP(O)(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCC\C=C/CCCCCCCC SNKAWJBJQDLSFF-YEUCEMRASA-O 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical group P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 239000004815 dispersion polymer Substances 0.000 description 1
- 238000009509 drug development Methods 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 238000005538 encapsulation Methods 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- FJUINRMQOFNLCW-SXAUZNKPSA-N ethyl 1-[3-(2-ethylpiperidin-1-yl)propyl]-5,5-bis[(Z)-heptadec-8-enyl]-2H-imidazole-2-carboxylate Chemical compound C(C)C1N(CCCC1)CCCN1C(N=CC1(CCCCCCC\C=C/CCCCCCCC)CCCCCCC\C=C/CCCCCCCC)C(=O)OCC FJUINRMQOFNLCW-SXAUZNKPSA-N 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 239000000835 fiber Substances 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 125000002485 formyl group Chemical group [H]C(*)=O 0.000 description 1
- 238000010362 genome editing Methods 0.000 description 1
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 239000010931 gold Substances 0.000 description 1
- 229910052737 gold Inorganic materials 0.000 description 1
- 239000001046 green dye Substances 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- 229920002674 hyaluronan Polymers 0.000 description 1
- 229960003160 hyaluronic acid Drugs 0.000 description 1
- 238000009396 hybridization Methods 0.000 description 1
- XMBWDFGMSWQBCA-UHFFFAOYSA-N hydrogen iodide Chemical compound I XMBWDFGMSWQBCA-UHFFFAOYSA-N 0.000 description 1
- WGCNASOHLSPBMP-UHFFFAOYSA-N hydroxyacetaldehyde Natural products OCC=O WGCNASOHLSPBMP-UHFFFAOYSA-N 0.000 description 1
- 125000004029 hydroxymethyl group Chemical group [H]OC([H])([H])* 0.000 description 1
- 229920000587 hyperbranched polymer Polymers 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 230000035992 intercellular communication Effects 0.000 description 1
- 230000010189 intracellular transport Effects 0.000 description 1
- IXQWNVPHFNLUGD-UHFFFAOYSA-N iron titanium Chemical compound [Ti].[Fe] IXQWNVPHFNLUGD-UHFFFAOYSA-N 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 239000002609 medium Substances 0.000 description 1
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 239000005543 nano-size silicon particle Substances 0.000 description 1
- 239000002539 nanocarrier Substances 0.000 description 1
- 229940031182 nanoparticles iron oxide Drugs 0.000 description 1
- 230000003959 neuroinflammation Effects 0.000 description 1
- VVGIYYKRAMHVLU-UHFFFAOYSA-N newbouldiamide Natural products CCCCCCCCCCCCCCCCCCCC(O)C(O)C(O)C(CO)NC(=O)CCCCCCCCCCCCCCCCC VVGIYYKRAMHVLU-UHFFFAOYSA-N 0.000 description 1
- 239000012299 nitrogen atmosphere Substances 0.000 description 1
- 229940023146 nucleic acid vaccine Drugs 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 210000003463 organelle Anatomy 0.000 description 1
- TWNQGVIAIRXVLR-UHFFFAOYSA-N oxo(oxoalumanyloxy)alumane Chemical compound O=[Al]O[Al]=O TWNQGVIAIRXVLR-UHFFFAOYSA-N 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 230000008782 phagocytosis Effects 0.000 description 1
- 239000002953 phosphate buffered saline Substances 0.000 description 1
- 150000008104 phosphatidylethanolamines Chemical class 0.000 description 1
- 229910052698 phosphorus Inorganic materials 0.000 description 1
- 239000011574 phosphorus Substances 0.000 description 1
- 230000001766 physiological effect Effects 0.000 description 1
- 229920001553 poly(ethylene glycol)-block-polylactide methyl ether Polymers 0.000 description 1
- 229920000141 poly(maleic anhydride) Polymers 0.000 description 1
- 229920002946 poly[2-(methacryloxy)ethyl phosphorylcholine] polymer Polymers 0.000 description 1
- 229930001119 polyketide Natural products 0.000 description 1
- 125000000830 polyketide group Chemical group 0.000 description 1
- 150000003132 pregnenolones Chemical class 0.000 description 1
- 150000003135 prenol lipids Chemical class 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 238000002331 protein detection Methods 0.000 description 1
- 238000004064 recycling Methods 0.000 description 1
- 150000003313 saccharo lipids Chemical class 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 229910052711 selenium Inorganic materials 0.000 description 1
- 239000011669 selenium Substances 0.000 description 1
- 230000009758 senescence Effects 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 238000010008 shearing Methods 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 239000002344 surface layer Substances 0.000 description 1
- 238000001308 synthesis method Methods 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 230000001839 systemic circulation Effects 0.000 description 1
- 102000013498 tau Proteins Human genes 0.000 description 1
- 108010026424 tau Proteins Proteins 0.000 description 1
- YLQBMQCUIZJEEH-UHFFFAOYSA-N tetrahydrofuran Natural products C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 239000010409 thin film Substances 0.000 description 1
- OGIDPMRJRNCKJF-UHFFFAOYSA-N titanium oxide Inorganic materials [Ti]=O OGIDPMRJRNCKJF-UHFFFAOYSA-N 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 230000005945 translocation Effects 0.000 description 1
- 150000003626 triacylglycerols Chemical class 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 238000002604 ultrasonography Methods 0.000 description 1
- 229920002554 vinyl polymer Polymers 0.000 description 1
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K19/00—Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
-
- 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
- C12N15/115—Aptamers, i.e. nucleic acids binding a target molecule specifically and with high affinity without hybridising therewith ; Nucleic acids binding to non-nucleic acids, e.g. aptamers
-
- 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
- C12N2310/16—Aptamers
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biochemistry (AREA)
- Molecular Biology (AREA)
- Immunology (AREA)
- Biomedical Technology (AREA)
- Biophysics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Hematology (AREA)
- Diabetes (AREA)
- Biotechnology (AREA)
- General Engineering & Computer Science (AREA)
- Epidemiology (AREA)
- Pain & Pain Management (AREA)
- Microbiology (AREA)
- Neurology (AREA)
- Rheumatology (AREA)
- Plant Pathology (AREA)
- Communicable Diseases (AREA)
- Oncology (AREA)
- Physics & Mathematics (AREA)
- Neurosurgery (AREA)
- Obesity (AREA)
- Gastroenterology & Hepatology (AREA)
Abstract
The invention provides a lipid-based protein degradation tool, application and a preparation method thereof. Wherein the lipid-based protein degradation means comprises: a POI recognition group, and a lipid hybrid attached to the POI recognition group. The POI recognition groups are modified on the surface of the lipid hybrid substance, and the POI recognition groups are exposed outside the lipid hybrid substance after being assembled, so that the degradation of target protein based on the lipid hybrid substance is realized, the synthesis difficulty for constructing a protein degradation tool is greatly reduced, and mass compounds, polypeptides, antibodies, nucleic acid aptamers and the like with binding force with the target protein can be upgraded into protein degradation drugs through a plug-and-play mode, so that new functions are exerted in the related fields of traditional liposomes and LNP, and the development of combined therapy is realized in scientific research and industrial application.
Description
Technical Field
The invention belongs to the technical field of targeted drugs, and particularly relates to a lipid-based protein degradation tool, application and a preparation method thereof.
Background
A targeted Protein degradation Tool (TPD) is a tool that specifically hijacks proteins of interest (POI) to an intracellular Protein recycling site to achieve targeted Protein degradation. TPD has become a powerful tool for biomedical research and the pharmaceutical industry.
Compared with the traditional enzyme inhibition/antagonism small molecule drugs, the proteolytic targeting chimera (PROTAC) as a representative member of the most representative first generation TPD can target and degrade the target spots which are traditionally difficult to form drugs, but the target spots are only limited by intracellular proteins. Extracellular and membrane proteins play an important role in the development of disease, with approximately 40% of the total protein encoded by a gene being non-intracellular. To extend the range of target protein degradation beyond the cytoplasm, previous research groups developed lysosome-targeting chimeras (LYTAC) linked through a cationic phosphooligomannose tail to antibodies/polypeptides that were endocytosed into the lysosome for degradation via the mannose-6-phosphate receptor. Subsequently, the group utilizes the characteristic that hepatic cells are rich in asialoglycoprotein receptor (ASGPR) to replace the lyTAC phosphomannose with a trigalactose structure, and further upgrades the lyTAC into liver-specific lyTAC, and although the LYTAC method has application potential, the synthetic method is complex. LYTAC is receptor dependent, requires special design on the LYTAC structure to help protein complexes enter cells, and also targets different cells, requires tail replacement, and is therefore tedious and difficult to design and synthesize.
The successful development of TPD, currently recognized, requires the design of (1) appropriate target protein recognition groups, (2) receptor-ligand matched pairs to hijack protein entry and intracellular trafficking, (3) initiation of appropriate protein degradation mechanisms, (4) design of targeting and biological barrier penetration specific to cell type from a pharmaceutical perspective. However, the mainstream TPD tools, including PROTAC, LYTAC, and the like, require laborious case design in developing tools for any new POI, where a de novo synthesis of compounds and extensive screening procedures are not rare for different diseases and cell types.
In conclusion, the existing targeted protein degradation tool has the defects of complex design and synthesis method, need of independent design aiming at each case of disease and cell type, poor flexibility, inconvenience in modification, inconvenience in-vivo targeted delivery, no crossing capability of biological barriers such as blood brain barrier and the like, and brings great inconvenience to a targeted treatment mode aiming at a patient.
Disclosure of Invention
To solve the above problems, the present invention provides a lipid-based protein degradation tool comprising:
a POI recognition group, and a lipid hybrid attached to the POI recognition group;
wherein the POI recognition group comprises an antibody, protein, polypeptide, aptamer or small molecule capable of specifically binding to the POI;
the lipid hybrid substance comprises liposome, exosome, cell membrane and LNP.
Preferably, when the POI recognition group is coupled to the lipid hybrid, the lipid-based protein degradation tool is a nanoparticle for protein degradation composed of the lipid hybrid in the core with the POI recognition group at the periphery.
Preferably, the lipid-based protein degradation means further comprises the lipid-based protein degradation means provided with a linking member between the POI recognition group and the lipid hybrid;
preferably, the linking member has a molecular weight of 0-1000kDa;
the connecting component is one of a polymer connecting arm and a lipid connecting arm;
preferably, the polymer linker arms comprise a hydrophilic polymer, a hydrophobic polymer, and an amphiphilic polymer;
preferably, the lipid linker arm is an amphiphilic lipid linker arm.
Preferably, when the POI recognition group is coupled to the lipid hybrid substance through the linking member, the POI recognition group and the linking member constitute a set of linking units; the lipid-based protein degradation tool is a lipid-based protein degradation tool having a multi-layered structure in which the lipid-hybridized substance is provided at the core, the plurality of sets of the linking members to which the lipid-hybridized substance is linked are provided at the intermediate layer, and the plurality of sets of linking units to which the POI recognition groups linked to the linking members are provided at the periphery.
Preferably, the lipid linking arm at least comprises two ends, one end is a lipophilic end which can be linked with the lipid hybrid substance, and the other end is a hydrophilic end;
preferably, the lipophilic terminus is a lipid molecule.
Preferably, the amphiphilic polymer is a polymer of a chain/branched molecular structure, which has at least one molecular end having hydrophilicity and one molecular end having hydrophobicity;
preferably, the amphiphilic polymer has a linear molecular structure, one end of which is a hydrophilic molecular end and the other end of which is a hydrophobic molecular end.
Preferably, further comprising a nanoparticle, wherein the nanoparticle is coated by the lipid hybrid substance at the core of the lipid-based protein degradation means;
the nanoparticles comprise hydrophilic particles, hydrophobic particles and inorganic nanoparticles;
preferably, the nanoparticles have a particle size of 5-1000nm.
In addition, in order to solve the above problems, the present invention also provides a lipid-based protein degradation tool as described above, and its use in the preparation of a medicament and a delivery system for treating and preventing diseases caused by abnormal accumulation of harmful proteins; wherein the diseases caused by abnormal accumulation of harmful proteins comprise tumors, immune system diseases, neurodegenerative diseases, blood system diseases and metabolic diseases.
In addition, in order to solve the above problems, the present invention also provides a lipid-based protein degradation tool as described above, and its application in the preparation of a detection product/kit for diseases caused by abnormal accumulation of harmful proteins; wherein the diseases caused by abnormal accumulation of harmful proteins comprise tumors, immune system diseases, neurodegenerative diseases, blood system diseases and metabolic diseases.
In addition, in order to solve the above problems, the present invention also provides a method for preparing the lipid-based protein degradation tool, wherein when the lipid-based protein degradation tool is a protein degradation tool in which the POI recognition group is coupled to the lipid hybrid, the method comprises: non-covalently bonding the POI recognition group to the lipid hybrid substance; alternatively, the POI recognition group is covalently bonded to the lipid hybrid based on a coupling group, thereby constituting the lipid-based protein degradation means.
Preferably, the lipid-based proteolytic means further comprises a proteolytic means in which the POI recognition group is linked to the lipid hybrid via a linking member;
wherein, when the lipid-based protein degradation tool is a protein degradation tool in which the POI recognition group is connected with the lipid hybrid through the connecting member, the preparation method comprises the following steps:
firstly, coupling the POI recognition group with the connecting member to form a coupling intermediate; then linking the coupling intermediate to the lipid hybrid substance to form the lipid-based protein degradation means;
or, constructing a nanocomposite structure with the connecting member as an outer layer and the lipid hybrid substance as a core; and coupling the POI recognition group with the nano-composite structure to form the lipid-based protein degradation tool.
The invention provides a lipid-based protein degradation tool, application and a preparation method thereof, wherein the lipid-based protein degradation tool comprises: a POI recognition group, and a lipid hybrid substance linked to the POI recognition group; wherein the POI recognition group comprises an antibody, protein, polypeptide, aptamer or small molecule capable of specifically binding to the POI; the lipid hybrid includes liposomes, exosomes, cell membranes and LNPs.
According to the invention, the POI recognition group is modified on the surface of the lipid hybrid substance, and the POI recognition group is exposed outside the lipid hybrid substance after being assembled, so that the degradation of the target protein based on the lipid hybrid substance is realized, the synthesis difficulty of constructing a protein degradation tool is greatly reduced, and through a plug-and-play mode, a large amount of compounds, polypeptides, antibodies, nucleic acid aptamers and the like which have binding force with the target protein can be upgraded into protein degradation drugs, so that new functions are exerted in the related fields of traditional liposomes (Liposome) and Lipid Nanoparticles (LNP), such as mRNA vaccines, nucleic acid delivery carriers and drug delivery, and the development of combined therapy in scientific research and industrial application is realized.
In addition, the application of ligand-targeted lipid nanoparticles (lipid hybrid substances) as protein degradation tools and the exploration of the mechanism thereof are still blank, the invention greatly expands the application range of the current lipid nanoparticles, provides basic knowledge for the TPD and nano delivery fields, and can degrade various human disease-related extracellular/membrane-related/intracellular proteins in vivo in principle.
Drawings
FIG. 1 is a schematic diagram of the structure of the POI recognition group of the present invention coupled to a lipid hybrid;
FIG. 2 is a schematic diagram of the structure of the present invention in which a POI recognition group containing a linking member is coupled to a lipid hybrid;
FIG. 3 is a schematic diagram of the structure of the present invention containing the POI recognition group of the linking member-lipid linking arm linked to the lipid hybrid substance;
FIG. 4 is a schematic diagram of the synthetic structure of NTZ-PEGlipo in example 1 of the present invention;
FIG. 5 shows the results of protein electrophoresis of the effect of NTZ-PEGlipo on the degradation of EGFR, a target protein, in M231 cells, in example 1 of the present invention;
FIG. 6 is a schematic view showing the synthesis of NTZ-lipo1, NTZ-lipo2, INE-lipo, palb-lipo, AV45-lipo in example 2 of the present invention;
FIG. 7 shows the results of protein electrophoresis of the effect of NTZ-lipo1 of different concentrations on the degradation of EGFR in M231 cells in example 2 of the present invention;
FIG. 8 shows the results of protein electrophoresis of the effect of NTZ-lipo1 of different lipid hybrid composition ratios on the degradation of the target protein EGFR in M231 cells in example 2 of the present invention;
FIG. 9 shows the results of protein electrophoresis of the effect of NTZ-lipo2 on the degradation of EGFR, a target protein, in M231 cells, according to example 2 of the present invention;
FIG. 10 shows the results of protein electrophoresis showing the effect of INE-lipo on the degradation of the target protein HER2 in M231 cells in example 2 of the present invention;
FIG. 11 shows the NMR results of the coupling of the lipid linker arm used in Palb-lipo with the POI recognition group in example 2 of the present invention;
FIG. 12 shows the NMR results of hydrogen spectra of the lipid linker arm used in AV45-lipo of example 2 coupled with the recognition group of POI;
FIG. 13 shows the results of protein electrophoresis of the effect of varying concentrations of Palb-lipo on the degradation of the target protein CDK4 in M231 cells in example 2 of the present invention;
FIG. 14 is a schematic view of the structure of AV45-lipo in embodiment 2 of the present invention;
FIG. 15 is a graph showing the results of confocal microscopy for detecting autofluorescence of AV45-lipo and beta-amyloid (A beta) oligomers labeled with FITC (fluorescent dye) for cells in a human glial cell culture medium after incubation of AV45-lipo with beta-amyloid (A beta) oligomers in example 2 of the present invention followed by PBS washing;
fig. 16 is a schematic structural diagram of NTZ-LNP1, NTZ-LNP1s, and NTZ-LNP2 in embodiment 3 of the present invention;
FIG. 17 is a graph showing the particle size distribution of the NTZ-LNP1, NTZ-LNP1s, and NTZ-LNP2 Dynamic Light Scattering (DLS) in example 3 of the present invention;
FIG. 18 shows the results of protein electrophoresis of the effect of NTZ-LNP1 in example 3 and NTZ-lipo1 in example 2 on the degradation of EGFR, a target protein, in M231 cells;
FIG. 19 shows the results of protein electrophoresis of the effect of NTZ-LNP1s on the degradation of the target protein EGFR in M231 cells in example 3 of the present invention;
FIG. 20 shows the results of protein electrophoresis of the effect of NTZ-LNP2 on the degradation of the target protein EGFR in M231 cells in example 3 of the present invention;
FIG. 21 is a graph of the DLS particle size distribution and the average dispersion coefficient PDI of NTZ-exo in example 4 of the present invention;
FIG. 22 shows the results of protein electrophoresis of the effect of NTZ-exo on the degradation of EGFR, a target protein, in M231 cells, in example 4 of the present invention;
FIG. 23 is a schematic view of the structure of NTZ-lipoP in example 5 of the present invention;
FIG. 24 shows the results of protein electrophoresis of the effect of NTZ-lipoP of different concentrations on the degradation of the target protein EGFR in M231 cells in example 5 of the present invention;
FIG. 25 is a schematic structural diagram of a CTX-RBCmD according to embodiment 5 of the present invention;
FIG. 26 is a graph showing the DLS particle size distribution results of CTX-RBCmD in example 5 of the present invention;
FIG. 27 shows the results of protein electrophoresis of the effect of CTX-RBCmD on the degradation of EGFR, a target protein, in M231 cells, according to example 5 of the present invention.
Reference numerals:
100, a lipid-based protein degradation means; 1,poi recognition group; 2, liposomes, exosomes or cell membranes in lipid hybrids; LNP in lipid hybrids; 4, a lipid linker arm; 5, a polymer linker arm; 6, a coupling group; 7, nano particles.
The implementation, functional features and advantages of the present invention will be further described with reference to the accompanying drawings.
Detailed Description
The technical solutions of the present invention will be described clearly and completely with reference to the following embodiments, and it should be understood that the described embodiments are some, but not all, embodiments of the present invention. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
Unless defined otherwise below, all technical and scientific terms used in the detailed description of the invention are intended to have the same meaning as commonly understood by one of ordinary skill in the art. While the following terms are believed to be well understood by those skilled in the art, the following definitions are set forth to better explain the present invention.
As used herein, the terms "comprising," "including," "having," "containing," or "involving" are inclusive or open-ended and do not exclude additional unrecited elements or method steps. The term "consisting of 823030a" is considered a preferred embodiment of the term "comprising". If in the following a certain group is defined to comprise at least a certain number of embodiments, this should also be understood as disclosing a group which preferably only consists of these embodiments.
Where an indefinite or definite article is used when referring to a singular noun e.g. "a" or "an", "the", this includes a plural of that noun.
The term "about" in the present invention denotes an interval of accuracy that can be understood by a person skilled in the art, which still guarantees the technical effect of the feature in question. The term generally denotes a deviation of ± 10%, preferably ± 5%, from the indicated value.
Furthermore, the terms first, second, third, (a), (b), (c), and the like in the description and in the claims, are used for distinguishing between similar elements and not necessarily for describing a sequential or chronological order. It is to be understood that the terms so used are interchangeable under appropriate circumstances and that the embodiments of the invention described herein are capable of operation in other sequences than described or illustrated herein.
The following is provided merely to aid in understanding the invention. These definitions should not be construed to have a scope less than understood by those skilled in the art.
The technical solution of the present invention is further described in detail by referring to the specific embodiments, but the present invention is not limited thereto, and any limited number of modifications made by anyone within the scope of the claims of the present invention are still within the scope of the claims of the present invention.
Referring to fig. 1, the present example provides a lipid-based protein degradation tool comprising:
a POI recognition group, and a lipid hybrid substance linked to the POI recognition group;
wherein the POI recognition group comprises an antibody, protein, polypeptide, aptamer or small molecule capable of specifically binding to the POI;
the lipid hybrid includes liposomes, exosomes, cell membranes and LNPs.
The POI recognition group is an antibody, protein, polypeptide or small molecule capable of specifically binding to POI.
The lipid hybrid substance includes Liposome (Liposome), exosome, cell membrane and LNP. Wherein LNP is lipid Nanoparticle (lipid Nanoparticle).
It should be noted that lipid nanoparticles are mainly used for in vivo drug delivery, and have been widely used in medical treatment in the fields of small molecule drug delivery, nucleic acid drug delivery, nano-vaccines and the like, from traditional liposomes (liposomes) to Lipid Nanoparticles (LNPs). The mRNA vaccine of COVID-19 mostly consists of LNP.
Lipid nanoparticles have the advantages of low cost and convenient preparation, the composition rule of the lipid nanoparticles is relatively clear, and the lipid nanoparticles for medical treatment and drug development have a relatively regular composition, and are mainly membrane skeletons and cholesterol auxiliary materials. The lipid nanoparticles for clinical use can be added with other components according to needs, and can include but are not limited to the following components:
1. the membrane skeleton, which constitutes the main component of lipid bilayer membrane.
2. Auxiliary materials: (1) Cholesterol (cholestrol) regulates membrane fluidity and improves particle stability, and (2) auxiliary phospholipid maintains liposome microscopic morphology to make lysosome membrane unstable.
3. The PEG lipid can reduce the combination of the particles and plasma protein in vivo and prolong the systemic circulation time.
4. Cationic lipids, highly efficient entrapment of nucleic acid drugs, positive charge, in vivo transfection, pH sensitivity (ionizable).
5. The stabilizer has the freeze-drying protection effect and maintains the stable structure of the liposome in the freeze-drying process.
The lipid hybrid substance comprises liposome, exosome, cell membrane, LNP, LPP and lipid nanoemulsion. Among them, liposomes (lipopome), also known as classical liposomes, are vacuolated and are mainly used to carry hydrophobic drugs. As the liposome is used as an amphiphilic membrane, hydrophobic drugs can be loaded in the hydrophobic vacuoles, and hydrophilic drugs can be loaded in the hydrophilic vacuoles. The LNP is a lipid nanoparticle, the interior of the LNP contains cationic lipid, the existing mRNA vaccine and some nucleic acid vaccines are the LNP used for the existing mRNA vaccine, the nucleic acid is negatively charged, the cationic lipid is positively charged, the entrapment efficiency can be effectively improved, and the LNP is also used for delivering CRISPR gene editing elements. The traditional liposome lipo has low efficiency of carrying nucleic acid, and generally carries hydrophobic and hydrophilic micromolecular drugs. The LPP has the characteristics of lipid and polymer due to the inclusion of the polymer core, and has better drug-loading compatibility and stability.
In addition, LNP and LPP have the advantage of being easily modified, liposomes, LNP are easily modified by amphiphilic pegylated lipids and similar structures, which can be used to bind to receptors via PEG-terminally linked ligands (ligands), possibly facilitating drug delivery to target organs, also known as actively targeted lipid nanocarriers.
The lipid hybrid substance is convenient for carrying drugs, and comprises hydrophilic and hydrophobic micromolecular drugs, nucleic acid drugs, protein drugs, gene editing vectors and other biotechnology drugs which can be effectively loaded and delivered. However, the application of the drug delivery system still focuses on drug delivery, the physiological and pathological properties of the structure of the drug delivery system still need to be further explored, and more therapeutic potential and possibility need to be explored.
In summary, in the embodiment, the POI identifying group is modified on the surface of the lipid hybrid substance, and the POI identifying group is exposed outside the lipid hybrid substance after being assembled, so that the degradation of the target protein based on the lipid hybrid substance is realized, the synthesis difficulty of constructing a protein degradation tool is greatly reduced, and a large amount of compounds, polypeptides, antibodies, aptamers and the like having binding force with the target protein can be upgraded into protein degradation drugs through a plug and play mode, and further, new functions are exerted in the related fields of traditional liposomes (Liposome) and Lipid Nanoparticles (LNP), such as mRNA vaccines, nucleic acid delivery vectors and drug delivery, so that the development of combination therapy in scientific research and industrial applications is realized.
In addition, the application of ligand-targeted lipid nanoparticles (lipid hybrid substances) as protein degradation tools and the exploration of the mechanism thereof are still blank, the invention greatly expands the application range of the current lipid nanoparticles, provides basic knowledge for the TPD and nano delivery fields, and can degrade various human disease-related extracellular/membrane-related/intracellular proteins in vivo in principle.
Further, when the POI recognition group is coupled to the lipid hybrid, the lipid-based protein degradation tool is a nanoparticle for protein degradation composed of the lipid hybrid in the core and the POI recognition group in the periphery.
The above shows the structure of the lipid-based protein degradation tool, wherein the whole body can be a granular structure, including a core part and a periphery, the core part can be a lipid hybrid substance, and the periphery is a plurality of POI recognition groups connected with the lipid hybrid substance, i.e. a layer of POI recognition groups connected with the lipid hybrid substance is arranged on the outer surface of the lipid hybrid substance, thereby forming the whole lipid-based protein degradation tool.
Furthermore, depending on the form of the lipid hybrid, in principle, if it is a classical liposome, its form is vacuolated.
Further, referring to fig. 2, the lipid-based protein degradation means may further include a lipid-based protein degradation means having a connection member between the POI recognition group and the lipid hybrid.
Preferably, the linking member has a molecular weight of 0-1000kDa;
the connecting component is one of a polymer connecting arm and a lipid connecting arm; wherein, the structure of the lipid-based protein degradation tool containing the lipid linker arm is shown with reference to fig. 3.
Preferably, the polymer linker arms comprise hydrophilic polymers, hydrophobic polymers and amphiphilic polymers.
Preferably, the lipid linker arm is an amphiphilic lipid linker arm.
In the above, another lipid-based protein degradation tool is provided, similar to the basic structure, with a linking member between the lipid hybrid and the POI recognition group. The sequential connection combination of the lipid hybrid substance, the connecting component and the POI recognition group is formed, wherein the connecting component is provided with a coupling group which can be connected with the POI recognition group, thereby realizing the combination of the two.
Further, when the POI recognition group is coupled to the lipid hybrid through the linking member, the POI recognition group and the linking member constitute a group of linking units; the lipid-based protein degradation tool is a lipid-based protein degradation tool having a multi-layered structure in which the lipid-hybridized substance is provided at the core, the plurality of sets of the linking members to which the lipid-hybridized substance is linked are provided at the intermediate layer, and the plurality of sets of linking units to which the POI recognition groups linked to the linking members are provided at the periphery.
The above shows that the lipid-based protein degradation tool has a spatial structure, the core of the lipid-based protein degradation tool is a lipid hybrid material (core), the surface layer of the lipid-based protein degradation tool is connected with a plurality of connecting members (middle layers), and each connecting member is connected with a lipid hybrid material (periphery), so that an inner layer and an outer layer of granular structures are formed, and the whole lipid-based protein degradation tool can be spherical in theory or in other shapes.
Further, the molecular weight of the linking member is 0-1000kDa.
The linking member is one of a polymer linking arm and a lipid linking arm.
Preferably, the polymer linker arms comprise hydrophilic polymers, hydrophobic polymers, and amphiphilic polymers.
TABLE 1 three structural forms of Polymer linker arms
Preferably, the lipid linker arm is an amphiphilic lipid linker arm.
Above, the characteristics of the connecting member are defined, which are divided into two broad categories, including: the polymer connecting arm and the lipid connecting arm can be any one according to the synthesis requirement and the water solubility requirement of a substance, and the lipid connecting arm is amphiphilic.
The hydrophilic polymer, as described above, may include, but is not limited to: polyethylene glycol (PEG), polyethylene oxide (PEO), poly (ethylene glycol) methacrylate (POEG), poly (2-methacryloyloxyethyl phosphoryl) (PMPC), polycarboxylic betaine (PCB), dextran, hyaluronic acid, chitosan, beta-cyclodextrin, hyperbranched polyglycidyl ether (HPG), poly N- (2-hydroxypropyl) methacrylamide (PHPMA), polyhydroxyethylmethacrylate (PHEMA), polyacrylamide (PAM), polyvinylpyrrolidone (PVP), polyacrylic acid (PAA), polymaleic anhydride (HPMA), polyquaternary ammonium salts and pharmaceutically acceptable polymer salts thereof, polyethyleneimine (PEI), N-dimethylaminoethyl methacrylate (PDMAEMA), polylysine (PLL), polyglutamic acid (PGu), polyaspartic acid (PASp) and other hydrophilic polyaminoacids, and derivatives of the above polymers and pharmaceutically acceptable salts thereof.
The hydrophobic polymer may include: polylactic acid-glycolic acid copolymer (PLGA), polylactic acid (PLA), polycaprolactone (PCL), polycarbonate (PMC) and derivatives thereof, glycolide/lactide/caprolactone/carbonate various combinations and copolymers of components, polyurethane (PU), polyether ether ketone (PEEK), polymethyl methacrylate (PMMA), polyvinyl alcohol (PVA), polyethylene (PE), poly (phenylalanine) and other various hydrophobic polyamino acids, derivatives of the polymers and pharmaceutically acceptable salts thereof.
Amphiphilic polymers include: PEG-PLGA, PEG-PCL, PEG-PLA, PEG-PMC, and various amphiphilic block polymers and derivatives of the combination of the hydrophilic polymer and the hydrophobic polymer, and various amphiphilic polymers and derivatives of the combination of the hydrophilic polymer and the hydrophobic molecule (such as lipid).
Furthermore, the lipid connecting arm at least comprises two ends, one end of the lipid connecting arm is a lipophilic end which can be connected with the lipid hybrid substance, and the other end of the lipid connecting arm is a hydrophilic end;
preferably, the lipophilic terminus is a lipid molecule.
When the linking member is a lipid linking arm, it may structurally comprise a multi-terminal structure, but at least two terminals, a lipophilic terminal (linked to the lipid hybrid) and a hydrophilic terminal, and the lipophilic terminal is a lipid molecule.
The lipid molecules include: fatty acids (fatty acids), glycerolipids (glycerolipids), glycerophospholipids (glycerophospholipids), sphingolipids (sphingolipids), sterol lipids (sterol lipids), pregnenolone lipids (prenol lipids), glycolipids (saccharolipids), polyvinyls (polyketides), cationic lipids (cationic lipids) and ionizable lipids (ionizable lipids).
Further, the lipid molecule may be DSPE (distearoylphosphatidylethanolamine), distearoylphosphatidylcholine (DSPC), 1,2-Dimyristoyl-SN-GLYCEROL (DMG, 1, 2-Dimyristoyl-SN-GLYCEROL), 1,2-DIPALMITOYL-SN-GLYCEROL (DPG, 1, 2-DIPALMITOYL-SN-GLYCEROL), 1,2-DIPALMITOYL-SN-GLYCEROL (DPyG, 1, 2-diphytoyl-SN-GLYCEROL), diacylglycerol (DAG); triglycerides (TAG), 1, 2-Dipalmitin (DPG), 1' - [ (1R) -1- (hydroxymethyl) -1, 2-ethanediyl]Octadecanoic acid ester (D)SG), dianhydroxanoyl phosphatidylcholine (DAPC), 1-palmitoyl-2-lauroyl-sn-glycero-3-phosphocholine (DLPC), dimyristoyl phosphatidylcholine (DMPC), 1, 2-dioleoyl lecithin (DOPC), dipalmitoyl phosphatidylcholine (DPPC), 1, 2-dipalmitoyl-sn-glycero-3-ethylphosphocholine (DPePC), phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylglycerol (PG), phosphatidylserine (PS), phosphatidylinositol (PI); ceramide (Cer), sphingomyelin (SM), cholesterol (Cho), cholesteryl Ester (CE), 1, 2-dimyristoyl-SN-glycero-3-phosphate (DMPA), dilauroylphosphatidic acid (DLPA), phosphocholine (Phosphocholine), trimethyl-2, 3-dioleoyloxypropylammonium bromide (DOTAP), ionizable lipids including DLin-MC3-DMA, A6, OF-02, A18-ISO5-2DC18, 98N12-5、9A1P9、C12-200、cKK-E12、7c1、G0-C14、L319、304O13、OF-Deg-Lin、306-OB12、306-Oi10And FTT5, and derivatives of the foregoing lipid molecules and pharmaceutically acceptable salts thereof. The PEG lipid can be PEG and conjugates of PEG derivatives and the aforementioned lipids.
Preferably, the lipid molecule may be a pegylated lipid, wherein PEG has a relative molecular weight of 2000 and the pegylated lipid may comprise: PEG-DSPE (polyethylene glycol-distearoylphosphatidylethanolamine), PEG-DMG (polyethylene glycol-dimyristoyl glyceride), conjugates of the aforementioned lipid molecules and PEG, and pharmaceutically acceptable salts thereof. The PEG comprises PEG and PEG with methoxy or other groups at the tail end of the PEG.
Further, the amphiphilic polymer is a polymer with a chain/branch molecular structure, and at least one molecular end with hydrophilicity and one molecular end with hydrophobicity exist in the polymer;
the structure of the amphiphilic polymer is defined as a chain/branched molecular structure as described above, and may have a plurality of branches and a plurality of chains because of the characteristics of the chain/branched molecular structure, but it has at least two ends, one being a molecular end having hydrophilicity and one being a molecular end having hydrophobicity.
Preferably, the amphiphilic polymer has a linear molecular structure, one end of which is a hydrophilic molecular end and the other end of which is a hydrophobic molecular end.
The amphiphilic polymer is further defined as having a linear molecular structure.
Preferably, the lipid-based protein degradation tool further comprises a nanoparticle, wherein the nanoparticle is coated at the core of the lipid-based protein degradation tool by the lipid hybrid substance;
the nanoparticles comprise hydrophilic particles, hydrophobic particles and inorganic nanoparticles;
preferably, the nanoparticles have a particle size of 5-1000nm.
In another embodiment, a special structure is provided, that is, the nanoparticle is at the core, and the lipid hybrid material wraps the nanoparticle to form a composite structure.
Thus, the lipid-based protein degradation means may comprise several morphological structures as follows:
TABLE 2 morphological Structure of lipid-based protein degradation tools
In addition, in another embodiment, a special structure is provided, namely, the nano-particles are at the core, the lipid hybrid substance is coated on the nano-particles to form a composite structure,
wherein the nanoparticle is coated by the lipid-hybrid substance at the core of the lipid-based protein degradation means;
the nanoparticle (nanoparticle coated at the core in the lipid hybrid) includes:
(1) The hydrophilic particles may be a variety of nanogels composed of a dendrimer, a hyperbranched polymer, and the hydrophilic polymer and derivatives thereof as claimed in claim 4, and a nano albumin.
(2) The hydrophobic particles may be nanoparticles prepared from various hydrophobic polyamino acids and derivatives thereof, such as polylactic acid-glycolic acid copolymer (PLGA), polylactic acid (PLA), polycaprolactone (PCL), polycarbonate (PMC) and derivatives thereof, various combinations and copolymers of glycolide/lactide/caprolactone/carbonate, polyurethane (PU), polyetheretherketone (PEEK), polymethylmethacrylate (PMMA), polyvinyl alcohol (PVA), polyethylene (PE), and poly (phenylalanine).
(3) Inorganic nanoparticles, which can be gold nanoparticles, carbon nanoparticles, silicon nanoparticles, iron oxide nanoparticles, calcium phosphate nanoparticles, barium sulfate and iodide contrast agents, aluminum nitride nanoparticles, aluminum oxide nanoparticles, titanium oxide nanoparticles, aluminum iron alloy particles, and titanium iron alloy particles, are all materials with stable and low biological toxicity.
(4) And mixed nanoparticles composed of the aforementioned nanoparticles.
Among them, when the nanoparticle is one of a hydrophilic particle and a hydrophobic particle, and is a polymer, after being coated with a lipid hybrid substance, it can also be called lipid polymer (LPP), i.e., a lipid film is coated on the surface of the nanoparticle of the polymer having hydrophilic or hydrophobic properties, and Cationic nanoemulsion (Cationic lipid) is also widely studied and applied in vaccine and drug delivery.
Further, in the POI recognition group, the antibody is a therapeutic monoclonal antibody, a multispecific antibody, a nano antibody, an antibody derivative or an antibody coupling drug;
the polypeptide is a polypeptide with POI specific binding capacity;
the small molecule is a compound with POI specific binding capacity;
preferably, the small molecule comprises a derivative of the CDK4/6 protein inhibitor Palbociclib (Palbociclib), a derivative of the BRD4 protein inhibitor JQ1, a derivative of the beta-Amyloid protein probe AV-45, a derivative of the Pittsburgh compound PiB, and a derivative of the Tau protein probes GTP1 and PBB 3.
Preferably, the polypeptide comprises a binding polypeptide of ACE2, CD13, beta-Amyloid;
preferably, the antibody comprises: CTX, NTZ, PTZ, CRLZ, INE, ATZ, and aducanumab, miltuximab.
In addition, the invention also provides a lipid-based protein degradation tool, and application of the lipid-based protein degradation tool in preparation of drugs, vaccines and delivery systems for treating and preventing diseases related to abnormal accumulation of proteins; wherein the diseases caused by abnormal accumulation of the protein comprise tumors, immune system diseases, inflammation and pathogen infection, neurodegenerative diseases, blood system diseases and metabolic diseases.
In addition, the invention also provides a protein degradation tool based on lipid, and the application of the protein degradation tool in preparing a detection product/kit aiming at the research of abnormal protein accumulation related diseases and protein interaction; wherein the diseases caused by abnormal accumulation of the protein comprise tumors, immune system diseases, inflammation and pathogen infection, neurodegenerative diseases, blood system diseases and metabolic diseases.
In addition, the invention also provides a preparation method of the lipid-based protein degradation tool, wherein when the lipid-based protein degradation tool is a protein degradation tool of which the POI recognition group is coupled with the lipid hybrid substance, the preparation method comprises two steps:
(1) Non-covalently bonding the POI recognition group to the lipid hybrid;
(2) Alternatively, the POI recognition group is covalently bonded to the lipid hybrid substance based on a coupling group, thereby constituting the lipid-based proteolytic tool.
Further, the lipid-based proteolytic means further comprises a proteolytic means in which the POI recognition group is linked to the lipid hybrid via a linking member;
wherein, when the lipid-based protein degradation tool is a protein degradation tool in which the POI recognition group is connected with the lipid hybrid through the connecting member, the preparation method comprises the following steps:
(1) Firstly, coupling the POI recognition group with the connecting member to form a coupling intermediate; then linking the coupling intermediate to the lipid hybrid substance to form the lipid-based protein degradation means;
(2) Or, constructing a nanocomposite structure with the connecting member as an outer layer and the lipid hybrid substance as a core; and coupling the POI recognition group with the nano-composite structure to form the lipid-based protein degradation tool.
The invention is further illustrated by the following specific examples, but it should be understood that these examples are included merely for purposes of illustration in more detail and are not intended to limit the invention in any way.
TABLE 3 structural morphology of the Nanoprotein degradation tools prepared in examples 1-5
Example 1
Referring to table 3, in this example, the degradation tool of example 1 was prepared by coupling the POI recognition group to a lipid hybrid, wherein the POI recognition group is: monoclonal antibody drug Nitol (NTZ); the lipid hybrid substance is PEG (polyethylene glycol) liposome, namely the liposome (liposome) contains PEG exposed on the surface of the lipid hybrid substance, and the tail end of the PEG is provided with an active reaction site NHS (N-hydroxysuccinimide) for coupling with the amino group of the POI recognition group NTZ antibody. The structure obtained was NTZ-PEGlipo.
The preparation method comprises the following steps:
(1) Pretreatment: the buffer of the original antibody was replaced with PBS. In this example, EGFR monoclonal beads (NTZ, nimotuzumab) that have been used for clinical treatment were used.
Taking a nimotuzumab solution, centrifuging the nimotuzumab solution for 2 minutes at 4000 Xg by using a 3kDa ultrafiltration tube, further concentrating the antibody, and then diluting the antibody by using PBS (phosphate buffer solution); after repeated concentration and dilution, the buffer was replaced with PBS, and after dilution, the protein concentration was measured by IgG mode a280 of NanoDrop one C instrument, and the result was verified by BCA protein quantification kit.
(2) Preparation of lipid hybrid: the lipid hybrid substance used is liposome, which is composed of hydrogenated soybean lecithin (HSPC), cholesterol (CHO, cholestrol), DSPE (distearoylphosphatidylethanolamine) -PEG (polyethylene glycol) (polyethylene glycol molecular weight is 2 kDa) by a classical film hydration method, wherein the mass ratio HSPC: DSPE-PEG = 56. HSPC, CHO was dissolved in chloroform and then evaporated to concentrate and dried to form a film. After the chloroform was completely evaporated, 1mL of DSPE-PEG-NHS PBS solution was added, followed by sonication at 100W for 3min at room temperature. The resulting solution was then passed through 800nm, 400nm and 200nm filters to allow insertion of DSPE-PEG-NHS onto the lipid membrane, which should be controlled to be completed within 10 min.
(3) Coupling of POI recognition group NTZ antibody to PEG lipid hybrid:
the coupling reaction was initiated in an ice-water mixture and protected with nitrogen. The lipid hybrid solution was then added to the antibody PBS solution stirred at 800rpm (revolutions per minute); the reaction was then stirred and incubated at 4 ℃ on a 20rpm rotator for 24 hours.
(4) And (3) purification: the reaction mixture was then concentrated by ultrafiltration 3 times using a 50kDa centrifugal ultrafiltration tube and subsequently made up to 100. Mu.L using PBS. Protein concentration was determined by a NanoDrop one C instrument by a280 absorbance of IgG and verified by BCA method.
The experimental results are as follows: referring to FIG. 4, a diagram of the NTZ-PEGlipo synthesis process is shown. FIG. 5 shows the result of protein electrophoresis detection of EGFR protein expression by collecting protein after MDA-MB-231 (M231) human breast cancer tumor cells are treated with 500nM NTZ-PEGlipo for 24 hours, which shows that NTZ-PEGlipo can effectively degrade target protein EGFR.
The cell culture medium volumes were the same in all examples, and the molar concentrations of the treatments were in units of μ M (μ Mol/L) and nM (nMol/L). In the experiment, the cell culture and treatment were carried out in 12-well plate cell culture dishes in 500. Mu.L of the medium. Cell culture was performed according to standard cell culture specifications. All control groups were used at the same molar concentration as the degradation tool group, for example, the NTZ group and NTZ-lipo group had the same molar concentration and volume of antibody NTZ, while the lipo group without antibody and NTZ-lipo group had the same molar concentration and volume of lipid-hybrid. Wherein the lipid-hybrid control group in each example refers to the lipid-hybrid in the example to which the POI recognition group is not attached. In all examples involving multi-concentration testing, the control POI recognition group or the control lipid hybrid group was treated at an equal molar concentration to the highest concentration treatment in the concentration test group. GAPDH or VIN (vinculin) of all protein electrophorograms are internal controls.
Example 2
Referring to table 3, in this example, the degradation tool of example 2 was prepared by coupling the POI recognition group to the amphiphilic lipid linker arm followed by self-assembly with the lipid hybrid species. Wherein the POI recognition groups in example 2a and example 2b are: monoclonal antibody drug Nitol (NTZ); the linker arm is NHS-PEG-DSPE with NHS group. The lipid hybrid substance comprises a membrane skeleton and cholesterol (CHO, cholestrol), the membrane skeleton is hydrogenated soybean lecithin (HSPC) in example 2a, and the degradation tool of the membrane skeleton is NTZ-lipo1, the membrane skeleton is distearoyl phosphatidylcholine (DSPC) in example 2b, and the degradation tool of the membrane skeleton is NTZ-lipo2. The POI recognition groups in example 2c are: therapeutic monoclonal antibody of HER2 Inetetamab (INE, inetetamab), HER2 is tumor proliferation signal receptor and marker, the connecting arm is NHS-PEG-DSPE with NHS group, the lipid hybrid substance is composed of HSPC membrane skeleton and CHO, and the degradation tool composed of the HSPC membrane skeleton and CHO is INE-lipo.
The POI recognition groups in example 2d are: the small molecular CDK4 inhibitor Palbociclib (Palb) is characterized in that a connecting arm is NHS-PEG-DSPE with NHS groups, palbociclib is coupled with NHS through amino reaction, a lipid hybrid substance consists of an HSPC membrane skeleton and CHO, and a degradation tool formed by Palb-lipo is Palb-lipo.
The preparation method comprises the following steps:
experiment 1: the POI recognition group is an antibody. Wherein the POI recognition groups in example 2a and example 2b are: the mab drug Nitol (NTZ). The POI recognition groups in example 2c are: monoclonal antibody drug Inunitol (INE).
(1) Pretreatment: the buffer of the original antibody was replaced with phosphate buffered saline PBS. In this example, the NTZ and INE monoclonal antibody solutions were centrifuged at 4000 xg for 2 minutes using a 3kDa ultrafiltration tube, and then the antibodies were concentrated and diluted with PBS; after repeated concentration and dilution 3 times, the buffer main component was replaced with PBS, and after dilution with an antibody solution, the protein concentration was measured by IgG mode a280 of NanoDrop one C instrument, and the result was verified by BCA protein quantification kit.
(2) Coupling reaction of the POI recognition group and the amphiphilic lipid connecting arm: the antibody was reacted with NHS-PEG (2 kDa) -DSPE by reacting the amino group on the antibody with NHS (N-Hydroxysuccinimide) group at the end of DSPE (distearoylphosphatidylethanolamine) -PEG. The coupling reaction was initiated in an ice water mixture and protected with nitrogen. To improve dispersibility, DSPE-PEG was optionally sonicated in PBS buffer at 40kHz for 3 minutes, followed by addition of equimolar concentrations and volumes (molar ratio is not limited to 1, reference antibody conjugated drug) of NHS-PEG-DSPE PBS solution to the antibody PBS solution stirred at 800rpm (revolutions per minute); the reaction was then incubated at 4 ℃ for 24 hours on a 20rpm rotator (incubation time and conditions were not specifically required, with lower rpm and 4 degrees being preferred to maintain antibody activity). Obtaining NTZ-PEG-DSPE reaction mixture or INE-PEG-DSPE reaction mixture.
The coupling method has no special requirement, and can be coupling methods of general antibody coupling drugs, including but not limited to non-site coupling and site-specific coupling, such as amino coupling, carboxyl coupling and bridging sulfhydryl coupling. Site-directed conjugation may be click chemistry, selenium bond conjugation, serine conjugation, cysteine conjugation, unnatural amino acid conjugation, coupling after enzymatic catalysis, and sugar site conjugation.
(3) And (3) purification: the reaction mixture containing NTZ-PEG-DSPE or INE-PEG-DSPE was concentrated by ultrafiltration 3 times using a 50kDa centrifugal ultrafiltration tube, and then made up to 100. Mu.L with PBS. Protein concentration was determined by a NanoDrop one C instrument by a280 absorbance of IgG and verified by BCA method.
(4) Preparation of lipid hybrid: the lipid hybrid used was a liposome consisting of hydrogenated soy lecithin (HSPC) or distearoyl phosphatidylcholine (DSPC), cholesterol (CHO, cholestrol), DSPE-PEG (2 kDa) by classical membrane hydration, where the mass ratio [ HSPC or DSPC ]: CHO: DSPE-PEG (without POI recognition group) = 56. HSPC or DSPC, CHO were dissolved in chloroform and then concentrated by evaporation to form a film.
Preparation of NTZ-lipo1, NTZ-lipo2, INE-lipo: after the solvent chloroform of the lipid hybrid substance solution was completely evaporated, 1mL of DSPE-PEG-antibody in PBS was added, followed by sonication at 100W for 3min at room temperature. The resulting solution was then passed through 800nm, 400nm and 200nm filters. Following further purification, the reaction mixture was concentrated by ultrafiltration 3 times using a 50kDa centrifugal ultrafiltration tube and then made up to 100. Mu.L using PBS. Protein concentration was determined by a NanoDrop one C instrument by a280 absorbance of IgG and verified by BCA method.
Cell experiments were then performed depending on antibody concentration.
Experiment 2: POI recognition groups are small molecules. In example 2d, the POI recognition group is Palbociclib small molecule drug or beta-amyloid probe AV-45.
Palbociclib is a clinical commercial CDK4 inhibitor, and the conjugate of the lipid hybrid substance is Palb-lipo in the embodiment. CDK4 is a cyclin-dependent kinase that is widely involved in cell senescence tumorigenesis.
Palbociclib was reacted with NHS-PEG (2 kDa) -DSPE via the amino group on Palbociclib with the NHS group at the end of DSPE-PEG (molar ratio 1.2. Stirring and reacting for 24 hours at room temperature under the protection of nitrogen to obtain a Palb-PEG-DSPE reaction mixture. Palb-PEG-DSPE was dialyzed for 24 hours and lyophilized, and the concentration was determined by a standard curve of ultraviolet absorption peaks.
Lipid hybrids were prepared as in experiment 1 of this example.
And (3) preparing Palb-lipo, adding 1mL of PBS solution of Palb-PEG-DSPE after the solvent chloroform of the lipid hybrid substance solution is completely evaporated, and then carrying out ultrasonic treatment at room temperature for 3min at 100W. The resulting solution was then passed through 800nm, 400nm and 200nm filters. The concentration was determined by a standard curve of the UV absorption peak and cell experiments were performed according to the Palb molarity.
AV-45 is a beta-amyloid probe of a classical marker of Alzheimer's disease, and the lipid hybrid substance conjugate is AV45-lipo.
Beta-amyloid is a potential pathogenic protein of Alzheimer's disease, which is an extracellular protein, a precursor of the beta-amyloid is expressed on a cell membrane, and the precursor is secreted to form an oligomer (beta-amyloid 1-42 oligomer) and fibers outside the cell after shearing, wherein the oligomer is considered to have the greatest toxicity, the accumulation of the extracellular protein beta-amyloid oligomer brings pathological risks such as neuroinflammation, and the isotope labeled AV-45 is a clinical commercial beta-amyloid probe. The isotope-free AV-45 derivative selected in this example is AV-45-SH, AV-45 has blue autofluorescence, and is convenient for cell tracking, and its sulfhydryl SH is used for coupling with Mal (maleimide) -PEG-DSPE.
The preparation method of AV45-PEG-DSPE comprises the following steps: combining the AV45-SH with the sulfydryl with Mal-PEG (molecular weight is 2 kDa) -DSPE, namely dissolving the AV45-SH and the Mal-PEG-DSPE in DMSO at a molar ratio of 1.2, and protecting water bath at 37 ℃ for 24 hours in a dark nitrogen atmosphere. Followed by lyophilization by dialysis. The concentration was determined by a standard curve through the uv absorption peak.
Lipid hybrids were prepared as in experiment 1 of this example. Preparation of AV 45-lipo: after the solvent chloroform of the lipid hybrid solution was completely evaporated, 1mL of PBS solution of AV45-PEG-DSPE was added, followed by sonication at 100W for 3min at room temperature. The resulting solution was then passed through 800nm, 400nm and 200nm filters. The concentration was determined by a standard curve of uv absorbance peaks and cell experiments were performed according to AV45 molarity.
The experimental results are as follows: referring to fig. 6, a schematic diagram of the synthesis process is shown. FIG. 7 shows that the protein expression result of EGFR protein expression detection by protein electrophoresis of protein harvested after M231 human breast cancer tumor cells are treated by NTZ-lipo1 with different concentrations for 24 hours shows that NTZ-lipo1 can effectively degrade the target protein EGFR. FIG. 8 shows the results of protein electrophoresis of the degradation effect of the target protein EGFR by treating M231 cells with NTZ-lipo1 whose lipid hybrid substances are composed of different HSPC and CHO ratios for 24 hours, showing that different compositions have the protein degradation effect, and the ratios of the membrane skeleton HSPC and the helper lipid cholesterol have an influence on the protein degradation effect. FIG. 9 shows that the protein expression result of EGFR protein detection by protein electrophoresis is obtained after M231 human breast cancer tumor cells are treated by 500nM NTZ-lipo2 for 24 hours, and the result shows that the NTZ-lipo2 can effectively degrade the target protein EGFR. FIG. 10 shows the results of protein electrophoresis detection of HER2 protein expression from protein harvest after M231 human breast cancer tumor cells were treated with 500nM INE-lipo for 24 hours, showing that INE-lipo can effectively degrade the target protein.
FIG. 11 shows the successful ligation of Palb-PEG-DSPE by hydrogen nuclear magnetic resonance of the lipid linker arm used in Palb-lipo coupled to the POI recognition group. FIG. 12 shows the successful ligation as a result of hydrogen nuclear magnetic resonance of AV45-PEG-DSPE after the lipid linker arm used in AV45-lipo is coupled with the POI recognition group. FIG. 13 shows the result of protein electrophoresis detection of CDK4 protein expression from protein collected from M231 human breast cancer tumor cells after being treated with Palb-lipo for 24 hours at different concentrations, which shows that Palb-lipo can effectively degrade target proteins. FIG. 14 shows a schematic diagram of the structure of AV45-lipo and the structure of AV 45-SH. FIG. 15 shows confocal microscopy imaging of human glial cells after 24 hours of treatment with different concentrations of AV45-lipo, i.e., detection of β -amyloid (Abeta) being hijacked into cells. mu.M of oligomers prepared from beta-amyloid 1-42 polypeptide labeled with FITC green dye was incubated with 10 mu.M of AV45-lipo for 24 hours, followed by washing away the non-cellular processed material with PBS and confocal microscopy imaging to detect the green fluorescence of beta-amyloid 1-42 oligomers, which indicates greater amount of hijacking into cells, theoretically greater hijacking and phagocytosis and better degradation of lysosomal translocation energy, to reduce the toxicity of extracellular beta-amyloid oligomers accumulated, and the lysosomal dye LySoTracker was used to label lysosomes at a scale of 10 μ M.
Example 3
Referring to table 3, in this example, the degradation tool of example 3 was prepared by coupling the POI recognition group to the amphiphilic lipid linker arm followed by self-assembly with the lipid hybrid. Wherein the POI recognition group is: monoclonal antibody drug nitol bead (NTZ); the connecting arm is NHS-PEG-DSPE or NHS-PEG-DMG (1, 2-dimyristoyl-rac-glycerol-3-methoxypolyethylene glycol, polyethylene glycol end-modified N-hydroxysuccinimide) with NHS group. The lipid hybrid material consists of a membrane backbone, HSPC, cholesterol, and the cationic lipid, DOTAP (trimethyl-2, 3-dioleoyloxypropylammonium bromide). Cationic Lipid DOTAP is a representative cationic Lipid that facilitates Lipid hybridization of substances to form Lipid Nanoparticles (LNPs) for the encapsulation of negatively charged nucleic acid drugs. When the composition of the lipid hybrid substance is PEG-DSPE/cholesterol/HSPC, the degradation tool of the composition is NTZ-LNP1, and when the NTZ-LNP1 carries nonsense sequence no-load small interfering RNA (siRNA), the degradation tool of the composition is NTZ-LNP1s; when the composition of the lipid hybrid substance is PEG-DMG/cholesterol/HSPC, the degradation tool of the composition is NTZ-LNP2.
The preparation method comprises the following steps:
pretreatment, attachment of POI recognition group and linker arm, purification, using the method used in example 2.
Preparation of lipid hybrid: the lipid hybrid used was lipid nanoparticles, consisting of hydrogenated soy lecithin (HSPC) or Distearoylphosphatidylcholine (DSPC), cholesterol (CHO, cholestrol), DSPE-PEG (2 kDa) by classical thin film hydration, where the mass ratio [ HSPC or DSPC ]: CHO: DSPE-PEG (without POI recognition group) = 56. HSPC, CHO was dissolved in chloroform and then evaporated to concentrate and dried to form a film.
After synthesis of the lipid hybrid described above, 1mL of DSPE-PEG-antibody in PBS solution was added while DOTAP solution (molar ratio HSPC: DOTAP = 1) after complete evaporation of chloroform and then sonicated at 100W for 3min at room temperature. The resulting solution was then passed through 800nm, 400nm and 200nm filters.
NTZ-LNP1s: after the synthesis of the lipid hybrid described above, 1mL of DSPE-PEG-antibody in PBS was added after the chloroform had evaporated completely, and the amount of siRNA was calculated from the nitrogen to phosphorus ratio to DOTAP (N/P = 3. Simultaneously adding a DOTAP solution (molar ratio HSPC: DOTAP = 1) and an siRNA solution. Then sonicated at 100W for 3min at room temperature. The resulting solution was then passed through 800nm, 400nm and 200nm filters.
And (3) purification: the reaction mixture was then concentrated by ultrafiltration 3 times using a 50kDa centrifugal ultrafiltration tube and subsequently made up to 100. Mu.L using PBS. Protein concentration was determined by a NanoDrop one C instrument by a280 absorbance of IgG and verified by BCA method.
The experimental results are as follows: referring to FIG. 16, it is a schematic structural diagram of NTZ-LNP1, NTZ-LNP1s, and NTZ-LNP2. Fig. 17, particle size distribution for Dynamic Light Scattering (DLS) detection of the lipid hybrid. FIG. 18 shows the results of protein electrophoresis of protein harvest after 24 hours of 500nM NTZ-LNP1 treatment of M231 human breast cancer tumor cells for detecting EGFR protein expression, in which lipo1 in example 2 is also used as a control. The results show that NTZ-LNP1 can effectively degrade the target protein EGFR. FIG. 19 shows that protein electrophoresis detection of EGFR protein expression by protein harvest after M231 human breast cancer tumor cells are treated with NTZ-LNP1s for 24 hours shows that NTZ-LNP1s can effectively degrade target protein EGFR. FIG. 20 shows that protein electrophoresis detection of EGFR protein expression by protein harvest after M231 human breast cancer tumor cells are treated with NTZ-LNP2 for 24 hours shows that NTZ-LNP2 can effectively degrade target protein EGFR.
Example 4
Referring to table 3, in this example, the degradation tool of example 4 was prepared by coupling the POI recognition group to the amphiphilic lipid linker arm followed by self-assembly with the lipid hybrid. Wherein the POI recognition group is: monoclonal antibody drug Nitol (NTZ); the linking arm is NHS-PEG-DSPE with NHS group. Lipid hybrids consist of exosomes (exosomes). The exosome is an extracellular vesicle secreted by eukaryotic organisms such as animal or plant cells, is spontaneously formed, can perform intercellular communication, generally contains protein and a small amount of nucleic acid, has high biocompatibility and drug-loading capacity, and has a phospholipid membrane structure on the surface.
The preparation method comprises the following steps:
pretreatment, attachment of POI recognition group and linker arm, purification, using the method used in example 2.
Preparation of lipid hybrid: the exosome is separated, purified and identified by DC2.4 cells according to standard steps, and the exosome is coupled with a connecting arm through two modes of ultrasound and membrane extrusion. Namely, 1mL of PBS solution of DSPE-PEG-antibody is added into the PBS solution of exosome,
the combination of the connecting arm and the lipid hybrid substance adopts a membrane extrusion method or an ultrasonic method. Sonication consists of adding a PBS solution of DSPE-PEG-antibody to a PBS solution of exosomes (e.g. mass ratio exosomes: DSPE-PEG = 25 μ g in a 1mL system, i.e. 40. The extrusion method comprises the following steps: to the exosomes were added 1mL of a PBS solution of DSPE-PEG-antibody (mass ratio exosomes: DSPE-PEG =1mg, i.e. 40. The resulting solution was then passed through 800nm, 400nm and 200nm filters.
And (3) purification: the reaction mixture was then concentrated by ultrafiltration 3 times using a 50kDa centrifugal ultrafiltration tube and subsequently made up to 100. Mu.L using PBS. Protein concentration was determined by a NanoDrop one C instrument by a280 absorbance of IgG and verified by BCA method.
The experimental results are as follows: referring to fig. 21, DLS-detected particle size and average dispersion coefficient PDI (Polymer dispersion index) of the lipid hybrid material, the smaller the PDI, the more uniform the particle size, and the result indicates that the particle size is uniform. FIG. 22 shows that the protein collected from M231 human breast cancer tumor cells treated with NTZ-exo of different concentrations for 24 hours is subjected to protein electrophoresis to detect EGFR protein expression, and the result shows that NTZ-exo can effectively degrade target protein EGFR.
Example 5
Referring to table 3, in this example, the degradation tool of example 4 was prepared by coating the surface of the nanoparticle with the lipid hybrid, coupling the POI recognition group to the amphiphilic lipid linker arm, and then self-assembling with the lipid hybrid, with the POI recognition group exposed to the outside. Wherein the POI recognition group is: EGFR-targeting mab drug Nitol (NTZ) or Cetuximab (CTX); the linking arm is NHS-PEG-DSPE with NHS group. When the lipid hybrid substance consists of HSPC/cholesterol and wraps PLGA nano-particles, the degradation tool is NTZ-lipoP. Polylactic-co-glycolic acid (PLGA) is a representative polymer nanoparticle, has hydrophobicity, high biocompatibility, stable properties, and is biodegradable, and is widely used in medical treatment. The lipid hybrid is composed of mouse red cell membrane (RBCm) and encapsulates acetalized Dextran (Dextran), and when the POI recognition group is composed of CTX, the degradation tool is CTX-RBCmd. Dextran is representative of hydrophilic biodegradable nanoparticles. The erythrocyte membrane is a representative of cell membrane and organelle membrane, is a lipid hybrid substance, and is convenient to separate and extract.
The preparation method comprises the following steps:
pretreatment, attachment of POI recognition group and linker arm, purification, using the method used in example 2.
Preparation of lipid hybrid:
preparation of NTZ-lipoP: the composition is prepared from HSPC and CHO by a classical film rehydration method, wherein the mass ratio of the HSPC to the CHO: PEG-DSPE (without POI recognition group) = 56. HSPCs and CHO were dissolved in chloroform and subsequently concentrated and evaporated to form a lipid film. After the chloroform was completely evaporated, 1mL of antibody-PEG-DSPE dissolved in PBS was added for resuspension, followed by sonication at 100W for 3min at room temperature. The resulting solution was then passed through 800nm, 400nm and 200nm filters to insert the DSPE-PEG-antibody onto the lipid membrane.
And (3) coating PLGA: PLGA (15 kDa) was dissolved in DMF (10. Mu.g/. Mu.L), 2. Mu.L was added to 1mL of PBS solution every 10 seconds, the mixture was stirred at 700rpm for 3 hours, after DMF was evaporated, 1mg of LIPO (referred to as 1mg total of HSPC + CHO) was coated on each 0.01. Mu. Mol PLGA, and appropriate amounts of HSPC and CHO were dissolved in chloroform, and after concentration by evaporation, the mixture was resuspended in DSPE-PEG-NTZ PBS solution and sonicated at 100W for 3min. Incubating for 30min in a shaking table at 37 ℃, performing ultrasonic treatment for 5min at room temperature of 100W, and passing through a membrane of 800nm, 400nm and 200 nm. Mixing the two mixtures with PLGA PBS solution, performing 100W ice bath ultrasonic treatment for 2min, passing through 800, 400 and 200nm membranes, and performing ultrafiltration concentration. IgG concentration was measured.
Preparation of CTX-RBCmD: the synthesis of CTX-PEG-DSPE is as in example 2.
1mg of acetalized dextran was dissolved in 200. Mu.L of tetrahydrofuran, 10. Mu.L of the solution was dropped every 10s one drop into 1mL of alkaline water with pH =8 under stirring, and the solution was stirred at 700rpm for 3 hours. 1mg of mouse erythrocyte membrane was taken and resuspended in 1mL of PBS. PBS solution of red cell membrane was mixed with PEG solution of CTX-PEG-DSPE (calculated from membrane: acetalized dextran =1mg, molar ratio of acetalized dextran core to DSPE-PEG is 10 1) and incubated at 200rpm at 37 ℃ for 30min in shaker, followed by 100W at room temperature for 5min, and then the resulting solution was filtered through 400nm and 200nm filter membranes in order to obtain uniform size. Then mixed with the acetalized dextran solution, sonicated in a 100W ice bath for 2min, and the resulting solution was filtered through 400nm and 200nm filters to obtain the desired product. Then, ultrafiltration concentration was carried out. Protein concentration was measured in IgG format by NanoDrop one C (thermoldissher) and verified by BCA protein assay kit.
TABLE 4 DLS particle size statistics and PDI statistics for NTZ-lipoP and lipo-PLGA not linked by NTZ-PEG-DSPE
The experimental results are as follows: referring to FIG. 23, it is a schematic diagram of NTZ-lipoP structure. The results of DLS particle size statistics and PDI statistics for NTZ-lipoP and lipo-PLGA not linked by NTZ-PEG-DSPE show that the particle size is uniform and the hydrated particle size is slightly increased after the NTZ-PEG-DSPE is linked. FIG. 24 shows that the protein obtained from M231 human breast cancer tumor cells treated with NTZ-lipoP of different concentrations for 24 hours is subjected to protein electrophoresis to detect EGFR protein expression, and the result shows that the EGFR protein expression can effectively degrade the target protein EGFR. FIG. 25 is a schematic diagram of the CTX-RBCmD structure. Fig. 26 is a graph showing the result of DLS particle size distribution. FIG. 27 shows that the EGFR protein expression is detected by protein electrophoresis of protein harvested after M231 human breast cancer tumor cells are treated by 500nM CTX-RBCmD for 24 hours, and the result shows that the EGFR protein expression can be effectively degraded.
While the preferred embodiment and the corresponding examples of the present invention have been described, it should be understood that various changes and modifications, including but not limited to, adjustments of proportions, flows and amounts, which are within the scope of the invention, may be made by those skilled in the art without departing from the inventive concept thereof. While the preferred embodiment and the corresponding examples have been described, it should be understood that various changes and modifications, including but not limited to, adjustments of proportions, flows and amounts, which are within the scope of this invention, may be made by those skilled in the art without departing from the inventive concept.
Claims (11)
1.A lipid-based protein degradation tool, comprising:
a POI recognition group, and a lipid hybrid substance linked to the POI recognition group;
wherein the POI recognition group comprises an antibody, protein, polypeptide, aptamer or small molecule capable of specifically binding to the POI;
the lipid hybrid substance comprises liposome, exosome, cell membrane and LNP.
2. The lipid-based protein degradation means of claim 1,
when the POI recognition group is coupled to the lipid hybrid, the lipid-based protein degradation tool is a nanoparticle for protein degradation composed of the lipid hybrid in the core and the POI recognition group in the periphery.
3. The lipid-based protein degradation means of claim 1,
the lipid-based protein degradation means further comprises the lipid-based protein degradation means provided with a linking member between the POI recognition group and the lipid hybrid;
preferably, the linking member has a molecular weight of 0-1000kDa;
the connecting component is one of a polymer connecting arm and a lipid connecting arm;
preferably, the polymer linker arms comprise hydrophilic polymers, hydrophobic polymers, and amphiphilic polymers;
preferably, the lipid linker arm is an amphiphilic lipid linker arm.
4. The lipid-based proteolytic tool of claim 3, wherein the POI recognition group and the linking member form a set of linking units when the POI recognition group is coupled to the lipid hybrid through the linking member; the lipid-based protein degradation tool is a lipid-based protein degradation tool having a multi-layered structure in which the lipid hybrid is disposed at the core, the plurality of sets of the connection members connected to the lipid hybrid are disposed at the intermediate layer, and the plurality of sets of the connection units connected to the connection members are disposed at the periphery.
5. The lipid-based protein degradation kit according to claim 3, wherein the lipid linker arm comprises at least two ends, one end being a lipophilic end capable of linking to the lipid hybrid substance, and the other end being a hydrophilic end;
preferably, the lipophilic terminus is a lipid molecule.
6. The lipid-based protein degradation means of claim 3,
the amphiphilic polymer is a polymer with a chain/branched molecular structure, and at least one hydrophilic molecular end and one hydrophobic molecular end exist in the amphiphilic polymer;
preferably, the amphiphilic polymer has a linear molecular structure, one end of which is a hydrophilic molecular end and the other end of which is a hydrophobic molecular end.
7. The lipid-based protein degradation tool of claim 1, further comprising a nanoparticle, wherein the nanoparticle is coated at the core of the lipid-based protein degradation tool by the lipid hybrid;
the nanoparticles comprise hydrophilic particles, hydrophobic particles and inorganic nanoparticles;
preferably, the nanoparticles have a particle size of 5-1000nm.
8. Use of a lipid-based protein degradation means according to any of claims 1 to 7 for the preparation of a medicament, vaccine and delivery system for the treatment and prevention of diseases associated with abnormal accumulation of proteins; wherein, the diseases caused by abnormal accumulation of the protein comprise tumors, immune system diseases, inflammation and pathogen infection, neurodegenerative diseases, blood system diseases and metabolic diseases.
9. Use of a lipid-based protein degradation means according to any of claims 1 to 7 for the preparation of a test product/kit for the study of diseases associated with abnormal accumulation of proteins and protein interactions; wherein, the diseases caused by abnormal accumulation of the protein comprise tumors, immune system diseases, inflammation and pathogen infection, neurodegenerative diseases, blood system diseases and metabolic diseases.
10. A method for preparing a lipid-based protein degradation means according to any of claims 1 to 7,
when the lipid-based protein degradation tool is a protein degradation tool in which the POI recognition group is coupled to the lipid hybrid, the preparation method thereof is: non-covalently bonding the POI recognition group to the lipid hybrid; alternatively, the POI recognition group is covalently bonded to the lipid hybrid substance based on a coupling group, thereby constituting the lipid-based proteolytic tool.
11. The method of claim 10, wherein the lipid-based protein degradation means further comprises a protein degradation means wherein the POI recognition group is linked to the lipid hybrid via a linking member;
wherein, when the lipid-based protein degradation tool is a protein degradation tool in which the POI recognition group is connected with the lipid hybrid through the connecting member, the preparation method comprises the following steps:
firstly, coupling the POI recognition group with the connecting member to form a coupling intermediate; then linking the coupling intermediate to the lipid hybrid substance to form the lipid-based protein degradation means;
or, constructing a nanocomposite structure with the connecting member as an outer layer and the lipid hybrid substance as a core; and coupling the POI recognition group with the nano-composite structure to form the lipid-based protein degradation tool.
Priority Applications (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202210911853.5A CN115260304A (en) | 2022-07-29 | 2022-07-29 | Lipid-based protein degradation tool, application and preparation method thereof |
PCT/CN2023/104199 WO2024022009A1 (en) | 2022-07-29 | 2023-06-29 | Nanoprotein degradation tool, use thereof and preparation method therefor, and lipid-based protein degradation tool, use thereof and preparation method therefor |
US18/223,962 US20240033374A1 (en) | 2022-07-29 | 2023-07-19 | Nano-structural Protein Degradation Tool, Use, and Preparation Method thereof, and Lipid-based Protein Degradation Tool, Use, and Preparation Method thereof |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202210911853.5A CN115260304A (en) | 2022-07-29 | 2022-07-29 | Lipid-based protein degradation tool, application and preparation method thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
CN115260304A true CN115260304A (en) | 2022-11-01 |
Family
ID=83748052
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202210911853.5A Pending CN115260304A (en) | 2022-07-29 | 2022-07-29 | Lipid-based protein degradation tool, application and preparation method thereof |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN115260304A (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2024022009A1 (en) * | 2022-07-29 | 2024-02-01 | 河南大学 | Nanoprotein degradation tool, use thereof and preparation method therefor, and lipid-based protein degradation tool, use thereof and preparation method therefor |
-
2022
- 2022-07-29 CN CN202210911853.5A patent/CN115260304A/en active Pending
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2024022009A1 (en) * | 2022-07-29 | 2024-02-01 | 河南大学 | Nanoprotein degradation tool, use thereof and preparation method therefor, and lipid-based protein degradation tool, use thereof and preparation method therefor |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7046872B2 (en) | Hybridsomes, compositions containing them, methods of their production, and their use. | |
Wu et al. | Extracellular vesicles: A bright star of nanomedicine | |
Mondal et al. | Hybrid exosomes, exosome-like nanovesicles and engineered exosomes for therapeutic applications | |
AU2022201963B2 (en) | Fusogenic liposome-coated porous silicon nanoparticles | |
Tahmasbi Rad et al. | Combinational effects of active targeting, shape, and enhanced permeability and retention for cancer theranostic nanocarriers | |
US11713359B2 (en) | Apolipoprotein nanodiscs with telodendrimer | |
Tang et al. | Soft materials as biological and artificial membranes | |
Jiang et al. | The in vivo fate and targeting engineering of crossover vesicle-based gene delivery system | |
Kotmakçı et al. | Extracellular vesicles as natural nanosized delivery systems for small-molecule drugs and genetic material: steps towards the future nanomedicines | |
Tang et al. | Synergistic targeted delivery of payload into tumor cells by dual-ligand liposomes co-modified with cholesterol anchored transferrin and TAT | |
Hu et al. | Redox-sensitive folate-conjugated polymeric nanoparticles for combined chemotherapy and photothermal therapy against breast cancer | |
ES2745528T3 (en) | Magnetic nanoparticles functionalized with catechol, production and use thereof | |
CN109288794B (en) | Melittin liposome nano preparation and preparation method and application thereof | |
Golkar et al. | Modulated cellular delivery of anti-VEGF siRNA (bevasiranib) by incorporating supramolecular assemblies of hydrophobically modified polyamidoamine dendrimer in stealth liposomes | |
CN113633625B (en) | Nano-drug of hybrid membrane loaded oxidative phosphorylation inhibitor and preparation method thereof | |
US20080317840A1 (en) | Compositions and methods for polymer-caged liposomes | |
CN115260304A (en) | Lipid-based protein degradation tool, application and preparation method thereof | |
B Goudoulas | Polymers and biopolymers as drug delivery systems in nanomedicine | |
CN115043932A (en) | Nano protein degradation tool, application and preparation method thereof | |
US20050180922A1 (en) | Block co-polymer worm micelles and methods of use therefor | |
CN118252815A (en) | Cell membrane coated nucleic acid lipid composite nanoparticle and preparation method and application thereof | |
WO2024022009A1 (en) | Nanoprotein degradation tool, use thereof and preparation method therefor, and lipid-based protein degradation tool, use thereof and preparation method therefor | |
Raj et al. | Multi-functional lipid-based polymer composites for in vivo imaging, tissue healing, cell rejuvenation and theranostic applications | |
Kim et al. | One-Pot, One-Step Synthesis of Drug-Loaded Magnetic Multimicelle Aggregates | |
KR20230013009A (en) | Extracellular endoplasmic reticulum and manufacturing method thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination |