WO2023174207A9 - A method of obtaining extracts of spatholobus suberectus dunn (ssd), fractions and compostions thereof and using against viral diseases - Google Patents
A method of obtaining extracts of spatholobus suberectus dunn (ssd), fractions and compostions thereof and using against viral diseases Download PDFInfo
- Publication number
- WO2023174207A9 WO2023174207A9 PCT/CN2023/081058 CN2023081058W WO2023174207A9 WO 2023174207 A9 WO2023174207 A9 WO 2023174207A9 CN 2023081058 W CN2023081058 W CN 2023081058W WO 2023174207 A9 WO2023174207 A9 WO 2023174207A9
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- ssp
- ssd
- extract
- solvent
- cov
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims abstract description 94
- 239000000284 extract Substances 0.000 title claims abstract description 50
- 230000003612 virological effect Effects 0.000 title claims abstract description 22
- 201000010099 disease Diseases 0.000 title claims abstract description 17
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 title claims abstract description 17
- 241000800405 Spatholobus suberectus Species 0.000 title claims abstract description 9
- 241001678559 COVID-19 virus Species 0.000 claims abstract description 48
- 229920002770 condensed tannin Polymers 0.000 claims abstract description 23
- 238000002360 preparation method Methods 0.000 claims abstract description 11
- 239000000203 mixture Substances 0.000 claims abstract description 7
- 230000002265 prevention Effects 0.000 claims abstract description 6
- 238000004519 manufacturing process Methods 0.000 claims abstract description 5
- 241000700605 Viruses Species 0.000 claims description 28
- 239000002904 solvent Substances 0.000 claims description 27
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 claims description 26
- 241000315672 SARS coronavirus Species 0.000 claims description 24
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 claims description 23
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 claims description 21
- 230000008569 process Effects 0.000 claims description 20
- 241001115402 Ebolavirus Species 0.000 claims description 19
- LRHPLDYGYMQRHN-UHFFFAOYSA-N N-Butanol Chemical compound CCCCO LRHPLDYGYMQRHN-UHFFFAOYSA-N 0.000 claims description 18
- 241001473385 H5N1 subtype Species 0.000 claims description 17
- 238000001035 drying Methods 0.000 claims description 17
- 241000713772 Human immunodeficiency virus 1 Species 0.000 claims description 14
- 239000007864 aqueous solution Substances 0.000 claims description 14
- 239000000843 powder Substances 0.000 claims description 10
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 claims description 8
- 229920002472 Starch Polymers 0.000 claims description 8
- 238000005325 percolation Methods 0.000 claims description 8
- 230000002829 reductive effect Effects 0.000 claims description 8
- 239000008107 starch Substances 0.000 claims description 8
- 235000019698 starch Nutrition 0.000 claims description 8
- 239000003480 eluent Substances 0.000 claims description 7
- 239000011347 resin Substances 0.000 claims description 7
- 229920005989 resin Polymers 0.000 claims description 7
- 238000000926 separation method Methods 0.000 claims description 7
- 229920000168 Microcrystalline cellulose Polymers 0.000 claims description 6
- 238000005520 cutting process Methods 0.000 claims description 6
- 239000008108 microcrystalline cellulose Substances 0.000 claims description 6
- 235000019813 microcrystalline cellulose Nutrition 0.000 claims description 6
- 229940016286 microcrystalline cellulose Drugs 0.000 claims description 6
- 239000003208 petroleum Substances 0.000 claims description 6
- 239000001267 polyvinylpyrrolidone Substances 0.000 claims description 6
- 229920000036 polyvinylpyrrolidone Polymers 0.000 claims description 6
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 claims description 6
- 239000007921 spray Substances 0.000 claims description 6
- 239000003795 chemical substances by application Substances 0.000 claims description 5
- 239000007922 nasal spray Substances 0.000 claims description 5
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 claims description 4
- 229920000881 Modified starch Polymers 0.000 claims description 4
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 claims description 4
- 229930006000 Sucrose Natural products 0.000 claims description 4
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 claims description 4
- 239000011230 binding agent Substances 0.000 claims description 4
- 238000004108 freeze drying Methods 0.000 claims description 4
- 238000010438 heat treatment Methods 0.000 claims description 4
- 239000003960 organic solvent Substances 0.000 claims description 4
- 238000002390 rotary evaporation Methods 0.000 claims description 4
- 229910052708 sodium Inorganic materials 0.000 claims description 4
- 239000011734 sodium Substances 0.000 claims description 4
- 239000005720 sucrose Substances 0.000 claims description 4
- 239000000375 suspending agent Substances 0.000 claims description 4
- 239000000654 additive Substances 0.000 claims description 3
- 235000009508 confectionery Nutrition 0.000 claims description 3
- IDGUHHHQCWSQLU-UHFFFAOYSA-N ethanol;hydrate Chemical compound O.CCO IDGUHHHQCWSQLU-UHFFFAOYSA-N 0.000 claims description 3
- 239000000945 filler Substances 0.000 claims description 3
- NWUYHJFMYQTDRP-UHFFFAOYSA-N 1,2-bis(ethenyl)benzene;1-ethenyl-2-ethylbenzene;styrene Chemical compound C=CC1=CC=CC=C1.CCC1=CC=CC=C1C=C.C=CC1=CC=CC=C1C=C NWUYHJFMYQTDRP-UHFFFAOYSA-N 0.000 claims description 2
- 229920001817 Agar Polymers 0.000 claims description 2
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 claims description 2
- 229920002101 Chitin Polymers 0.000 claims description 2
- 241000711573 Coronaviridae Species 0.000 claims description 2
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 claims description 2
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 claims description 2
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 claims description 2
- 229930195725 Mannitol Natural products 0.000 claims description 2
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 claims description 2
- 239000008272 agar Substances 0.000 claims description 2
- 238000007664 blowing Methods 0.000 claims description 2
- 239000001913 cellulose Substances 0.000 claims description 2
- 229920002678 cellulose Polymers 0.000 claims description 2
- 235000010980 cellulose Nutrition 0.000 claims description 2
- 238000005119 centrifugation Methods 0.000 claims description 2
- 238000004587 chromatography analysis Methods 0.000 claims description 2
- 238000002425 crystallisation Methods 0.000 claims description 2
- 230000008025 crystallization Effects 0.000 claims description 2
- 239000003405 delayed action preparation Substances 0.000 claims description 2
- 239000000839 emulsion Substances 0.000 claims description 2
- 239000000499 gel Substances 0.000 claims description 2
- 239000008187 granular material Substances 0.000 claims description 2
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 claims description 2
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 claims description 2
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 claims description 2
- UFVKGYZPFZQRLF-UHFFFAOYSA-N hydroxypropyl methyl cellulose Chemical compound OC1C(O)C(OC)OC(CO)C1OC1C(O)C(O)C(OC2C(C(O)C(OC3C(C(O)C(O)C(CO)O3)O)C(CO)O2)O)C(CO)O1 UFVKGYZPFZQRLF-UHFFFAOYSA-N 0.000 claims description 2
- 238000002347 injection Methods 0.000 claims description 2
- 239000007924 injection Substances 0.000 claims description 2
- 239000003456 ion exchange resin Substances 0.000 claims description 2
- 229920003303 ion-exchange polymer Polymers 0.000 claims description 2
- 239000008101 lactose Substances 0.000 claims description 2
- 239000007788 liquid Substances 0.000 claims description 2
- 229940031703 low substituted hydroxypropyl cellulose Drugs 0.000 claims description 2
- 239000007937 lozenge Substances 0.000 claims description 2
- DKXULEFCEORBJK-UHFFFAOYSA-N magnesium;octadecanoic acid Chemical compound [Mg].CCCCCCCCCCCCCCCCCC(O)=O DKXULEFCEORBJK-UHFFFAOYSA-N 0.000 claims description 2
- 239000000594 mannitol Substances 0.000 claims description 2
- 235000010355 mannitol Nutrition 0.000 claims description 2
- 229910052757 nitrogen Inorganic materials 0.000 claims description 2
- 239000006187 pill Substances 0.000 claims description 2
- 238000001556 precipitation Methods 0.000 claims description 2
- 238000010992 reflux Methods 0.000 claims description 2
- 238000004366 reverse phase liquid chromatography Methods 0.000 claims description 2
- 239000000377 silicon dioxide Substances 0.000 claims description 2
- 235000015424 sodium Nutrition 0.000 claims description 2
- 235000019333 sodium laurylsulphate Nutrition 0.000 claims description 2
- 238000000638 solvent extraction Methods 0.000 claims description 2
- 238000001694 spray drying Methods 0.000 claims description 2
- 239000003826 tablet Substances 0.000 claims description 2
- 239000000454 talc Substances 0.000 claims description 2
- 229910052623 talc Inorganic materials 0.000 claims description 2
- 208000025721 COVID-19 Diseases 0.000 abstract description 10
- 229930003935 flavonoid Natural products 0.000 abstract description 6
- 150000002215 flavonoids Chemical class 0.000 abstract description 6
- 235000017173 flavonoids Nutrition 0.000 abstract description 6
- 210000004027 cell Anatomy 0.000 description 69
- 241001112090 Pseudovirus Species 0.000 description 30
- 230000000840 anti-viral effect Effects 0.000 description 29
- 208000015181 infectious disease Diseases 0.000 description 20
- 241000700159 Rattus Species 0.000 description 19
- 241000699670 Mus sp. Species 0.000 description 18
- 238000002474 experimental method Methods 0.000 description 16
- 102000053723 Angiotensin-converting enzyme 2 Human genes 0.000 description 14
- 108090000975 Angiotensin-converting enzyme 2 Proteins 0.000 description 14
- 241000711975 Vesicular stomatitis virus Species 0.000 description 14
- 230000003013 cytotoxicity Effects 0.000 description 14
- 231100000135 cytotoxicity Toxicity 0.000 description 14
- 238000012360 testing method Methods 0.000 description 14
- 239000003814 drug Substances 0.000 description 13
- 230000000694 effects Effects 0.000 description 13
- 229940079593 drug Drugs 0.000 description 12
- 238000002203 pretreatment Methods 0.000 description 11
- 108060001084 Luciferase Proteins 0.000 description 9
- 239000005089 Luciferase Substances 0.000 description 9
- 230000005764 inhibitory process Effects 0.000 description 9
- 230000009385 viral infection Effects 0.000 description 9
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 8
- 108010003533 Viral Envelope Proteins Proteins 0.000 description 8
- 230000002401 inhibitory effect Effects 0.000 description 8
- 241001465754 Metazoa Species 0.000 description 7
- 229940087603 grape seed extract Drugs 0.000 description 7
- 235000002532 grape seed extract Nutrition 0.000 description 7
- 210000000056 organ Anatomy 0.000 description 7
- 238000001228 spectrum Methods 0.000 description 7
- 210000001519 tissue Anatomy 0.000 description 7
- 239000001717 vitis vinifera seed extract Substances 0.000 description 7
- 102100034349 Integrase Human genes 0.000 description 6
- 208000036142 Viral infection Diseases 0.000 description 6
- 238000000605 extraction Methods 0.000 description 6
- 238000001727 in vivo Methods 0.000 description 6
- 239000003112 inhibitor Substances 0.000 description 6
- 210000004072 lung Anatomy 0.000 description 6
- 238000000159 protein binding assay Methods 0.000 description 6
- 238000004704 ultra performance liquid chromatography Methods 0.000 description 6
- 230000035899 viability Effects 0.000 description 6
- 230000007502 viral entry Effects 0.000 description 6
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical group [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 5
- 230000037396 body weight Effects 0.000 description 5
- PEASPLKKXBYDKL-FXEVSJAOSA-N enfuvirtide Chemical compound C([C@@H](C(=O)N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](C)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(N)=O)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(C)=O)[C@@H](C)O)[C@@H](C)CC)C1=CN=CN1 PEASPLKKXBYDKL-FXEVSJAOSA-N 0.000 description 5
- 239000012091 fetal bovine serum Substances 0.000 description 5
- 239000013641 positive control Substances 0.000 description 5
- 239000011780 sodium chloride Substances 0.000 description 5
- 238000010186 staining Methods 0.000 description 5
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 description 4
- 102100031673 Corneodesmosin Human genes 0.000 description 4
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 4
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 description 4
- 230000007059 acute toxicity Effects 0.000 description 4
- 231100000403 acute toxicity Toxicity 0.000 description 4
- 210000004369 blood Anatomy 0.000 description 4
- 239000008280 blood Substances 0.000 description 4
- 150000001875 compounds Chemical class 0.000 description 4
- 239000000463 material Substances 0.000 description 4
- 231100000682 maximum tolerated dose Toxicity 0.000 description 4
- 210000004779 membrane envelope Anatomy 0.000 description 4
- 229920000642 polymer Polymers 0.000 description 4
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 4
- 102000005962 receptors Human genes 0.000 description 4
- 108020003175 receptors Proteins 0.000 description 4
- 238000004809 thin layer chromatography Methods 0.000 description 4
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 3
- 101710139375 Corneodesmosin Proteins 0.000 description 3
- 241000196324 Embryophyta Species 0.000 description 3
- 208000031886 HIV Infections Diseases 0.000 description 3
- 201000003176 Severe Acute Respiratory Syndrome Diseases 0.000 description 3
- 230000005856 abnormality Effects 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 230000007541 cellular toxicity Effects 0.000 description 3
- 239000003067 chemokine receptor CCR5 antagonist Substances 0.000 description 3
- 239000002576 chemokine receptor CXCR4 antagonist Substances 0.000 description 3
- 229940121384 cxc chemokine receptor type 4 (cxcr4) antagonist Drugs 0.000 description 3
- 230000037406 food intake Effects 0.000 description 3
- 235000012631 food intake Nutrition 0.000 description 3
- VEXZGXHMUGYJMC-UHFFFAOYSA-N hydrochloric acid Substances Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 3
- 150000002500 ions Chemical class 0.000 description 3
- 230000007794 irritation Effects 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- 229940097496 nasal spray Drugs 0.000 description 3
- 229930014626 natural product Natural products 0.000 description 3
- 239000013642 negative control Substances 0.000 description 3
- 238000006116 polymerization reaction Methods 0.000 description 3
- 150000008442 polyphenolic compounds Chemical class 0.000 description 3
- 235000013824 polyphenols Nutrition 0.000 description 3
- 230000003389 potentiating effect Effects 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 238000003908 quality control method Methods 0.000 description 3
- 230000010076 replication Effects 0.000 description 3
- -1 such as Substances 0.000 description 3
- 230000001988 toxicity Effects 0.000 description 3
- 231100000419 toxicity Toxicity 0.000 description 3
- 102100035875 C-C chemokine receptor type 5 Human genes 0.000 description 2
- 101710149870 C-C chemokine receptor type 5 Proteins 0.000 description 2
- 102100038132 Endogenous retrovirus group K member 6 Pro protein Human genes 0.000 description 2
- 101710091045 Envelope protein Proteins 0.000 description 2
- 241000725303 Human immunodeficiency virus Species 0.000 description 2
- 108010061833 Integrases Proteins 0.000 description 2
- 108060004795 Methyltransferase Proteins 0.000 description 2
- 238000011887 Necropsy Methods 0.000 description 2
- 108091005804 Peptidases Proteins 0.000 description 2
- 229920002873 Polyethylenimine Polymers 0.000 description 2
- 239000004365 Protease Substances 0.000 description 2
- 101710188315 Protein X Proteins 0.000 description 2
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 2
- 101000629318 Severe acute respiratory syndrome coronavirus 2 Spike glycoprotein Proteins 0.000 description 2
- 244000269722 Thea sinensis Species 0.000 description 2
- 229940118555 Viral entry inhibitor Drugs 0.000 description 2
- 230000001154 acute effect Effects 0.000 description 2
- 230000002155 anti-virotic effect Effects 0.000 description 2
- 239000003443 antiviral agent Substances 0.000 description 2
- 229930101531 artemisinin Natural products 0.000 description 2
- BLUAFEHZUWYNDE-NNWCWBAJSA-N artemisinin Chemical compound C([C@](OO1)(C)O2)C[C@H]3[C@H](C)CC[C@@H]4[C@@]31[C@@H]2OC(=O)[C@@H]4C BLUAFEHZUWYNDE-NNWCWBAJSA-N 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- 239000000470 constituent Substances 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 239000000539 dimer Substances 0.000 description 2
- 230000002222 downregulating effect Effects 0.000 description 2
- 238000009509 drug development Methods 0.000 description 2
- 238000010828 elution Methods 0.000 description 2
- 125000001033 ether group Chemical group 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 239000012634 fragment Substances 0.000 description 2
- 230000004927 fusion Effects 0.000 description 2
- 241000411851 herbal medicine Species 0.000 description 2
- 210000004969 inflammatory cell Anatomy 0.000 description 2
- 230000003993 interaction Effects 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 231100001252 long-term toxicity Toxicity 0.000 description 2
- 230000007774 longterm Effects 0.000 description 2
- 229960004710 maraviroc Drugs 0.000 description 2
- GSNHKUDZZFZSJB-QYOOZWMWSA-N maraviroc Chemical compound CC(C)C1=NN=C(C)N1[C@@H]1C[C@H](N2CC[C@H](NC(=O)C3CCC(F)(F)CC3)C=3C=CC=CC=3)CC[C@H]2C1 GSNHKUDZZFZSJB-QYOOZWMWSA-N 0.000 description 2
- 239000002398 materia medica Substances 0.000 description 2
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 239000000178 monomer Substances 0.000 description 2
- 210000003928 nasal cavity Anatomy 0.000 description 2
- 229950010131 puromycin Drugs 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 231100000161 signs of toxicity Toxicity 0.000 description 2
- 210000002784 stomach Anatomy 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 239000006228 supernatant Substances 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 238000005406 washing Methods 0.000 description 2
- JPFCOVZKLAXXOE-XBNSMERZSA-N (3r)-2-(3,5-dihydroxy-4-methoxyphenyl)-8-[(2r,3r,4r)-3,5,7-trihydroxy-2-(4-hydroxyphenyl)-3,4-dihydro-2h-chromen-4-yl]-3,4-dihydro-2h-chromene-3,5,7-triol Chemical compound C1=C(O)C(OC)=C(O)C=C1C1[C@H](O)CC(C(O)=CC(O)=C2[C@H]3C4=C(O)C=C(O)C=C4O[C@@H]([C@@H]3O)C=3C=CC(O)=CC=3)=C2O1 JPFCOVZKLAXXOE-XBNSMERZSA-N 0.000 description 1
- OSWFIVFLDKOXQC-UHFFFAOYSA-N 4-(3-methoxyphenyl)aniline Chemical compound COC1=CC=CC(C=2C=CC(N)=CC=2)=C1 OSWFIVFLDKOXQC-UHFFFAOYSA-N 0.000 description 1
- 208000030507 AIDS Diseases 0.000 description 1
- 206010002091 Anaesthesia Diseases 0.000 description 1
- 235000003826 Artemisia Nutrition 0.000 description 1
- 235000001405 Artemisia annua Nutrition 0.000 description 1
- 240000000011 Artemisia annua Species 0.000 description 1
- 235000003261 Artemisia vulgaris Nutrition 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 241000258920 Chilopoda Species 0.000 description 1
- 201000011001 Ebola Hemorrhagic Fever Diseases 0.000 description 1
- 108010032976 Enfuvirtide Proteins 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- GRRNUXAQVGOGFE-UHFFFAOYSA-N Hygromycin-B Natural products OC1C(NC)CC(N)C(O)C1OC1C2OC3(C(C(O)C(O)C(C(N)CO)O3)O)OC2C(O)C(CO)O1 GRRNUXAQVGOGFE-UHFFFAOYSA-N 0.000 description 1
- 239000012901 Milli-Q water Substances 0.000 description 1
- 206010033799 Paralysis Diseases 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 229920001991 Proanthocyanidin Polymers 0.000 description 1
- 229940096437 Protein S Drugs 0.000 description 1
- 101150010882 S gene Proteins 0.000 description 1
- 208000037847 SARS-CoV-2-infection Diseases 0.000 description 1
- 238000012300 Sequence Analysis Methods 0.000 description 1
- 229930182558 Sterol Natural products 0.000 description 1
- 210000001744 T-lymphocyte Anatomy 0.000 description 1
- 241000711973 Vesicular stomatitis Indiana virus Species 0.000 description 1
- 108070000030 Viral receptors Proteins 0.000 description 1
- 108010031318 Vitronectin Proteins 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000006978 adaptation Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 230000037005 anaesthesia Effects 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 150000004056 anthraquinones Chemical class 0.000 description 1
- 239000002259 anti human immunodeficiency virus agent Substances 0.000 description 1
- 230000003110 anti-inflammatory effect Effects 0.000 description 1
- 238000002832 anti-viral assay Methods 0.000 description 1
- 239000003430 antimalarial agent Substances 0.000 description 1
- 229940121357 antivirals Drugs 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 244000030166 artemisia Species 0.000 description 1
- 235000009052 artemisia Nutrition 0.000 description 1
- 229960004191 artemisinin Drugs 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 230000017531 blood circulation Effects 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 210000000621 bronchi Anatomy 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 238000012200 cell viability kit Methods 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 238000013375 chromatographic separation Methods 0.000 description 1
- 238000012761 co-transfection Methods 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 238000002784 cytotoxicity assay Methods 0.000 description 1
- 231100000263 cytotoxicity test Toxicity 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 239000000645 desinfectant Substances 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 238000007876 drug discovery Methods 0.000 description 1
- 238000002330 electrospray ionisation mass spectrometry Methods 0.000 description 1
- 229960002062 enfuvirtide Drugs 0.000 description 1
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- 235000019253 formic acid Nutrition 0.000 description 1
- 210000003918 fraction a Anatomy 0.000 description 1
- 238000003304 gavage Methods 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 235000008216 herbs Nutrition 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- GRRNUXAQVGOGFE-NZSRVPFOSA-N hygromycin B Chemical compound O[C@@H]1[C@@H](NC)C[C@@H](N)[C@H](O)[C@H]1O[C@H]1[C@H]2O[C@@]3([C@@H]([C@@H](O)[C@@H](O)[C@@H](C(N)CO)O3)O)O[C@H]2[C@@H](O)[C@@H](CO)O1 GRRNUXAQVGOGFE-NZSRVPFOSA-N 0.000 description 1
- 229940097277 hygromycin b Drugs 0.000 description 1
- 208000006278 hypochromic anemia Diseases 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 230000007365 immunoregulation Effects 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 230000001524 infective effect Effects 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 210000003127 knee Anatomy 0.000 description 1
- 210000000867 larynx Anatomy 0.000 description 1
- 229920005610 lignin Polymers 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 201000004792 malaria Diseases 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 210000004379 membrane Anatomy 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 210000000214 mouth Anatomy 0.000 description 1
- 210000004400 mucous membrane Anatomy 0.000 description 1
- 231100000862 numbness Toxicity 0.000 description 1
- 238000003305 oral gavage Methods 0.000 description 1
- 244000045947 parasite Species 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 239000013612 plasmid Substances 0.000 description 1
- 239000002798 polar solvent Substances 0.000 description 1
- 231100000683 possible toxicity Toxicity 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 108090000623 proteins and genes Proteins 0.000 description 1
- 230000004850 protein–protein interaction Effects 0.000 description 1
- 230000005180 public health Effects 0.000 description 1
- 239000008213 purified water Substances 0.000 description 1
- 229930183339 qinghaosu Natural products 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 239000003419 rna directed dna polymerase inhibitor Substances 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 238000013223 sprague-dawley female rat Methods 0.000 description 1
- 238000013222 sprague-dawley male rat Methods 0.000 description 1
- 150000003432 sterols Chemical class 0.000 description 1
- 235000003702 sterols Nutrition 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 150000003505 terpenes Chemical class 0.000 description 1
- 235000007586 terpenes Nutrition 0.000 description 1
- 238000007079 thiolysis reaction Methods 0.000 description 1
- 231100000816 toxic dose Toxicity 0.000 description 1
- 238000002723 toxicity assay Methods 0.000 description 1
- 235000013619 trace mineral Nutrition 0.000 description 1
- 239000011573 trace mineral Substances 0.000 description 1
- 210000003437 trachea Anatomy 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 230000010415 tropism Effects 0.000 description 1
- 238000001195 ultra high performance liquid chromatography Methods 0.000 description 1
- 238000001946 ultra-performance liquid chromatography-mass spectrometry Methods 0.000 description 1
- 238000005199 ultracentrifugation Methods 0.000 description 1
- 229960005486 vaccine Drugs 0.000 description 1
- 230000029812 viral genome replication Effects 0.000 description 1
- 230000017613 viral reproduction Effects 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
Classifications
-
- 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
- A61P31/12—Antivirals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K36/00—Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
- A61K36/18—Magnoliophyta (angiosperms)
- A61K36/185—Magnoliopsida (dicotyledons)
- A61K36/48—Fabaceae or Leguminosae (Pea or Legume family); Caesalpiniaceae; Mimosaceae; Papilionaceae
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2236/00—Isolation or extraction methods of medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicine
Definitions
- Disclosed is a method of preparing a natural product of antiviral extract from Spatholobus suberectus Dunn (SSD) , which contains total flavonoid and proanthocyanidins.
- the disclosure relates to a method for discovering an active proanthocyanidins of Spatholobus suberectus (SSP) as viral entry inhibitor.
- a method of preventing and treating viral diseases such as coronavirus disease 2019 (COVID-19) .
- Herbal medicines and purified natural products provide a rich resource for novel antiviral drug development. Identification of the antiviral mechanisms from these natural agents has shed light on where they interact with the viral life cycle, such as viral entry, replication, assembly, and release.
- Traditional Chinese Medicine TCM
- Artemisia annua is the representative herb.
- Qinghaosu an antimalarial drug from Artemisia annual, and subsequent development of several therapies to inhibit the malaria parasite has saved millions of lives.
- the plant SSD Slightly sweet, warm in nature, belongs to Chinese medicine liver and kidney. "Compendium of Materia Medica” claims that it can nourish the stomach and dry stomach; “Compendium of Compendium of Materia Medica” claims that it can promote blood circulation and warm the waist and knees.
- Centipede to treat diseases caused by viruses, but it has not been reported in Covid-19. Till now, no publication has indicated SSP as a viral entry inhibitor.
- the chemical constituents of SSD mainly include flavonoids, terpenes, sterols, lignin, anthraquinones, polyphenols, proanthocyanidins and some trace elements, among which the content of flavonoids is the most.
- the research on the extraction method of content mostly uses the total flavonoid extraction rate as an indicator and does not consider the effect of the extraction method on the drug efficacy of the extract.
- a method of obtaining an extract of SSD comprising: (i) cutting SSD into pieces; (ii) immersing and treating SSD pieces in a solvent at room temperature to obtain an extract; (iii) concentrating the extract; and (iv) drying the concentrated extract to obtain the powder of the extract of SSD (SSP) .
- a method of obtaining an active fraction from SSD comprising: (i) cutting SSD into pieces; (ii) immersing and treating SSD pieces in a solvent at room temperature to obtain an extract; (iii) concentrating the extract; (iv) drying the concentrated extract to obtain the powder of the extract of SSD (SSP) ; (v) dispersing SSP in water to obtain an aqueous solution; (vi) extracting the SSP aqueous solution with a series of solvent with different polarities at equal volume to obtain parts of SSP corresponding to each solvent, the series of solvent comprising n-butanol; and (vii) separating the n-butanol part (SSP-n-BuOH) with eluents to obtain the active fraction.
- the treating process in (ii) comprises percolation.
- an extract of SSD and an active fraction from SSD obtained by the above methods as well as a composition comprising the extract or the active fraction.
- a method of preventing and/or treating a viral disease comprising administering an effective amount of an extract of SSD or an active fraction from SSD obtained by the above methods to a subject in need thereof.
- the viral disease is caused by SARS-CoV-2 including Omicron variants, Ebola virus (EBOV) , HIV-1 with either CCR5 or CXCR4 tropism, SARS-CoV, H5N1.
- SARS-CoV-2 including Omicron variants, Ebola virus (EBOV) , HIV-1 with either CCR5 or CXCR4 tropism, SARS-CoV, H5N1.
- EBOV Ebola virus
- the effective dose is 0.5 ⁇ g/ml to 5000 ⁇ g/ml.
- an extract of SSD or an active fraction from SSD obtained by the above methods in the manufacture of preparations for prevention and/or treatment of a viral disease.
- Figure 1 Flow chart of the study. Preparation and quality-controlled of SSP which is used to determine the broad-spectrum antiviral activity and its underlying mechanism. The powder SSP was further extracted to obtain the effective active parts.
- FIG. 1 Representative pictures of SSP's quality control.
- FIG. 3 Antiviral activity of SSP against SARS-CoV-2. Serially diluted SSP was added to HEK293T-ACE2 cells infected with SARS-CoV-2 (a) and VSV (b) respectively. The luciferase level was measured 2 or 3 days post-infection. To test SSP cytotoxicity, cells viability (c) was measured. (d) Pre-treatment of SARS-CoV-2 and target cells inhibited viral infection. (e) Binding of RBD to ACE2 expressing 293T cells, but not 293T control cells. (f) SSP inhibited RBD binding to target cells.
- SSP pre-treated HEK293T-ACE2 cells were incubated with RBD-PD1 for 30 mins on ice, followed by antibody staining of ACE2 (upper panel) and RBD (lower panel) and flow cytometry analysis.
- the data represent the mean ⁇ SEM of triplicate experiments.
- FIG. 4 Antiviral activity of SSP against SARS-CoV-1, H5N1, and HIV, and EBOV viruses.
- Serially diluted SSP was added to HEK293T-ACE2, MDCK, GHOST-CCR5, and GHOST-CXCR4 infected with SARS-CoV-1 (a) H5N1 (b) HIV ADA (c) , and HIV HXB2 (d) respectively.
- the luciferase level was measured 2 or 3 days post-infection. Cells viability (e) was measured to test SSP cytotoxicity.
- Serially diluted SSP was added to 293T cells infected with EBOV. The luciferase level was measured 2 days post infection. Cells viability was measured to test SSP cytotoxicity against the 293T cells. The data represent the mean ⁇ SEM of triplicate experiments.
- FIG. 1 Mechanisms of SSP mediated virus entry inhibition.
- (a-b) Pre-treatment of SARS-CoV-1 (a) , H5N1 (b) and target cells inhibited viral infection.
- (c-d) Post-entry assay.
- GHOST-CD4-CCR5 or CXCR4 cells were co-incubated with pseudovirus for 2 hrs, washed, and then treated with the presence of 50 mg/ml SSP and 1 mM AZT as a positive control for 48 hr.
- SSP does not inhibit either HIV-1 ADA or HIV HxB2 virus gene replication after the viral entry is achieved.
- (e-f) SSP-virus interaction assay. HIV-1 ADA and HIV HxB2 pseudovirus pre-treated with 50 mg/ml SSP and entry inhibitor enfuvirtide (T-20) as a positive control.
- SSP pre-treatment inhibited both HIV ADA and HIV HxB2 pseudovirus infection to a similar degree as T-20.
- FIG. 7 Antiviral activity of SSP and their extracted parts against SARS-CoVs, and VSV.
- serially diluted SSP, SSP-n-BuOH, Fr. B, F, and G were added to HEK293T-ACE2 cells infected with SARS-CoV-2 (a) , SARS-CoV (b) , and VSV (c) .
- the luciferase level was measured 2 or 3 days post-infection. To test their cytotoxicity, cells viability (d) was also measured.
- e-h Pre-treatment of SARS-CoV-2 and target cells inhibited viral infection.
- HEK293T-ACE2 cells were infected with SARS-CoV-2 pseudovirus and treated with gradient diluted SSP simultaneously. The data represent the mean ⁇ SEM of triplicate experiments.
- FIG. 8 SSP did not show significant cytotoxicity in multiple cell lines, or long-term in vivo toxicity in rats.
- (a ⁇ d) To test SSP cytotoxicity, cells viability was measured in HEK293T-ACE2 (a) , MDCK (b) , GHOST (3) -CD4-CXCR4 (c) /CCR5 (d) .
- (e) Effects of SSP administration for 4 weeks on the bodyweight of SD rats.
- (f) Effects of SSP administration for 4 weeks on food intake of SD rats.
- FIG. 9 SSP shows higher potency and broader spectrum against SARS-CoV-2 variants entry in comparison with GSE and TP.
- A After keeping SSP solution for 6 and 13 days, the anti-viral activity and cyto-toxicity were tested.
- B SSP had a fairly strong inhibitory effect against major SARS-CoV-2 variants [including Alpha (B. 1.1.7) , Beta (B. 1.351) , Gamma (P1) , Delta (B. 1.617.2) , and Omicron (BA. 1, BA. 2, BA2.1.2.1 and BA. 4/5) ] .
- C ⁇ D The anti-viral efficacy of grape seed extract (GSE) and tea polyphenol (TP) against SARS-CoV-2 and Omicron variants, as well as their cyto-toxicity.
- FIG. 10 SSP shows efficacy in protection against nasal challenge of SARS_CoV-2 omicron in mice.
- A Flowchart of the in vivo experiments.
- B Omicron BA. 2 virus-infected cells (green) and inflammatory cells (red) were significantly reduced in the lung tissues of mice treated with SSP compared with the saline group.
- C In the lung tissue of mice treated with SSP, the expression level of RNA-dependent RNA polymerase and the virus titer were significantly lower than those of mice treated with saline. *P ⁇ 0.05.
- the present invention is related to a method of obtaining an extract of SSD, said method comprising: (i) cutting SSD into pieces; (ii) immersing and treating SSD pieces in a solvent at room temperature to obtain an extract; (iii) concentrating the extract; and (iv) drying the concentrated extract to obtain the powder of the extract of SSD, the extract of SSD including proanthocyanidins, named SSP.
- SSD Spatholobus suberectus Dunn.
- SSP refers to the extract of SSD including proanthocyanidins.
- SSP can be obtained by the above method comprising steps (i) to (iv) .
- suitable treating process can comprise percolation, ultrasonic, heating and refluxing, decoction, and/or solvent extraction.
- the treating process in (ii) is percolation.
- the solvent used can comprise water, and/or an organic solvent.
- the solvent in (ii) comprises water and ethanol, such as 60%ethanol/water (v/v) .
- step (ii) comprises treating SSD pieces via a percolation process with a 60%ethanol (in water, v/v) at room temperature.
- the percolation process can be performed for 12 hours.
- the concentrating process can comprise reduced pressure rotary evaporation, centrifugation, and/or nitrogen blowing.
- the concentrating process in (iii) is reduced pressure rotary evaporation.
- the concentrating process can be performed at a temperature under 50°C.
- the drying process can comprise freeze drying, spray drying, microwave drying, and/or infrared heating drying.
- the drying process in (iv) is freeze drying.
- the invention also relates to the SSP obtained by the above method.
- SSP exhibited significant inhibitory ability against SARS-CoV-2 IC 50 values of 3.574 ⁇ g/mL and 3.648 ⁇ g/mL.
- SSP uniquely inhibited the entry of SARS-CoV-2, HIV-1, Ebola virus (EBOV) , SARS-CoV-1 and influeunza H5N1 infections but failed to block vesicular stomatitis virus (VSV) .
- VSV vesicular stomatitis virus
- SSP had no effects on post-entry events of replication. It blocked SARS-CoV-2, HIV-1, H5N1, EBOV and SARS-CoV-1 entry by acting on viral envelope directly.
- SSP is a novel entry inhibitor, which has a potential for preventing and treating COVID-19, also can fight EBOV, HIV-1, H5N1 and SARS-CoV infection, and stockpiling for possible use against future pandemic caused by related virus.
- the invention is related to a method of obtaining an active fraction from SSD, the active fractions including proanthocyanidins, said method comprising: (i) cutting SSD into pieces; (ii) immersing and treating SSD pieces in a solvent at room temperature to obtain an extract; (iii) concentrating the extract; (iv) drying the concentrated extract to obtain the powder of the extract of SSD (SSP) ; (v) dispersing SSP in water to obtain an aqueous solution; (vi) extracting the SSP aqueous solution with a series of solvent with different polarities at equal volume to obtain parts of SSP corresponding to each solvent, the series of solvent comprising n-butanol; and (vii) separating the n-butanol part (SSP-n-BuOH) with eluents to obtain the active fraction.
- SSP n-butanol part
- the term “active fraction” refers to the fraction extracted from SSD which is active as entry inhibitor and/or has anti-virus activity.
- one or more active fractions can be obtained by the above method.
- the active fraction (s) can include concentrated proanthocyanidins than SSP.
- the active fraction (s) exhibits entry inhibitory, anti-virus, immunoregulation and/or anti-inflammatory effects as SSP.
- the active fraction (s) can have different mean degrees of polymerization (mDP) .
- the active fraction (s) can have higher mDP than SSP.
- one active fraction can have a mDP of 7 ⁇ 10.
- Each of the steps (i) - (iv) can have any of the features as described above in the method of obtaining an extract of SSD.
- the series of solvent can further comprise water, ethanol, petroleum ether (PE) , and/or ethyl acetate.
- the series solvent is petroleum ether (PE) , ethyl acetate and n-butanol.
- the parts of SSP corresponding to each solvent comprise petroleum ether part (SSP-PE) , ethyl acetate part (SSP-EA) , ethyl acetate insoluble part (SSP-EAin) , and n-butanol part (SSP-n-BuOH) .
- suitable separating process can comprise macroporous resin columns separation, fractional precipitation, crystallization, normal phase chromatography, reverse phase chromatography, and/or ion exchange resin columns separation.
- the separating process is macroporous resin columns separation.
- suitable eluents can comprise water, and/or an organic solvent.
- the eluents in (vii) are gradient ethanol-water from 10%to 95%.
- the method of obtaining an active fraction from SSD can further comprise: (viii) concentrating the active fraction; and (ix) drying the concentrated active fraction.
- the concentrating process in step (viii) can have any of the features of the corresponding process in the above step (iii) .
- the drying process in step (ix) can have any of the features of the corresponding process in the above step (iv) .
- the invention also relates to the active fraction obtained by the above method.
- the above active fraction from SSD related nature products from SSD may be useful in this disclosure.
- the prescript or formula similar to SSP can also have anti-viral function against COVID-19.
- the invention further relates to a composition comprising the SSP or the active fraction obtained by the above methods.
- the composition can further comprise auxiliary materials.
- the auxiliary materials can include, but are not limited to fillers, such as starch, pregelatinized starch, lactose, mannitol, chitin, microcrystalline cellulose, sucrose, etc. ; disintegration agents, such as, starch, pregelatinized starch, microcrystalline cellulose, sodium carboxymethyl starch, cross-linked polyvinylpyrrolidone, low-substituted hydroxypropyl cellulose, cross-linked polymethyl cellulose sodium, etc.
- composition can further comprise herbs other than SSD.
- SSP is an entry inhibitor for SARS-CoV-2 via direct binding to viral envelope and downregulating viral receptor ACE2 on target cell surface.
- the invention relates to a method of preventing and/or treating a viral disease comprising administering an effective amount of the SSP or the active fraction obtained by the above methods to a subject in need thereof.
- the effect dose of SSP can be ranging from 0.5 ⁇ g/mL to 5000 ⁇ g/mL.
- the invention also relates to use of SSP or the active fraction obtained by the above methods in the manufacture of preparations for prevention and/or treatment of a viral disease.
- the preparations can be in the form of injections, tablets, granules, emulsions, gels, sustained-release preparations, nasal wash/spray, oral liquids, throat spray, like-tea pills/candy, nano preparations, “throat lozenges” , “throat sprays” , “nasal washes” , and/or “nasal sprays” .
- the viral disease include that caused by coronaviruses, e.g., SARS-CoV-2 and SARS-CoV-1 and their variants, Ebola virus (EBOV) , HIV-1, H5N1 and other enveloped viruses except for VSV.
- coronaviruses e.g., SARS-CoV-2 and SARS-CoV-1 and their variants, Ebola virus (EBOV) , HIV-1, H5N1 and other enveloped viruses except for VSV.
- SSP Dried SS stems were purchased from KangMei Pharmaceutical Company Ltd (Guang Xizhou, China) and the plants were authenticated by inspectors in the School of Chinese Medicine, the University of Hong Kong, Hong Kong. Briefly, SS was cut into pieces and extracted using a percolating device with 10 times volume (v/w) of 60%ethanol with 12 hours at room temperature. The extract was then concentrated by reduced pressure under 50°C and freeze-dried by vacuum freeze dryer to obtain the percolation powder, named SSP ( Figure 1) . SSP can be dissolved in dimethyl sulfoxide (DMSO) to reach a concentration of 40 mg/mL for future use.
- DMSO dimethyl sulfoxide
- the quality control methods for SSP extract contains: 1) the contents of proanthocyanidins by vanillin-hydrochloric acid method; 2) content determination of references compounds by UPLC; 3) TLC identification.
- GHOST (3) -CD4-CCR5/CXCR4 cell lines were cultivated in DMEM with 10%FBS, 1%P/S, and 100 ⁇ g/mL hygromycin B, 500 ⁇ g/mL G418, and 1 ⁇ g/mL puromycin (Sigma-Aldrich, St.Louis, MO, USA) .
- pseudoviruses were generated by co-transfection of 293T cells with HIV-1 NL4-3 ⁇ Env Vpr Luciferase (pNL4-3. Luc. R-E-) or HIV-1 NL4-3 ⁇ Env EGFP Reporter Vector (NIH AIDS Reagent Program, Cat #3418 and 11100) and envelope protein from different strains of viruses (polyethyleneimine [PEI] ; Polysciences Inc., Warrington, PA) . Cell-free supernatant was collected 48h post-transfection and frozen at -80 °C. To determine the viral titer, around 10,000 HEK293T-ACE2 cells per well in 96-well plates were seeded in 10%FBS-containing media.
- TCID50 50%tissue culture infective dose
- the inhibitory activities of the SSP against viruses are evaluated as previously described (L. Liu et al., 2007; X. Lu et al., 2012) . Briefly, serially diluted SSP was tested against 100TCID50 viral infection. HEK 293FT-ACE2 were used for SARS-CoV-1/2; MDCK was used for H5N1; GHOST (3) -CD4-CCR5/CXCR4 were used for HIV ADA and HIV HXB2 infection. On day three, the viral infection was determined by measuring the reporter luciferase activity in target cells post-infection using commercially available kits (Promega, Wisconsin, USA) . Antiviral data are reported as the concentration of drugs required to inhibit viral replication by 50% (EC 50 ) .
- the different SARA-CoV-2 variants including Alpha (B. 1.1.7) , Beta (B. 1.351) , Gamma (P1) , Delta (B.1.617.2) , and Omicron (BA. 1, BA. 2, BA2.1.2.1 and BA. 4/5) were also used to test the inhibition rate of SSP.
- the anti-viral activity of grape seed extract (GSE) and tea polyphenol (TP, CAS: 84650-60-2) (Shanghai Yuanye Company) were also tested by the same method.
- Acute toxicity experiment observe and study the toxicity of SD rats and NIH mice given single or multiple times by intragastric administration of SSP within 24 hours to determine the acute toxic dose and the maximum tolerated dose and possible toxicity for target organs for its clinical safety.
- Method Several SD rats and NIH mice were selected, half male and female, and randomly divided into vehicle control group and SSP group. The rats were fasted for 12h-16h before administration and weighed.
- the SSP group was given SSP powder 0.25g /mL was given to rats by 20mL /kg, and the vehicle control group was given an equal volume of purified water, orally or twice a day (each dosing interval 8h) , continuous observation for 4 hours after each administration , D1 ⁇ D14 were observed once a day in the morning and afternoon; D1, D4, D7, D14 weighed the rats and their situations were also recorded. After animals sacrifice and visual observation, any changes in volume, color, and texture of any tissues and organs were recorded and histopathological checked.
- Nasal spray irritation test-Rabbits This experiment was completed in the CFDA-certified GLP-compliant Drug Safety Evaluation Research Center (Shandong Xinbo Drug Research Co., Ltd. ) , and the code of this experiment was KY22233.
- the stability of SSP powder is determined by vanillin-hydrochloric acid method as mentioned in 3-1) to detect the content of proanthocyanidins, and the detection samples are the SSP aqueous solution prepared immediately, and the SSP aqueous solution after keeping at room temperature for over 1 month.
- Efficacy of SSP against live intranasal SARS-CoV-2 Omicron BA. 2 challenge were tested in K18-hACE2 mice. The units of 2*10 ⁇ 4 moi of viruses were administered to the nasal cavity of the mice. As shown in Figure 10a, one dose of SSP intervention (40 mg/ml *20 ⁇ L, 800 ⁇ g) was given at the time of 30 minutes before virus infection, and the control group was saline group (n 5 per group) . After 3 days of virus infection, the lung tissues of the mice were collected.
- SSP SARS-CoV-2 pseudoviruses were generated (L. L. Liu et al., 2019; X. ; ) .
- HE293T-ACE2 cells were then infected with 100 TCID50 SARS-CoV-2 pseudoviruses in the presence of serially diluted SSP.
- Virus pseudotyped with a second, unrelated viral envelope glycoprotein of VSV-G was included as a control to reduce the false positives.
- SSP displayed an EC 50 of 3.5 ⁇ g/mL in the inhibition of SARS-CoV-2 pseudovirus infection (Figure 3a) and was devoid of overt cytotoxicity (Figure 3c) .
- SARS-CoV-2 and VSV pseudoviruses share the common genetic HIV backbone expressing protease (PR) , reverse transcriptase (RT) , and integrase (IN) and differ only in their glycoproteins on the surface of the viruses.
- PR protease
- RT reverse transcriptase
- I integrase
- SSP antagonizes SARS-CoV-2 pseudovirus entry rather than post-entry events (e.g., reverse transcription) .
- the lack of antiviral activity against VSV also ruled out the possibility that SSP simply inactivates the SARS-CoV-2 virus by acting on viral lipids as a disinfectant.
- SSP acts by blocking the attachment of SARS-CoV spike envelope protein to entry receptor ACE2
- SARS-CoV-2 enter cells in the host through a virus surface-anchored S-protein.
- the S protein mediates viral entry through the RBD in the S1 subunit that specifically recognizes ACE2 as its receptor and then fuses the viral into host membranes through the S2 subunit.
- Entry inhibitors usually target protein-protein interactions within the viral envelope proteins or between viral envelope proteins and host cells or inhibit protein-lipid interactions.
- the viruses and HEK-293T-ACE2 cells were then recovered and subsequently subjected to infect HEK293T-ACE2 or incubated with untreated SARS-CoV-2 pseudovirus (Rapista et al., 2011) .
- HEK293T-ACE2 cells were infected with SARS-CoV-2 pseudovirus and then treated with gradient diluted SSP immediately or 2-hours post-infection (hpi) .
- SSP treatment at 2-hours post-infection showed limited anti-SARS-CoV-2 activity compared with the virus and SSP being added simultaneously. This result confirmed the antiviral entry activity of SSP against SARS-CoV-2.
- pre-treatment of pseudovirus with SSP resulted in increased inhibition activities with the EC 50 value of 2.3 ⁇ g/mL, suggesting that SSP targets SARS-CoV-2 viral envelop S protein to inhibit SARS-CoV-2 entry.
- pre-treatment of target cells with SSP halted SARS-CoV-2 infection with the EC 50 value of 2.1 ⁇ g/mL, suggesting that SSP also targets cellular components to inhibit SARS-CoV-2 entry.
- HEK-293T-ACE2 cells were treated with serially diluted SSP for 2 hours at 37°C, washed, and were then incubated with supernatant collected from RBD-PD1 expressing plasmid transfected HEK293T cells or a goat polyclonal antibody against ACE2 for 30 min on ice. Cells were then washed and stained with a fluorescent-labeled secondary antibody against goat-IgG or PD-1, respectively.
- SSP shows broad-spectrum antiviral activities against SARS-CoV, H5N1, EBOV and HIV-1
- SSP broad-spectrum antiviral activities
- a panel of pseudoviruses was generated in our previous studies, including SARS-CoV (L. Liu et al., 2019) , H5N1 Turkey (Xiao et al., 2013) , CCR5-tropic HIV-1 ADA , and CXCR4-tropic HIV-1 HXB2 (Liang et al., 2013) , were utilized to test the inhibition rate of SSP.
- SSP inhibited their infection with EC 50 around 3.64, 5.13, 3.61, and 8.15 ⁇ g/mL, respectively, and was devoid of overt cytotoxicity (Figure 4e) .
- SSP blocks SARS-CoV-1 and H5N1 entry by binding to viral envelope and its target cells.
- SSP-virus binding and SSP-cell binding assays were performed.
- pseudovirus was initially pre-treated with 50 ⁇ g/mL of SSP. The viruses were then recovered by ultracentrifugation and subjected to infect target cells. We found that SSP inhibited SARS-CoV-1 pseudovirus infection at a similar degree as when the virus and SSP were added into cells simultaneously ( Figure 5) .
- the SSP-cell binding assays were performed by pre-treatment of the target cells with SSP for 1 hour at 37°C. After washing by PBS, cells were incubated with SARS-CoV-1 at 37 °C for 48 hrs (Rapista et al., 2011) .
- pre-treatment of SSP on the target cells inhibited virus infection ( Figure 5) .
- the same approach was also used to test the inhibitory effect of SSP against H5N1 with a single high dose of 50 ⁇ g/mL. Similar to SARS-CoV-1, SSP treatment with virus particles or their target cells efficiently blocked H5N1 virus entry (Figure 5b) .
- SSP blocks HIV-1 entry by binding to the viral envelope.
- AZT but not SSP, significantly inhibited HIV-1 infection when the treatment was initiated at 2 hours post-infection.
- pre-treatment of the virus with 50 ⁇ g/mL of SSP significantly inhibited HIV-1 infection at a similar degree as T-20, which blocks HIV-1 fusion (Figure 5c ⁇ 5d) .
- the SSP-cell binding assays were performed by treating the target GHOST cells with SSP or with control compounds the CCR5 antagonist Marvaroc (MVC) or the CXCR4 antagonist JM2987 for 2 hours at 37°C. After washing by PBS, cells were subjected to HIV-1 ADA or HIV-1 HXB2 infection and cultivated at 37 °C for 48 hrs (Rapista et al., 2011) .
- MVC and JM2987 showed potent viral inhibition against the respective ADA and HXB2 pseudoviruses at 1 ⁇ M as expected ( Figures 5e ⁇ 5f) .
- This evidence demonstrated that SSP inhibited HIV-1 infection by actions on the viral envelope glycoprotein gp160, which mediates the viral entry into the host target cells.
- Fr. G inhibited viral entry with lower EC 50 value than adding the virus and fragments simultaneously, suggesting that those fragments target both SARS-CoV-2 viral envelop S protein and target cells to inhibit SARS-CoV-2 entry. More importantly, Fr. G exhibited more outstanding bioactivity when compared to Fr. B, suggesting that Part II of SSP contains more effective antiviral activities than Part I. These results revealed that Fr. G contains an intermediate polymer of proanthocyanidins, which may have better antiviral effects.
- the maximum tolerated dose for one-time administration is 67 g crude drug /kg in mice (Table 1) .
- the maximum tolerated dose for one-time administration is 107 g crude drug /kg in rats (Table 2) .
- the proanthocyanidins contained in the immediately prepared SSP aqueous solution were 794.794 ⁇ 13.619 mg/g, while the content in the SSP aqueous solution after being placed for 1 month was 778.964 ⁇ 63.589mg/g, which was within the quality standard range of 5%, so the SSP aqueous solution is stable in terms of proanthocyanidin content.
Landscapes
- Health & Medical Sciences (AREA)
- Natural Medicines & Medicinal Plants (AREA)
- Life Sciences & Earth Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical & Material Sciences (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Medicinal Chemistry (AREA)
- Biotechnology (AREA)
- Communicable Diseases (AREA)
- Microbiology (AREA)
- Epidemiology (AREA)
- Medical Informatics (AREA)
- Botany (AREA)
- Alternative & Traditional Medicine (AREA)
- Engineering & Computer Science (AREA)
- Virology (AREA)
- Mycology (AREA)
- Oncology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Medicines Containing Plant Substances (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Disclosed is a method of obtaining preparing an extract of Spatholobus suberectus Dunn, which contains total flavonoid and proanthocyanidins. The disclosure relates to a method for extracting an active fraction from Spatholobus suberectus. Also provided is the SSP, the active fraction, and a composition thereof. Further provided is a method of treating viral diseases such as COVID-19 disease caused by SARS-CoV-2, and use of the SSP and the active fraction in the manufacture of preparations for prevention and/or treatment of a viral disease.
Description
Disclosed is a method of preparing a natural product of antiviral extract from Spatholobus suberectus Dunn (SSD) , which contains total flavonoid and proanthocyanidins. The disclosure relates to a method for discovering an active proanthocyanidins of Spatholobus suberectus (SSP) as viral entry inhibitor. Also provided is a method of preventing and treating viral diseases such as coronavirus disease 2019 (COVID-19) .
Recent epidemic outbreaks caused by emerging and re-emerging viruses in the advent of globalization and rapid urbanization have underscored their prevention and treatment as a critical threat to public health. Examples include the recent outbreak of COVID-19 caused by severe acute respiratory syndrome virus type 2 (SARS-CoV-2) . Despite the progress made in immunization and drug development, many viruses lack preventive vaccines and efficient antiviral therapies. Thus, identifying novel agents especially natural products that are highly efficacious and cost-effective for the management and control of viral infections is of critical importance. Broad-spectrum antivirals will be particularly beneficial to combat new variants of related viruses.
Herbal medicines and purified natural products provide a rich resource for novel antiviral drug development. Identification of the antiviral mechanisms from these natural agents has shed light on where they interact with the viral life cycle, such as viral entry, replication, assembly, and release. Traditional Chinese Medicine (TCM) is particularly appreciated. With the existence of between 250,000 to 300,000 plant species in the world, Chinese herbal medicine provides a fast track and important source for drug discovery 6-10. Artemisia annua is the representative herb. The discovery of Qinghaosu (artemisinin) , an antimalarial drug from Artemisia annual, and subsequent development of several therapies to inhibit the malaria parasite has saved millions of lives.
The plant SSD. Slightly sweet, warm in nature, belongs to Chinese medicine liver and kidney. "Compendium of Materia Medica" claims that it can nourish the stomach and dry stomach; "Compendium of Compendium of Materia Medica" claims that it can promote blood circulation and warm the waist and knees. There is a large medical demand for the treatment of patients with closedness, rheumatism, numbness, paralysis, blood deficiency and chlorosis. In recent years, there have been some reports on the common use of Centipede to treat diseases caused by viruses, but it has not been reported in Covid-19. Till now, no publication has indicated SSP as a viral entry inhibitor.
The chemical constituents of SSD mainly include flavonoids, terpenes, sterols, lignin, anthraquinones, polyphenols, proanthocyanidins and some trace elements, among which the content of flavonoids is the most. The research on the extraction method of content mostly uses the total flavonoid extraction rate as an indicator and does not consider the effect of the extraction method on the drug efficacy of the extract.
Provided herein is a method of obtaining an extract of SSD, said method comprising: (i) cutting SSD into pieces; (ii) immersing and treating SSD pieces in a solvent at room temperature to obtain an extract; (iii) concentrating the extract; and (iv) drying the concentrated extract to obtain the powder of the extract of SSD (SSP) .
Provided herein is a method of obtaining an active fraction from SSD, the active fraction including proanthocyanidins, said method comprising: (i) cutting SSD into pieces; (ii) immersing and treating SSD pieces in a solvent at room temperature to obtain an extract; (iii) concentrating the extract; (iv) drying the concentrated extract to obtain the powder of the extract of SSD (SSP) ; (v) dispersing SSP in water to obtain an aqueous solution; (vi) extracting the SSP aqueous solution with a series of solvent with different polarities at equal volume to obtain parts of SSP corresponding to each solvent, the series of solvent comprising n-butanol; and (vii) separating the n-butanol part (SSP-n-BuOH) with eluents to obtain the active fraction.
In one embodiment, the treating process in (ii) comprises percolation.
Provided herein are an extract of SSD and an active fraction from SSD obtained by the above methods, as well as a composition comprising the extract or the active fraction.
Provided herein is a method of preventing and/or treating a viral disease comprising administering an effective amount of an extract of SSD or an active fraction from SSD obtained by the above methods to a subject in need thereof.
In one embodiment, the viral disease is caused by SARS-CoV-2 including Omicron variants, Ebola virus (EBOV) , HIV-1 with either CCR5 or CXCR4 tropism, SARS-CoV, H5N1.
In one embodiment, the effective dose is 0.5 μg/ml to 5000 μg/ml.
Provided herein is use of an extract of SSD or an active fraction from SSD obtained by the above methods in the manufacture of preparations for prevention and/or treatment of a viral disease.
Figure 1. Flow chart of the study. Preparation and quality-controlled of SSP which is used to determine the broad-spectrum antiviral activity and its underlying mechanism. The powder SSP was further extracted to obtain the effective active parts.
Figure 2. Representative pictures of SSP's quality control. (a) HPLC chromatogram of different batches of SSP after ethyl acetate processing. (b) TLC chromatogram of different batches of SSP and positive control of SS.
Figure 3. Antiviral activity of SSP against SARS-CoV-2. Serially diluted SSP was added to HEK293T-ACE2 cells infected with SARS-CoV-2 (a) and VSV (b) respectively. The luciferase level was measured 2 or 3 days post-infection. To test SSP cytotoxicity, cells viability (c) was measured. (d) Pre-treatment of SARS-CoV-2 and target cells inhibited viral infection. (e) Binding of RBD to ACE2 expressing 293T cells, but not 293T control cells. (f) SSP inhibited RBD binding to target cells. SSP pre-treated HEK293T-ACE2 cells were incubated with RBD-PD1 for 30 mins on ice, followed by antibody staining of ACE2 (upper panel) and RBD (lower panel) and flow cytometry analysis. The data represent the mean ± SEM of triplicate experiments.
Figure 4. Antiviral activity of SSP against SARS-CoV-1, H5N1, and HIV, and EBOV viruses. Serially diluted SSP was added to HEK293T-ACE2, MDCK, GHOST-CCR5, and GHOST-CXCR4 infected with SARS-CoV-1 (a) H5N1 (b) HIVADA (c) , and HIVHXB2 (d) respectively. The luciferase level was measured 2 or 3 days post-infection. Cells viability (e) was measured to test SSP cytotoxicity. (f) Serially diluted SSP was added to 293T cells infected with EBOV. The luciferase level was measured 2 days post infection. Cells viability was measured to test SSP cytotoxicity against the 293T cells. The data represent the mean ± SEM of triplicate experiments.
Figure 5. Mechanisms of SSP mediated virus entry inhibition. (a-b) . Pre-treatment of SARS-CoV-1 (a) , H5N1 (b) and target cells inhibited viral infection. SARS-CoV-1 or H5N1 pseudovirus and HEK293T-ACE2 (a) or MDCK (b) cells pre-treated with serial diluted SSP were recovered and subsequently subjected to infect target cells, or incubated with untreated SARS-CoV-1 pseudovirus for 48 hours, respectively. The luciferase level was measured 2 or 3 days post-infection. (c-d) . Post-entry assay. GHOST-CD4-CCR5 or CXCR4 cells were co-incubated with pseudovirus for 2 hrs, washed, and then treated with the presence of 50 mg/ml SSP and 1 mM AZT as a positive control for 48 hr. SSP does not inhibit either HIV-1ADA or HIVHxB2 virus gene replication after the viral entry is achieved. (e-f) . SSP-virus interaction assay. HIV-1ADA and HIVHxB2 pseudovirus pre-treated with 50 mg/ml SSP and entry inhibitor enfuvirtide (T-20) as a positive control. SSP pre-treatment inhibited both HIVADA and HIVHxB2 pseudovirus infection to a similar degree as T-20. (g-h) SSP-cell binding assay. GHOST cells were pretreated with SSP and the CCR5 antagonist maraviroc and the CXCR4 antagonist JM2987 as positive controls for 1 hr at 37℃ prior to being infected. SSP pretreatment with the cells had no antiviral effect, whereas maraviroc and JM2987 pre-treatment showed strong inhibition, as expected. Dimethyl sulfoxide (DMSO) was used as the negative control. The data represent the mean ± SD of triplicate experiments.
Figure 6. Isolation of SSP and UPLC-MS analysis. (a) UPLC-DAD spectra of SSP; (b) UPLC spectra of Fr. B (Part I) ; (c) UPLC spectra of Fr. G (Part II) ; (d) Extracted ion chromatogram (EIC) of monomer and dimer of the proanthocyanidins in SSP; (e) Extracted ion chromatogram (EIC) of polymers with mDP between 3 and 7 of the proanthocyanidins in SSP; (f) Extracted ion chromatogram (EIC) of polymers with mDP between 8 and 10 of the proanthocyanidins in SSP.
Figure 7. Antiviral activity of SSP and their extracted parts against SARS-CoVs, and VSV. As shown in a~d, serially diluted SSP, SSP-n-BuOH, Fr. B, F, and G were added to HEK293T-ACE2 cells infected with SARS-CoV-2 (a) , SARS-CoV (b) , and VSV (c) . The luciferase level was measured 2 or 3 days post-infection. To test their cytotoxicity, cells viability (d) was also measured. (e-h) Pre-treatment of SARS-CoV-2 and target cells inhibited viral infection. SARS-CoV-2 pseudovirus and HEK293T-ACE2 pre-treated with serial diluted SSP-n-BuOH (e) , Fr. B (f) , F (g) , and G (h) were recovered and subsequently subjected to infect HEK293T-ACE2, or incubated with untreated SARS-CoV-2 pseudovirus for 48 hr, respectively. As controls, HEK293T-ACE2 cells were infected with SARS-CoV-2 pseudovirus and treated with gradient diluted SSP simultaneously. The data represent the mean ± SEM of triplicate experiments.
Figure 8. SSP did not show significant cytotoxicity in multiple cell lines, or long-term in vivo toxicity in rats. (a~ d) To test SSP cytotoxicity, cells viability was measured in HEK293T-ACE2 (a) , MDCK (b) , GHOST (3) -CD4-CXCR4 (c) /CCR5 (d) . (e) Effects of SSP administration for 4 weeks on the bodyweight of SD rats. (f) Effects of SSP administration for 4 weeks on food intake of SD rats. (g) Effect of SSP administration for 4 weeks on the organ coefficient of SD rats.
Figure 9. SSP shows higher potency and broader spectrum against SARS-CoV-2 variants entry in comparison with GSE and TP. (A) After keeping SSP solution for 6 and 13 days, the anti-viral activity and cyto-toxicity were tested. (B) SSP had a fairly strong inhibitory effect against major SARS-CoV-2 variants [including Alpha (B. 1.1.7) , Beta (B. 1.351) , Gamma (P1) , Delta (B. 1.617.2) , and Omicron (BA. 1, BA. 2, BA2.1.2.1 and BA. 4/5) ] . (C~D) The anti-viral efficacy of grape seed extract (GSE) and tea polyphenol (TP) against SARS-CoV-2 and Omicron variants, as well as their cyto-toxicity.
Figure 10. SSP shows efficacy in protection against nasal challenge of SARS_CoV-2 omicron in mice. (A) Flowchart of the in vivo experiments. (B) Omicron BA. 2 virus-infected cells (green) and inflammatory cells (red) were significantly reduced in the lung tissues of mice treated with SSP compared with the saline group. (C) In the lung tissue of mice treated with SSP, the expression level of RNA-dependent RNA polymerase and the virus titer were significantly lower than those of mice treated with saline. *P<0.05.
The present invention is related to a method of obtaining an extract of SSD, said method comprising: (i) cutting SSD into pieces; (ii) immersing and treating SSD pieces in a solvent at room temperature to obtain an extract; (iii) concentrating the extract; and (iv) drying the concentrated extract to obtain the powder of the extract of SSD, the extract of SSD including proanthocyanidins, named SSP.
As used herein, the term “SSD” refers to Spatholobus suberectus Dunn. As used herein, the term “SSP” refers to the extract of SSD including proanthocyanidins. Herein, SSP can be obtained by the above method comprising steps (i) to (iv) .
In step (ii) , suitable treating process can comprise percolation, ultrasonic, heating and refluxing, decoction, and/or solvent extraction. Suitably, the treating process in (ii) is percolation.
In step (ii) , the solvent used can comprise water, and/or an organic solvent. In one embodiment, the solvent in (ii) comprises water and ethanol, such as 60%ethanol/water (v/v) .
For example, step (ii) comprises treating SSD pieces via a percolation process with a 60%ethanol (in water, v/v) at room temperature. The percolation process can be performed for 12 hours.
In step (iii) , the concentrating process can comprise reduced pressure rotary evaporation, centrifugation, and/or nitrogen blowing. In one embodiment, the concentrating process in (iii) is reduced pressure rotary evaporation. The concentrating process can be performed at a temperature under 50℃.
In step (iv) , the drying process can comprise freeze drying, spray drying, microwave drying, and/or infrared heating drying. In one embodiment, the drying process in (iv) is freeze drying.
The invention also relates to the SSP obtained by the above method. SSP exhibited significant inhibitory ability against SARS-CoV-2 IC50 values of 3.574 μg/mL and 3.648 μg/mL. Innovatively, SSP uniquely inhibited the entry of SARS-CoV-2, HIV-1, Ebola virus (EBOV) , SARS-CoV-1 and influeunza H5N1 infections but failed to block vesicular stomatitis virus (VSV) . Furthermore, we showed that SSP had no effects on post-entry events of replication. It blocked SARS-CoV-2, HIV-1, H5N1, EBOV and SARS-CoV-1 entry by acting on viral envelope directly. Our findings suggested SSP is a novel entry inhibitor, which has a potential for preventing and treating COVID-19, also can fight EBOV, HIV-1, H5N1 and SARS-CoV infection, and stockpiling for possible use against future pandemic caused by related virus.
Our findings provide scientific evidence for the potential use of SSP for preventing &treating COVID-19 (SARS-CoV-2) infection, and stockpiling for possible use against future pandemic caused by related virus, as well as for the search of ingredient small molecular compounds as entry inhibitor.
Further, the invention is related to a method of obtaining an active fraction from SSD, the active fractions including proanthocyanidins, said method comprising: (i) cutting SSD into pieces; (ii) immersing and treating SSD pieces in a solvent at room temperature to obtain an extract; (iii) concentrating the extract; (iv) drying the concentrated extract to obtain the powder of the extract of SSD (SSP) ; (v) dispersing SSP in water to obtain an aqueous solution; (vi) extracting the SSP aqueous solution with a series of solvent with different polarities at equal volume to obtain parts of SSP corresponding to each solvent, the series of solvent comprising n-butanol; and (vii) separating the n-butanol part (SSP-n-BuOH) with eluents to obtain the active fraction.
As used herein, the term “active fraction” refers to the fraction extracted from SSD which is active as entry inhibitor and/or has anti-virus activity. Herein, one or more active fractions can be obtained by the above method. The active fraction (s) can include concentrated proanthocyanidins than SSP. Thus, the active fraction (s) exhibits entry inhibitory, anti-virus, immunoregulation and/or anti-inflammatory effects as SSP.
The active fraction (s) can have different mean degrees of polymerization (mDP) . The active fraction (s) can have higher mDP than SSP. For example, one active fraction can have a mDP of 7~10.
Each of the steps (i) - (iv) can have any of the features as described above in the method of obtaining an extract of SSD.
In step (vi) , the series of solvent can further comprise water, ethanol, petroleum ether (PE) , and/or ethyl acetate. In one embodiment, the series solvent is petroleum ether (PE) , ethyl acetate and n-butanol. In this case, the parts of SSP corresponding to each solvent comprise petroleum ether part (SSP-PE) , ethyl acetate part (SSP-EA) , ethyl acetate insoluble part (SSP-EAin) , and n-butanol part (SSP-n-BuOH) .
In step (vii) , suitable separating process can comprise macroporous resin columns separation, fractional precipitation, crystallization, normal phase chromatography, reverse phase chromatography, and/or ion exchange resin columns separation. In one embodiment, the separating process is macroporous resin columns separation.
In step (vii) , suitable eluents can comprise water, and/or an organic solvent. In one embodiment, the eluents in (vii) are gradient ethanol-water from 10%to 95%.
The method of obtaining an active fraction from SSD can further comprise: (viii) concentrating the active fraction; and (ix) drying the concentrated active fraction. The concentrating process in step (viii) can have any of the features of the corresponding process in the above step (iii) . The drying process in step (ix) can have any of the features of the corresponding process in the above step (iv) .
The invention also relates to the active fraction obtained by the above method.
Other than the above SSP extract, the above active fraction from SSD, related nature products from SSD may be useful in this disclosure. Meanwhile, the prescript or formula similar to SSP can also have anti-viral function against COVID-19.
The invention further relates to a composition comprising the SSP or the active fraction obtained by the above methods. The composition can further comprise auxiliary materials. The auxiliary materials can include, but are not limited to fillers, such as starch, pregelatinized starch, lactose, mannitol, chitin, microcrystalline cellulose, sucrose, etc. ; disintegration agents, such as, starch, pregelatinized starch, microcrystalline cellulose, sodium carboxymethyl starch, cross-linked polyvinylpyrrolidone, low-substituted hydroxypropyl cellulose, cross-linked polymethyl cellulose sodium, etc. ; additives, such as, stearic acid magnesium, sodium lauryl sulfate, talc, and/or silica suspending agents, binders, etc. ; suspending agents, such as, polyvinylpyrrolidone, microcrystalline cellulose, sucrose, agar, hydroxypropyl methylcellulose, etc. ; binders, such as, starch Pulp, polyvinylpyrrolidone, etc. The composition can further comprise herbs other than SSD.
Disclosed herein is that SSP is an entry inhibitor for SARS-CoV-2 via direct binding to viral envelope and downregulating viral receptor ACE2 on target cell surface. The invention relates to a method of preventing and/or treating a viral disease comprising administering an effective amount of the SSP or the active fraction obtained by the above methods to a subject in need thereof. Suitably, the effect dose of SSP can be ranging from 0.5 μg/mL to 5000 μg/mL.
The invention also relates to use of SSP or the active fraction obtained by the above methods in the manufacture of preparations for prevention and/or treatment of a viral disease. The preparations can be in the form of injections, tablets, granules, emulsions, gels, sustained-release preparations, nasal wash/spray, oral liquids, throat spray, like-tea pills/candy, nano preparations, “throat lozenges” , “throat sprays” , “nasal washes” , and/or “nasal sprays” .
The viral disease include that caused by coronaviruses, e.g., SARS-CoV-2 and SARS-CoV-1 and their variants, Ebola virus (EBOV) , HIV-1, H5N1 and other enveloped viruses except for VSV.
The following Examples further illustrate the findings of the present disclosure.
EXAMPLES
The following examples are intended to illustrate the present invention, but are not intended to limit the scope of the present invention.
Materials and methods
1. SSP Extract of SSD
Dried SS stems were purchased from KangMei Pharmaceutical Company Ltd (Guang Xi Province, China) and the plants were authenticated by inspectors in the School of Chinese Medicine, the University of Hong Kong, Hong Kong. Briefly, SS was cut into pieces and extracted using a percolating device with 10 times volume (v/w) of 60%ethanol with 12 hours at room temperature. The extract was then concentrated by reduced pressure under 50℃ and freeze-dried by vacuum freeze dryer to obtain the percolation powder, named SSP (Figure 1) . SSP can be dissolved in dimethyl sulfoxide (DMSO) to reach a concentration of 40 mg/mL for future use.
2. Active fractions extraction
About 200 g SSP extract was weighed and dispersed evenly in an appropriate amount of water to obtain the aqueous solution, followed by extraction with equal volumes of petroleum ether (PE) , ethyl acetate (EA) , and n-butanol (n-BuOH) to obtain the following parts: 2 g petroleum ether part (SSP-PE) , 21 g ethyl acetate part (SSP-EA) , 8.2 g EA insoluble part (SSP-EAin) , 62 g n-butanol part (SSP-n-BuOH) and 113 g water layer residue (SSP-W) . Then the n-butanol part (SSP-n-BuOH) was subjected to macroporous resin columns for gradient elution ethanol-water to obtain fraction A~J (Figure 1) .
3. Quality control
The quality control methods for SSP extract contains: 1) the contents of proanthocyanidins by vanillin-hydrochloric acid method; 2) content determination of references compounds by UPLC; 3) TLC identification.
1) The contents of PACs (proanthocyanidins) in SSP were determined by the vanillin-hydrochloric acid method with minor modification (Y. CHENG et al., 2011) . 2) UHPLC of SSP was performed by Ultimate 3000 system (Thermo Fisher Scientific Inc., Waltham, Massachusetts, U.S. ) with a diode array detector (DAD) . An ACE Excel 2 C18 column (100 mm × 2.1 mm id, Scotland, UK) was used for the chromatographic separation, and the mobile phase was composed of acetonitrile (solvent A) and Milli-Q water with 0.1%formic acid (solvent B) , using a gradient elution of 0 min 95%B, 3 min 92%B, 20 min 85%B, 26 min 70%B, 30 min, 40%B, 35 min 5%B and keep at 5%B for five more minutes. The solvent flow rate was set as 0.4 mL/min, and the wavelength for analysis was conducted at 280 nm (Figure 2a) . This method was validated. Flavonoids, and proanthocyanidins were present in the SSP extract UPLC spectrum. 3) Identification of SSP by thin-layer chromatography (TLC) according to the Chinese Pharmacopoeia (version of the Year 2015) . Based on the color spot on the chromatogram, the compounds of SSP were identified according to the corresponding position of the control (Figure 2b) .
4. Cell Culture
Highly sensitive and transferable cell lines, 293T, TZM-bl, and MDCK, which were obtained from ATCC company (American Type Culture Collection) and passage numbers were within 10 times, were cultivated in Dulbecco’s modified Eagle medium (DMEM, Gibco, Grand Island, NY, USA) with 10%inactivated fetal bovine serum (FBS, Gibco, Grand Island, NY, USA) , 1%mixture of penicillin and streptomycin (P/S, Sigma-Aldrich, St. Louis, MO, USA) . HEK 293FT-ACE2 was cultivated in DMEM with 10%FBS, 1%P/S, and 1 μg/mL puromycin (Sigma-Aldrich, St. Louis, MO, USA) . GHOST (3) -CD4-CCR5/CXCR4 cell lines were cultivated in DMEM with 10%FBS, 1%P/S, and 100 μg/mL hygromycin B, 500 μg/mL G418, and 1 μg/mL puromycin (Sigma-Aldrich, St.Louis, MO, USA) .
5. Pseudovirus generation
We inserted an optimized full-length S gene of SARS-CoV-2 (QHR63250) into the pVAX-1 vector, namely pVax-1-S-COVID19. The construction was confirmed by sequence analysis. Single-cycle luciferase SARS-CoV-2, SARS-CoV-1, HIVADA, HIVHXB2, H5N1, and Vesicular stomatitis Indiana virus (VSV) pseudoviruses were constructed as previously described (L. Liu et al., 2007; Yi, Ba, Zhang, Ho, &Chen, 2005) . Briefly, pseudoviruses were generated by co-transfection of 293T cells with HIV-1 NL4-3 ΔEnv Vpr Luciferase (pNL4-3. Luc. R-E-) or HIV-1 NL4-3 ΔEnv EGFP Reporter Vector (NIH AIDS Reagent Program, Cat #3418 and 11100) and envelope protein from different strains of viruses (polyethyleneimine [PEI] ; Polysciences Inc., Warrington, PA) . Cell-free supernatant was collected 48h post-transfection and frozen at -80 ℃. To determine the viral titer, around 10,000 HEK293T-ACE2 cells per well in 96-well plates were seeded in 10%FBS-containing media. Cells were infected with serially diluted SARS-CoV-2 pseudovirus in a final volume of 200 μL. After 48 hours, infected cells were lysed to measure luciferase activity using a commercial kit (Promega, E1500, Madison, WI) . The 50%tissue culture infective dose (TCID50) was calculated using the "TCID" macro as described previously (Richards &Clapham, 2006) .
6. Cytotoxicity assays
Cells were incubated in the presence or absence of serially diluted SSP at 37 ℃ in 5%CO2 for 48 hours. The viability of the cells was then measured using theLuminescent Cell Viability Assay kit (Promega, Wisconsin, USA) . The cytotoxicity was generated according to the sample concentrations and luminescent values. The result was analyzed based on the formula:
7. Antiviral assays
The inhibitory activities of the SSP against viruses are evaluated as previously described (L. Liu et al., 2007; X. Lu et al., 2012) . Briefly, serially diluted SSP was tested against 100TCID50 viral infection. HEK 293FT-ACE2 were used for SARS-CoV-1/2; MDCK was used for H5N1; GHOST (3) -CD4-CCR5/CXCR4 were used for HIVADA and HIVHXB2 infection. On day three, the viral infection was determined by measuring the reporter luciferase activity in target cells post-infection using commercially available kits (Promega, Wisconsin, USA) . Antiviral data are reported as the concentration of drugs required to inhibit viral replication by 50% (EC50) . Similarly, the different SARA-CoV-2 variants including Alpha (B. 1.1.7) , Beta (B. 1.351) , Gamma (P1) , Delta (B.1.617.2) , and Omicron (BA. 1, BA. 2, BA2.1.2.1 and BA. 4/5) were also used to test the inhibition rate of SSP. The anti-viral activity of grape seed extract (GSE) and tea polyphenol (TP, CAS: 84650-60-2) (Shanghai Yuanye Company) were also tested by the same method.
8. Safety evaluation
Model: Acute Toxicity-Mice and rats; Long-term Toxicity-Rats; Nasal spray irritation test-Rabbits
Acute toxicity experiment: observe and study the toxicity of SD rats and NIH mice given single or multiple times by intragastric administration of SSP within 24 hours to determine the acute toxic dose and the maximum tolerated dose and possible toxicity for target organs for its clinical safety. Method: Several SD rats and NIH mice were selected, half male and female, and randomly divided into vehicle control group and SSP group. The rats were fasted for 12h-16h before administration and weighed. The SSP group was given SSP powder 0.25g /mL was given to rats by 20mL /kg, and the vehicle control group was given an equal volume of purified water, orally or twice a day (each dosing interval 8h) , continuous observation for 4 hours after each administration , D1 ~ D14 were observed once a day in the morning and afternoon; D1, D4, D7, D14 weighed the rats and their situations were also recorded. After animals sacrifice and visual observation, any changes in volume, color, and texture of any tissues and organs were recorded and histopathological checked.
Long-term toxicity: Twenty-four male and female Sprague-Dawley rats (150-180 g) were used to determine long time adverse effect of SSP. All experiments were approved by the Institutional guidelines of Laboratory Animal Care and Committee on the Use of Live Animals in Teaching and Research, South China Agricultural University, Guangzhou, China. The animals were randomly divided into four groups, with three males and three females per group. SSP was dissolved in water and administered via gavage once a day at doses of 2, 4, 6 g/kg, or water. Their body weights, general behavior, signs of toxicity, food intake, and mortality were recorded at an interval of every two-four days. After 25 days, the animals were sacrificed by excessive anesthesia. Gross necropsy, calculation of organ index (such as brain, heart, lung, liver, kidney, etc. ) , pathological examinations of the organs, blood collection, and analysis were performed.
Nasal spray irritation test-Rabbits: This experiment was completed in the CFDA-certified GLP-compliant Drug Safety Evaluation Research Center (Shandong Xinbo Drug Research Co., Ltd. ) , and the code of this experiment was KY22233.
9. Stability
The stability of SSP powder is determined by vanillin-hydrochloric acid method as mentioned in 3-1) to detect the content of proanthocyanidins, and the detection samples are the SSP aqueous solution prepared immediately, and the SSP aqueous solution after keeping at room temperature for over 1 month.
Besides, it was found that the anti-viral activity of the SSP aqueous solution after being placed for 6 and 13 days.
10. In vivo anti-viral experiments
Efficacy of SSP against live intranasal SARS-CoV-2 Omicron BA. 2 challenge were tested in K18-hACE2 mice. The units of 2*10^4 moi of viruses were administered to the nasal cavity of the mice. As shown in Figure 10a, one dose of SSP intervention (40 mg/ml *20 μL, 800 μg) was given at the time of 30 minutes before virus infection, and the control group was saline group (n = 5 per group) . After 3 days of virus infection, the lung tissues of the mice were collected.
Results
11. SSP inhibits SARS-CoV-2 entry
To determine the antiviral activity of SSP, SSP was weighed and dissolved in 40 mg/mL as described. As previously described, the SARS-CoV-2 pseudoviruses were generated (L. L. Liu et al., 2019; X. ; ) . To examine the antiviral activity of SSP against SARS-CoV-2, HE293T-ACE2 cells were then infected with 100 TCID50 SARS-CoV-2 pseudoviruses in the presence of serially diluted SSP. Virus pseudotyped with a second, unrelated viral envelope glycoprotein of VSV-G was included as a control to reduce the false positives. As shown in Figure 3, SSP displayed an EC50 of 3.5 μg/mL in the inhibition of SARS-CoV-2 pseudovirus infection (Figure 3a) and was devoid of overt cytotoxicity (Figure 3c) . No inhibition was observed when SSP was tested against VSV pseudovirus (Figure 3b) . Because SARS-CoV-2 and VSV pseudoviruses share the common genetic HIV backbone expressing protease (PR) , reverse transcriptase (RT) , and integrase (IN) and differ only in their glycoproteins on the surface of the viruses. This result suggested that SSP antagonizes SARS-CoV-2 pseudovirus entry rather than post-entry events (e.g., reverse transcription) . Moreover, the lack of antiviral activity against VSV also ruled out the possibility that SSP simply inactivates the SARS-CoV-2 virus by acting on viral lipids as a disinfectant.
12. SSP acts by blocking the attachment of SARS-CoV spike envelope protein to entry receptor ACE2
As known, SARS-CoV-2 enter cells in the host through a virus surface-anchored S-protein. The S protein mediates viral entry through the RBD in the S1 subunit that specifically recognizes ACE2 as its receptor and then fuses the viral into host membranes through the S2 subunit. Entry inhibitors usually target protein-protein interactions within the viral envelope proteins or between viral envelope proteins and host cells or inhibit protein-lipid interactions. To determine whether SSP act by targeting SARS-CoV-2 S protein or host cells, we pre-treated pseudovirus and HEK-293T-ACE2 cells with serially diluted SSP for 2 hours at 37 ℃. The viruses and HEK-293T-ACE2 cells were then recovered and subsequently subjected to infect HEK293T-ACE2 or incubated with untreated SARS-CoV-2 pseudovirus (Rapista et al., 2011) . As controls, HEK293T-ACE2 cells were infected with SARS-CoV-2 pseudovirus and then treated with gradient diluted SSP immediately or 2-hours post-infection (hpi) . As shown in Figure 3d, SSP treatment at 2-hours post-infection showed limited anti-SARS-CoV-2 activity compared with the virus and SSP being added simultaneously. This result confirmed the antiviral entry activity of SSP against SARS-CoV-2. Moreover, pre-treatment of pseudovirus with SSP resulted in increased inhibition activities with the EC50 value of 2.3 μg/mL, suggesting that SSP targets SARS-CoV-2 viral envelop S protein to inhibit SARS-CoV-2 entry. Notably, pre-treatment of target cells with SSP halted SARS-CoV-2 infection with the EC50 value of 2.1 μg/mL, suggesting that SSP also targets cellular components to inhibit SARS-CoV-2 entry.
To determine whether SSP targets ACE2 receptor to block virus entry, RBD binding assay was performed. HEK-293T-ACE2 cells were treated with serially diluted SSP for 2 hours at 37℃, washed, and were then incubated with supernatant collected from RBD-PD1 expressing plasmid transfected HEK293T cells or a goat polyclonal antibody against ACE2 for 30 min on ice. Cells were then washed and stained with a fluorescent-labeled secondary antibody against goat-IgG or PD-1, respectively. As shown in Figure 3e, untreated HEK-293T-ACE2 cells were positive for both ACE2 and RBD-PD1 staining but negative for the staining of antibodies against goat-IgG and PD-1, which suggested that HEK-293T-ACE2 cells were having a high expression level of ACE2 and can bind to RBD of SARS-CoV-2 S protein. SSP-treated cells showed significantly reduced geometric mean signal for ACE2 staining but not the percentage of ACE2+ cells, suggesting the likelihood that SSP partially blocked ACE2 specific antibody binding rather than downregulating ACE2 expression. In contrast, SSP treatment deducted both geometric mean signal and percentage of positive cells of RBD staining when compared to untreated cells, suggesting that SSP treatment partially blocked binding of RBD to ACE2 protein (Figure 3f) .
13. SSP shows broad-spectrum antiviral activities against SARS-CoV, H5N1, EBOV and HIV-1
To determine whether SSP possesses broad-spectrum antiviral activities, a panel of pseudoviruses was generated in our previous studies, including SARS-CoV (L. Liu et al., 2019) , H5N1Turkey (Xiao et al., 2013) , CCR5-tropic HIV-1ADA, and CXCR4-tropic HIV-1HXB2 (Liang et al., 2013) , were utilized to test the inhibition rate of SSP. As shown in Figure 4a-d, SSP inhibited their infection with EC50 around 3.64, 5.13, 3.61, and 8.15 μg/mL, respectively, and was devoid of overt cytotoxicity (Figure 4e) . This result suggested that SSP has a broad-spectrum antiviral activity against entry of SARS-CoV, H5N1, and HIV-1. Additionally, recent re-emergence of Ebola virus disease in West Africa has led us to exam whether S SP also inhibit EBOV infection. EBOV pseudovirus was produced in 293T cells as described previously. We tested pseudoviral EBOV and VSV as control. 293 T cells were infected with EBOV and VSV, respectively, in the presence of serial diluted SSP. We found that SSP inhibited EBOV with EC50 value around 4 μg/ml without cell toxicity observed (Figure 4f) . Consistently, SSP did not inhibit VSV infection, indicating that SSP blocked EBOV entry to the target cells.
SSP blocks SARS-CoV-1 and H5N1 entry by binding to viral envelope and its target cells. To determine whether SSP inhibits viral entry by binding to the SARS-COV-1 envelope or its receptor on target cells, SSP-virus binding and SSP-cell binding assays were performed. In the SSP-virus binding assay, pseudovirus was initially pre-treated with 50 μg/mL of SSP. The viruses were then recovered by ultracentrifugation and subjected to infect target cells. We found that SSP inhibited SARS-CoV-1 pseudovirus infection at a similar degree as when the virus and SSP were added into cells simultaneously (Figure 5) . Meantime, the SSP-cell binding assays were performed by pre-treatment of the target cells with SSP for 1 hour at 37℃. After washing by PBS, cells were incubated with SARS-CoV-1 at 37 ℃ for 48 hrs (Rapista et al., 2011) . We found that pre-treatment of SSP on the target cells inhibited virus infection (Figure 5) . The same approach was also used to test the inhibitory effect of SSP against H5N1 with a single high dose of 50 μg/mL. Similar to SARS-CoV-1, SSP treatment with virus particles or their target cells efficiently blocked H5N1 virus entry (Figure 5b) .
SSP blocks HIV-1 entry by binding to the viral envelope. We tested HIV-1 with a similar but slightly modified experimental protocol by including four anti-HIV drugs: AZT, a potent NRTI, HIV-1 fusion blocker T20, CCR5 antagonist Marvaroc (MVC) , and the CXCR4 antagonist JM2987 as positive controls. As shown in Figure 5, AZT, but not SSP, significantly inhibited HIV-1 infection when the treatment was initiated at 2 hours post-infection. In contrast, pre-treatment of the virus with 50 μg/mL of SSP significantly inhibited HIV-1 infection at a similar degree as T-20, which blocks HIV-1 fusion (Figure 5c~5d) . Subsequently, the SSP-cell binding assays were performed by treating the target GHOST cells with SSP or with control compounds the CCR5 antagonist Marvaroc (MVC) or the CXCR4 antagonist JM2987 for 2 hours at 37℃. After washing by PBS, cells were subjected to HIV-1ADA or HIV-1HXB2 infection and cultivated at 37 ℃ for 48 hrs (Rapista et al., 2011) . We found that pre-treating the target cells with SSP had virtually no antiviral effect, while MVC and JM2987 showed potent viral inhibition against the respective ADA and HXB2 pseudoviruses at 1 μM as expected (Figures 5e~5f) . This evidence demonstrated that SSP inhibited HIV-1 infection by actions on the viral envelope glycoprotein gp160, which mediates the viral entry into the host target cells.
14. Antiviral activity of SSP and its effective constituents
To interpret the materials containing SS, a feasible UPLC method was used to separate SSP, in which SSP was divided into three parts, namely Part I, II, and III (Figure 6a) . UPLC-ESI-MS/MSn experiment was also performed under the same method (Figure 6d~6f) , and found that: (1) Monomer and dimer were mainly enriched in the 3~9 min (Part I) ; (2) 8~10mer were mainly enriched in the 23~33 min (Part II) ; (3) 3~7mer were both enriched in the two parts. Nevertheless, polymers with a degree of polymerization (DP) above 10 were undetected due to the limited range of molecular weight. Based on this result, different polar solvents were used for the extraction, and the n-butanol part (SSP-n-BuOH) was selected for further separation by using the macroporous resin columns. Through UPLC's detection, Fr. B and Fr. G were separated into two parts, Part I and Part II, respectively, as mentioned above (Figure 6b~6c) . In this study, we extracted and greatly enriched proanthocyanidins in SSP, reaching about 60%concentration. The thiolysis experiments also proved that the mean degrees of polymerization (mDP) of SSP, SSP-n-BuOH, Fr. B, and Fr. G were 3.49, 4.57, 2.95, and 7.52 respectively, which meant macroporous resin columns could effectively separate and obtain parts with different mDPs. Using the method as previously described, we found that SSP, SSP-n-BuOH, and Fr. G exhibited lower EC50 in the inhibition of SARS-CoV and SARS-CoV-2 pseudovirus infection than Fr. B (Figure 7a~7b) . No significant inhibitory effects on VSV pseudovirus (Figure 7c) or cytotoxicity were observed (Figure 7d) . Moreover, pre-treatment of pseudovirus or cells with SSP-n-BuOH, Fr. F, or Fr. G inhibited viral entry with lower EC50 value than adding the virus and fragments simultaneously, suggesting that those fragments target both SARS-CoV-2 viral envelop S protein and target cells to inhibit SARS-CoV-2 entry. More importantly, Fr. G exhibited more outstanding bioactivity when compared to Fr. B, suggesting that Part II of SSP contains more effective antiviral activities than Part I. These results revealed that Fr. G contains an intermediate polymer of proanthocyanidins, which may have better antiviral effects.
15. Safety evaluation result
To evaluate the safety of SSP, the cytotoxicity in multiple cell lines was determined by cell toxicity assay (IC50 = 181.2~339.8 μg/mL as shown in Figure 8a~8d) .
To test the acute toxicity in vivo, NIH mice and SD rats were employed to obtain the maximum tolerated dose. The maximum tolerated dose for one-time administration is 67 g crude drug /kg in mice (Table 1) . The maximum tolerated dose for one-time administration is 107 g crude drug /kg in rats (Table 2) .
To further examine the long-term toxicity in vivo, 24 male and female SD rats were divided into four groups: water (blank control) , 2 g/kg, 4 g/kg, and 6 g/kg SSP. Drug or water was administered via oral gavage once a day for 25 days. No mortality was observed in any group of rats during the monitoring period (Figure 8) . The body weight and food intake were shown in Figures 8e-8f. Administration of SSP at the doses did not manifest any signs of toxicity to rats except the loss of body weight in the high dose group. No significant differences in organ indexes (ratio of organ weight to body weight) were observed compared with the control mice (Figure 8g) . No abnormality was observed in levels of blood biochemical parameters were not harmed, indicating that SSP has no apparent toxicity to the experimental rats (Table 3) .
Mice result:
Table 1 acute toxicity of SSP in mice (NIH)
Rat result:
Table 2 acute toxicity of SSP in rat (SD)
Table 3 Blood biochemical index testing items in male and female rats
16. Stability results
According to the same method, the proanthocyanidins contained in the immediately prepared SSP aqueous solution were 794.794 ± 13.619 mg/g, while the content in the SSP aqueous solution after being placed for 1 month was 778.964 ± 63.589mg/g, which was within the quality standard range of 5%, so the SSP aqueous solution is stable in terms of proanthocyanidin content.
Besides, it was found that the anti-viral activity of the SSP aqueous solution after being placed for 6 and 13 days were no significant reduction, and cyto-toxicity was also no changes (Figure 9a) .
17. In vitro anti-viral experiments against SARS-CoV-2 virus variants
As shown in Figure 9a-9b, the inhibitory activity of SSP against SARS-CoV-2 virus variants was also quite potent, meanwhile, the cell toxicity was minor. Compared with SSP, the GSE (Figure 9c) and TP (Figure 9d) , which also contained PACs, exhibited much weaker inhibitory effect and not all variants were inhibited. SSP shows higher potency and broader spectrum against SARS-CoV-2 variants entry, which is different from GSE and TP. To sum up, SSP is the unique with higher potency and broader spectrum against virus, which deserves deeper exploration.
18. In vivo anti-viral experiments
As shown in Figure 10b, Omicron BA. 2 virus-infected cells (green) and inflammatory cells (red) were significantly reduced in the lung tissue of mice treated with SSP compared with the saline group. Also, in the lung tissue of mice treated with SSP, the expression level of RNA-dependent RNA polymerase and the virus titer were significantly lower than those of mice treated with saline (Figure 10c) . Overall, SSP showed efficacy in protection against nasal challenge of SARS-CoV-2 omicron variant virus in mice.
19. Nasal spray irritation test
This experiment was completed in the CFDA-certified GLP-compliant Drug Safety Evaluation Research Center (Shandong Xinbo Drug Research Co., Ltd. ) , and the code of this experiment was KY22233. During the test period, the animals were generally in good condition, and no death occurred; no obvious abnormalities were observed immediately after each administration. 24 hours after the last administration and 14 days after recovery, the necropsy was performed to observe the local mucous membrane tissue (oral cavity, nasal cavity, larynx, trachea, and bronchi) with the naked eye. No obvious abnormalities were found in the negative control group and the test product group. The results of histopathological examination also showed that 24 days after the last administration and the end of the recovery period, no drug-related histopathological changes were found in all animal parts of the negative control group and the test product group.
REFERENCES
1. Liang, J., et al., Extracts of the medicinal herb Sanguisorba officinalis inhibit the entry of human immunodeficiency virus-1. Journal of Food and Drug Analysis. 21 (4) : p. S52-S58.
2. Chu, Y. and H. Liu, Advances of research on anti-HIV agents from traditional Chinese herbs. Adv Dent Res, 2011. 23 (1) : p. 67-75.
3. Wu, X.F., et al., Thoughts on intervention in HIV/AIDS with traditional Chinese medicine. J Tradit Chin Med, 2011. 31 (4) : p. 265-8.
4. Lu, C.J., et al., A preliminary study on the medical expenditure of Chinese medicine and integrative medicine treatment for influenza A (H1N1) in the fever clinics. Chin J Integr Med, 2010. 16 (6) : p. 493-7.
5. Liu, J., H. McIntosh, and H. Lin, Chinese medicinal herbs for chronic hepatitis B: a systematic review. Liver, 2001. 21 (4) : p. 280-6.
6. CHENG, Y., et al., Determination on the Contents of Condensed Tannins in Spatholobus suberectus Dunn. Extracts and Primary Study on their Anti-tumor Activities. Acta Scientiarum Naturalium Universitatis Sunyatseni, 2011. 50 (2) : p. 75-80.
7. Liu, L., et al., Natural mutations in the receptor binding domain of spike glycoprotein determine the reactivity of cross-neutralization between palm civet coronavirus and severe acute respiratory syndrome coronavirus. J Virol, 2007. 81 (9) : p. 4694-700.
8. Yi, C.E., et al., Single amino acid substitutions in the severe acute respiratory syndrome coronavirus spike glycoprotein determine viral entry and immunogenicity of a major neutralizing domain. J Virol, 2005. 79 (18) : p. 11638-46.
9. Richards, K.H. and P.R. Clapham, Human immunodeficiency viruses: propagation, quantification, and storage. Curr Protoc Microbiol, 2006. Chapter 15: p. Unit15J 1.
10. Lu, X., et al., F18, a novel small-molecule nonnucleoside reverse transcriptase inhibitor, inhibits HIV-1 replication using distinct binding motifs as demonstrated by resistance selection and docking analysis. Antimicrob Agents Chemother, 2012. 56 (1) : p. 341-51.
11. Liang, J., et al., Extracts of medicinal herb Sanguisorba officinalis inhibit the entry of
human immunodeficiency virus type one. Yao Wu Shi Pin Fen Xi, 2013. 21 (4) : p. S52-S58.
11. Liang, J., et al., Extracts of medicinal herb Sanguisorba officinalis inhibit the entry of
human immunodeficiency virus type one. Yao Wu Shi Pin Fen Xi, 2013. 21 (4) : p. S52-S58.
12. Liu, L., et al., Anti-spike IgG causes severe acute lung injury by skewing macrophage responses during acute SARS-CoV infection. JCI Insight, 2019. 4 (4) .
13. Rapista, A., et al., Human defensins 5 and 6 enhance HIV-1 infectivity through promoting HIV attachment. Retrovirology, 2011. 8: p. 45.
14. Xiao, H., et al., A replicating modified vaccinia tiantan strain expressing an avian- derived influenza H5N1 hemagglutinin induce broadly neutralizing antibodies and cross-clade protective immunity in mice. PLoS One, 2013. 8 (12) : p. e83274.
The foregoing description of the specific embodiments will so fully reveal the general nature of the disclosure that others can, by applying knowledge within the skill of the relevant art (s) (including the contents of the documents cited and incorporated by reference herein) , readily modify and/or adapt for various applications such specific embodiments, without undue experimentation, without departing from the general concept of the present disclosure. Such adaptations and modifications are therefore intended to be within the meaning and range of equivalents of the disclosed embodiments, based on the teaching and guidance presented herein. It is to be understood that the phraseology or terminology herein is for the purpose of description and not of limitation, such that the terminology or phraseology of the present specification is to be interpreted by the skilled artisan in light of the teachings and guidance presented herein, in combination with the knowledge of one skilled in the relevant art (s) .
While various embodiments of the present disclosure have been described above, it should be understood that they have been presented by way of examples, and not limitation. It would be apparent to one skilled in the relevant art (s) that various changes in form and detail could be made therein without departing from the spirit and scope of the disclosure. Thus, the present disclosure should not be limited by any of the above-described exemplary embodiments but should be defined only in accordance with the following claims and their equivalents.
All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.
Claims (35)
- A method of obtaining an extract of Spatholobus suberectus Dunn (SSD) , said method comprising: (i) cutting SSD into pieces; (ii) immersing and treating SSD pieces in a solvent at room temperature to obtain an extract; (iii) concentrating the extract; and (iv) drying the concentrated extract to obtain the powder of the extract of SSD, the extract of SSD including proanthocyanidins (SSP) .
- A method of obtaining an active fraction from Spatholobus suberectus Dunn (SSD) , the active fraction including proanthocyanidins, said method comprising: (i) cutting SSD into pieces; (ii) immersing and treating SSD pieces in a solvent at room temperature to obtain an extract; (iii) concentrating the extract; (iv) drying the concentrated extract to obtain the powder of the extract of SSD (SSP) ; (v) dispersing SSP in water to obtain an aqueous solution; (vi) extracting the SSP aqueous solution with a series of solvent with different polarities at equal volume to obtain parts of SSP corresponding to each solvent, the series of solvent comprising n-butanol; and (vii) separating the n-butanol part (SSP-n-BuOH) with eluents to obtain the active fraction.
- The method of claim 1 or 2, wherein the treating process in (ii) comprises percolation, ultrasonic, heating and refluxing, decoction, and/or solvent extraction.
- The method of claim 3, wherein the treating process in (ii) comprises percolation.
- The method of claim 1 or 2, wherein the solvent in (ii) comprises water, and/or an organic solvent.
- The method of claim 5, wherein the solvent in (ii) comprises water and ethanol.
- The method of claim 6, wherein the solvent in (ii) is 60%ethanol/water (v/v) .
- The method of claim 1 or 2, wherein the concentrating process in (iii) comprises reduced pressure rotary evaporation, centrifugation, and/or nitrogen blowing.
- The method of claim 8, wherein the concentrating process in (iii) comprises reduced pressure rotary evaporation.
- The method of claim 1 or 2, wherein the concentrating process is performed at a temperature under 50℃.
- The method of claim 1 or 2, wherein the drying process in (iv) comprises freeze drying, spray drying, microwave drying, and/or infrared heating drying.
- The method of claim 11, wherein the drying process in (iv) comprises freeze drying.
- The method of claim 1 or 2, wherein the series of solvent in (vi) further comprises water, ethanol, petroleum ether (PE) , and/or ethyl acetate.
- The method of claim 1 or 2, wherein the separating process in (vii) comprises macroporous resin columns separation, fractional precipitation, crystallization, normal phase chromatography, reverse phase chromatography, and/or ion exchange resin columns separation.
- The method of claim 14, wherein the separating process in (vii) comprises macroporous resin columns separation.
- The method of claim 1 or 2, wherein the eluents in (vii) comprises water, and/or an organic solvent.
- The method of claim 16, wherein the eluents in (vii) are gradient ethanol-water from 10%to 95%.
- The method of claim 2, further comprising: (viii) concentrating the active fractions.
- The method of claim 18, further comprising: (ix) drying the concentrated active fractions.
- An extract of SSD obtained by the method of any one of claims 1 and 3-17.
- An active fraction from SSD obtained by the method of any one of claims 2-19.
- A composition comprising an extract of SSD obtained by the method of any one of claims 1 and 3-17, or an active fraction from SSD obtained by the method of any one of claims 2-19.
- A method of preventing and/or treating a viral disease comprising administering an effective amount of an extract of SSD obtained by the method of any one of claims 1 and 3-17 to a subject in need thereof.
- A method of preventing and/or treating a viral disease comprising administering an effective amount of an active fraction from SSD obtained by the method of any one of claims 2-19 to a subject in need thereof.
- The method of claim 23 and 24, wherein the effective dose is ranging from 0.5 μg/mL to 5000 μg/mL.
- Use of an extract of SSD obtained by the method of any one of claims 1 and 3-17 in the manufacture of preparations for prevention and/or treatment of a viral disease.
- Use of an active fraction from SSD obtained by the method of any one of claims 2-19 in the manufacture of preparations for prevention and/or treatment of a viral disease.
- The method/use of any one of claims 23-26, wherein the viral disease is caused by coronaviruses including SARS-CoV-2 and SARS-CoV-1 and their variants, Ebola virus (EBOV) , HIV-1, H5N1, and other enveloped viruses except for VSV.
- The use of claim 26 or 27, wherein the preparations are in the form of injections, tablets, granules, emulsions, gels, sustained-release preparations, nasal wash/spray, oral liquids, throat spray, like-tea pills/candy, nano preparations, “throat lozenges” , “throat sprays” , “nasal washes” , and/or “nasal sprays” .
- The use of claim 26 or 27, where in the preparations further comprise fillers, disintegration agents, additives, suspending agents, and/or binders.
- The use of claim 30, wherein the fillers comprise starch, pregelatinized starch, lactose, mannitol, chitin, microcrystalline cellulose, and/or sucrose.
- The use of claim 30, wherein the disintegration agents comprise starch, pregelatinized starch, microcrystalline cellulose, sodium carboxymethyl starch, cross-linked polyvinylpyrrolidone, low-substituted hydroxypropyl cellulose, and/or cross-linked polymethyl cellulose sodium.
- The use of claim 30, wherein the additives comprise stearic acid magnesium, sodium lauryl sulfate, talc, and/or silica.
- The use of claim 30, wherein the suspending agents comprise polyvinylpyrrolidone, microcrystalline cellulose, sucrose, agar, and/or hydroxypropyl methylcellulose.
- The use of claim 30, wherein the binders comprise starch Pulp, and/or polyvinylpyrrolidone.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202263319940P | 2022-03-15 | 2022-03-15 | |
US63/319,940 | 2022-03-15 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2023174207A1 WO2023174207A1 (en) | 2023-09-21 |
WO2023174207A9 true WO2023174207A9 (en) | 2024-10-24 |
Family
ID=88022252
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2023/081058 WO2023174207A1 (en) | 2022-03-15 | 2023-03-13 | A method of obtaining extracts of spatholobus suberectus dunn (ssd), fractions and compostions thereof and using against viral diseases |
Country Status (2)
Country | Link |
---|---|
TW (1) | TW202342082A (en) |
WO (1) | WO2023174207A1 (en) |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2005314316A (en) * | 2004-04-30 | 2005-11-10 | Kikkoman Corp | Anti-sars coronavirus agent |
CN101474247A (en) * | 2008-12-26 | 2009-07-08 | 中国人民解放军军事医学科学院卫生学环境医学研究所 | Application of spatholobus stem extract in preparing medicament for resisting influenza virus and enterovirus |
CN113925890A (en) * | 2020-06-29 | 2022-01-14 | 香港大学 | Extraction method of caulis spatholobi extract, caulis spatholobi extract and application thereof |
-
2023
- 2023-03-13 WO PCT/CN2023/081058 patent/WO2023174207A1/en unknown
- 2023-03-14 TW TW112109368A patent/TW202342082A/en unknown
Also Published As
Publication number | Publication date |
---|---|
WO2023174207A1 (en) | 2023-09-21 |
TW202342082A (en) | 2023-11-01 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Jahan et al. | Potentials of plant-based substance to inhabit and probable cure for the COVID-19 | |
Bhuiyan et al. | Plants metabolites: possibility of natural therapeutics against the COVID-19 pandemic | |
Law et al. | Antiviral effect of forsythoside A from Forsythia suspensa (Thunb.) Vahl fruit against influenza A virus through reduction of viral M1 protein | |
Papies et al. | Antiviral and immunomodulatory effects of Pelargonium sidoides DC. root extract EPs® 7630 in SARS-CoV-2-infected human lung cells | |
Komolafe et al. | Coronavirus disease 2019 and herbal therapy: pertinent issues relating to toxicity and standardization of phytopharmaceuticals | |
RU2505306C2 (en) | Composition for preventing and treating viral infections | |
Liu et al. | Broad‐spectrum antiviral activity of Spatholobus suberectus Dunn against SARS‐CoV‐2, SARS‐CoV‐1, H5N1, and other enveloped viruses | |
Döring et al. | Insights into the direct anti-influenza virus mode of action of Rhodiola rosea | |
Rathinavel et al. | Virtual screening of COVID-19 drug from three Indian traditional medicinal plants through in silico approach | |
Velásquez et al. | Effectiveness of Drug Repurposing and Natural Products Against SARS-CoV-2: A Comprehensive Review | |
Bhattacharya et al. | Recent advances on the therapeutic potential with Ocimum species against COVID-19: A review | |
WO2023174207A9 (en) | A method of obtaining extracts of spatholobus suberectus dunn (ssd), fractions and compostions thereof and using against viral diseases | |
Siew et al. | Fighting nature with nature: Antiviral compounds that target retroviruses | |
Watanabe | Drug‐Repositioning Approach for the Discovery of Anti‐Influenza Virus Activity of Japanese Herbal (Kampo) Medicines In Vitro: Potent High Activity of Daio‐Kanzo‐To | |
Palshetkar et al. | Natural Product-derived Phytochemicals as Potential Inhibitors of Angiotensin Converting Enzyme 2 (ACE2): Promising Drug Candidates for COVID-19 | |
CN118900695A (en) | Method for obtaining extract of Sparassis crispa (SSD), fraction and composition thereof and use for combating viral diseases | |
Sharma et al. | Therapeutic options for the SARS-CoV-2 virus: is there a key in herbal medicine? | |
Bagde et al. | Effects of plant metabolites on the growth of COVID-19 (Coronavirus Disease-19) including omicron strain | |
Oktavianawati et al. | Recent progress on drugs discovery study for treatment of COVID-19: repurposing existing drugs and current natural bioactive molecules | |
Barkat et al. | Phytoconstituents in the management of Covid-19: Demystifying the fact | |
CN103565970B (en) | A kind of Radix Sangusorbae extract for the treatment of acquired immune deficiency syndrome (AIDS) and preparation method thereof | |
Chattopadhyay et al. | Ethnopharmacology: A new engine for the development of antivirals from naturaceuticals | |
Hemaiswarya et al. | Synergistic Herb-Drug Interactions Against Viral Diseases | |
Venkateswaran et al. | INTERNATIONAL JOURNAL OF RESEARCH IN PHARMACEUTICAL SCIENCES | |
Victoriano-Belvis et al. | A Preliminary Investigation on the Antiviral Activities of the Philippine Marshmint (Mentha arvensis) Leaf Extracts against Dengue Virus Serotype 2 In Vitro |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 23769710 Country of ref document: EP Kind code of ref document: A1 |