WO2010019698A2 - Inhibition of calicivirus (norovirus) - Google Patents
Inhibition of calicivirus (norovirus) Download PDFInfo
- Publication number
- WO2010019698A2 WO2010019698A2 PCT/US2009/053591 US2009053591W WO2010019698A2 WO 2010019698 A2 WO2010019698 A2 WO 2010019698A2 US 2009053591 W US2009053591 W US 2009053591W WO 2010019698 A2 WO2010019698 A2 WO 2010019698A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- ifn
- cells
- ribavirin
- genome
- virus
- Prior art date
Links
- 241001263478 Norovirus Species 0.000 title claims description 23
- 230000005764 inhibitory process Effects 0.000 title description 11
- IWUCXVSUMQZMFG-AFCXAGJDSA-N Ribavirin Chemical compound N1=C(C(=O)N)N=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 IWUCXVSUMQZMFG-AFCXAGJDSA-N 0.000 claims abstract description 102
- 229960000329 ribavirin Drugs 0.000 claims abstract description 102
- HZCAHMRRMINHDJ-DBRKOABJSA-N ribavirin Natural products O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1N=CN=C1 HZCAHMRRMINHDJ-DBRKOABJSA-N 0.000 claims abstract description 102
- VVOAZFWZEDHOOU-UHFFFAOYSA-N honokiol Natural products OC1=CC=C(CC=C)C=C1C1=CC(CC=C)=CC=C1O VVOAZFWZEDHOOU-UHFFFAOYSA-N 0.000 claims abstract description 78
- 108010047761 Interferon-alpha Proteins 0.000 claims abstract description 50
- 102000006992 Interferon-alpha Human genes 0.000 claims abstract description 50
- 238000011282 treatment Methods 0.000 claims abstract description 45
- OHDXDNUPVVYWOV-UHFFFAOYSA-N n-methyl-1-(2-naphthalen-1-ylsulfanylphenyl)methanamine Chemical compound CNCC1=CC=CC=C1SC1=CC=CC2=CC=CC=C12 OHDXDNUPVVYWOV-UHFFFAOYSA-N 0.000 claims abstract description 43
- 108010074328 Interferon-gamma Proteins 0.000 claims abstract description 42
- 102100037850 Interferon gamma Human genes 0.000 claims abstract description 38
- 238000000034 method Methods 0.000 claims abstract description 28
- BYTORXDZJWWIKR-UHFFFAOYSA-N Hinokiol Natural products CC(C)c1cc2CCC3C(C)(CO)C(O)CCC3(C)c2cc1O BYTORXDZJWWIKR-UHFFFAOYSA-N 0.000 claims abstract description 20
- 150000001875 compounds Chemical class 0.000 claims abstract description 20
- FVYXIJYOAGAUQK-UHFFFAOYSA-N honokiol Chemical compound C1=C(CC=C)C(O)=CC=C1C1=CC(CC=C)=CC=C1O FVYXIJYOAGAUQK-UHFFFAOYSA-N 0.000 claims abstract description 20
- 208000006339 Caliciviridae Infections Diseases 0.000 claims abstract description 13
- 238000011321 prophylaxis Methods 0.000 claims abstract description 9
- WDXRGPWQVHZTQJ-OSJVMJFVSA-N (8r,9s,10r,13s,14s,17z)-17-ethylidene-10,13-dimethyl-1,2,6,7,8,9,11,12,14,15-decahydrocyclopenta[a]phenanthrene-3,16-dione Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC(=O)\C(=C/C)[C@@]1(C)CC2 WDXRGPWQVHZTQJ-OSJVMJFVSA-N 0.000 claims abstract description 8
- 229960003130 interferon gamma Drugs 0.000 claims abstract description 8
- WDXRGPWQVHZTQJ-UHFFFAOYSA-N trans-guggulsterone Natural products C1CC2=CC(=O)CCC2(C)C2C1C1CC(=O)C(=CC)C1(C)CC2 WDXRGPWQVHZTQJ-UHFFFAOYSA-N 0.000 claims abstract description 8
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 5
- 239000003937 drug carrier Substances 0.000 claims abstract 3
- 241000714209 Norwalk virus Species 0.000 claims description 142
- 241000700605 Viruses Species 0.000 claims description 23
- 239000000203 mixture Substances 0.000 claims description 14
- 208000015181 infectious disease Diseases 0.000 claims description 10
- 239000003814 drug Substances 0.000 claims description 7
- 241000369733 Lagovirus Species 0.000 claims description 6
- 241000369757 Sapovirus Species 0.000 claims description 6
- 241000369696 Vesivirus Species 0.000 claims description 6
- 239000007894 caplet Substances 0.000 claims description 3
- 239000002775 capsule Substances 0.000 claims description 3
- 238000009472 formulation Methods 0.000 claims description 3
- 239000012729 immediate-release (IR) formulation Substances 0.000 claims description 3
- 239000007937 lozenge Substances 0.000 claims description 3
- 238000013268 sustained release Methods 0.000 claims description 3
- 239000012730 sustained-release form Substances 0.000 claims description 3
- 239000003826 tablet Substances 0.000 claims description 3
- 210000004027 cell Anatomy 0.000 description 131
- 230000000694 effects Effects 0.000 description 45
- 230000010076 replication Effects 0.000 description 45
- 108090000623 proteins and genes Proteins 0.000 description 33
- 230000014509 gene expression Effects 0.000 description 30
- 102000004169 proteins and genes Human genes 0.000 description 29
- NYHBQMYGNKIUIF-UUOKFMHZSA-N Guanosine Chemical compound C1=NC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O NYHBQMYGNKIUIF-UUOKFMHZSA-N 0.000 description 23
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 19
- 238000011529 RT qPCR Methods 0.000 description 19
- 230000009467 reduction Effects 0.000 description 19
- 241000272327 Murine norovirus 1 Species 0.000 description 18
- 230000000840 anti-viral effect Effects 0.000 description 18
- 102000014150 Interferons Human genes 0.000 description 14
- 108010050904 Interferons Proteins 0.000 description 14
- 101800000789 Protease-polymerase p70 Proteins 0.000 description 14
- 101800000786 Protease-polymerase p76 Proteins 0.000 description 14
- 230000007246 mechanism Effects 0.000 description 14
- 230000035772 mutation Effects 0.000 description 14
- 230000002829 reductive effect Effects 0.000 description 14
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 13
- 229940079322 interferon Drugs 0.000 description 13
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 13
- 238000001262 western blot Methods 0.000 description 13
- 231100000673 dose–response relationship Toxicity 0.000 description 12
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 12
- 229960000951 mycophenolic acid Drugs 0.000 description 12
- 239000002609 medium Substances 0.000 description 11
- 208000005577 Gastroenteritis Diseases 0.000 description 10
- 239000003443 antiviral agent Substances 0.000 description 10
- MIKUYHXYGGJMLM-GIMIYPNGSA-N Crotonoside Natural products C1=NC2=C(N)NC(=O)N=C2N1[C@H]1O[C@@H](CO)[C@H](O)[C@@H]1O MIKUYHXYGGJMLM-GIMIYPNGSA-N 0.000 description 9
- NYHBQMYGNKIUIF-UHFFFAOYSA-N D-guanosine Natural products C1=2NC(N)=NC(=O)C=2N=CN1C1OC(CO)C(O)C1O NYHBQMYGNKIUIF-UHFFFAOYSA-N 0.000 description 9
- 101000959820 Homo sapiens Interferon alpha-1/13 Proteins 0.000 description 9
- 102100040019 Interferon alpha-1/13 Human genes 0.000 description 9
- 229940029575 guanosine Drugs 0.000 description 9
- 238000011534 incubation Methods 0.000 description 9
- 241000711549 Hepacivirus C Species 0.000 description 8
- 230000003612 virological effect Effects 0.000 description 8
- 241000283707 Capra Species 0.000 description 6
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 6
- 241000283973 Oryctolagus cuniculus Species 0.000 description 6
- 238000004113 cell culture Methods 0.000 description 6
- 238000011278 co-treatment Methods 0.000 description 6
- 229940027941 immunoglobulin g Drugs 0.000 description 6
- 230000002401 inhibitory effect Effects 0.000 description 6
- 239000000126 substance Substances 0.000 description 6
- 108010025815 Kanamycin Kinase Proteins 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 229930193140 Neomycin Natural products 0.000 description 5
- 238000012300 Sequence Analysis Methods 0.000 description 5
- 238000011161 development Methods 0.000 description 5
- 230000018109 developmental process Effects 0.000 description 5
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 5
- 229960004927 neomycin Drugs 0.000 description 5
- 102000007469 Actins Human genes 0.000 description 4
- 108010085238 Actins Proteins 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- 102000008070 Interferon-gamma Human genes 0.000 description 4
- 241001529936 Murinae Species 0.000 description 4
- 241000700159 Rattus Species 0.000 description 4
- 230000009471 action Effects 0.000 description 4
- 230000004913 activation Effects 0.000 description 4
- 238000003556 assay Methods 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 230000001413 cellular effect Effects 0.000 description 4
- 230000007541 cellular toxicity Effects 0.000 description 4
- 239000003112 inhibitor Substances 0.000 description 4
- 230000003389 potentiating effect Effects 0.000 description 4
- 239000000047 product Substances 0.000 description 4
- 238000003786 synthesis reaction Methods 0.000 description 4
- 108091093088 Amplicon Proteins 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 241000714201 Feline calicivirus Species 0.000 description 3
- 101000599940 Homo sapiens Interferon gamma Proteins 0.000 description 3
- 241001105894 Murine norovirus Species 0.000 description 3
- 241000282898 Sus scrofa Species 0.000 description 3
- 108010067390 Viral Proteins Proteins 0.000 description 3
- 230000000996 additive effect Effects 0.000 description 3
- 239000003795 chemical substances by application Substances 0.000 description 3
- 231100000433 cytotoxic Toxicity 0.000 description 3
- 230000001472 cytotoxic effect Effects 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 230000036541 health Effects 0.000 description 3
- 230000006698 induction Effects 0.000 description 3
- 238000011835 investigation Methods 0.000 description 3
- 230000001404 mediated effect Effects 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 230000037361 pathway Effects 0.000 description 3
- 239000013612 plasmid Substances 0.000 description 3
- 230000000861 pro-apoptotic effect Effects 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 238000003757 reverse transcription PCR Methods 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 230000009385 viral infection Effects 0.000 description 3
- 102100038495 Bile acid receptor Human genes 0.000 description 2
- 241000714198 Caliciviridae Species 0.000 description 2
- 108090000565 Capsid Proteins Proteins 0.000 description 2
- 241000700199 Cavia porcellus Species 0.000 description 2
- 102100023321 Ceruloplasmin Human genes 0.000 description 2
- 240000003890 Commiphora wightii Species 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 2
- 206010061166 Gastroenteritis bacterial Diseases 0.000 description 2
- 101000603876 Homo sapiens Bile acid receptor Proteins 0.000 description 2
- 101000617830 Homo sapiens Sterol O-acyltransferase 1 Proteins 0.000 description 2
- GRSZFWQUAKGDAV-KQYNXXCUSA-N IMP Chemical compound O[C@@H]1[C@H](O)[C@@H](COP(O)(O)=O)O[C@H]1N1C(NC=NC2=O)=C2N=C1 GRSZFWQUAKGDAV-KQYNXXCUSA-N 0.000 description 2
- 108010087227 IMP Dehydrogenase Proteins 0.000 description 2
- 102000006674 IMP dehydrogenase Human genes 0.000 description 2
- 101710200424 Inosine-5'-monophosphate dehydrogenase Proteins 0.000 description 2
- 241001673966 Magnolia officinalis Species 0.000 description 2
- 108060004795 Methyltransferase Proteins 0.000 description 2
- 206010028980 Neoplasm Diseases 0.000 description 2
- 108700026244 Open Reading Frames Proteins 0.000 description 2
- 206010035226 Plasma cell myeloma Diseases 0.000 description 2
- 108010076039 Polyproteins Proteins 0.000 description 2
- 241000725643 Respiratory syncytial virus Species 0.000 description 2
- 102100021993 Sterol O-acyltransferase 1 Human genes 0.000 description 2
- 101000697584 Streptomyces lavendulae Streptothricin acetyltransferase Proteins 0.000 description 2
- 108091027544 Subgenomic mRNA Proteins 0.000 description 2
- 238000011053 TCID50 method Methods 0.000 description 2
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 description 2
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 2
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 2
- -1 Z-gugglesteron Chemical compound 0.000 description 2
- 150000001413 amino acids Chemical class 0.000 description 2
- 230000000843 anti-fungal effect Effects 0.000 description 2
- 230000003110 anti-inflammatory effect Effects 0.000 description 2
- 230000000975 bioactive effect Effects 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 210000003618 cortical neuron Anatomy 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 241001493065 dsRNA viruses Species 0.000 description 2
- 239000012636 effector Substances 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- 238000007710 freezing Methods 0.000 description 2
- 230000008014 freezing Effects 0.000 description 2
- 235000013902 inosinic acid Nutrition 0.000 description 2
- 230000003993 interaction Effects 0.000 description 2
- 230000003834 intracellular effect Effects 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 239000000546 pharmaceutical excipient Substances 0.000 description 2
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 238000012216 screening Methods 0.000 description 2
- 230000035945 sensitivity Effects 0.000 description 2
- 238000012163 sequencing technique Methods 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
- 239000000758 substrate Substances 0.000 description 2
- 239000013589 supplement Substances 0.000 description 2
- 230000002195 synergetic effect Effects 0.000 description 2
- 238000010257 thawing Methods 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 230000029812 viral genome replication Effects 0.000 description 2
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 2
- SJCDBQHCQSIZHN-UHFFFAOYSA-N 1,2-dihydrotriazole-3-carboxamide Chemical compound NC(=O)N1NNC=C1 SJCDBQHCQSIZHN-UHFFFAOYSA-N 0.000 description 1
- 102100027962 2-5A-dependent ribonuclease Human genes 0.000 description 1
- 108010000834 2-5A-dependent ribonuclease Proteins 0.000 description 1
- 201000001320 Atherosclerosis Diseases 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 101150071146 COX2 gene Proteins 0.000 description 1
- 101100114534 Caenorhabditis elegans ctc-2 gene Proteins 0.000 description 1
- 101100180402 Caenorhabditis elegans jun-1 gene Proteins 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 239000004099 Chlortetracycline Substances 0.000 description 1
- 206010009944 Colon cancer Diseases 0.000 description 1
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- 108010037462 Cyclooxygenase 2 Proteins 0.000 description 1
- 241000450599 DNA viruses Species 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 101710088194 Dehydrogenase Proteins 0.000 description 1
- 239000012983 Dulbecco’s minimal essential medium Substances 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 208000010201 Exanthema Diseases 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 102000027484 GABAA receptors Human genes 0.000 description 1
- 108091008681 GABAA receptors Proteins 0.000 description 1
- 101710121996 Hexon protein p72 Proteins 0.000 description 1
- 101100369992 Homo sapiens TNFSF10 gene Proteins 0.000 description 1
- 241000712003 Human respirovirus 3 Species 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 102100034170 Interferon-induced, double-stranded RNA-activated protein kinase Human genes 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 241000218378 Magnolia Species 0.000 description 1
- 101710125418 Major capsid protein Proteins 0.000 description 1
- 101710157639 Minor capsid protein Proteins 0.000 description 1
- 208000034578 Multiple myelomas Diseases 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 108010057466 NF-kappa B Proteins 0.000 description 1
- 102000003945 NF-kappa B Human genes 0.000 description 1
- 239000000020 Nitrocellulose Substances 0.000 description 1
- 206010067482 No adverse event Diseases 0.000 description 1
- 241001617329 Norovirus isolates Species 0.000 description 1
- 101150000187 PTGS2 gene Proteins 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 241000009328 Perro Species 0.000 description 1
- 102100038280 Prostaglandin G/H synthase 2 Human genes 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 102000001253 Protein Kinase Human genes 0.000 description 1
- 101710136297 Protein VP2 Proteins 0.000 description 1
- 241000714203 Rabbit hemorrhagic disease virus Species 0.000 description 1
- 108010017324 STAT3 Transcription Factor Proteins 0.000 description 1
- 206010039491 Sarcoma Diseases 0.000 description 1
- 102100024040 Signal transducer and activator of transcription 3 Human genes 0.000 description 1
- 102000001494 Sterol O-Acyltransferase Human genes 0.000 description 1
- 108010054082 Sterol O-acyltransferase Proteins 0.000 description 1
- 101710172711 Structural protein Proteins 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 102000046283 TNF-Related Apoptosis-Inducing Ligand Human genes 0.000 description 1
- 108700012411 TNFSF10 Proteins 0.000 description 1
- GLNADSQYFUSGOU-GPTZEZBUSA-J Trypan blue Chemical compound [Na+].[Na+].[Na+].[Na+].C1=C(S([O-])(=O)=O)C=C2C=C(S([O-])(=O)=O)C(/N=N/C3=CC=C(C=C3C)C=3C=C(C(=CC=3)\N=N\C=3C(=CC4=CC(=CC(N)=C4C=3O)S([O-])(=O)=O)S([O-])(=O)=O)C)=C(O)C2=C1N GLNADSQYFUSGOU-GPTZEZBUSA-J 0.000 description 1
- 102000004142 Trypsin Human genes 0.000 description 1
- 108090000631 Trypsin Proteins 0.000 description 1
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 description 1
- 206010046865 Vaccinia virus infection Diseases 0.000 description 1
- 108020000999 Viral RNA Proteins 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 241000710772 Yellow fever virus Species 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 208000038016 acute inflammation Diseases 0.000 description 1
- 230000006022 acute inflammation Effects 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000001142 anti-diarrhea Effects 0.000 description 1
- 230000002785 anti-thrombosis Effects 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 230000003078 antioxidant effect Effects 0.000 description 1
- 229940121357 antivirals Drugs 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 239000003613 bile acid Substances 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 238000009937 brining Methods 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 108091092328 cellular RNA Proteins 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- CYDMQBQPVICBEU-UHFFFAOYSA-N chlorotetracycline Natural products C1=CC(Cl)=C2C(O)(C)C3CC4C(N(C)C)C(O)=C(C(N)=O)C(=O)C4(O)C(O)=C3C(=O)C2=C1O CYDMQBQPVICBEU-UHFFFAOYSA-N 0.000 description 1
- 229960004475 chlortetracycline Drugs 0.000 description 1
- CYDMQBQPVICBEU-XRNKAMNCSA-N chlortetracycline Chemical compound C1=CC(Cl)=C2[C@](O)(C)[C@H]3C[C@H]4[C@H](N(C)C)C(O)=C(C(N)=O)C(=O)[C@@]4(O)C(O)=C3C(=O)C2=C1O CYDMQBQPVICBEU-XRNKAMNCSA-N 0.000 description 1
- 235000019365 chlortetracycline Nutrition 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 230000001149 cognitive effect Effects 0.000 description 1
- 208000029742 colonic neoplasm Diseases 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 238000011284 combination treatment Methods 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 238000013329 compounding Methods 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 238000002784 cytotoxicity assay Methods 0.000 description 1
- 231100000263 cytotoxicity test Toxicity 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- 239000012153 distilled water Substances 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 229960004679 doxorubicin Drugs 0.000 description 1
- 238000009509 drug development Methods 0.000 description 1
- 238000007876 drug discovery Methods 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 201000005884 exanthem Diseases 0.000 description 1
- 238000013265 extended release Methods 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 108010064833 guanylyltransferase Proteins 0.000 description 1
- 229940098322 guggul lipid Drugs 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 208000014674 injury Diseases 0.000 description 1
- 230000015788 innate immune response Effects 0.000 description 1
- 239000002348 inosinate dehydrogenase inhibitor Substances 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 229940047124 interferons Drugs 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 208000020816 lung neoplasm Diseases 0.000 description 1
- 108010026228 mRNA guanylyltransferase Proteins 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 239000002207 metabolite Substances 0.000 description 1
- 231100000324 minimal toxicity Toxicity 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 210000000822 natural killer cell Anatomy 0.000 description 1
- 229930014626 natural product Natural products 0.000 description 1
- 230000014511 neuron projection development Effects 0.000 description 1
- 230000000324 neuroprotective effect Effects 0.000 description 1
- 229920001220 nitrocellulos Polymers 0.000 description 1
- 244000309711 non-enveloped viruses Species 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 230000007918 pathogenicity Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 102000013415 peroxidase activity proteins Human genes 0.000 description 1
- 108040007629 peroxidase activity proteins Proteins 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 229910052697 platinum Inorganic materials 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000001023 pro-angiogenic effect Effects 0.000 description 1
- 230000001915 proofreading effect Effects 0.000 description 1
- 230000000644 propagated effect Effects 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 150000003815 prostacyclins Chemical class 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 235000019833 protease Nutrition 0.000 description 1
- 108060006633 protein kinase Proteins 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 206010037844 rash Diseases 0.000 description 1
- 238000012827 research and development Methods 0.000 description 1
- 239000000523 sample Substances 0.000 description 1
- 239000012723 sample buffer Substances 0.000 description 1
- 238000013207 serial dilution Methods 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 208000010110 spontaneous platelet aggregation Diseases 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 230000005100 tissue tropism Effects 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 230000001960 triggered effect Effects 0.000 description 1
- 239000012588 trypsin Substances 0.000 description 1
- 230000036967 uncompetitive effect Effects 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 210000003932 urinary bladder Anatomy 0.000 description 1
- 201000005112 urinary bladder cancer Diseases 0.000 description 1
- 208000007089 vaccinia Diseases 0.000 description 1
- 208000019553 vascular disease Diseases 0.000 description 1
- 239000005723 virus inoculator Substances 0.000 description 1
- 230000006394 virus-host interaction Effects 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 150000003722 vitamin derivatives Chemical class 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 229940051021 yellow-fever virus Drugs 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/21—Interferons [IFN]
- A61K38/212—IFN-alpha
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/045—Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
- A61K31/05—Phenols
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/56—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7042—Compounds having saccharide radicals and heterocyclic rings
- A61K31/7052—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
- A61K31/7056—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing five-membered rings with nitrogen as a ring hetero atom
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/21—Interferons [IFN]
- A61K38/217—IFN-gamma
-
- 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
Definitions
- Caliciviruses are small, nonenveloped viruses of 27-35 nm in diameter (Green, K. Y. Chancock, R.M. & Kapikian, A.Z. (2001) in Fields Virology, eds. Knipe, D.M. & Howley, P.M. (Lippincott, Williams & Wilkins, Philidelphia), Vol. 1, pp. 841-874)). They possess a single-strand, plus-sense genomic RNA of 7-8 kb. Calicivirus replication proceeds through a minus strand RNA intermediate, which is used as the template for the synthesis of positive-sense full length genome and subgenomic RNA.
- This replication is catalyzed by the nonstructural proteins, including the viral RNA dependent RNA polymerase (RdRp), and occurs in cytoplasmic membrane-associated replicase complexes (Green, K. Y. Chancock, R.M. & Kapikian, A.Z. (2001) in Fields Virology, eds. Knipe, D.M. & Howley, P.M. (Lippincott, Williams & Wilkins, Philidelphia), Vol. 1, pp. 841-874)).
- RdRp viral RNA dependent RNA polymerase
- Norwalk virus (NV, a prototype norovirus) was originally identified in stools collected from an outbreak of gastroenteritis at a school in Norwalk, Ohio in 1968 (Kapikian et al., J Virol 10, 1075-81 (1972)).
- the viral genome is -7.7 kb in length and organized into three open reading frames (ORPs 1, 2 and 3) that encode a ⁇ 1800-amino acid polyprotein, the major capsid protein (VPl), and a minor capsid protein (VP2), respectively.
- ORPs 1, 2 and 3 that encode a ⁇ 1800-amino acid polyprotein, the major capsid protein (VPl), and a minor capsid protein (VP2), respectively.
- the structural proteins, VPl and VP2 are synthesized from a subgenomic RNA containing ORFs 2 and 3.
- Studies of NV replication and the development of antiviral drugs have been severely hampered by the absence of cell culture systems and animal models (Duizer et al., J Gen Virol 85, 79-87 (2004)).
- Studies of noroviruses associated with disease in humans have been hampered by the continued absence of a cell culture system (Duizer et al., J Gen Virol 85, 79-87 (2004)).
- RNA replicon-bearing cells expressed NV proteins and RNA, and could be maintained after multiple passages in the presence of G418. Importantly, the replicon- bearing cells could be examined for the effects of potential viral inhibitors, and they provided evidence that NV replication is sensitive to the effects of exogenous interferon (IFN).
- IFN exogenous interferon
- Virus replicon systems (especially viruses which are fastidious in cells) have proved an important tool in the investigation of virus-host interactions and anti-viral drug developments (Blight, et al., Science 290, 1972-4 (2000); Foy et al., Science 300(5622): 1145-8 (2003); Gale and Foy, Nature 436(7053):939-45 (2005)), and the availability of a NV replicon provides a new system in which such interactions can be assessed for the human noroviruses.
- NV replicon- bearing cells were shown to be an excellent platform to screen antivirals (Chang et al., Virology 353, 463-473 (2006)).
- norovirus infection is generally considered self-limiting and a short- term illness
- recent findings showed that the infection could last longer than several days or even several months especially in immunocompromised patients.
- the treatment options for norovirus infection are limited partly due to the absence of screening systems for antiviral drugs.
- Caliciviruses are plus strand RNA viruses in the family Caliciviridae that consists of four genera, Norovirus, Sapovirus, Lagovirus, and Vesivirus (Green et al., J Infect. Dis. 181, Suppl. 2, S322-330 (2000)). Caliciviruses are important pathogens in humans and animals with wide variety of pathogenicity that ranges gastroenteritis and system infections (Green et al., J Infect. Dis. 181, Suppl. 2, S322-330 (2000)). Viruses in genera Vesivirus and Lagovirus include animal viruses such as vesicular exanthema swine virus, feline calicivirus and rabbit hemorrhagic disease virus.
- Viruses in the genera Norovirus and Sapovirus cause gastroenteritis in humans and animals (Green et al., Human caliciviruses, p. 841-874. In D. M. Knipe and P. M. Howley (ed.), Fields Virology, vol. 1. Lippincott Williams & Wilkins, Philadelphia, PA (2001).
- MNV-I has a tissue tropism of macrophage-like cells in vivo and in vitro, but it is not clear if human noroviruses target to such cells at present.
- NV Norwalk virus
- IFN Interferon
- the replicon-bearing cells were generated by transfecting RNA transcripts derived from a plasmid containing the full length NV genome and neomycin resistant gene (neomycin phosphotransferase II, NPT II) in the place of VPl region (pNV-Neo) (Chang et al., Virology 353, 463-473 (2006)).
- the replicon bearing cells provide an excellent tool to study the replication of noroviruses and serve a platform to screen potential antiviral drugs.
- U.S. Patent 6,923,992 describes the use of compounds of Bao-Ji-Wan, for anti- diarrhea and relieving gastrointestinal symptoms.
- this patent does not disclose the treatment of any such condition due to Calicivirus. Accordingly, there remains a need for methods for treating or for prophylaxis of Calicivirus infections.
- the invention provides a method of treatment or prophylaxis of Calicivirus infection, comprising administering a pharmaceutical composition comprising a compound selected from the group of compounds consisting of interferon-alpha (IFN- ⁇ ), interferon- gamma (IFN- ⁇ ), honokiol, magnolol, ribavirin, z-guggulsterone, and a combination of any or all thereof (such as 2, 3, 4, 5, or all 6 of the compounds) and a pharmaceutically- acceptable carrierto a patient having a Calicivirus infection, or at risk for becoming infected with a Calicivirus.
- the invention provides the use of such compounds or such combinations thereof for preparing a medicament for treatment or prophylaxis of Calcivirus infection.
- the invention provides a method of making an inhibitor of a Calicivirus, brining into contact a Calicivirus-cell based replicon system with a compound, identifying a compound that has an inhibitory effect on the Calicivirus replicon, and synthesizing the compound with the inhibitory effect on the Calicivirus replicon.
- Fig. 1 The effect of IFN- ⁇ and IFN- ⁇ on the NV replication in HG23 cells (A). The effect of IFN- ⁇ and IFN- ⁇ on the expression of NV genome.
- One-day old semi-confluent NV replicon-bearing HG23 cells were incubated with various concentrations of IFN- ⁇ and IFN- ⁇ for 72 h, and then total RNA was prepared for real time qRT-PCR for detecting NV genome.
- NV genome by IFNs was calculated by the comparison to that with mock-treatment.
- B The effect of IFN- ⁇ on the expression of NV ProPol or neomycin phosphotranferase in HG23 cells.
- One-day old semi-confluent HG23 cells were incubated with 0 (mock) or 100 units/ml of IFN- ⁇ for 72 h. The cells were then fixed with 100% methanol, and stained with antibodies to NV ProPol (upper panel) or neomycin phosphotranferase (lower panel) and FITC-conjugated secondary antibody.
- the negative control includes parental Huh-7 cells with the same staining.
- FIG. 2 The effect of ribavirin on the NV replication in HG23 cells
- A The effect of ribavirin on the expression of NV genome.
- One-day old semi-confluent HG23 cells were incubated with various concentrations ranging from 0 (mock) to 100 ⁇ M of ribavirin for 48, 72 and 96 h, and then total RNA was prepared for real time qRT-PCR for detecting NV genome. The reduction of NV genome by IFNs was calculated by the comparison to that with mock-treatment.
- B The effect of ribavirin on the expression of neomycin phosphotranferase in HG23 cells.
- HG23 cells were incubated with various concentrations ranging from 0 (mock) to 200 ⁇ M of ribavirin for 72 h. The cells were then fixed with 100% methanol, and stained with antibody to neomycin phosphotranferase and FITC-conjugated secondary antibody.
- the negative control includes parental Huh-7 cells with the same staining.
- FIG. 3 The combination effects of IFN- ⁇ and ribavirin on NV replication in HG23 cells.
- A The effect of ribavirin alone or co-treatment with IFN- ⁇ and ribavirin on the expression of NV genome.
- One-day old semi-confluent HG23 cells were incubated with various concentrations ranging from 0 (mock) to 100 ⁇ M of ribavirin with or without 2 units/ml of IFN- ⁇ . After 72 h of the incubation, total RNA was prepared for real time qRT- PCR for detecting NV genome. The reduction of NV genome by the treatments was calculated by the comparison to that with mock-treatment.
- B The reduction of NV genome by the treatments was calculated by the comparison to that with mock-treatment.
- ribavirin alone or co-treatment with IFN- ⁇ and ribavirin on the expression of NPT II detected by Western blot analysis.
- One-day old semi-confluent HG23 cells were incubated with various concentrations ranging from 0 (mock) to 100 ⁇ M of ribavirin with or without 2 units/ml of IFN- ⁇ . After 72 h of the incubation, cell lysate was prepared for Western blot analysis with antibody to neomycin phosphotransferase.
- Fig. 4 The effect of ribavirin on MNV-I in RAW267.2 cells.
- Confluent RAW267.2 cells in 6- well plates were treated with varying concentrations (0-100 ⁇ M) of ribavirin for 6 hr before MNV-I was added to the same medium at a MOI of 5.
- the virus infected cells were further incubated for 24 h, and the virus replication was measured by the TCID50 assay after the plates were freezing and thawing 3 times.
- Fig. 5 The effect of supplement of guanosine in ribavirin treatment and of MPA on NV replication in HG23 cells.
- A The effect of ribavirin with or without of guanosine (100 ⁇ M) on NV genome in HG23 cells.
- One-day old semi-confluent HG23 cells were incubated with various concentrations ranging from 0 (mock) to 100 ⁇ M of ribavirin with or without 100 ⁇ M of guanosine. After 72 h of the incubation, total RNA was prepared for real time qRT-PCR for detecting NV genome. The reduction of NV genome by the treatments was calculated by the comparison to that with mock-treatment.
- B The effect of supplement of guanosine in ribavirin treatment and of MPA on NV replication in HG23 cells.
- HG23 cells were incubated with various concentrations ranging from 0 (mock) to 2 ⁇ M of MPA for 72 h. After the incubation, total RNA was prepared for real time qRT-PCR for detecting NV genome. The reduction of NV genome by the treatments was calculated by the comparison to that with mock-treatment.
- Fig. 6. The effect of magnolol on the NV replication in HG23 cells (A). The effect of magnolol on the expression of NV genome.
- HG23 cells were incubated with various concentrations ranging from 0 (mock) to 20 ⁇ M of magnolol for 48, 72 and 96 h, and then total RNA was prepared for real time qRT-PCR for detecting NV genome. The reduction of NV genome by magnolol was calculated by the comparison to that with mock-treatment.
- Fig. 7. Chemical structure of z-gugglesterone .
- Fig. 8. Chemical structure of rivabirin.
- Fig. 9 Chemical structure of IFN- ⁇ .
- Fig. 10. Chemical structure of magnolol.
- Fig. 11. Chemical structure of honokiol.
- Ribavirn (l- ⁇ -D-ribofuranosyl-l,2,4-triazole-3-carboxamide) is a synthetic guanosine analogue and has shown antiviral actions against a range of DNA and RJSfA viruses including hepatitis C virus (HCV) and respiratory syncytial virus (De Clercq E, J. Clin Virol. 30(2): 115-33 (2004)).
- HCV hepatitis C virus
- RJSfA viruses hepatitis C virus (De Clercq E, J. Clin Virol. 30(2): 115-33 (2004).
- HCV hepatitis C virus
- syncytial virus De Clercq E, J. Clin Virol. 30(2): 115-33 (2004).
- combination therapy of IFN- ⁇ and ribavirin is the most effective treatment.
- Interferon- ⁇ is synthesized by various cells including NK cells during the acute inflammations and plays an essential role as a frontier fighter against the invading microbes before specific immune responses elicit (Samuel, Clin Microbiol Rev. 14(4):778-809 (2001)).
- IFN- ⁇ inhibited the replication of NV in the replicon bearing cells showing the reduction of NV genome and proteins in a dose-dependent manner.
- the effective dose for reducing 50% (ED50) of NV genome and protein was calculated as 50 units/ml.
- ED50 50%
- ribavirin was applied to the cells, it effectively reduced NV genome and protein with the ED50 calculated as approximately 40 ⁇ M.
- IFN- ⁇ and ribavirin showed additive effects on the inhibition of NV replication.
- Z guggulsteron as antiviral drugs to treat the gastroenteritis in humans and animals by enteric caliciviruses including noroviruses.
- Z guggulsteron is available freely at local vitamin stores as a natural product extracted from Indian guggl tree (named as guggul herb, guggul lipid or guggul gum).
- Z gugglesteron which is an antagonist of bile acids on the farnesoid X receptor (FXR) can block the replication of norovirus.
- Z- gugglesteron inhibited the replication of NV in the replicon bearing cells showing the reduction of NV genome and proteins in a dose-dependent manner.
- Magnolol is a bioactive compound found in the bark of the Houpu magnolia (Magnolia officinalis) and is available commercially (e.g., from Sigma-Aldrich). It is known to act on the GABAA receptors in rats, as well as having antifungal properties. Magnolol is a bioactive plant component isolated from the root and stem bark of Magnolia officinalis.
- Magnolol displays an array of activities including antifungal, 1 antibacterial, and antioxidant effects.2 Magnolol has also been shown act as a natural inhibitor to acyl-CoA: cholesterol acyltransferase (ACAT).3 Magnolol has been shown to demonstrate anti-inflammatory activity by interfering with NF-kB signaling. It also appears to have anti-inflammatory properties related to vascular disorders such as atherosclerosis due to its ability to inhibit IL- 6-induced STAT3 activation.4 Magnolol inhibited the replication of NV in the replicon bearing cells showing the reduction of NV genome and proteins in a dose-dependent manner. The effective dose for reducing 50% (ED50) of NV genome and protein was calculated as 20 ⁇ M.
- Honokiol is a structural isomer of Magnolol, the purification and synthesis of has been reported (Amblard et al., J Med Chem. 2006 Jun 1;49(11):3426-7; Esumi et al, Bioorg Med Chem Lett. 2004 May 17;14(10):2621-5; Takeya et al., Chemical & Pharmaceutical Bulletin (1986), 34(5), 2066-70) and is available commercially (e.g., from Sigma-Aldrich).
- Honokiol has shown pro-apoptotic effects in melanoma, sarcoma, myeloma, leukemia, bladder, lung, prostate, and colon cancer cell lines (Shigemura et al., Cancer. 2007 Apr 1;109(7): 1279-89; Ishitsuka et al., Blood. 2005 Sep l;106(5):1794-800; Battle et al., Blood. 2005 JuI 15;106(2):690-7; Bai et al., J Biol Chem. 2003 Sep 12;278(37):35501-7). Honokiol inhibits phosphorylation of Akt, p44/42mitogen-activated protein kinase (MAPK), and src.
- MAPK p44/42mitogen-activated protein kinase
- honokiol modulates the nuclear factor kappa beta (NfKB) activation pathway, an upstream effector of vascular endothelial growth factor (VEGF), cyclooxygenase 2 (COX- 2), and MCLl, all significant pro-angiogenic and survival factors.
- Honokiol induces caspase- dependent apoptosis in a TRAIL-mediated manner, and potentiates the pro-apoptotic effects of doxorubicin and other etoposides. So potent is honokiol's pro-apoptotic effects that it overcomes even notoriously drug resistant neoplasms such as multiple myeloma and chronic B-cell leukemia.
- Honokiol has been shown to promote neurite outgrowth and have neuroprotective effects in rat cortical neurons. Additionally, honokiol increases free cytoplasmic Ca 2+ in rat cortical neurons (Fukuyama et al., Bioorg Med Chem Lett. 2002 Apr 22;12(8):1163-6). Honokiol inhibits platelet aggregation in rabbits in a dose-dependent manner, and protects cultured RAEC against oxidized low density liproptein injury. Honokiol significantly increases the prostacyclin metabolite 6-keto-PGFl alpha, potentially the key factor in honokiol's anti-thrombotic activity (Hu et al., Acta Pharmacol Sin. 2005 Sep;26(9): 1063-8.).
- this invention in one aspect, relates to the development for potential anti-noroviral agents.
- a pharmaceutical composition comprising a compound selected from the group of compounds consisting of interferon-alpha (IFN- ⁇ ), interferon-gamma (IFN- ⁇ ), honokiol, magnolol, ribavirin, z-guggulsterone, and a combination thereof is administered to a patient for Calicivirus prophylaxis or treatment of a Calicivirus infection.
- the composition can be formulated for administration to the patient by any desired route.
- the composition can be formulated for administration topically (e.g., a cream or ointment), via cubcutaneous, intramuscular, or parenteral injection, inhalation, orally, or via any other traditional route of administration.
- compositions are formulated for oral administration
- such forms can be immediate-release, sustained-release, or extended release forms, such as capsules, caplets, tablets, lozenges, etc.
- the composition also can be formulated as a powder for oral administration.
- the composition can include suitable binders, diluents, glidants, budders, flavoring agents, preservatives, and other common excipients. (see Rowe et al., "Handbook of Pharmaceutical Excipients" (Pharmaceutical Press), 5 th Ed. 2006).
- the dose of interferon-alpha (IFN- ⁇ ), interferon-gamma (IFN- ⁇ ), honokiol, magnolol, ribavirin, z-guggulsterone (or combination thereof) can vary depending on the size of the patient and dosage form, and the selection of an appropriate dose is within the skill of treating physician. However, typically, the dose will range from about 1 ⁇ g/kg to about 100 mg/kg.
- the inventive method can be employed prophylactically or therapeutically against any Calicivirus, such as Norovirus (e.g., Norwalk virus and others), Sapovirus, Lagovirus,
- Calicivirus such as Norovirus (e.g., Norwalk virus and others), Sapovirus, Lagovirus,
- composition is administered to the patent suitable for effecting prophylaxis or therapeutic treatment of such infections.
- prophylactic use it is not necessary for the inventive method to completely prevent infection, although this is a desired outcome. It is sufficient for prophylaxis to reduce the risk of infection or reduce the severity of any infection.
- therapeutic use of the inventive method need not cure or eliminate the
- Calicivirus infection although this is a preferred result. It is sufficient for the inventive method to reduce the severity or duration of the infection or its symptoms.
- inventive method it is, of course, possible for the inventive method to be performd in repetition, such as daily dosing for a period of time (e.g., a week or so), or dosing 2, 3, or 4 times daily as directed by a health care professional.
- a period of time e.g., a week or so
- dosing 2, 3, or 4 times daily as directed by a health care professional.
- the inventive method is contemplated for use with human patients, the patent also can be a non-human animal, such as a mouse, rat, dog, cat, pig, horse, goat, cow, monkey or ape.
- a non-human animal such as a mouse, rat, dog, cat, pig, horse, goat, cow, monkey or ape.
- HG23 the NV replicon-bearing cells, cells
- a murine macrophage-like RAW267.2 cells were maintained in Dulbecco's minimal essential medium containing 10% fetal bovine serum and antibiotics [chlortetracycline (25 ⁇ g/ml), penicillin (250 U/ml), and streptomycin (250 ⁇ g/ml)] (DMEM- C).
- Murine norovirus- 1 (MNV-I) was kindly provided by Dr. Virgin (Washington University at St Louis, MO), and maintained in RAW267.2 cells.
- Recombinant IFN type 1 human IFN- ⁇ A+IFN- ⁇ D fusion protein
- recombinant human IFN- ⁇ were purchased from Serotec Inc. (Raleigh, NC).
- Polyclonal Antibody specific to NV ProPol was described in previous report ⁇ Chang, 2006 #152 ⁇ .
- Antibodies specific for NPT II or ⁇ -actin were obtained from Santa Cruz biotech (Santa Cruz, CA) or Cell Signaling Tech (Danvers, MA), respectively.
- Ribavirin, Z-gugglesteron, magnolol and MPA were purchased from Sigma (St. Louis, MO), dissolved in distilled water or DMSO, respectively.
- Detection of Norwalk virus RNA and proteins Immunofluorescence assay (IFA).
- the ProPol serum was added to methanol-fixed monolayers of cells, and the binding of antibodies was detected with fluorescein isothiocyanate (FITC)-conjugated, affinity- purified goat antibodies to guinea pig immunoglobulin G (IgG) (ICN Biomedicals, Aurora, OH) as described previously (Chang, Virology 304, 302-10 (2002)).
- FITC fluorescein isothiocyanate
- IgG immunoglobulin G
- RNA in cells was extracted with the RNeasy kit (Qiagen, Valencia, CA).
- RNA transcripts derived from pNV-Neo A standard concentration curve was generated with serial dilutions of RNA transcripts derived from pNV-Neo in each experiment to calculate the total number of genome copies present. The relative genome levels in cells with various treatments were calculated after the RNA levels were normalized with those of ⁇ - actin.
- IFN- ⁇ or IFN- ⁇ Treatment of NV-harboring cells with IFN- ⁇ or IFN- ⁇ .
- NV protein or genome were analyzed with IFA and Western blot analysis, or qRT-PCR, respectively.
- IFA immunofluorescence blot analysis
- qRT-PCR Western blot analysis
- HG23 cells were treated with 2 units/ml (ED50) of IFN- ⁇ and varying concentrations of ribavirin (0-100 ⁇ M), and the reduction of NV protein and genome were compared to that by the ribavirin treatment alone.
- the nonspecific cytotoxic effects in HG23 cells by ribavirin were monitored by measuring the rates of cell propagation in the presence of various concentrations of ribavirin after 24, 48 and 72 hrs of incubation.
- the cells were digested with trypsin and counted after staining with trypan blue for examining the cell toxicity by ribavirin.
- MNV-I can be cultured in the murine macrophage-like cell line RAW267.2 cells
- MNV-I as a surrogate system to examine the effects of ribavirin on norovirus replication in cells.
- Confluent RAW267.2 cells in 6-well plates were treated with varying concentrations (0-100 ⁇ M) of ribavirin for 6 hr before MNV-I was added to the same medium at a MOI of 5. The virus infected cells were further incubated for 24 and 48 h.
- HG23 cells were pre-incubated with 100 ⁇ M of guanosine for 6 hr before ribavirin was added to the medium at various final concentrations (0 [mock-medium] -100 ⁇ M). After 24 or 48 hr of the treatment, the NV protein or genome was analyzed with Western blot analysis or realtime qRT-PCR, respectively, as described above.
- MPA mycophenolic acid
- Various concentrations ranging 0 (mock-medium) to 2 ⁇ M of MPA were added in the medium of semi-confluent HG23 cells for 48 h, and total RNA was extracted for real-time qRT-PCR.
- the comparable expressions of NV genome by MPA were calculated by that of mock treatment.
- HG23 cells were incubated with 0 (mock-medium) or 100 ⁇ M of ribavirin for 48 hr.
- NV-4kF GAGAATGCTAAACATATGAAACCC
- NV-5kR GGGTCCAGAAGATTTGGCGTTC (SEQ ID NO:2)
- HG23 cells were treated with increasing concentrations of human IFN- ⁇ (up to 200 units/ml), and its effect on NV genome and protein expression was monitored by real time qRT-PCR, and IFA and Western blot analysis, respectively.
- IFN- ⁇ the addition of IFN- ⁇ to HG23 cells inhibited NV genome and protein expression in a dose-dependent manner (Fig.l), while the cells themselves showed no toxic effects of the treatment (data not shown).
- the presence of 100 units /ml of IFN- ⁇ for 72 h resulted in nearly complete clearance of the replicon proteins and RNA (Fig. IA and B).
- the reduction rates of NV genome in the presence of IFN- ⁇ was 81%, 73%, 43% and 8% at 2, 10, 50 and 100 units/ml, respectively for 72 h incubation (Fig IA).
- the ED50 of IFN- ⁇ for reducing NV protein (ProPol) and genome copies in HG23 cells at 72 h was calculated to be approximately 40 units/ml (Fig. 1).
- the recombinant human IFN- ⁇ did not show any effects on the expression of NV proteins and genome in the BHK21 -based NV replicon-bearing cells (G3 cells) (data not shown).
- the expression of NPT II was also examined using IFA and Western blot analysis with the antibody against the protein for the evaluation of NV replication. By IFA, the expression of NPT II was well correlated with that of NV ProPol (Fig. IB), indicated that the commercial antibody could be a good tool to study the expression of NV replication in NV replicon bearing cells.
- ribavirin on NV replication.
- a known antiviral agent ribavirin on NV replicon in HG23 cells.
- the HG23 cells were treated with increasing concentrations of ribavirin (up to 400 ⁇ M) for up to 96 h, and its effect on NV protein expression was monitored by IFA, and Western blot analysis using antibody to NV ProPol and the neomycin phosphotransferase.
- qRT-PCR assay was used for detecting NV genome in mock-treated or ribavirin-treated NV replicon-bearing cells.
- HG23 cells treated with ribavirin up to 200 ⁇ M for up to 96 h Minimal cell toxicity was observed in HG23 cells treated with ribavirin up to 200 ⁇ M for up to 96 h.
- the treatment of ribavirin at the concentrations above 10 ⁇ M in HG23 cells reduced the expression of NV genome in a dose-dependent manner (Fig.2A).
- the reduction of NV genome was observed at 24 h of the treatment and increased to 72h.
- the ED50 was calculated to be approximately 40 ⁇ M at 72 h of the ribvirin treatment (Fig. 2A).
- the expression of NV ProPol and the NPT II was reduced in the presence of ribavirin in a dose-dependent manner (Fig.2B and 3B) without nonspecific toxic effects up to 200 ⁇ M.
- the HG23 cells were incubated with ribavirin and IFN- ⁇ (2 unit/ml) to examining any enhanced effects by the co-treatment.
- the treatment of IFN- ⁇ alone at the concentration of 2 units/ml reduced the expression of NPT II and NV genome to about 50% (Fig. 3 A and B).
- ribavirin treatment showed the reduction of NV geneome to 43%, 35% and 18% of the mock-treated cells at 20 ⁇ M, 50 ⁇ M and 100 ⁇ M, repectively, co-treatment with IFN- ⁇ (2 units/ml) resulted in the reduction to 38%, 28% and 10% at the same concentrations of ribavirin (Fig. 3A).
- qRT-PCR assay was used for detecting NV genome in mock-treated or Z-gugglesterone -treated NV replicon-bearing cells.
- Minimal cell toxicity was observed in HG23 cells treated with Z-gugglesterone up to 100 ⁇ M for up to 96 h.
- the treatment of Z-gugglesterone at the concentrations above 10 ⁇ M in HG23 cells reduced the expression of NV genome in a dose-dependent manner.
- the reduction of NV genome was observed at 24 h of the treatment and increased to 72h.
- the ED50 was calculated to be approximately 40 ⁇ M at 72 h of the Z-gugglesterone treatment.
- the expression of NV ProPol and the NPT II was reduced in the presence of Z-gugglesterone in a dose-dependent manner.
- HG23 cells were treated with increasing concentrations of magnolol (up to 200 ⁇ M) for up to 96 h, and its effect on NV protein expression was monitored by IFA, and Western blot analysis using antibody to NV ProPol and the neomycin phosphotransferase. Also qRT-PCR assay was used for detecting NV genome in NV replicon-bearing cells. Minimal cell toxicity was observed in HG23 cells treated with magnolol up to 100 ⁇ M for up to 96 h. The treatment of magnolol at the concentrations above 5 ⁇ M in HG23 cells reduced the expression of NV genome in a dose- dependent manner.
- NV genome The reduction of NV genome was observed at 24 h of the treatment and increased to 72h.
- the ED50 was calculated to be approximately 20 ⁇ M at 72 h of the magnolol treatment.
- the expression of NV ProPol and the NPT II was reduced in the presence of magnolol in a dose-dependent manner.
- ribavirin The effects of ribavirin on the growth of MNV-I.
- Murine norovirus-1 can be cultured in RAW267.2 cells and we examined if ribavirin could reduce the replication of MNV-I.
- nonspecific cytotoxicity of ribavirin in RAW267.2 cells was measured, and there was minimal toxicity at the concentrations below 200 ⁇ M determined by the methods described in the Method section.
- the treatment of ribavirin above 100 ⁇ M reduced the titer of MNV-I at least one log at 24 hr after the virus inoculation (Fig. 4). At 48 h of the treatment, however the virus titers of ribavirin and mock-treated groups were similar each other (data not shown).
- guanosine While the incubation of ribavirin alone for 48 h resulted in the reduction of NV genome level to 90%, 42%, 37% and 8% at 10 ⁇ M, 20 ⁇ M, 50 ⁇ M and 100 ⁇ M, respectively, the addition of guanosine to ribavirin reduced the levels to 100%, 62%, 51% and 34% at the same concentrations of ribavirin (Fig. 5A).
- the potent IMP dehydrogenase inhibitor, MPA also effectively reduced the NV genome levels at the concentrations above 0.2 ⁇ M (Fig. 5B).
- NV genome (the Pol region) in HG23 cells treated and mock-medium or 100 ⁇ M ribavirin for 48 h showed that there is no mutation in the RT-PCR products detected by the direct sequence of the amplicon.
- point mutations occurred 12 or 13 plasmids in the ribavirin treated or non-treated groups, respectively Also total numbers of mutations were 22 or 23 in the ribavirin treated or non-treated groups, respectively C. Discussion
- Ribavirn is a synthetic guanosine analogue and has shown antiviral actions against a range of DNA and RNA viruses including hepatitis C virus and respiratory syncytial virus.
- ribavirin was also very effective to reduce NV replication in the replicon-bearing cells, as ED50 was calculated as approximately 40 ⁇ M.
- Ribavirin also showed the inhibitory effects on the murine norovirsus-1 (MNV-I) in RAW267.2 cells. In the presence of 100 ⁇ M of ribvirin, the titer of MNV-I dropped approximately 10 fold at 24 h after virus infection.
- ribavirin could trigger catastrophic mutations (including fetal mutations) for the mechanism of the antiviral effect (Crotty et al., Nat Med 6:1375-9 (2000)). Sequencing analysis of the conserved polymerase region of NV in the ribavirin-treated (100 ⁇ M) or non-treated groups showed that both groups produced similar rates of mutation. Interestingly, although we did not find any mutations in RT-PCR products, we found high rates of mutations in the clones even without ribavirin. The NV polymerase without proofreading functions may be responsible for the high mutations. This data suggested that the antiviral effects of ribavirin on NV may not be associated with catastrophic mutations in the replicon-bearing cells.
- B-CLL B-cell chronic lymphocytic leukemia
- the broad-spectrum antiviral ribonucleoside ribavirin is an RNA virus mutagen. Nat Med 6:1375-9 (2000)
- Honokiol overcomes conventional drug resistance in human multiple myeloma by induction of caspase-dependent and -independent apoptosis. Blood.
- RNA in Huh-7 cells ribavirin induces mutagenesis in HCV RNA. J Viral Hepat 11 :479-
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Zoology (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Immunology (AREA)
- Virology (AREA)
- Molecular Biology (AREA)
- Communicable Diseases (AREA)
- Oncology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
The invention provides a method of treatment or prophylaxis of Calicivirus infection, comprising administering a pharmaceutical composition comprising a compound selected from the group of compounds consisting of interferon-alpha (IFN-α), interferon-gamma (IFN-γ), honokiol, magnolol, ribavirin, z-guggulsterone, and a combination thereof and a pharmaceutically-acceptable carrier to a patient having a Calicivirus infection, or at risk for becoming infected with a Calicivirus.
Description
INHIBITION OF CALICIVIRUS (NOROVIRUS)
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This patent application claims the benefit of U.S. Provisional Patent Application No. 61/088,187, filed August 12, 2008, which is incorporated by reference.
STATEMENT REGARDING
FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT [0002] This invention was made with government support under COBRE grant 2 P20 RRO 16443 -07 awarded by the National Institutes of Health and under Animal Health Project No. KS481846 awarded by the United States Department of Agriculture. The government has certain rights in the invention.
BACKGROUND OF THE INVENTION
[0003] Caliciviruses (Family Caliciviridae) are small, nonenveloped viruses of 27-35 nm in diameter (Green, K. Y. Chancock, R.M. & Kapikian, A.Z. (2001) in Fields Virology, eds. Knipe, D.M. & Howley, P.M. (Lippincott, Williams & Wilkins, Philidelphia), Vol. 1, pp. 841-874)). They possess a single-strand, plus-sense genomic RNA of 7-8 kb. Calicivirus replication proceeds through a minus strand RNA intermediate, which is used as the template for the synthesis of positive-sense full length genome and subgenomic RNA. This replication is catalyzed by the nonstructural proteins, including the viral RNA dependent RNA polymerase (RdRp), and occurs in cytoplasmic membrane-associated replicase complexes (Green, K. Y. Chancock, R.M. & Kapikian, A.Z. (2001) in Fields Virology, eds. Knipe, D.M. & Howley, P.M. (Lippincott, Williams & Wilkins, Philidelphia), Vol. 1, pp. 841-874)). [0004] Four genera have been established in the family: Norovirus, Sapovirus, Lagovirus, and Vesivirus. (Green et al, J Infect. Dis. 181, Suppl. 2, S322-330 (2000)). These genera have a similar genomic structure, but differ significantly in sequence. Overall amino acid identities between capsid proteins of viruses from different genera are below 30%. There are at least five genogroups (genogroup 1 to 5) in the Norovirus genus. Overall amino acid identities between capsid proteins from same genogroup or different genogroup viruses are about 70% or about 40-50%, repectively. Recent studies showed that noroviruses are responsible for greater than 90% of non-bacterial gastroenteritis outbreaks and are associated
with an estimated 23 million cases of gastroenteritis in the U.S. each year (Mead et al., Emerg Infect Dis 5, 607-25 (1999)).
[0005] Norwalk virus (NV, a prototype norovirus) was originally identified in stools collected from an outbreak of gastroenteritis at a school in Norwalk, Ohio in 1968 (Kapikian et al., J Virol 10, 1075-81 (1972)). The viral genome is -7.7 kb in length and organized into three open reading frames (ORPs 1, 2 and 3) that encode a ~1800-amino acid polyprotein, the major capsid protein (VPl), and a minor capsid protein (VP2), respectively. (Xi et al., Science 250, 1580-3 (1990)). The polyprotein from ORFl is processed by a viral proteinase into final products including the viral RdRp. The structural proteins, VPl and VP2, are synthesized from a subgenomic RNA containing ORFs 2 and 3. Studies of NV replication and the development of antiviral drugs have been severely hampered by the absence of cell culture systems and animal models (Duizer et al., J Gen Virol 85, 79-87 (2004)). Studies of noroviruses associated with disease in humans have been hampered by the continued absence of a cell culture system (Duizer et al., J Gen Virol 85, 79-87 (2004)). Recent advances in norovirus research include the discovery of a murine norovirus that grows in a murine macrophage-like cell line (Karst et al., Science 299(5612): 1575-8 (2003)) and the demonstration that transfection of a full-length cDNA clone of the NV genome (under control of the T7 promoter) into modified vaccinia Ankara (MVA)-T7 infected cells allowed the expression of viral proteins and subsequent NV RNA replication (Asanaka et al., Proc Natl AcadSci USA. 102(29): 10327-32 (2005)). Chang et al. reported the generation of a stable RNA replicon system for NV that functions in both human Huh-7 cells and hamster BHK21 cells, and that circumvents the need for a helper virus (Chang et al., Virology 353, 463-473 (2006)). The replicon-bearing cells expressed NV proteins and RNA, and could be maintained after multiple passages in the presence of G418. Importantly, the replicon- bearing cells could be examined for the effects of potential viral inhibitors, and they provided evidence that NV replication is sensitive to the effects of exogenous interferon (IFN). Virus replicon systems (especially viruses which are fastidious in cells) have proved an important tool in the investigation of virus-host interactions and anti-viral drug developments (Blight, et al., Science 290, 1972-4 (2000); Foy et al., Science 300(5622): 1145-8 (2003); Gale and Foy, Nature 436(7053):939-45 (2005)), and the availability of a NV replicon provides a new system in which such interactions can be assessed for the human noroviruses. NV replicon- bearing cells were shown to be an excellent platform to screen antivirals (Chang et al., Virology 353, 463-473 (2006)).
[0006] Although norovirus infection is generally considered self-limiting and a short- term illness, recent findings showed that the infection could last longer than several days or even several months especially in immunocompromised patients. (Nilsson, M. et al., J Virol 11, 13117-24 (2003)). The treatment options for norovirus infection are limited partly due to the absence of screening systems for antiviral drugs.
[0007] Caliciviruses are plus strand RNA viruses in the family Caliciviridae that consists of four genera, Norovirus, Sapovirus, Lagovirus, and Vesivirus (Green et al., J Infect. Dis. 181, Suppl. 2, S322-330 (2000)). Caliciviruses are important pathogens in humans and animals with wide variety of pathogenicity that ranges gastroenteritis and system infections (Green et al., J Infect. Dis. 181, Suppl. 2, S322-330 (2000)). Viruses in genera Vesivirus and Lagovirus include animal viruses such as vesicular exanthema swine virus, feline calicivirus and rabbit hemorrhagic disease virus. Viruses in the genera Norovirus and Sapovirus cause gastroenteritis in humans and animals (Green et al., Human caliciviruses, p. 841-874. In D. M. Knipe and P. M. Howley (ed.), Fields Virology, vol. 1. Lippincott Williams & Wilkins, Philadelphia, PA (2001).
[0008] Recent studies estimate that human enteric caliciviruses are responsible for more than 90% of non-bacterial gastroenteritis outbreaks (Fankhauser et al., J Infect Dis. 178(6):1571-8 (1998)) and as many as 23 million cases of gastroenteritis in the U.S. each year (Mead, P.S. et al., Emerg Infect Dis 5, 607-25 (1999)). Norwalk virus (NV) is a prototype strain of the noro viruses, and was associated with an outbreak of gastroenteritis in Norwalk, Ohio in 1968 (Kapikian et al., J Virol 10, 1075-81 (1972)). [0009] Studies of the replication of human enteric caliciviruses have been severely hampered by the absence of a cell culture system (Duizer, et al., J Gen Virol 85, 79-87 (2004)). Among the noroviruses, only murine norovirus (strain MNV-I) (Karst et al., Science. 299(5612):1575-8 (2003)) has been successfully propagated in cell culture (Wobus et al., PLoS BiolA2:e432 (2004)). Murine noroviruses present widely in laboratory mouse colonies without apparent clinical symptoms (Hsu et al., Comp Med. 56(4):247-51 (2006); Wobus et al., PLoS Biol Λ2:e432 (2004)). Interestingly MNV-I has a tissue tropism of macrophage-like cells in vivo and in vitro, but it is not clear if human noroviruses target to such cells at present.
[0010] Recently the generation of Norwalk virus (NV) replicon-bearing cells in BHK21 and Huh-7 cells was reported. Interferon (IFN)-α effectively inhibited the replication of NV in the (Chang et al., Virology 353, 463-473 (2006)) was demonstrated. The replicon-bearing
cells were generated by transfecting RNA transcripts derived from a plasmid containing the full length NV genome and neomycin resistant gene (neomycin phosphotransferase II, NPT II) in the place of VPl region (pNV-Neo) (Chang et al., Virology 353, 463-473 (2006)). The replicon bearing cells provide an excellent tool to study the replication of noroviruses and serve a platform to screen potential antiviral drugs.
[0011] U.S. Patent 6,923,992 describes the use of compounds of Bao-Ji-Wan, for anti- diarrhea and relieving gastrointestinal symptoms. However, this patent does not disclose the treatment of any such condition due to Calicivirus. Accordingly, there remains a need for methods for treating or for prophylaxis of Calicivirus infections.
BRIEF SUMMARY OF THE INVENTION
[0012] The invention provides a method of treatment or prophylaxis of Calicivirus infection, comprising administering a pharmaceutical composition comprising a compound selected from the group of compounds consisting of interferon-alpha (IFN-α), interferon- gamma (IFN-γ), honokiol, magnolol, ribavirin, z-guggulsterone, and a combination of any or all thereof (such as 2, 3, 4, 5, or all 6 of the compounds) and a pharmaceutically- acceptable carrierto a patient having a Calicivirus infection, or at risk for becoming infected with a Calicivirus. In another aspect, the invention provides the use of such compounds or such combinations thereof for preparing a medicament for treatment or prophylaxis of Calcivirus infection.
[0013] In another aspect, the invention provides a method of making an inhibitor of a Calicivirus, brining into contact a Calicivirus-cell based replicon system with a compound, identifying a compound that has an inhibitory effect on the Calicivirus replicon, and synthesizing the compound with the inhibitory effect on the Calicivirus replicon. [0014] The present invention may be understood more readily by reference to the following detailed description of preferred embodiments of the invention in conjunction with the accompanying figures.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWING(S) [0015] The accompanying drawings and figures, which are incorporated in and constitute a part of this specification, illustrate several embodiments and together with the description, serve to explain the principles of the invention.
[0016] Fig. 1. The effect of IFN-α and IFN-γ on the NV replication in HG23 cells (A). The effect of IFN-α and IFN-γ on the expression of NV genome. One-day old semi-confluent NV replicon-bearing HG23 cells were incubated with various concentrations of IFN-α and IFN-γ for 72 h, and then total RNA was prepared for real time qRT-PCR for detecting NV genome. The reduction of NV genome by IFNs was calculated by the comparison to that with mock-treatment. (B). The effect of IFN-γ on the expression of NV ProPol or neomycin phosphotranferase in HG23 cells. One-day old semi-confluent HG23 cells were incubated with 0 (mock) or 100 units/ml of IFN-γ for 72 h. The cells were then fixed with 100% methanol, and stained with antibodies to NV ProPol (upper panel) or neomycin phosphotranferase (lower panel) and FITC-conjugated secondary antibody. The negative control includes parental Huh-7 cells with the same staining.
[0017] Fig. 2. The effect of ribavirin on the NV replication in HG23 cells (A). The effect of ribavirin on the expression of NV genome. One-day old semi-confluent HG23 cells were incubated with various concentrations ranging from 0 (mock) to 100 μM of ribavirin for 48, 72 and 96 h, and then total RNA was prepared for real time qRT-PCR for detecting NV genome. The reduction of NV genome by IFNs was calculated by the comparison to that with mock-treatment. (B). The effect of ribavirin on the expression of neomycin phosphotranferase in HG23 cells. One-day old semi-confluent HG23 cells were incubated with various concentrations ranging from 0 (mock) to 200 μM of ribavirin for 72 h. The cells were then fixed with 100% methanol, and stained with antibody to neomycin phosphotranferase and FITC-conjugated secondary antibody. The negative control includes parental Huh-7 cells with the same staining.
[0018] Fig. 3. The combination effects of IFN-α and ribavirin on NV replication in HG23 cells. (A). The effect of ribavirin alone or co-treatment with IFN-α and ribavirin on the expression of NV genome. One-day old semi-confluent HG23 cells were incubated with various concentrations ranging from 0 (mock) to 100 μM of ribavirin with or without 2 units/ml of IFN-α. After 72 h of the incubation, total RNA was prepared for real time qRT- PCR for detecting NV genome. The reduction of NV genome by the treatments was calculated by the comparison to that with mock-treatment. (B). The effect of ribavirin alone or co-treatment with IFN-α and ribavirin on the expression of NPT II detected by Western blot analysis. One-day old semi-confluent HG23 cells were incubated with various concentrations ranging from 0 (mock) to 100 μM of ribavirin with or without 2 units/ml of
IFN-α. After 72 h of the incubation, cell lysate was prepared for Western blot analysis with antibody to neomycin phosphotransferase.
[0019] Fig. 4. The effect of ribavirin on MNV-I in RAW267.2 cells. Confluent RAW267.2 cells in 6- well plates were treated with varying concentrations (0-100 μM) of ribavirin for 6 hr before MNV-I was added to the same medium at a MOI of 5. The virus infected cells were further incubated for 24 h, and the virus replication was measured by the TCID50 assay after the plates were freezing and thawing 3 times.
[0020] Fig. 5. The effect of supplement of guanosine in ribavirin treatment and of MPA on NV replication in HG23 cells. (A) The effect of ribavirin with or without of guanosine (100 μM) on NV genome in HG23 cells. One-day old semi-confluent HG23 cells were incubated with various concentrations ranging from 0 (mock) to 100 μM of ribavirin with or without 100 μM of guanosine. After 72 h of the incubation, total RNA was prepared for real time qRT-PCR for detecting NV genome. The reduction of NV genome by the treatments was calculated by the comparison to that with mock-treatment. (B). The effects of MPA on NV replication. One-day old semi-confluent HG23 cells were incubated with various concentrations ranging from 0 (mock) to 2 μM of MPA for 72 h. After the incubation, total RNA was prepared for real time qRT-PCR for detecting NV genome. The reduction of NV genome by the treatments was calculated by the comparison to that with mock-treatment. [0021] Fig. 6. The effect of magnolol on the NV replication in HG23 cells (A). The effect of magnolol on the expression of NV genome. One-day old semi-confluent HG23 cells were incubated with various concentrations ranging from 0 (mock) to 20 μM of magnolol for 48, 72 and 96 h, and then total RNA was prepared for real time qRT-PCR for detecting NV genome. The reduction of NV genome by magnolol was calculated by the comparison to that with mock-treatment.
[0022] Fig. 7. Chemical structure of z-gugglesterone . [0023] Fig. 8. Chemical structure of rivabirin. [0024] Fig. 9 Chemical structure of IFN-γ. [0025] Fig. 10. Chemical structure of magnolol. [0026] Fig. 11. Chemical structure of honokiol.
DETAILED DESCRIPTION OF THE INVENTION
[0027] Before the present compounds, compositions, articles, devices, and/or methods are disclosed and described, it is to be understood that they are not limited to specific synthetic
methods or specific recombinant biotechnology methods unless otherwise specified, or to particular reagents unless otherwise specified, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
[0028] IFN-γ and ribavirin effectively inhibited the replication of NV in the replicon- bearing cells. Ribavirn (l-β-D-ribofuranosyl-l,2,4-triazole-3-carboxamide) is a synthetic guanosine analogue and has shown antiviral actions against a range of DNA and RJSfA viruses including hepatitis C virus (HCV) and respiratory syncytial virus (De Clercq E, J. Clin Virol. 30(2): 115-33 (2004)). In the chronic HCV infection, combination therapy of IFN-α and ribavirin is the most effective treatment. Similar studies were done in the HCV replicon- bearing cells for the antiviral effects of IFNs, ribavirin and other specific viral inhibitors (Bartenschlager, et al., Antiviral Res. 60(2):91-102 (2003); Pietschmann et al., Curr Opin Drug Discov Devel. 4(5):657-64 (2001)). Interferon-γ is synthesized by various cells including NK cells during the acute inflammations and plays an essential role as a frontier fighter against the invading microbes before specific immune responses elicit (Samuel, Clin Microbiol Rev. 14(4):778-809 (2001)). However, many viruses develop mechanisms against the IFN system to avoid its antiviral effects, which includes blocking STAT pathways and inhibiting the synthesis of IFNs by viral proteins or genome (Samuel, Clin Microbiol Rev. 14(4):778-809 (2001)). Using the replicon-bearing cells, Chang et al demonstrated that NV replicon lacked such anti-IFN mechanism, which may be a reason for the high sensitivity to IFN-α treatment (Chang et al., Virology 353, 463-473 (2006)). This is the first report that IFNs and ribavirin could be excellent antiviral drugs against norovirus replication. Also this report confirms that NV replicon-bearing cells as a significant tool for studying basic research and drug discovery relevant to the control of norovirus gastroenteritis. [0029] The development of effective therapies for noroviral gastroenteritis has been hampered by the absence of a cell culture system. Recently we reported the generation of Norwalk virus (NV) replicon bearing cells in BHK21 and Huh-7 cells, and demonstrated that interferon (IFN)-α effectively inhibited the replication of NV in the cells. In continuing studies for screening potential anti-noroviral agents, we tested IFN-γ, ribavirin, Z- gugglesterone, magnolol for their effects on NV replication in the cells. Like IFN-α, IFN-γ inhibited the replication of NV in the replicon bearing cells showing the reduction of NV genome and proteins in a dose-dependent manner. The effective dose for reducing 50% (ED50) of NV genome and protein was calculated as 50 units/ml. When ribavirin was applied
to the cells, it effectively reduced NV genome and protein with the ED50 calculated as approximately 40 μM. When combined together, IFN-α and ribavirin showed additive effects on the inhibition of NV replication.
[0030] Z guggulsteron as antiviral drugs to treat the gastroenteritis in humans and animals by enteric caliciviruses including noroviruses. Z guggulsteron is available freely at local vitamin stores as a natural product extracted from Indian guggl tree (named as guggul herb, guggul lipid or guggul gum). In this invention, Z gugglesteron which is an antagonist of bile acids on the farnesoid X receptor (FXR) can block the replication of norovirus. Z- gugglesteron inhibited the replication of NV in the replicon bearing cells showing the reduction of NV genome and proteins in a dose-dependent manner. The effective dose for reducing 50% (ED50) of NV genome and protein was calculated as 40 μM. [0031] Magnolol is a bioactive compound found in the bark of the Houpu magnolia (Magnolia officinalis) and is available commercially (e.g., from Sigma-Aldrich). It is known to act on the GABAA receptors in rats, as well as having antifungal properties. Magnolol is a bioactive plant component isolated from the root and stem bark of Magnolia officinalis. Magnolol displays an array of activities including antifungal, 1 antibacterial, and antioxidant effects.2 Magnolol has also been shown act as a natural inhibitor to acyl-CoA: cholesterol acyltransferase (ACAT).3 Magnolol has been shown to demonstrate anti-inflammatory activity by interfering with NF-kB signaling. It also appears to have anti-inflammatory properties related to vascular disorders such as atherosclerosis due to its ability to inhibit IL- 6-induced STAT3 activation.4 Magnolol inhibited the replication of NV in the replicon bearing cells showing the reduction of NV genome and proteins in a dose-dependent manner. The effective dose for reducing 50% (ED50) of NV genome and protein was calculated as 20 μM.
[0032] Honokiol is a structural isomer of Magnolol, the purification and synthesis of has been reported (Amblard et al., J Med Chem. 2006 Jun 1;49(11):3426-7; Esumi et al, Bioorg Med Chem Lett. 2004 May 17;14(10):2621-5; Takeya et al., Chemical & Pharmaceutical Bulletin (1986), 34(5), 2066-70) and is available commercially (e.g., from Sigma-Aldrich). Honokiol has shown pro-apoptotic effects in melanoma, sarcoma, myeloma, leukemia, bladder, lung, prostate, and colon cancer cell lines (Shigemura et al., Cancer. 2007 Apr 1;109(7): 1279-89; Ishitsuka et al., Blood. 2005 Sep l;106(5):1794-800; Battle et al., Blood. 2005 JuI 15;106(2):690-7; Bai et al., J Biol Chem. 2003 Sep 12;278(37):35501-7). Honokiol inhibits phosphorylation of Akt, p44/42mitogen-activated protein kinase (MAPK), and src.
Additionally, honokiol modulates the nuclear factor kappa beta (NfKB) activation pathway, an upstream effector of vascular endothelial growth factor (VEGF), cyclooxygenase 2 (COX- 2), and MCLl, all significant pro-angiogenic and survival factors. Honokiol induces caspase- dependent apoptosis in a TRAIL-mediated manner, and potentiates the pro-apoptotic effects of doxorubicin and other etoposides. So potent is honokiol's pro-apoptotic effects that it overcomes even notoriously drug resistant neoplasms such as multiple myeloma and chronic B-cell leukemia. Honokiol has been shown to promote neurite outgrowth and have neuroprotective effects in rat cortical neurons. Additionally, honokiol increases free cytoplasmic Ca2+ in rat cortical neurons (Fukuyama et al., Bioorg Med Chem Lett. 2002 Apr 22;12(8):1163-6). Honokiol inhibits platelet aggregation in rabbits in a dose-dependent manner, and protects cultured RAEC against oxidized low density liproptein injury. Honokiol significantly increases the prostacyclin metabolite 6-keto-PGFl alpha, potentially the key factor in honokiol's anti-thrombotic activity (Hu et al., Acta Pharmacol Sin. 2005 Sep;26(9): 1063-8.).
[0033] In accordance with the purpose of this invention, as embodied broadly described herein, this invention, in one aspect, relates to the development for potential anti-noroviral agents.
[0034] In conjunction with the inventive method, a pharmaceutical composition comprising a compound selected from the group of compounds consisting of interferon-alpha (IFN-α), interferon-gamma (IFN-γ), honokiol, magnolol, ribavirin, z-guggulsterone, and a combination thereof is administered to a patient for Calicivirus prophylaxis or treatment of a Calicivirus infection. The composition can be formulated for administration to the patient by any desired route. Thus, for example, the composition can be formulated for administration topically (e.g., a cream or ointment), via cubcutaneous, intramuscular, or parenteral injection, inhalation, orally, or via any other traditional route of administration. Where the compositions are formulated for oral administration, such forms can be immediate-release, sustained-release, or extended release forms, such as capsules, caplets, tablets, lozenges, etc. The composition also can be formulated as a powder for oral administration. [0035] It will be observed that standard practice in pharmaceutical compounding can be employed to make a composition suitable for use in the inventive method. Thus, the composition can include suitable binders, diluents, glidants, budders, flavoring agents, preservatives, and other common excipients. (see Rowe et al., "Handbook of Pharmaceutical Excipients" (Pharmaceutical Press), 5th Ed. 2006). Moreover, to the extent that formulations
containing IFN-α, IFN-γ, honokiol, magnolol, ribavirin, or z-guggulsterone (or any combination thereof) are commercially available, such can be employed in the inventive method.
[0036] The dose of interferon-alpha (IFN-α), interferon-gamma (IFN-γ), honokiol, magnolol, ribavirin, z-guggulsterone (or combination thereof) can vary depending on the size of the patient and dosage form, and the selection of an appropriate dose is within the skill of treating physician. However, typically, the dose will range from about 1 μg/kg to about 100 mg/kg.
[0037] The inventive method can be employed prophylactically or therapeutically against any Calicivirus, such as Norovirus (e.g., Norwalk virus and others), Sapovirus, Lagovirus,
Vesivirus. The composition is administered to the patent suitable for effecting prophylaxis or therapeutic treatment of such infections. For prophylactic use, it is not necessary for the inventive method to completely prevent infection, although this is a desired outcome. It is sufficient for prophylaxis to reduce the risk of infection or reduce the severity of any infection. Similarly, therapeutic use of the inventive method need not cure or eliminate the
Calicivirus infection, although this is a preferred result. It is sufficient for the inventive method to reduce the severity or duration of the infection or its symptoms.
[0038] It is, of course, possible for the inventive method to be performd in repetition, such as daily dosing for a period of time (e.g., a week or so), or dosing 2, 3, or 4 times daily as directed by a health care professional.
[0039] While the inventive method is contemplated for use with human patients, the patent also can be a non-human animal, such as a mouse, rat, dog, cat, pig, horse, goat, cow, monkey or ape.
EXAMPLE
A. Materials and Methods
[0040] Cells, viruses, and reagents. The Huh-7, HG23 (the NV replicon-bearing cells, cells) and a murine macrophage-like RAW267.2 cells were maintained in Dulbecco's minimal essential medium containing 10% fetal bovine serum and antibiotics [chlortetracycline (25 μg/ml), penicillin (250 U/ml), and streptomycin (250 μg/ml)] (DMEM- C). Murine norovirus- 1 (MNV-I) was kindly provided by Dr. Virgin (Washington University at St Louis, MO), and maintained in RAW267.2 cells. Recombinant IFN type 1 (human IFN- αA+IFN-αD fusion protein) and recombinant human IFN-γ were purchased from Serotec Inc.
(Raleigh, NC). Polyclonal Antibody specific to NV ProPol was described in previous report {Chang, 2006 #152}. Antibodies specific for NPT II or β-actin were obtained from Santa Cruz biotech (Santa Cruz, CA) or Cell Signaling Tech (Danvers, MA), respectively. Ribavirin, Z-gugglesteron, magnolol and MPA were purchased from Sigma (St. Louis, MO), dissolved in distilled water or DMSO, respectively.
[0041] Detection of Norwalk virus RNA and proteins. Immunofluorescence assay (IFA). The ProPol serum was added to methanol-fixed monolayers of cells, and the binding of antibodies was detected with fluorescein isothiocyanate (FITC)-conjugated, affinity- purified goat antibodies to guinea pig immunoglobulin G (IgG) (ICN Biomedicals, Aurora, OH) as described previously (Chang, Virology 304, 302-10 (2002)). We also used rabbit antiserum specific for the NPT II and FITC-conjugated goat antibodies to rabbit IgG for detecting the presence of NV replicon in cells.
[0042] Western blot analysis. Protein samples of Huh-7 and HG23 cells were prepared in SDS-PAGE sample buffer containing 2% 2-mercaptoethanol, and sonicated for 20 s. The proteins were resolved in a 10% No vex Tris-Bis gel (Invitrogen) and transferred to a nitrocellulose membrane. The membranes were probed with guinea pig antibodies specific for the ProPol protein, and the binding of the antibodies was detected with peroxidase- conjugated, goat anti-guinea pig IgG (Sigma, St .Louis, MO). Or the membranes were probed with rabbit antiserum specific for NPT II and peroxidase-conjugated, goat anti-rabbit IgG. Following incubation with a chemiluminescent substrate (SuperSignal West Pico Chemiluminescent Substrate, Pierce biotechnology, Rockford, IL), the signals were detected with X-ray film. We also used rabbit antiserum specific for the NPT II and peroxidase- conjugated goat antibodies to rabbit IgG for detecting the presence of NV replicon in cells. [0043] Real-Time qRT-PCR. The quantity of NV genome in the replicon-bearing cells was measured by real-time qRT-PCR with the One-step Platinum qRT-PCR kit (Invitrogen), following an established protocol with GI-specific primers and FAM-labeled Gl probes (Kageyama et al., J Clin Microbiol. 41(4): 1548-57 (2003)). For quantity control of cellular RNA, qRT-PCR for the β-actin was performed as described previously (Spann et al., J Virol. 78(8):4363-9 (2004)). For qRT-PCR, the total RNA in cells (in 6-well plates) was extracted with the RNeasy kit (Qiagen, Valencia, CA). A standard concentration curve was generated with serial dilutions of RNA transcripts derived from pNV-Neo in each experiment to calculate the total number of genome copies present. The relative genome levels in cells with
various treatments were calculated after the RNA levels were normalized with those of β- actin.
[0044] Treatment of NV-harboring cells with IFN-α or IFN-γ. The effects of IFN-α or IFN -γ on the replication of NV in the replicon-bearing cells were examined at concentrations ranging from 1 to 20 units/ml (for IFN-α) or 1 to 100 units/ml (for IFN-γ). Varying concentrations of IFN-α or IFN-γ were added to one-day old, 80-90% confluent HG23, and the cells were analyzed for viral protein and genome expression at 24, 48, 72 or 96 h after treatment. The NV protein or genome expression levels were examined by IFA and Western blot analysis, or qRT-PCR, respectively, as described above. Western blot analysis included the detection of β-actin using the specific antibody for the loading control. The inhibitory effect of IFN-α or IFN-γ on the NV replicon was calculated as the concentration of IFN-α or IFN-γ that resulted in 50% reduction of NV genome (ED50) as detected by qRT-PCR. [0045] The effects of ribavirin, gugglesteron, magnolol on the replication of NV in the replicon-bearing cells. One-day old, 80-90% confluent HG23 cells were treated with varying concentrations of ribavirin (0-100 μM) to examine its effects on the replication of NV. After the desired time points, the NV protein or genome were analyzed with IFA and Western blot analysis, or qRT-PCR, respectively. To examine the combinatory effects of ribavirin and IFN-α, one-day old HG23 cells were treated with 2 units/ml (ED50) of IFN-α and varying concentrations of ribavirin (0-100 μM), and the reduction of NV protein and genome were compared to that by the ribavirin treatment alone. The nonspecific cytotoxic effects in HG23 cells by ribavirin were monitored by measuring the rates of cell propagation in the presence of various concentrations of ribavirin after 24, 48 and 72 hrs of incubation. The cells were digested with trypsin and counted after staining with trypan blue for examining the cell toxicity by ribavirin. In addition, we used to cell cytotoxicity assay kit (Promega) for the non-specific cytotoxic effects by ribavirin.
[0046] The effects of ribavirin on the replication of MNV-I in RAW267.2 cells. Because MNV-I can be cultured in the murine macrophage-like cell line RAW267.2 cells, we used MNV-I as a surrogate system to examine the effects of ribavirin on norovirus replication in cells. Confluent RAW267.2 cells in 6-well plates were treated with varying concentrations (0-100 μM) of ribavirin for 6 hr before MNV-I was added to the same medium at a MOI of 5. The virus infected cells were further incubated for 24 and 48 h. The replication of MNV-I in the presence of ribavirin was measured by the TCID50 assay after
the plates were freezing and thawing 3 times. The nonspecific cytotoxic effects in RAW267.2 cells by ribavirin were monitored by the method described above. [0047] The potential mechanisms of ribavirin on the replication of NV. Several mechanisms of actions for the antiviral effects of ribavirin have been suggested and those include the depletion of intracellular guanosine triphosphosphate (GTP) through the inhibition of the cellular inosine monophosphate (IMP) dehydrogenase and triggering catastrophic mutations on virus genome by ribavirin. To examine the potential mechanisms, we performed two experiments: 1) supplement of guanosine in the medium with ribavirin treatment; 2) sequence analysis of NV genome in the presence of absence of ribavirin. First, HG23 cells were pre-incubated with 100 μM of guanosine for 6 hr before ribavirin was added to the medium at various final concentrations (0 [mock-medium] -100 μM). After 24 or 48 hr of the treatment, the NV protein or genome was analyzed with Western blot analysis or realtime qRT-PCR, respectively, as described above. We also tested mycophenolic acid (MPA) which is a potent inhibitor of the cellular IMP dehydrogenase for the expression of NV genome in the replicon-bearing cells. Various concentrations ranging 0 (mock-medium) to 2 μM of MPA were added in the medium of semi-confluent HG23 cells for 48 h, and total RNA was extracted for real-time qRT-PCR. The comparable expressions of NV genome by MPA were calculated by that of mock treatment. To examine if the treatment of ribavirin triggered catastrophic mutagenesis, HG23 cells were incubated with 0 (mock-medium) or 100 μM of ribavirin for 48 hr. After the incubation, total RNA was extracted and RT-PCR was performed to amplify the region encoding NV pol (nucleotide number 4298-5101) with primers of NV-4kF (GAGAATGCTAAACATATGAAACCC (SEQ ID NO: I)) and NV-5kR (GCGGGTCCAGAAGATTTGGCGTTC (SEQ ID NO:2)). Sequence analysis of the amplicon was done either directly or after the products were cloned into pCR2.1 vector (Invitrogen). For the sequencing of the gene in the recombinant vector, we selected 18 clones per each group, and analyzed mutations in the region. The sequence analysis was performed using the GenomeLab DTCS-Quick Start kit (Beckman-Coulter, Fullerton, CA). Sequences were resolved on an automated sequencer (Beckman Coulter).
[0048] Statistical analysis. The effects of IFNs, ribavirin, MPA, and combination of IFN-α and ribavirin on NV replication were analyzed by Student's t test. Results were considered statistically significant when the P value was < 0.05. B. Results
[0049] Effect of IFN-α and IFN-γ on the NV replicon. Previously we reported that IFN-α effectively reduced the expression of NV proteins and genome: the effective dose for IFN-α for reducing NV protein (ProPol) and genome copies in HG23 cells to 50% of that observed in the non-treated (mock) control at 72 h was calculated to be approximately 2 units/ml. Similarly, we examined the effects of IFN-γ on the replication of NV in the cells. The HG23 cells were treated with increasing concentrations of human IFN-γ (up to 200 units/ml), and its effect on NV genome and protein expression was monitored by real time qRT-PCR, and IFA and Western blot analysis, respectively. Like IFN-α, the addition of IFN- γ to HG23 cells inhibited NV genome and protein expression in a dose-dependent manner (Fig.l), while the cells themselves showed no toxic effects of the treatment (data not shown). The presence of 100 units /ml of IFN-γ for 72 h resulted in nearly complete clearance of the replicon proteins and RNA (Fig. IA and B). The reduction rates of NV genome in the presence of IFN-γ was 81%, 73%, 43% and 8% at 2, 10, 50 and 100 units/ml, respectively for 72 h incubation (Fig IA). The ED50 of IFN-γ for reducing NV protein (ProPol) and genome copies in HG23 cells at 72 h was calculated to be approximately 40 units/ml (Fig. 1). However, the recombinant human IFN-γ did not show any effects on the expression of NV proteins and genome in the BHK21 -based NV replicon-bearing cells (G3 cells) (data not shown). The expression of NPT II was also examined using IFA and Western blot analysis with the antibody against the protein for the evaluation of NV replication. By IFA, the expression of NPT II was well correlated with that of NV ProPol (Fig. IB), indicated that the commercial antibody could be a good tool to study the expression of NV replication in NV replicon bearing cells.
[0050] The effects of ribavirin on NV replication. Next, we examined a known antiviral agent, ribavirin on NV replicon in HG23 cells. The HG23 cells were treated with increasing concentrations of ribavirin (up to 400 μM) for up to 96 h, and its effect on NV protein expression was monitored by IFA, and Western blot analysis using antibody to NV ProPol and the neomycin phosphotransferase. Also qRT-PCR assay was used for detecting NV genome in mock-treated or ribavirin-treated NV replicon-bearing cells. Minimal cell toxicity was observed in HG23 cells treated with ribavirin up to 200 μM for up to 96 h. The treatment of ribavirin at the concentrations above 10 μM in HG23 cells reduced the expression of NV genome in a dose-dependent manner (Fig.2A). The reduction of NV genome was observed at 24 h of the treatment and increased to 72h. The ED50 was calculated to be approximately 40 μM at 72 h of the ribvirin treatment (Fig. 2A). Like NV
genome, the expression of NV ProPol and the NPT II was reduced in the presence of ribavirin in a dose-dependent manner (Fig.2B and 3B) without nonspecific toxic effects up to 200 μM. The HG23 cells were incubated with ribavirin and IFN-α (2 unit/ml) to examining any enhanced effects by the co-treatment. The treatment of IFN-α alone at the concentration of 2 units/ml reduced the expression of NPT II and NV genome to about 50% (Fig. 3 A and B). While ribavirin treatment showed the reduction of NV geneome to 43%, 35% and 18% of the mock-treated cells at 20 μM, 50 μM and 100 μM, repectively, co-treatment with IFN-α (2 units/ml) resulted in the reduction to 38%, 28% and 10% at the same concentrations of ribavirin (Fig. 3A). In addition, the similar reduction rates of NTP II in Western blot analysis were observed by the co-treatment (Fig. 3B). These results indicated that there was additive effect by the co-treatment rather than synergistic effects in HG23 cells. [0051] The effects of Z-gugglesteron on NV replication. The HG23 cells were treated with increasing concentrations of Z-gugglesterone (up to 100 μM) for up to 96 h, and its effect on NV protein expression was monitored by IFA, and Western blot analysis using antibody to NV ProPol and the neomycin phosphotransferase. Also qRT-PCR assay was used for detecting NV genome in mock-treated or Z-gugglesterone -treated NV replicon-bearing cells. Minimal cell toxicity was observed in HG23 cells treated with Z-gugglesterone up to 100 μM for up to 96 h. The treatment of Z-gugglesterone at the concentrations above 10 μM in HG23 cells reduced the expression of NV genome in a dose-dependent manner. The reduction of NV genome was observed at 24 h of the treatment and increased to 72h. The ED50 was calculated to be approximately 40 μM at 72 h of the Z-gugglesterone treatment. Like NV genome, the expression of NV ProPol and the NPT II was reduced in the presence of Z-gugglesterone in a dose-dependent manner.
[0052] The effects of magnolol on NV replication. The HG23 cells were treated with increasing concentrations of magnolol (up to 200 μM) for up to 96 h, and its effect on NV protein expression was monitored by IFA, and Western blot analysis using antibody to NV ProPol and the neomycin phosphotransferase. Also qRT-PCR assay was used for detecting NV genome in NV replicon-bearing cells. Minimal cell toxicity was observed in HG23 cells treated with magnolol up to 100 μM for up to 96 h. The treatment of magnolol at the concentrations above 5 μM in HG23 cells reduced the expression of NV genome in a dose- dependent manner. The reduction of NV genome was observed at 24 h of the treatment and increased to 72h. The ED50 was calculated to be approximately 20 μM at 72 h of the
magnolol treatment. Like NV genome, the expression of NV ProPol and the NPT II was reduced in the presence of magnolol in a dose-dependent manner.
[0053] The effects of ribavirin on the growth of MNV-I. Murine norovirus-1 can be cultured in RAW267.2 cells and we examined if ribavirin could reduce the replication of MNV-I. First, nonspecific cytotoxicity of ribavirin in RAW267.2 cells was measured, and there was minimal toxicity at the concentrations below 200 μM determined by the methods described in the Method section. The treatment of ribavirin above 100 μM, reduced the titer of MNV-I at least one log at 24 hr after the virus inoculation (Fig. 4). At 48 h of the treatment, however the virus titers of ribavirin and mock-treated groups were similar each other (data not shown).
[0054] Potential mechanisms of antinoroviral effects by ribavirin. When we examined if the replenishment of guanosine affected the antiviral effects of ribavirin, we found that the addition of 100 μM of guanosin in the ribavirin treated cells, moderately reversed the antiviral effects by ribavirin (Fig. 5A). While the incubation of ribavirin alone for 48 h resulted in the reduction of NV genome level to 90%, 42%, 37% and 8% at 10 μM, 20 μM, 50 μM and 100 μM, respectively, the addition of guanosine to ribavirin reduced the levels to 100%, 62%, 51% and 34% at the same concentrations of ribavirin (Fig. 5A). The potent IMP dehydrogenase inhibitor, MPA also effectively reduced the NV genome levels at the concentrations above 0.2 μM (Fig. 5B). Sequence analysis of NV genome (the Pol region) in HG23 cells treated and mock-medium or 100 μM ribavirin for 48 h showed that there is no mutation in the RT-PCR products detected by the direct sequence of the amplicon. The sequence analysis of the NV Pol region after the amplicon was cloned into pCR2.1 indicated there were no different mutation rates between ribavirin treated and non-treated groups. Among 18 recombinant plasmids each group, point mutations occurred 12 or 13 plasmids in the ribavirin treated or non-treated groups, respectively Also total numbers of mutations were 22 or 23 in the ribavirin treated or non-treated groups, respectively C. Discussion
[0055] Without a cell culture system it is extremely difficult (if not impossible) to study the development of effective therapies for noroviral gastroenteritis. Recent development of NV-replicon bearing cells enabled us to pursue potential anti-noroviral agents. In previous works, we demonstrated that IFN-α was effective to inhibit NV replication, and in this invention, we discovered that IFN-γ also reduced the expression of NV genome and proteins in a dose-dependent manner.
[0056] Type I (IFN-α) and II (IFN-γ) play a central role in innate immunity against virus infections before adapted immunity arises. The functions of IFNs (type I and II) mediated through the interaction with IFN receptors which are expressed in all nucleated cells. The binding of IFNs to their cognitive receptors triggers activation of signal transducers and activator of transcriptions (STATs) and cascade events, which results in the inductions of various antiviral proteins such as PKR and RNase L. However, many viruses armed with anti-IFN mechanism, such as those counteracting the STATs and inhibiting the IFN synthesis. Previously we demonstrated that NV did not have strong anti-IFN mechanism in the replicon-bearing cells and this may be a reason for its high sensitivity to the treatment of IFN-α or IFN-γ in this study. Recently, Marcello et al. reported that both IFN-α and IFN-γ effectively reduced the replication of HCV in the replicon-harboring cells, but with different pathways. They demonstrated that the anti-HCV effect of IFN-γ was independent of type 1 and II IFN receptors and the kinetics of IFN-γ-mediated STAT activation and induction of potential effector genes were distinct from those of IFN-α (Marcello et al., Interferons alpha and lambda inhibit hepatitis C virus replication with distinct signal transduction and gene regulation kinetics. Gastroenterology 131 : 1887-98 (2006)). We plan to conduct similar studies to elucidate the mechanism of anti-NV effects of IFN-α and IFN-γ in HG23 cells. [0057] Ribavirn is a synthetic guanosine analogue and has shown antiviral actions against a range of DNA and RNA viruses including hepatitis C virus and respiratory syncytial virus. In this investigation, we found that ribavirin was also very effective to reduce NV replication in the replicon-bearing cells, as ED50 was calculated as approximately 40 μM. Ribavirin also showed the inhibitory effects on the murine norovirsus-1 (MNV-I) in RAW267.2 cells. In the presence of 100 μM of ribvirin, the titer of MNV-I dropped approximately 10 fold at 24 h after virus infection. However, at 48 h of virus infection, the titers of MNV-I in the presence of absence ribavirin were similar. It was reported that ribavirin was not effective on the replication of feline calicivirus. We are not sure the reason of the discrepancy between MNV- 1 and FCV. It may be related to different viruses or cell types. Ribavirin requires to be metabolized in cells to elicit antiviral activity, and it is possible that there are different metabolite rates of ribavirin among different cells. It has been shown that the effects of combination treatment of IFN and ribavirin were synergistic in various viruses including HCV in the replicon-bearing cells (Kanda et al., J Viral Hepat 11:479-87 (2004)); Tanabe et al., J Infect Dis 189:1129-39 (2004)). However, in this study, we found that when combined together, IFN-α and ribavirin showed only additive effects on the inhibition of NV replication
in the replicon-bearing cells. See Chang, K.O. and George D.W., J. Virol. 81, 12111-12118 (2007).
[0058] Several mechanisms of action for the antiviral activity of ribavirin have been suggested and these include: 1) depletion of intracellular GTP pool by inhibition of the cellular IMP dehydrogenase; 2) induction of catastrophe mutation on viral genome; 3) inhibition of viral polymerase activity, 4) inhibition of viral capping by inhibition of viral or cellular guanylyltransferase activity. The mechanisms of actions may be different in different viruses and it can also be combinational effects with one or two mechanisms predominant. To elucidate potential mechanism of antiviral activity of ribavirin in NV, we tested the first two possibilities in this study. First, we replenish GTP in cells by adding guanosin in the medium in the ribavirin treated cells. In certain viruses (yellow fever virus, human parainfluenza virus 3), addition of guanosin in the medium efficiently reversed the antiviral effects of ribavirin (Leyssen et al., J. Virol. 79:1943-7 (2005)).
[0059] In this investigation, the addition of guanosine in the ribavirin treatment moderately reversed the antiviral effects in the cells (Fig. 5A). We also examined MPA which is a potent uncompetitive inhibitor of IMPDH, and found that MPA effectively inhibit NV replication at the concentrations above 0.2 μM. While the active form of ribavirin is 5'- monophosphate to elicit antiviral activities, the action of MPA is not required to be metabolically activated in the cells. The strong inhibition of NV replication by MPA suggested that the antiviral effects by ribavirin are associated with the inhibition of IMPDH. It has been suggested that ribavirin could trigger catastrophic mutations (including fetal mutations) for the mechanism of the antiviral effect (Crotty et al., Nat Med 6:1375-9 (2000)). Sequencing analysis of the conserved polymerase region of NV in the ribavirin-treated (100 μM) or non-treated groups showed that both groups produced similar rates of mutation. Interestingly, although we did not find any mutations in RT-PCR products, we found high rates of mutations in the clones even without ribavirin. The NV polymerase without proofreading functions may be responsible for the high mutations. This data suggested that the antiviral effects of ribavirin on NV may not be associated with catastrophic mutations in the replicon-bearing cells.
[0060] All references, including publications, patent applications, and patents, cited herein (including the following list) are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
Amblard F, Delinsky D, Arbiser JL, and Schinazi RF. Facile purification of honokiol and its antiviral and cytotoxic properties. J Med Chem. 49(11):3426-7 (2006). Asanaka M, Atmar RL, Ruvolo V, Crawford SE, Neill FH, and Estes MK. Replication and packaging of Norwalk virus RNA in cultured mammalian cells. Proc Natl Acad Sci US
A. 102(29): 10327-32 (2005). Bartenschlager R, and Kaul A, Sparacio S.Replication of the hepatitis C virus in cell culture.
Antiviral Res. 60(2):91-102 (2003). Battle TE, Arbiser J, and Frank DA. The natural product honokiol induces caspase-dependent apoptosis in B-cell chronic lymphocytic leukemia (B-CLL) cells. Blood. 15;106(2):690-
7 (2005). Belliot, G., S. V. Sosnovtsev, T. Mitra, C. Hammer, M. Garfield, and K. Y. Green. In vitro proteolytic processing of the MD 145 noro virus ORFl nonstructural polyprotein yields stable precursors and products similar to those detected in calicivirus-infected cells. J
Virol 77:10957-74 (2003). Blight, K. J., Kolykhalov, A. A. and Rice, CM. Efficient initiation of HCV RNA replication in cell culture. Science 290, 1972-4 (2000). Bradford, M. M. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72:248-54 (1976). Chang, K.O., Kim, Y., Green, K. Y. and Saif, LJ. Cell-culture propagation of porcine enteric calicivirus mediated by intestinal contents is dependent on the cyclic AMP signaling pathway. Virology 304, 302-10 (2002). Chang KO, Sosnovtsev SV, Belliot G, King AD, and Green KY. Stable expression of a
Norwalk virus RNA replicon in a human hepatoma cell line. Virology 353, 463-473
(2006). Chang, K.O. and George D.W. Interferons and ribavirin effectively inhibit Norwalk virus replication in replicon-bearing cells. J. Virol. 81, 12111-12118 (2007) Crotty, S., D. Maag, J. J. Arnold, W. Zhong, J. Y. Lau, Z. Hong, R. Andino, and C. E.
Cameron. The broad-spectrum antiviral ribonucleoside ribavirin is an RNA virus mutagen. Nat Med 6:1375-9 (2000)
De Clercq E. Antiviral drugs in current clinical use. JCHn Virol 30(2): 115-33 (2004). Duizer E, Schwab KJ, Neill FH, Atmar RL, Koopmans MP, and Estes MK. Laboratory efforts to cultivate noroviruses. J Gen Virol 85, 79-87 (2004)
Esumi T, Makado G, Zhai H, Shimizu Y, Mitsumoto Y, and Fukuyama Y. Efficient synthesis and structure-activity relationship of honokiol, a neurotrophic biphenyl-type neolignan.
Bioorg Med Chem Lett. 14(10): 2621-5 (2004). Fankhauser RL, Noel JS, Monroe SS, Ando T, and Glass RI. Molecular epidemiology of
"Norwalk-like viruses" in outbreaks of gastroenteritis in the United States. J Infect Dis.
178(6):1571-8 (1998). Fernandez- Vega, V., Sosnovtsev, SV., Belliot, G., King, A.D., Mitra, T., Gorbalenya, A., and
Green, KY. Norwalk virus N-terminal nonstructural protein is associated with disassembly of the Golgi complex in transfected cells. J Virol. 78, 4827-37 (2004). Foy E, Li K, Wang C, Sumpter R Jr, Ikeda M, Lemon SM, and Gale M Jr. Regulation of interferon regulatory factor-3 by the hepatitis C virus serine protease. Science
300(5622): 1145-8 (2003). Fukuyama Y, Nakade K, Minoshima Y, Yokoyama R, Zhai H, and Mitsumoto Y.
Neurotrophic activity of honokiol on the cultures of fetal rat cortical neurons. Bioorg
Med Chem Lett. 22; 12(8): 1163-6 (2002). Gale M Jr, and Foy EM. Evasion of intracellular host defence by hepatitis C virus. Nature
436(7053):939-45 (2005). Gohara, D. W., C. S. Ha, S. Kumar, B. Ghosh, J. J. Arnold, T. J. Wisniewski, and C. E.
Cameron. Production of "authentic" polio virus RNA-dependent RNA polymerase
(3D(pol)) by ubiquitin-protease-mediated cleavage in Escherichia coli. Protein Expr
Purif. 17:128-38 (1999). Green, K. Y., Ando, T., Balayan, M.S., Berke, T., Clarke, I.N., Estes, M.K., Matson, D.O.,
Nakata, S., Neill, J.D., Studdert, MJ. and Thiel, HJ. J. Infect. Dis. 181, Suppl. 2, S322-
330 (2000). Green, K. Y., G. Belliot, J. L. Taylor, J. Valdesuso, J. F. Lew, A. Z. Kapikian, and F. Y. Lin.
A predominant role for Norwalk-like viruses as agents of epidemic gastroenteritis in
Maryland nursing homes for the elderly. J Infect Dis. 185:133-46 (2002). Green, K. Y., R. M. Chanock, and A. Z. Kapikian. Human caliciviruses, p. 841-874. In D. M.
Knipe and P. M. Howley (ed.), Fields Virology, vol. 1. Lippincott Williams & Wilkins,
Philadelphia, PA (2001). Hsu CC, Riley LK, Wills HM, and Livingston RS. Persistent infection with and serologic cross-reactivity of three novel murine noroviruses. Comp Med. 56(4):247-51 (2006).
Hu H, Zhang XX, Wang YY, and Chen SZ. Honokiol inhibits arterial thrombosis through endothelial cell protection and stimulation of prostacyclin. Acta Pharmacol Sin.
26(9): 1063-8 (2005). Ishitsuka K, Hideshima T, Hamasaki M, Raje N, Kumar S, Hideshima H, Shiraishi N, Yasui
H, Roccaro AM, Richardson P, Podar K, Le Gouill S, Chauhan D, Tamura K, Arbiser J, and Anderson KC. Honokiol overcomes conventional drug resistance in human multiple myeloma by induction of caspase-dependent and -independent apoptosis. Blood.
106(5):1794-800 (2005). Kageyama T, Kojima S, Shinohara M, Uchida K, Fukushi S, Hoshino FB, TakedaN,
Katayama K. Broadly reactive and highly sensitive assay for Norwalk-like viruses based on real-time quantitative reverse transcription-PCR. J Clin Microbiol. 41 (4): 1548-57
(2003). Kanda, T., O. Yokosuka, F. Imazeki, M. Tanaka, Y. Shino, H. Shimada, T. Tomonaga, F.
Nomura, K. Nagao, T. Ochiai, and H. Saisho. Inhibition of subgenomic hepatitis C virus
RNA in Huh-7 cells: ribavirin induces mutagenesis in HCV RNA. J Viral Hepat 11 :479-
87 (2004) Kapikian AZ, Wyatt RG, Dolin R, Thornhill TS, Kalica AR, Chanock RM. Visualization by immune electron microscopy of a 27-nm particle associated with acute infectious nonbacterial gastroenteritis. J Virol 10, 1075-81 (1972) Karst SM, Wobus CE, Lay M, Davidson J, Virgin HW 4th. STATl -dependent innate immunity to a Norwalk-like virus. Science. 299(5612): 1575-8 (2003). Leyssen, P., J. Balzarini, E. De Clercq, and J. Neyts. The predominant mechanism by which ribavirin exerts its antiviral activity in vitro against flaviviruses and paramyxoviruses is mediated by inhibition of IMP dehydrogenase. J Virol 79:1943-7 (2005) Liu, B. L., G. J. Viljoen, I. N. Clarke, and P. R. Lambden. Identification of further proteolytic cleavage sites in the Southampton calicivirus polyprotein by expression of the viral protease in E. coli. J Gen Virol 80:291-6 (1999). Lohmann, V. et al. Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line. Science 285, 110-3 (1999). Lohmann, V., Korner, F., Dobierzewska, A. & Bartenschlager, R. Mutations in hepatitis C virus RNAs conferring cell culture adaptation. J Virol 75, 1437-49 (2001),
Lopez Vazquez, A., J. M. Martin Alonso, R. Casais, J. A. Boga, and F. Parra. Expression of enzymatically active rabbit hemorrhagic disease virus RNA- dependent RNA polymerase in Escherichia coli. J Virol 72:2999-3004 (1998). Lopez Vazquez, A. L., J. M. Martin Alonso, and F. Parra. Characterisation of the RNA- dependent RNA polymerase from Rabbit hemorrhagic disease virus produced in
Escherichia coli. Arch Virol 146:59-69 (2001). Marcello, T., A. Grakoui, G. Barba-Spaeth, E. S. Machlin, S. V. Kotenko, M. R. MacDonald, and C. M. Rice. Interferons alpha and lambda inhibit hepatitis C virus replication with distinct signal transduction and gene regulation kinetics. Gastroenterology 131 :1887-98.
(2006). Mead PS, Slutsker L, Dietz V, McCaig LF, Bresee JS, Shapiro C, Griffin PM, Tauxe RV.
Food-related illness and death in the United States. Emerg Infect Dis 5, 607-25 (1999). Nilsson, M. et al. Evolution of human calicivirus RNA in vivo: accumulation of mutations in the protruding P2 domain of the capsid leads to structural changes and possibly a new phenotype. J Virol 77, 13117-24 (2003). Pietschmann T. and Bartenschlager R. The hepatitis C virus replicon system and its application to molecular studies. Curr Opin Drug Discov Devel. 4(5):657-64 2001. Rowe R, Sheskey P. and Weller P., Handbook of pharmaceutical excipients (Pharmaceutical
Press), 5th Ed. (2006)
Samuel CE. Antiviral actions of interferons. Clin Microbiol Rev. 14(4):778-809 (2001). Sankar, S., and A. G. Porter. Expression, purification, and properties of recombinant encephalomyocarditis virus RNA-dependent RNA polymerase. J Virol 65:2993-3000
(1991). Shigemura K, Arbiser JL, Sun SY, Zayzafoon M, Johnstone PA, Fujisawa M, Gotoh A,
Weksler B, Zhau HE, Chung LW.Honokiol, a natural plant product, inhibits the bone metastatic growth of human prostate cancer cells. Cancer 109(7): 1279-89 (2007). Spann KM, Tran KC, Chi B, Rabin RL, Collins PL. Suppression of the induction of alpha, beta, and lambda interferons by the NSl and NS2 proteins of human respiratory syncytial virus in human epithelial cells and macrophages [corrected]. J Virol.
78(8):4363-9 (2004). Sosnovtsev, S.V., Sosnovtseva, S.A. & Green, K. Y. Cleavage of the feline calicivirus capsid precursor is mediated by a virus-encoded proteinase. J Virol. 72, 3051-9 (1998).
Sosnovtsev, S., and K. Y. Green. RNA transcripts derived from a cloned full-length copy of the feline calicivirus genome do not require VpG for infectivity. Virology 210:383-90
(1995). Takeya T, Takeuchi N, Kasama T, Mayuzumi K, Fukaya H, Tobinaga S. Biphenyls, a new class of compound that inhibits platelet-activating factors. Chem Pharm Bull (Tokyo).
38(2):559-61 (1990). Tanabe, Y., N. Sakamoto, N. Enomoto, M. Kurosaki, E. Ueda, S. Maekawa, T. Yamashiro,
M. Nakagawa, C. H. Chen, N. Kanazawa, S. Kakinuma, and M. Watanabe. 2004.
Synergistic inhibition of intracellular hepatitis C virus replication by combination of ribavirin and interferon- alpha. J Infect Dis 189: 1129-39 Thumfart, J.O. & Meyers, G. Feline calicivirus: recovery of wild-type and recombinant viruses after transfection of cRNA or cDNA constructs. J Virol 76, 6398-407 (2002). Wei, L., J. S. Huhn, A. Mory, H. B. Pathak, S. V. Sosnovtsev, K. Y. Green, and C. E.
Cameron. Proteinase-polymerase precursor as the active form of feline calicivirus RNA- dependent RNA polymerase. J Virol 75:1211-9 (2001). Wobus CE, Karst SM, Thackray LB, Chang KO, Sosnovtsev SV, Belliot G, Krug A,
Mackenzie JM, Green KY, Virgin HW. Replication of Norovirus in cell culture reveals a tropism for dendritic cells and macrophages. PLoS Biol Λ2:e432 (2004). Wyatt, L. S., Moss, B. & Rozenblatt, S. Replication-deficient vaccinia virus encoding bacteriophage T7 RNA polymerase for transient gene expression in mammalian cells.
Virology 210, 202-5 (1995). Xi, J.N., Graham, D.Y., Wang, K.N. & Estes, M.K. Norwalk virus genome cloning and characterization. Science 250, 1580-3 (1990).
[0061] The use of the terms "a" and "an" and "the" and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms "comprising," "having," "including," and "containing" are to be construed as open-ended terms (i.e., meaning "including, but not limited to,") unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods
described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
[0062] Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.
Claims
1. Use of a compound selected from the group of compounds consisting of interferon- alpha (IFN-α), interferon-gamma (IFN-γ), honokiol, magnolol, ribavirin, z-guggulsterone, and a combination of any or all thereof for preparing a medicament for treatment or prophylaxis of Calcivirus infection.
2. The use of claim 1, wherein the medicament is formulated for oral administration.
3. The use of claim 2, wherein the medicament is an immediate-release or a sustained release formulation.
4. The use of claim 2 or 3, wherein the medicament is a tablet, capsule, caplet, or lozenge.
5. The use of any of claims 1-4, wherein the Calicivirus infection comprises Noro virus, Sapovirus, Lagovirus, Vesivirus or any combination thereof.
6. The use of claim 5, wherein the virus is Norwalk Virus.
7. A method for treatment of Calicivirus infection, comprising administering to a patient infected with a Calicivirus, a pharmaceutical composition comprising a compound selected from the group of compounds consisting of interferon-alpha (IFN-α), interferon-gamma (IFN-γ), honokiol, magnolol, ribavirin, z-guggulsterone, and a combination of any or all thereof and a pharmaceutically-acceptable carrier.
8. A method of prophylaxis of Calicivirus infection, comprising administering to a patient at risk for becoming infected with a Calicivirus, a pharmaceutical composition comprising a compound selected from the group of compounds consisting of interferon-alpha (IFN-α), interferon-gamma (IFN-γ), honokiol, magnolol, ribavirin, z-guggulsterone, and a combination of any or all thereof and a pharmaceutically-acceptable carrier.
9. The method of claim 7 or 8, wherein the composition is formulated for oral administration.
10. The method of claim 9, wherein the composition is an immediate-release or a sustained release formulation.
11. The method of claim 9 or 10, wherein the composition is a tablet, capsule, caplet, or lozenge.
12. The method of any of claims 7-11, wherein the Calicivirus infection comprises Norovirus, Sapovirus, Lagovirus, Vesivirus or any combination thereof.
13. The method of claim 12, wherein the virus is Norwalk Virus.
14. The method of any of claims 7-13, wherein the patient is human.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US8818708P | 2008-08-12 | 2008-08-12 | |
US61/088,187 | 2008-08-12 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2010019698A2 true WO2010019698A2 (en) | 2010-02-18 |
WO2010019698A3 WO2010019698A3 (en) | 2010-07-08 |
Family
ID=41669655
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2009/053591 WO2010019698A2 (en) | 2008-08-12 | 2009-08-12 | Inhibition of calicivirus (norovirus) |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2010019698A2 (en) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9959073B1 (en) | 2016-03-30 | 2018-05-01 | EMC IP Holding Company LLC | Detection of host connectivity for data migration in a storage system |
WO2019154958A1 (en) * | 2018-02-08 | 2019-08-15 | Enyo Pharma | Use of modulators of neet proteins for the treatment of infection |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060057159A1 (en) * | 2004-09-13 | 2006-03-16 | Wyeth | Hemorrhagic feline calicivirus, calicivirus vaccine and method for preventing calicivirus infection or disease |
-
2009
- 2009-08-12 WO PCT/US2009/053591 patent/WO2010019698A2/en active Application Filing
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060057159A1 (en) * | 2004-09-13 | 2006-03-16 | Wyeth | Hemorrhagic feline calicivirus, calicivirus vaccine and method for preventing calicivirus infection or disease |
Non-Patent Citations (4)
Title |
---|
AMBLARD F. ET AL: 'Synthesis, cytotoxicity, and antiviral activities of new neolignans related to honokiol and magnolol' BIOORG. MED. CHEM. LETT. vol. 17, no. 16, 15 August 2007, pages 4428 - 4431 * |
BALDWIN S.L. ET AL: 'The biological effects of five feline IFN-alpha subtypes' VET. IMMUNOL. IMMUNOPATHOL. vol. 99, no. 3-4, June 2004, pages 153 - 167 * |
CHANG KO ET AL: 'Bile acids promote the expression of hepatitis C virus in replicon-harboring cells' J. VIROL. vol. 81, no. 18, September 2007, pages 9633 - 9640 * |
SCHIJNS V.E. ET AL: 'Facilitation of antibody forming responses to viral vaccine antigens in young cats by recombinant baculovirus-expressed feline IFN- gamma' VACCINE vol. 20, no. 13-14, 15 March 2002, pages 1718 - 1724 * |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9959073B1 (en) | 2016-03-30 | 2018-05-01 | EMC IP Holding Company LLC | Detection of host connectivity for data migration in a storage system |
WO2019154958A1 (en) * | 2018-02-08 | 2019-08-15 | Enyo Pharma | Use of modulators of neet proteins for the treatment of infection |
Also Published As
Publication number | Publication date |
---|---|
WO2010019698A3 (en) | 2010-07-08 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Todt et al. | The natural compound silvestrol inhibits hepatitis E virus (HEV) replication in vitro and in vivo | |
Kaddoura et al. | COVID-19 therapeutic options under investigation | |
Oo et al. | Deciphering the potential of baicalin as an antiviral agent for Chikungunya virus infection | |
Abdelnabi et al. | Towards antivirals against chikungunya virus | |
Faisal et al. | Alkaloids as potential antivirals. A comprehensive review | |
Zhu et al. | Inhibition of coronavirus infection by a synthetic STING agonist in primary human airway system | |
Duan et al. | Ergosterol peroxide exhibits antiviral and immunomodulatory abilities against porcine deltacoronavirus (PDCoV) via suppression of NF-κB and p38/MAPK signaling pathways in vitro | |
Fan et al. | Activation of interleukin-1β release by the classical swine fever virus is dependent on the NLRP3 inflammasome, which affects virus growth in monocytes | |
EA028605B1 (en) | Combination of oseltamivir and a mek inhibitor for the treatment of viral diseases | |
Wang et al. | Advances in anti-EV-A71 drug development research | |
Dinda et al. | Some natural compounds and their analogues having potent anti-SARS-CoV-2 and anti-proteases activities as lead molecules in drug discovery for COVID-19 | |
CA3152450A1 (en) | Mek inhibitors for the treatment of hantavirus infections | |
Li et al. | Inhibition of porcine epidemic diarrhea virus (PEDV) replication by A77 1726 through targeting JAK and Src tyrosine kinases | |
Zhou et al. | Bergamottin, a bioactive component of bergamot, inhibits SARS-CoV-2 infection in golden Syrian hamsters | |
JP2023521403A (en) | Use of macitinib for the treatment of coronavirus disease 2019 (COVID-19) | |
CN112261946B (en) | Broad-spectrum enterovirus-resistant polypeptide and application thereof | |
US20230295236A1 (en) | Compounds for use in inflammatory conditions | |
Alfajaro et al. | Feline calicivirus-and murine norovirus-induced COX-2/PGE2 signaling pathway has proviral effects | |
WO2010019698A2 (en) | Inhibition of calicivirus (norovirus) | |
Tsai et al. | Targeting heat shock factor 1 as an antiviral strategy against dengue virus replication in vitro and in vivo | |
KR20150048132A (en) | Heterocyclyl carboxamides for treating viral diseases | |
Zhou et al. | Rottlerin plays an antiviral role at early and late steps of Zika virus infection | |
CN105664166A (en) | Composition for treating enterovirus infection and drug combination method | |
EP3860598B1 (en) | Vemurafenib and salts thereof for use in the treatment of enteroviral infections | |
CN112618542A (en) | Use of HSP70 inhibitors for broad spectrum anti-flavivirus activity |
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: 09807244 Country of ref document: EP Kind code of ref document: A2 |
|
NENP | Non-entry into the national phase in: |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 09807244 Country of ref document: EP Kind code of ref document: A2 |