US20190255097A1 - Treatment of alphavirus-induced inflammation - Google Patents
Treatment of alphavirus-induced inflammation Download PDFInfo
- Publication number
- US20190255097A1 US20190255097A1 US16/304,550 US201616304550A US2019255097A1 US 20190255097 A1 US20190255097 A1 US 20190255097A1 US 201616304550 A US201616304550 A US 201616304550A US 2019255097 A1 US2019255097 A1 US 2019255097A1
- Authority
- US
- United States
- Prior art keywords
- pps
- rrv
- virus
- treatment
- alphavirus
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Abandoned
Links
- 206010061218 Inflammation Diseases 0.000 title claims abstract description 32
- 230000004054 inflammatory process Effects 0.000 title claims abstract description 31
- 241000710929 Alphavirus Species 0.000 title claims description 31
- 238000011282 treatment Methods 0.000 title description 72
- 229940043138 pentosan polysulfate Drugs 0.000 claims abstract description 136
- 238000000034 method Methods 0.000 claims abstract description 25
- 241001502567 Chikungunya virus Species 0.000 claims abstract description 24
- 210000003205 muscle Anatomy 0.000 claims abstract description 19
- 206010007710 Cartilage injury Diseases 0.000 claims abstract description 16
- 208000007887 Alphavirus Infections Diseases 0.000 claims abstract description 13
- 206010066919 Epidemic polyarthritis Diseases 0.000 claims abstract description 9
- 241000710942 Ross River virus Species 0.000 claims abstract description 8
- 241000710946 Barmah Forest virus Species 0.000 claims abstract description 6
- 241000700605 Viruses Species 0.000 claims description 12
- 230000037396 body weight Effects 0.000 claims description 6
- 150000003839 salts Chemical class 0.000 claims description 4
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 claims description 2
- 238000007918 intramuscular administration Methods 0.000 claims description 2
- 239000011734 sodium Substances 0.000 claims description 2
- 229910052708 sodium Inorganic materials 0.000 claims description 2
- 159000000000 sodium salts Chemical class 0.000 claims description 2
- 238000010255 intramuscular injection Methods 0.000 claims 2
- 239000007927 intramuscular injection Substances 0.000 claims 2
- 230000002829 reductive effect Effects 0.000 abstract description 20
- 230000003612 virological effect Effects 0.000 abstract description 17
- 230000009467 reduction Effects 0.000 abstract description 13
- 210000001519 tissue Anatomy 0.000 abstract description 13
- 241000699670 Mus sp. Species 0.000 description 59
- 201000010099 disease Diseases 0.000 description 57
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 57
- 208000015181 infectious disease Diseases 0.000 description 38
- 210000000845 cartilage Anatomy 0.000 description 29
- 102000016611 Proteoglycans Human genes 0.000 description 18
- 108010067787 Proteoglycans Proteins 0.000 description 18
- 206010003246 arthritis Diseases 0.000 description 16
- 210000001503 joint Anatomy 0.000 description 16
- 210000004027 cell Anatomy 0.000 description 15
- 238000012360 testing method Methods 0.000 description 14
- 210000001188 articular cartilage Anatomy 0.000 description 13
- 230000000770 proinflammatory effect Effects 0.000 description 13
- 102100036601 Aggrecan core protein Human genes 0.000 description 12
- 108010067219 Aggrecans Proteins 0.000 description 12
- 238000011740 C57BL/6 mouse Methods 0.000 description 12
- 108010035532 Collagen Proteins 0.000 description 11
- 102000008186 Collagen Human genes 0.000 description 11
- 241000699666 Mus <mouse, genus> Species 0.000 description 11
- 229920001436 collagen Polymers 0.000 description 11
- 239000011159 matrix material Substances 0.000 description 11
- 108090000623 proteins and genes Proteins 0.000 description 11
- 108091005664 ADAMTS4 Proteins 0.000 description 10
- 210000005067 joint tissue Anatomy 0.000 description 10
- 210000002540 macrophage Anatomy 0.000 description 10
- 102100027400 A disintegrin and metalloproteinase with thrombospondin motifs 4 Human genes 0.000 description 9
- 238000004458 analytical method Methods 0.000 description 9
- 230000015572 biosynthetic process Effects 0.000 description 9
- 230000006378 damage Effects 0.000 description 9
- 210000002966 serum Anatomy 0.000 description 9
- 108090001005 Interleukin-6 Proteins 0.000 description 8
- 102000004889 Interleukin-6 Human genes 0.000 description 8
- 230000003110 anti-inflammatory effect Effects 0.000 description 8
- 239000003085 diluting agent Substances 0.000 description 8
- 238000002474 experimental method Methods 0.000 description 8
- 230000002458 infectious effect Effects 0.000 description 8
- 230000007774 longterm Effects 0.000 description 8
- 238000003753 real-time PCR Methods 0.000 description 8
- 206010039073 rheumatoid arthritis Diseases 0.000 description 8
- OARRHUQTFTUEOS-UHFFFAOYSA-N safranin Chemical compound [Cl-].C=12C=C(N)C(C)=CC2=NC2=CC(C)=C(N)C=C2[N+]=1C1=CC=CC=C1 OARRHUQTFTUEOS-UHFFFAOYSA-N 0.000 description 8
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 7
- 108091005663 ADAMTS5 Proteins 0.000 description 7
- 102000051389 ADAMTS5 Human genes 0.000 description 7
- 102000004127 Cytokines Human genes 0.000 description 7
- 108090000695 Cytokines Proteins 0.000 description 7
- 241001465754 Metazoa Species 0.000 description 7
- 239000003814 drug Substances 0.000 description 7
- 210000001616 monocyte Anatomy 0.000 description 7
- 210000003314 quadriceps muscle Anatomy 0.000 description 7
- 238000007492 two-way ANOVA Methods 0.000 description 7
- CVYPRDPBCXSVBN-WDZFZDKYSA-N (5z)-5-[[5-[(4-chlorophenyl)methylsulfanyl]-1-methyl-3-(trifluoromethyl)pyrazol-4-yl]methylidene]-2-sulfanylidene-1,3-thiazolidin-4-one Chemical compound C=1C=C(Cl)C=CC=1CSC=1N(C)N=C(C(F)(F)F)C=1\C=C1/SC(=S)NC1=O CVYPRDPBCXSVBN-WDZFZDKYSA-N 0.000 description 6
- 201000002481 Myositis Diseases 0.000 description 6
- 229930040373 Paraformaldehyde Natural products 0.000 description 6
- 108020000999 Viral RNA Proteins 0.000 description 6
- 230000001154 acute effect Effects 0.000 description 6
- 238000003556 assay Methods 0.000 description 6
- 230000000694 effects Effects 0.000 description 6
- 229920002866 paraformaldehyde Polymers 0.000 description 6
- 230000008961 swelling Effects 0.000 description 6
- 206010065687 Bone loss Diseases 0.000 description 5
- RZSYLLSAWYUBPE-UHFFFAOYSA-L Fast green FCF Chemical compound [Na+].[Na+].C=1C=C(C(=C2C=CC(C=C2)=[N+](CC)CC=2C=C(C=CC=2)S([O-])(=O)=O)C=2C(=CC(O)=CC=2)S([O-])(=O)=O)C=CC=1N(CC)CC1=CC=CC(S([O-])(=O)=O)=C1 RZSYLLSAWYUBPE-UHFFFAOYSA-L 0.000 description 5
- 230000001684 chronic effect Effects 0.000 description 5
- 230000003247 decreasing effect Effects 0.000 description 5
- 208000027866 inflammatory disease Diseases 0.000 description 5
- 230000007246 mechanism Effects 0.000 description 5
- 238000010186 staining Methods 0.000 description 5
- 208000024891 symptom Diseases 0.000 description 5
- 238000003786 synthesis reaction Methods 0.000 description 5
- 230000001225 therapeutic effect Effects 0.000 description 5
- 108090000654 Bone morphogenetic protein 1 Proteins 0.000 description 4
- 102100028728 Bone morphogenetic protein 1 Human genes 0.000 description 4
- 229920002683 Glycosaminoglycan Polymers 0.000 description 4
- 108010072582 Matrilin Proteins Proteins 0.000 description 4
- 102000055008 Matrilin Proteins Human genes 0.000 description 4
- 108010015302 Matrix metalloproteinase-9 Proteins 0.000 description 4
- 102100030412 Matrix metalloproteinase-9 Human genes 0.000 description 4
- 102100026261 Metalloproteinase inhibitor 3 Human genes 0.000 description 4
- 101800000515 Non-structural protein 3 Proteins 0.000 description 4
- 238000011529 RT qPCR Methods 0.000 description 4
- 108010031429 Tissue Inhibitor of Metalloproteinase-3 Proteins 0.000 description 4
- 210000003423 ankle Anatomy 0.000 description 4
- 230000008355 cartilage degradation Effects 0.000 description 4
- 230000008859 change Effects 0.000 description 4
- 239000002975 chemoattractant Substances 0.000 description 4
- 230000007423 decrease Effects 0.000 description 4
- 229940079593 drug Drugs 0.000 description 4
- 230000002757 inflammatory effect Effects 0.000 description 4
- 210000000629 knee joint Anatomy 0.000 description 4
- 210000000265 leukocyte Anatomy 0.000 description 4
- 238000004519 manufacturing process Methods 0.000 description 4
- 210000000822 natural killer cell Anatomy 0.000 description 4
- 238000001543 one-way ANOVA Methods 0.000 description 4
- 230000037361 pathway Effects 0.000 description 4
- VKUYLANQOAKALN-UHFFFAOYSA-N 2-[benzyl-(4-methoxyphenyl)sulfonylamino]-n-hydroxy-4-methylpentanamide Chemical compound C1=CC(OC)=CC=C1S(=O)(=O)N(C(CC(C)C)C(=O)NO)CC1=CC=CC=C1 VKUYLANQOAKALN-UHFFFAOYSA-N 0.000 description 3
- 101800001631 3C-like serine proteinase Proteins 0.000 description 3
- 208000006820 Arthralgia Diseases 0.000 description 3
- 201000009182 Chikungunya Diseases 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 101000896557 Homo sapiens Eukaryotic translation initiation factor 3 subunit B Proteins 0.000 description 3
- 101000988834 Homo sapiens Hypoxanthine-guanine phosphoribosyltransferase Proteins 0.000 description 3
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 3
- 102100029098 Hypoxanthine-guanine phosphoribosyltransferase Human genes 0.000 description 3
- 102000002274 Matrix Metalloproteinases Human genes 0.000 description 3
- 108010000684 Matrix Metalloproteinases Proteins 0.000 description 3
- 102000008108 Osteoprotegerin Human genes 0.000 description 3
- 108010035042 Osteoprotegerin Proteins 0.000 description 3
- 241001111421 Pannus Species 0.000 description 3
- 101800004803 Papain-like protease Proteins 0.000 description 3
- 101800002227 Papain-like protease nsp3 Proteins 0.000 description 3
- 101800001074 Papain-like proteinase Proteins 0.000 description 3
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 102100030416 Stromelysin-1 Human genes 0.000 description 3
- 101710108790 Stromelysin-1 Proteins 0.000 description 3
- 210000001744 T-lymphocyte Anatomy 0.000 description 3
- 230000009471 action Effects 0.000 description 3
- 230000004913 activation Effects 0.000 description 3
- 230000004075 alteration Effects 0.000 description 3
- 210000000544 articulatio talocruralis Anatomy 0.000 description 3
- 230000015556 catabolic process Effects 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 210000001612 chondrocyte Anatomy 0.000 description 3
- 238000011161 development Methods 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- 238000000684 flow cytometry Methods 0.000 description 3
- KIUKXJAPPMFGSW-MNSSHETKSA-N hyaluronan Chemical compound CC(=O)N[C@H]1[C@H](O)O[C@H](CO)[C@@H](O)C1O[C@H]1[C@H](O)[C@@H](O)[C@H](O[C@H]2[C@@H](C(O[C@H]3[C@@H]([C@@H](O)[C@H](O)[C@H](O3)C(O)=O)O)[C@H](O)[C@@H](CO)O2)NC(C)=O)[C@@H](C(O)=O)O1 KIUKXJAPPMFGSW-MNSSHETKSA-N 0.000 description 3
- 230000008595 infiltration Effects 0.000 description 3
- 238000001764 infiltration Methods 0.000 description 3
- 208000018937 joint inflammation Diseases 0.000 description 3
- 238000011866 long-term treatment Methods 0.000 description 3
- XXUPLYBCNPLTIW-UHFFFAOYSA-N octadec-7-ynoic acid Chemical compound CCCCCCCCCCC#CCCCCCC(O)=O XXUPLYBCNPLTIW-UHFFFAOYSA-N 0.000 description 3
- 238000011002 quantification Methods 0.000 description 3
- 210000002027 skeletal muscle Anatomy 0.000 description 3
- 239000003981 vehicle Substances 0.000 description 3
- 210000003501 vero cell Anatomy 0.000 description 3
- 230000029812 viral genome replication Effects 0.000 description 3
- 230000009385 viral infection Effects 0.000 description 3
- 230000004580 weight loss Effects 0.000 description 3
- 238000012935 Averaging Methods 0.000 description 2
- 102000007350 Bone Morphogenetic Proteins Human genes 0.000 description 2
- 108010007726 Bone Morphogenetic Proteins Proteins 0.000 description 2
- 102000012422 Collagen Type I Human genes 0.000 description 2
- 108010022452 Collagen Type I Proteins 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 108700039887 Essential Genes Proteins 0.000 description 2
- 108010002350 Interleukin-2 Proteins 0.000 description 2
- 102000000588 Interleukin-2 Human genes 0.000 description 2
- 206010023232 Joint swelling Diseases 0.000 description 2
- 102000005741 Metalloproteases Human genes 0.000 description 2
- 108010006035 Metalloproteases Proteins 0.000 description 2
- 208000023178 Musculoskeletal disease Diseases 0.000 description 2
- 102000014128 RANK Ligand Human genes 0.000 description 2
- 108010025832 RANK Ligand Proteins 0.000 description 2
- 102000005876 Tissue Inhibitor of Metalloproteinases Human genes 0.000 description 2
- 108010005246 Tissue Inhibitor of Metalloproteinases Proteins 0.000 description 2
- 102000046299 Transforming Growth Factor beta1 Human genes 0.000 description 2
- 101800002279 Transforming growth factor beta-1 Proteins 0.000 description 2
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 2
- 102100040247 Tumor necrosis factor Human genes 0.000 description 2
- 239000000427 antigen Substances 0.000 description 2
- 108091007433 antigens Proteins 0.000 description 2
- 102000036639 antigens Human genes 0.000 description 2
- 230000002917 arthritic effect Effects 0.000 description 2
- 210000000988 bone and bone Anatomy 0.000 description 2
- 229940112869 bone morphogenetic protein Drugs 0.000 description 2
- 230000024203 complement activation Effects 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 230000000875 corresponding effect Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 239000003651 drinking water Substances 0.000 description 2
- 235000020188 drinking water Nutrition 0.000 description 2
- 229940088598 enzyme Drugs 0.000 description 2
- 210000002745 epiphysis Anatomy 0.000 description 2
- 210000002683 foot Anatomy 0.000 description 2
- 150000004676 glycans Chemical class 0.000 description 2
- 239000003102 growth factor Substances 0.000 description 2
- 210000004349 growth plate Anatomy 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 210000000548 hind-foot Anatomy 0.000 description 2
- 229920002674 hyaluronan Polymers 0.000 description 2
- 229940099552 hyaluronan Drugs 0.000 description 2
- 230000006698 induction Effects 0.000 description 2
- 210000004969 inflammatory cell Anatomy 0.000 description 2
- 230000004968 inflammatory condition Effects 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 210000003141 lower extremity Anatomy 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 108020004999 messenger RNA Proteins 0.000 description 2
- 239000000203 mixture Substances 0.000 description 2
- 208000017445 musculoskeletal system disease Diseases 0.000 description 2
- 210000000440 neutrophil Anatomy 0.000 description 2
- 230000008520 organization Effects 0.000 description 2
- 201000008482 osteoarthritis Diseases 0.000 description 2
- 210000000963 osteoblast Anatomy 0.000 description 2
- 230000008506 pathogenesis Effects 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 230000001737 promoting effect Effects 0.000 description 2
- XJMOSONTPMZWPB-UHFFFAOYSA-M propidium iodide Chemical compound [I-].[I-].C12=CC(N)=CC=C2C2=CC=C(N)C=C2[N+](CCC[N+](C)(CC)CC)=C1C1=CC=CC=C1 XJMOSONTPMZWPB-UHFFFAOYSA-M 0.000 description 2
- 102000004169 proteins and genes Human genes 0.000 description 2
- 230000008439 repair process Effects 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 239000000523 sample Substances 0.000 description 2
- 238000013207 serial dilution Methods 0.000 description 2
- 230000009885 systemic effect Effects 0.000 description 2
- 210000002303 tibia Anatomy 0.000 description 2
- 230000032258 transport Effects 0.000 description 2
- 230000008957 viral persistence Effects 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- WZUVPPKBWHMQCE-XJKSGUPXSA-N (+)-haematoxylin Chemical compound C12=CC(O)=C(O)C=C2C[C@]2(O)[C@H]1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-XJKSGUPXSA-N 0.000 description 1
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 1
- MTHORRSSURHQPZ-UHFFFAOYSA-N 2-[(1-benzylindazol-3-yl)methoxy]-2-methylpropanoic acid Chemical compound C12=CC=CC=C2C(COC(C)(C)C(O)=O)=NN1CC1=CC=CC=C1 MTHORRSSURHQPZ-UHFFFAOYSA-N 0.000 description 1
- 108091022879 ADAMTS Proteins 0.000 description 1
- 208000030090 Acute Disease Diseases 0.000 description 1
- 206010001842 Alphaviral infections Diseases 0.000 description 1
- 201000001320 Atherosclerosis Diseases 0.000 description 1
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 1
- 206010051728 Bone erosion Diseases 0.000 description 1
- 108700012434 CCL3 Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 102000000013 Chemokine CCL3 Human genes 0.000 description 1
- 102000001327 Chemokine CCL5 Human genes 0.000 description 1
- 108010055166 Chemokine CCL5 Proteins 0.000 description 1
- 102000019034 Chemokines Human genes 0.000 description 1
- 108010012236 Chemokines Proteins 0.000 description 1
- 102000029816 Collagenase Human genes 0.000 description 1
- 108060005980 Collagenase Proteins 0.000 description 1
- 101100506674 Danio rerio hey2 gene Proteins 0.000 description 1
- 102000007260 Deoxyribonuclease I Human genes 0.000 description 1
- 108010008532 Deoxyribonuclease I Proteins 0.000 description 1
- 208000007342 Diabetic Nephropathies Diseases 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 206010063045 Effusion Diseases 0.000 description 1
- 108010008165 Etanercept Proteins 0.000 description 1
- 208000010201 Exanthema Diseases 0.000 description 1
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 1
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 1
- 101710089384 Extracellular protease Proteins 0.000 description 1
- 206010016654 Fibrosis Diseases 0.000 description 1
- 229920002306 Glycocalyx Polymers 0.000 description 1
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 1
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 1
- 102100034221 Growth-regulated alpha protein Human genes 0.000 description 1
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Natural products C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 1
- 101001069921 Homo sapiens Growth-regulated alpha protein Proteins 0.000 description 1
- 101001046686 Homo sapiens Integrin alpha-M Proteins 0.000 description 1
- 102100034343 Integrase Human genes 0.000 description 1
- 102100022338 Integrin alpha-M Human genes 0.000 description 1
- 102000000589 Interleukin-1 Human genes 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- 108010002335 Interleukin-9 Proteins 0.000 description 1
- 102000000585 Interleukin-9 Human genes 0.000 description 1
- 208000005615 Interstitial Cystitis Diseases 0.000 description 1
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 102100026553 Mannose-binding protein C Human genes 0.000 description 1
- 206010027926 Monoplegia Diseases 0.000 description 1
- 101150065958 NR3C1 gene Proteins 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 208000037581 Persistent Infection Diseases 0.000 description 1
- 206010036030 Polyarthritis Diseases 0.000 description 1
- 208000002387 Popliteal pterygium syndrome Diseases 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 1
- 108010090804 Streptavidin Proteins 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 208000004760 Tenosynovitis Diseases 0.000 description 1
- 208000007536 Thrombosis Diseases 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 239000000853 adhesive Substances 0.000 description 1
- 230000001070 adhesive effect Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 239000003146 anticoagulant agent Substances 0.000 description 1
- 229960004676 antithrombotic agent Drugs 0.000 description 1
- 238000003491 array Methods 0.000 description 1
- 230000003286 arthritogenic effect Effects 0.000 description 1
- 238000003149 assay kit Methods 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 229950009949 bindarit Drugs 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 230000005540 biological transmission Effects 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 210000000601 blood cell Anatomy 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 230000022159 cartilage development Effects 0.000 description 1
- 230000006041 cell recruitment Effects 0.000 description 1
- 230000036755 cellular response Effects 0.000 description 1
- 210000000038 chest Anatomy 0.000 description 1
- 230000009816 chondrogenic differentiation Effects 0.000 description 1
- 229960002424 collagenase Drugs 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 230000002089 crippling effect Effects 0.000 description 1
- 230000001351 cycling effect Effects 0.000 description 1
- MSJQCBORNZDNDU-UHFFFAOYSA-D decasodium 3-methoxy-6-[2-(6-methoxy-4,5-disulfonatooxyoxan-3-yl)oxy-5-[5-(5-methoxy-3,4-disulfonatooxyoxan-2-yl)oxy-3,4-disulfonatooxyoxan-2-yl]oxy-4-sulfonatooxyoxan-3-yl]oxy-4,5-disulfonatooxyoxane-2-carboxylate Chemical compound [Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].COC1COC(OC2COC(OC3COC(OC4COC(OC)C(OS([O-])(=O)=O)C4OS([O-])(=O)=O)C(OC4OC(C(OC)C(OS([O-])(=O)=O)C4OS([O-])(=O)=O)C([O-])=O)C3OS([O-])(=O)=O)C(OS([O-])(=O)=O)C2OS([O-])(=O)=O)C(OS([O-])(=O)=O)C1OS([O-])(=O)=O MSJQCBORNZDNDU-UHFFFAOYSA-D 0.000 description 1
- 230000005860 defense response to virus Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000000593 degrading effect Effects 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 208000033679 diabetic kidney disease Diseases 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- BNIILDVGGAEEIG-UHFFFAOYSA-L disodium hydrogen phosphate Chemical compound [Na+].[Na+].OP([O-])([O-])=O BNIILDVGGAEEIG-UHFFFAOYSA-L 0.000 description 1
- 229910000397 disodium phosphate Inorganic materials 0.000 description 1
- 235000019800 disodium phosphate Nutrition 0.000 description 1
- BFMYDTVEBKDAKJ-UHFFFAOYSA-L disodium;(2',7'-dibromo-3',6'-dioxido-3-oxospiro[2-benzofuran-1,9'-xanthene]-4'-yl)mercury;hydrate Chemical compound O.[Na+].[Na+].O1C(=O)C2=CC=CC=C2C21C1=CC(Br)=C([O-])C([Hg])=C1OC1=C2C=C(Br)C([O-])=C1 BFMYDTVEBKDAKJ-UHFFFAOYSA-L 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 229940000406 drug candidate Drugs 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 238000009511 drug repositioning Methods 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 230000008482 dysregulation Effects 0.000 description 1
- 229940043249 elmiron Drugs 0.000 description 1
- 229940073621 enbrel Drugs 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 201000005884 exanthem Diseases 0.000 description 1
- 210000002744 extracellular matrix Anatomy 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 230000004761 fibrosis Effects 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 210000004517 glycocalyx Anatomy 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 230000006749 inflammatory damage Effects 0.000 description 1
- 210000002074 inflammatory monocyte Anatomy 0.000 description 1
- 230000004941 influx Effects 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 229940025708 injectable product Drugs 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 230000015788 innate immune response Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 208000017169 kidney disease Diseases 0.000 description 1
- 210000002414 leg Anatomy 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 210000002901 mesenchymal stem cell Anatomy 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 230000009456 molecular mechanism Effects 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 229940021182 non-steroidal anti-inflammatory drug Drugs 0.000 description 1
- 238000001422 normality test Methods 0.000 description 1
- 210000002997 osteoclast Anatomy 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 208000030428 polyarticular arthritis Diseases 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 101150043515 pps gene Proteins 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 230000000644 propagated effect Effects 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 206010037844 rash Diseases 0.000 description 1
- 108091006084 receptor activators Proteins 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 238000003757 reverse transcription PCR Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 231100000279 safety data Toxicity 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 230000022379 skeletal muscle tissue development Effects 0.000 description 1
- 229910000162 sodium phosphate Inorganic materials 0.000 description 1
- 239000001488 sodium phosphate Substances 0.000 description 1
- 235000011008 sodium phosphates Nutrition 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 210000001258 synovial membrane Anatomy 0.000 description 1
- 201000004595 synovitis Diseases 0.000 description 1
- 229940124598 therapeutic candidate Drugs 0.000 description 1
- 230000000451 tissue damage Effects 0.000 description 1
- 231100000827 tissue damage Toxicity 0.000 description 1
- 230000017423 tissue regeneration Effects 0.000 description 1
- 238000004448 titration Methods 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 1
- 238000002604 ultrasonography Methods 0.000 description 1
- 238000012762 unpaired Student’s t-test Methods 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 239000008215 water for injection Substances 0.000 description 1
Images
Classifications
-
- 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
- A61K31/715—Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
- A61K31/737—Sulfated polysaccharides, e.g. chondroitin sulfate, dermatan sulfate
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0019—Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/02—Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- Y—GENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
- Y02—TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
- Y02A—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
- Y02A50/00—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
- Y02A50/30—Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
Definitions
- This invention relates to the treatment of animals, especially humans, infected with alphaviruses and more particularly to a treatment directed to the reduction of inflammation and/or cartilage damage arising from the infection.
- Arthropod-borne arthritogenic alphaviruses such as Ross River virus (RRV) and chikungunya virus (CHIKV) cause large epidemics of severe musculoskeletal disease. They have been progressively expanding their global distribution, regularly emerging in new regions of the world (1, 2). The hallmark of alphavirus disease is crippling joint pain and arthritis, which often has an extended duration leaving patients bed-ridden and incapacitated. In 2014-2015, CHIKV further expanded its global distribution by entering the Americas and is circulating in several Caribbean islands.
- RRV Ross River virus
- CHIKV chikungunya virus
- RRV antigen has been detected by immunofluorescence in synovial monocytes and macrophages during the early phase of illness (7), and in basal epidermal and eccrine duct epithelia three days after the onset of RRV exanthem (8).
- RRV has also been detected in synovial effusions more than one month after the onset of symptoms, providing evidence of persistent infection in the inflamed synovium (9).
- the synovial space of joints is glycan-rich, containing high levels of glycosaminoglycan (GAGs) frequently linked to protein backbones that form proteoglycan structures.
- GAGs glycosaminoglycan
- Chondrocytes are the major cell type producing the matrix of articular cartilage that is rich in proteoglycans (14).
- proteoglycans 14
- the present invention consists in a method of treating a subject having an alphavirus infection comprising administering parenterally an amount of pentosan polysulfate or a salt thereof effective to reduce alphavirus induced inflammation and/or alphavirus induced cartilage damage in the subject.
- the present invention consists in the use of pentosan polysulfate or a salt thereof in the preparation of a medicament for the treatment of alphavirus induced inflammation and/or to reduce alphavirus induced cartilage damage in a subject having an alphavirus infection.
- the present invention consists in a composition comprising pentosan polysulfate and a pharmaceutically acceptable carrier for use in treating alphavirus induced inflammation and/or to reducing alphavirus induced cartilage damage in a subject having an alphavirus infection.
- the subject to be treated may be an animal, preferably the subject is a human infected with an alphavirus.
- FIG. 1 RRV-infection results in damages to the cartilage in joint tissues.
- FIG. 1 Annotations; (B) bone, (C) cartilage, (P) pannus. Images are representative of at least 5 mice per group (magnification 100 ⁇ ).
- C The width of cartilage from mock- and RRV-infected mice were measured in five areas per mouse and averaged with each point representing one mouse. Cartilage degradation from the medial femoral condyle (MFC) and the medial tibia plateau (MTP) of RRV mice was assessed as outlined in the methods, mock control mice show a score of 1. Data represent mean ⁇ SEM of 5 mice per group.
- D Similarly, at both early time points (i) and at peak disease (ii) total RNA from ankle joint tissues was isolated and analysed for mRNA expression by qRT-PCR.
- FIG. 2 Pentosan polysulfate reduces the severity of acute RRV-induced inflammatory disease.
- PPS treatment reduced the level of disease signs, prevented severe weight loss and reduced the level of inflammatory infiltrates into the joint and muscle tissues protecting the muscle tissue from extensive RRV-induced damage.
- 20-day-old C57BL/6 mice were infected s.c. with 10 4 pfu RRV or mock-infected with diluent alone then either treated daily i.p. with PPS at 3 mg/kg in 100 ⁇ L PBS or mock-treated with PBS alone.
- D Quadriceps muscles were removed from RRV-infected ‘pentosan’ and ‘PBS’ treated mice at day 7 and 10 p.i..
- CD45 hi Total leukocyte
- inflammatory monocyte Gr1 hi CD11b hi
- NK/NKT CD45 hi pan NK hi
- T cell CD3 hi
- FIG. 3 PPS-treatment does not alter the kinetics of viral replication.
- FIG. 4 PPS-treatment alters the expression of soluble factors in RRV-inflammatory disease.
- FIG. 5 Pentosan polysulfate treatment protects the joints from RRV-induced cartilage damage.
- mice 20-day-old C57BL/6 mice were infected s.c. with 10 4 pfu RRV or mock-infected with diluent alone then either treated daily i.p. with PPS at 3 mg/kg in 100 ⁇ L PBS or mock-treated with PBS alone.
- mice were sacrificed at peak disease at 10 days p.i., perfused with 4% PFA, whole legs removed, paraffin-embedded and 5 pm sections generated. Sections were stained with (A) Masson's Trichrome or (B) Safranin 0/fast green and showed an increase in collagen fibres, improvement in the skeletal muscle tissue morphology and protection of the proteoglycan matrix with treatment.
- FIG. 6 Pentosan polysulfate treatment counteracts the dysregulation of the cartilage matrix components caused by RRV-infection.
- joint tissues were removed, RNA extracted and real time PCR performed to evaluate gene regulation of key mediators of the proteoglycan matrix of joint cartilage.
- Results were normalised to the housekeeping gene HPRT1 and are expressed as fold of change compared to the mock control samples. Each data point represents the mean +/ ⁇ standard error of 5 to 6 mice and is representative of two independent experiments. *p ⁇ 0.05, ** p ⁇ 0.01, ***p ⁇ 0.001 using two-way ANOVA with Bonferroni post test.
- FIG. 7 Pentosan-polysulfate reduces the severity of acute CHIKV- inflammation without affecting the kinetics of viral infection.
- mice 25-day-old C57BL/6 mice were infected s.c. with CHIKV or mock-infected with diluent alone then either treated daily i.p. with PPS at 3 mg/kg in 100 ⁇ L PBS or mock-treated with PBS alone.
- CHIKV-induced footpad swelling was assessed daily by measuring the height and width of the perimetatarsal area of the hind foot. PPS treatment resulted in a significant reduction in swelling.
- B H&E stained histological analysis showed PPS treatment decreased the level of inflammatory infiltrates in CHIKV-infected mouse joints at peak swelling 3 days p.i.. Both infectious virus and viral RNA levels were measured indicating PPS-treatment did not affect viral clearance.
- the serum, quadriceps and ankle tissues were harvested, homogenised and the viral load determined by (C) plaque assay on Vero cells for infectious virus or (D) by qPCR for viral RNA in joint tissues with CHIKV E2 specific primers. Each data point represents a single mouse, line indicates the median value. ** p ⁇ 0.01 using two-way ANOVA with Bonferroni post test for foot swelling and plaque assay and Mann Whitney for PCR.
- FIG. 8 PPS-treatment alters soluble factors in CHIKV-inflammatory disease.
- the inflammation arising out of the alphavirus infection is in the joints and/or muscles.
- the inflammation may be confined to the joints or it may be confined to the muscles.
- the inflammation will be in both the joints and the muscles.
- the alphavirus infection will be selected from the group consisting of Ross River virus, chikungunya virus and Barmah Forest virus.
- the present invention in one embodiment is directed towards Ross River virus.
- the present invention in another embodiment is directed towards chikungunya virus.
- the present invention in another embodiment is directed towards Barmah Forest virus.
- administration is intra-articular.
- administration is intra-muscular.
- a daily dose of pentosan polysulfate will be administered. Such dosing may be into one or more joints, one or more muscles, or into both one or more joints and muscles. Alternatively, a single dose may be administered intravenously to treat inflammation in both joints and muscles.
- the amount to be administered is within the range of from 0.1 to 5.0 mg/kg body weight of subject.
- the amount to be administered is within the range of from 1.0 to 5.0 mg/kg body weight of subject.
- the amount to be administered is within the range of from 2.0 to 5.0 mg/kg body weight of subject.
- pentosan polysulfate Treatment of subjects is by administration to the subject of pentosan polysulfate. Owing to its solubility and ready availability, preferably the sodium salt of pentosan polysulfate is used. However, other salts such as magnesium and calcium may also be used.
- Bene-PharmaChem has supplied their PPS in 1 ml glass ampoules containing 100mg PPS/ml. Because of the ready availability of this sterile injectable product it is preferred to be used in the present invention for treating humans.
- the Bene-PharmaChem product comprises: sodium pentosan polysulfate (PPS) 100 mg, sodium phosphate 2.2mg, sodium hydrogen phosphate 6.8mg, adjusted to pH 6.5 with sodium hydroxide and water for injection, USP, qs. 1mL.
- PPS pentosan polysulfate
- a product such as Cartrophen Vet® (Biopharm Australia) may be used.
- mice C57BL/6 wild type mice were obtained from the Animal Resources Centre (Perth, Australia) and bred in- house. All animal experiments were performed in accordance with the guidelines set out by the Griffith University Animal Ethics Committee. C57BL/6 mice of twenty- to twenty-five day old, were inoculated subcutaneously (s.c.) with 10 4 pfu virus. Injections of RRV in PBS to a volume of 50 ⁇ L was in the thorax as described previously (11), and CHIKV in PBS to a volume of 20 ⁇ L in the ventral side of the footpad as described previously (10). Mock-infected mice were inoculated with PBS alone. Mice were weighed and scored for disease signs every 24 h.
- RRV disease scores were assessed based on animal strength and hind-leg paralysis as outlined previously (22). Swelling of the footpad induced by CHIKV was assessed by measuring the height and width of the perimetatarsal area of the hind foot using Kincrome digital vernier calipers.
- PPS treatment was delivered orally by adding to drinking water at a concentration of 100 mg/L which is equivalent to a dose of 25 mg/kg/day based on the daily water consumption of a C57BL/6 mouse as previously reported (23).
- 25 mg/kg/day is the equivalent of the human dose of 2 mg/kg which is the recommended dose for Elmiron®.
- mice were sacrificed and perfused with 4% paraformaldehyde (PFA). Tissues were collected and fixed in 4% PFA, followed by paraffin embedding. Ankles and knee joints were decalcified prior to embedding. Sagittal sections of five micron thickness were prepared and stained with haematoxylin and eosin (H & E), Masson's trichrome or Safranin O/Fast Green.
- PFA paraformaldehyde
- Cartilage thickness and damage was measured at 200 ⁇ magnification from the medial femoral condyle (MFC) and the medial tibial plateau (MTP) by averaging five random points of measurement (separated by at least 20 ⁇ M of distance) per region per mouse and graphed as the mean ⁇ SEM of 5 mice per group.
- Epiphyseal thickness was measured from central sagittal sections by averaging five random points of measurement (separated by at least 20 ⁇ M of distance) per region per mouse and graphed as the mean ⁇ SEM of 5 mice per group system.
- the level of serum cytokines was determined using multiplex bead arrays kits according to the manufacturer's instructions (Bio-Plex Pro Mouse Cytokine 23-plex kits) (Biorad, Hercules, CA). Data was acquired using a Luminex 200TM (Biorad) and analysed using the Bio-plex Manager' 6.1 software (Biorad).
- RNA was prepared from cell pellets using TRIzol (Life Technologies, Victoria, Australia) according to the manufacturer's instructions. Quantification of total RNA was measured by NanoDrop 1000 spectrophotometer (Thermo Scientific, Victoria, Australia). Extracted total RNA (20 ng/ ⁇ L) was reverse-transcribed using an oligo (dT) primer and reverse transcriptase (Sigma Aldrich, Sydney, Australia) according to the manufacturer's instructions.
- TRIzol Life Technologies, Victoria, Australia
- Quantification of total RNA was measured by NanoDrop 1000 spectrophotometer (Thermo Scientific, Victoria, Australia). Extracted total RNA (20 ng/ ⁇ L) was reverse-transcribed using an oligo (dT) primer and reverse transcriptase (Sigma Aldrich, Sydney, Australia) according to the manufacturer's instructions.
- Gene expression- SYBR® Green Real-time PCR was performed using 10 ng of template cDNA on a CFX96 TouchTM Real-Time PCR System in 96-well plates, using QuantiTect Primer Assay kits (Qiagen, Hilden, Germany) for HPRT1, or purchased from primers from Sigma-Aldrich with the sequences outlined in table 1.
- Viral load quantification-Standard curve was generated using serial dilutions of RRV T48 infectious plasmid DNA as described previously (26). Quantification of viral load was performed using SsoAdvanced Universal Probes Supermix (BioRad) in 12.5 ⁇ L reaction volume to detect nsP3 region RNA (table 1) (26).
- the fold change for each gene was calculated as 2 ⁇ Ct .
- Quadriceps muscles were removed and processed as described previously (10). Briefly, tissues were incubated with 3 mg/Ml collagenase IV and 1 mg/mL DNase I in 100 ⁇ L RPMI 1640 at 37° C. for 1.5 h then resuspended in 5 mL RPMI and passed through a 40 ⁇ m cell strainer. Cells were washed, pelleted and treated with 1 ⁇ RBC lysis buffer for 5 min, and counted.
- Fc Block (2.4G2; BD) for 5 min at 4° C.and labelled with fluorochrome-conjugated anti-mouse antibodies, including anti-CD3-FITC (145-2C11, BD), anti-CD19-APC (MB19-1, eBioscience), anti-CD11b-PE (M1/70, BD), anti-Grl-APC (RB6-8C5, eBioscience) and anti-pan-NK/NKT antigen-PE (U5A2-13, BD) in various combinations in the presence of biotinylated anti-CD45 (30-F11, eBioscience), followed by treatment with streptavidin PE-Cy7 at 4° C.for 30 min.
- fluorochrome-conjugated anti-mouse antibodies including anti-CD3-FITC (145-2C11, BD), anti-CD19-APC (MB19-1, eBioscience), anti-CD11b-PE (M1/70, BD), anti-Grl-APC (RB
- Ross River virus infection stimulates the production of proteases ADAMTS-4, MMP-3 and MMP-9 and causes damage to the articular cartilage in joints.
- FIG. 1D i shows that RRV infection results in cartilage degradation and thinning which is associated with arthritic disease symptoms.
- Pentosan polysulfate reduces the severity of RRV-induced disease and inflammation
- mice were infected with RRV or mock-infected with PBS alone and then treated i.p. with either PPS at 3 mg/kg or with vehicle daily.
- PPS treatment resulted in a 65% decrease (p>0.05) in clinical disease score in RRV-infected mice ( FIG. 2A ) and a corresponding protection from disease-associated weight loss (p>0.001) ( FIG. 2B ).
- RRV-infected mock and PPS-treated mice Tissues from RRV-infected mice were collected at the start and end of peak disease (day 7 and 10 p.i.) for histological analysis and flow cytometry. No inflammation was observed in the quadriceps muscle or ankle joint of control mock-infected mock-treated or mock-infected PPS-treated mice ( FIG. 2C ). Consistent with previous studies, RRV-infected mock-treated mice showed extensive inflammation and myositis in quadriceps muscle at day 7 p.i. ( FIG. 2C ,D) and around the ankle joint ( FIG. 2C ) (11, 21, 22, 28). In contrast, RRV-infected PPS-treated mice showed markedly reduced numbers of infiltrating cells in both the muscle and the joint tissues ( FIG. 2C ).
- a range of pro-inflammatory factors and chemoattractants mediate or contribute to alphaviral disease (29).
- PPS-treatment affects the production of soluble immune mediators during RRV infection
- sera from PPS-treated and mock-treated mice were analysed using multiplex and compared to mock-infected, PPS-treated or mock-treated mice.
- RRV infection resulted in an increase (p ⁇ 0.05) of pro-inflammatory factors (both cytokines and chemoattractants) at peak disease ( FIG. 4 ).
- PPS-treatment also altered the levels of the M2 anti-inflammatory cytokine IL-10.
- IL-10 kinetics corresponded to the kinetics of disease with serum levels increasing over time in RRV-infected, mock-treated mice.
- PPS-treatment resulted in an early surge of IL-10 in RRV-infected mice, being significantly elevated at both days 1 and 3 p.i. (p ⁇ 0.001 and 0.01 respectively) ( FIG. 4A ).
- PPS treatment significantly reduced the serum levels of IL-1 a, IL-2, IL-6, CCL-2 and MIP-1 ⁇ at peak disease (p ⁇ 0.05) ( FIG. 4B ).
- Pentosan polysulfate treatment protects the joints from cartilage damage associated with RRV infection.
- PPS treatment also prevented RRV-induced thinning of the epiphyseal plate, protecting against cartilage loss ( FIG. 5A , C).
- Safranin O/Fast Green staining of the cartilage in the knee joint revealed that PPS treatment protected the proteoglycan matrix of the articular cartilage, preventing the loss of articular cartilage observed in RRV-infected mock-treated mice as well as maintaining chondrocyte morphology ( FIG. 5B , C).
- ADAMTS-4 enzyme degradation of cartilage
- ADAMTS-5 ADAMTS-5, MMP-3 and MMP-9
- TGF- ⁇ 1 and BMP-1 stimulation of cartilage protection and synthesis
- aggrecan cartilage matrix proteins
- FIG. 1D RRV-infection caused an increase in ADAMTS-4, which remains elevated at day 3 p.i (p ⁇ 0.01), but drops by the time of peak disease (day 10 p.i), where there was a surge in ADMATS-5.
- RRV- infection also resulted in a late rise (at peak disease) of tissue inhibitor of metalloproteinases (TIMP)-3 (p ⁇ 0.001); known to inhibit both ADAMTS-4 and ADAMTS-5 ( FIG. 6A, 6C ).
- TGF- ⁇ 1 and BMP-1 tissue inhibitor of metalloproteinases
- Pentosan polysulfate treatment is a safe long-term treatment strategy for chronic RRV disease.
- mice were RRV-infected and treated orally with PPS or mock-treated in drinking water.
- Long-term PPS treatment resulted in no adverse clinical signs in the mice for the 3-month duration of the experiment.
- RRV-infected mock-treated mice showed extended disruption of the cartilage components with a three-fold elevation of aggrecan (p ⁇ 0.01) (Table 2).
- PPS-treated mice showed less joint damage and significantly decreased the expression of aggrecan back to base-line levels (p ⁇ 0.001).
- the levels of ADAMTS-4 expression were also reduced (p ⁇ 0.01).
- Pentosan polysulfate is a potential treatment for CHIKV-induced inflammation, reducing disease by altering the cytokine response.
- Alphavirus-induced disease has many similarities to rheumatoid arthritis (RA) including common inflammatory pathways and the key involvement of macrophages (11,12).
- RA rheumatoid arthritis
- the innate immune response is critical in the pathogenesis of alphaviral disease, mediating cell recruitment, viral clearance and inflammation (28, 29, 34).
- monocytes and macrophages are the major cellular contributors to disease progression and severity (29).
- monocytes play a significant role in disease development and cartilage destruction through the production of pro-inflammatory factors. (35).
- RRV-infection leads to an immune poly-arthritis that causes cartilage thinning that contributes to clinical signs associated with alphaviral disease.
- glycans as novel therapeutics has developed momentum in recent years (37).
- pathogens including viruses, exploit host glycans to cause infection.
- the therapeutic potential of this class of molecule to alleviate viral-induced arthritis and inflammatory disease has not been studied and currently remains unknown.
- Pentosan polysulfate is a semisynthetic polysaccharide derivative that chemically and structurally resembles other GAGs, including heparin.
- PPS is bioavailable in both its injectable and oral forms and produces limited toxic side effects, even when administered in high doses (39).
- PPS has been used as an anti-thrombotic agent for several decades, due to its ability to bind preferentially to the glycocalyx of circulating blood cells (40).
- PPS has been identified as having anti-inflammatory properties and is currently approved in the United States for the management of patients with interstitial cystitis, having an excellent long-term safety profile (15).
- injectable forms of PPS are currently used to treat osteoarthritis in veterinary medicine (19).
- IL-10 is part of a repair signal that activates specific cellular and molecular cascades to facilitate tissue recovery (43).
- PPS-treatment altered the kinetics of RRV-induced soluble pro- and anti-inflammatory factors, promoting a swing to anti-inflammatory cytokines with an early induction of IL-10 that enhances myogenesis (43).
- IL-10 inhibits the synthesis of pro-inflammatory soluble factors including, IL-1 ⁇ , IL-2, IL-6, TNF- ⁇ and CCL-2 (45), previously associated with increased severity of alphaviral disease.
- pro-inflammatory soluble factors including, IL-1 ⁇ , IL-2, IL-6, TNF- ⁇ and CCL-2 (45)
- PPS inhibits both the alternative and classical pathways of complement activation (46).
- complement activation is essential for the development of severe RRV disease (34).
- this is specific to RRV-activation of complement via the MBL pathway and is independent of the classical and alternative pathways (28), and therefore it is unlikely that the reduction in RRV disease observed with PPS treatment is due to its effect on complement.
- PPS protection against RRV-induced bone loss is due to its ability to inhibit both IL-6 and CCL-2.
- PPS also stimulates hyaluronan synthesis by synovial fibroblasts and proteoglycan synthesis by chondrocytes (49). This may explain the observed protection of both the articular cartilage and the proteoglycan matrix in PPS-treated RRV-infected mice.
- PPS also promotes the proliferation and chondrogenic differentiation of adult human bone marrow mesenchymal stem cells (50), further explaining the PPS protection of articular cartilage thinning that we see in RRV-infection.
- PPS can repress MMP, including ADAMTS expression and inflammation, as well as NF- ⁇ activation. It also enhances proteoglycan synthesis, including the production of aggrecan and hyaluronan (49) and has been shown to be efficacious as both a treatment and a prophylactic (51).
- MMP including ADAMTS expression and inflammation
- NF- ⁇ activation NF- ⁇ activation
- PPS In humans, long-term clinical use of PPS is extremely well tolerated and highly efficacious for periods greater than 12 months (53). Alphaviruses can produce chronic musculoskeletal ailments over a prolonged period of time. A long-term therapeutic strategy is therefore required for effective treatment of alphavirus-induced arthritis.
- PPS not only alleviates the acute signs of RRV-induced arthritis but also protects the cartilage over the long-term without compromising host viral clearance. Given that PPS promotes an anti- inflammatory immune state without promoting viral persistence, it is an attractive drug-repurposing candidate for the long-term treatment of RRV-associated inflammation and disease.
- non-steroidal anti-inflammatory drugs are the primary therapeutic means to alleviate the symptoms of alphavirus-associated inflammatory disease. These drugs can cause a variety of undesired side effects and may compromise immunity in treated patients (54).
- Studies by our group in the past have examined a number of drug candidates for the treatment of alphavirus disease. Bindarit (a CCL-2 inhibitor), while effective in reducing alphavirus induced arthritis and myositis, is currently not a drug that is available for human use (30, 55).
- Enbrel while available for human use, was found to suppress the antiviral response and enhance viral replication thereby worsen disease (56).
- methotrexate a licensed drug for the treatment of RA
- PPS treatment significantly reduced both the acute clinical signs and the inflammation in the muscle (myositis) and the joint (arthritis) in alphavirus disease.
- PPS has positive and extensive long-term human safety data, and is available as an approved drug by a number of regulatory authorities globally. We therefore conclude that PPS is a promising therapeutic candidate for alphaviral disease; and may also be effective in other infectious inflammatory conditions.
- Pentosan polysulfate promotes proliferation and chondrogenic differentiation of adult human bone marrow-derived mesenchymal precursor cells. Arthritis Res Ther 12:R28.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Chemical & Material Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Organic Chemistry (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Molecular Biology (AREA)
- Virology (AREA)
- Dermatology (AREA)
- Epidemiology (AREA)
- Rheumatology (AREA)
- Physical Education & Sports Medicine (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Communicable Diseases (AREA)
- Oncology (AREA)
- Pain & Pain Management (AREA)
- Immunology (AREA)
- Neurology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
Disclosed is a method of treating alphavirus infections, particularly in humans, in which pentosan polysulfate is administered to an infected subject. Whilst not effecting the viral load in a subject, the pentosan polysulfate acts to reduce inflammation in tissues, such as the muscles, and in the joints of a subject. In addition, cartilage damage in the joints may be reduced. The reduction in inflammation and/or cartilage damage acts to reduce the severe pain experienced by subjects suffering from alphavirus infections, such as Ross River virus, chikungunya virus and Barmah Forest virus.
Description
- This invention relates to the treatment of animals, especially humans, infected with alphaviruses and more particularly to a treatment directed to the reduction of inflammation and/or cartilage damage arising from the infection.
- Arthropod-borne arthritogenic alphaviruses such as Ross River virus (RRV) and chikungunya virus (CHIKV) cause large epidemics of severe musculoskeletal disease. They have been progressively expanding their global distribution, regularly emerging in new regions of the world (1, 2). The hallmark of alphavirus disease is crippling joint pain and arthritis, which often has an extended duration leaving patients bed-ridden and incapacitated. In 2014-2015, CHIKV further expanded its global distribution by entering the Americas and is circulating in several Caribbean islands. As of 23rd May 2016, the Pan American Health Organization (PAHO) reported an estimated total of over 1 5 million cases since 2014, with 100,000 reported in 2016 so far; additionally, the first report of local autochthonous CHIKV transmission in mainland USA was reported in July 2014 (3, 4). Due to the expanding range of alphaviral infections, understanding the mechanisms by which alphaviruses cause debilitating arthritic disease has become increasingly important, especially as there are no specific treatments available (5).
- The severe arthralgia/arthritis in the joints caused by alphaviruses can be both acute and chronic. Ultrasonography of CHIKV patients with joint pain reveals striking tenosynovitis, bone erosion and synovial thickening (6). RRV antigen has been detected by immunofluorescence in synovial monocytes and macrophages during the early phase of illness (7), and in basal epidermal and eccrine duct epithelia three days after the onset of RRV exanthem (8). Using antigen staining and RT-PCR, RRV has also been detected in synovial effusions more than one month after the onset of symptoms, providing evidence of persistent infection in the inflamed synovium (9).
- The synovial space of joints is glycan-rich, containing high levels of glycosaminoglycan (GAGs) frequently linked to protein backbones that form proteoglycan structures. Chondrocytes are the major cell type producing the matrix of articular cartilage that is rich in proteoglycans (14). However, there have been no studies to elucidate the impact of alphaviruses on cartilage and the proteoglycan matrix of the joint.
- In the work described herein, we show RRV infection results in similar histopathology of the joint to that observed in rheumatoid arthritis (RA). This includes pannus-like formation, immune infiltration and cartilage damage. We further show that treatment with pentosan polysulfate (PPS) ameliorates the severity of both RRV and CHIKV clinical disease, overall reduction in both immune infiltrates and soluble pro- inflammatory factors. We also observed a change in the kinetics of the soluble factors involved in macrophage activation. In RRV-infection treatment also reduced the loss of articular cartilage and protected the level of proteoglycans in the cartilage matrix, altering the expression of cartilage components including aggrecan and collagen in. Overall we show that PPS is a safe and effective treatment for both acute and chronic RRV-infection.
- Accordingly the present invention consists in a method of treating a subject having an alphavirus infection comprising administering parenterally an amount of pentosan polysulfate or a salt thereof effective to reduce alphavirus induced inflammation and/or alphavirus induced cartilage damage in the subject.
- In another aspect, the present invention consists in the use of pentosan polysulfate or a salt thereof in the preparation of a medicament for the treatment of alphavirus induced inflammation and/or to reduce alphavirus induced cartilage damage in a subject having an alphavirus infection.
- In a further aspect, the present invention consists in a composition comprising pentosan polysulfate and a pharmaceutically acceptable carrier for use in treating alphavirus induced inflammation and/or to reducing alphavirus induced cartilage damage in a subject having an alphavirus infection.
- Whilst the subject to be treated may be an animal, preferably the subject is a human infected with an alphavirus.
-
FIG. 1 . RRV-infection results in damages to the cartilage in joint tissues. - 20-day-old C57BL/6 mice were infected s.c. With 104 pfu RRV or mock-infected with diluent alone. Infection resulted in extensive inflammation, pannus formation, articular cartilage thinning, disruption of the proteoglycans and upregulation of cartilage associated genes. (A, B) For histological analysis the joints of RRV-infected mice were sacrificed at peak disease at 10 days p.i., perfused with 4% PFA, knee joint tissues removed, paraffin-embedded and 5 μm sections generated. Sections were stained with (A) H&E or (B) Safranin O/fast green. Annotations; (B) bone, (C) cartilage, (P) pannus. Images are representative of at least 5 mice per group (
magnification 100×). (C) The width of cartilage from mock- and RRV-infected mice were measured in five areas per mouse and averaged with each point representing one mouse. Cartilage degradation from the medial femoral condyle (MFC) and the medial tibia plateau (MTP) of RRV mice was assessed as outlined in the methods, mock control mice show a score of 1. Data represent mean ±SEM of 5 mice per group. (D) Similarly, at both early time points (i) and at peak disease (ii) total RNA from ankle joint tissues was isolated and analysed for mRNA expression by qRT-PCR. Data was normalised to the housekeeping gene HPRT1 and expressed as relative expression compared to mock-infected controls (as represented by the dashed line). Each bar represents the mean +/− the SEM for 5-6 mice per group, *p<0.05, ***p<0.001 one-way ANOVA Dunnett's post test. -
FIG. 2 . Pentosan polysulfate reduces the severity of acute RRV-induced inflammatory disease. - PPS treatment reduced the level of disease signs, prevented severe weight loss and reduced the level of inflammatory infiltrates into the joint and muscle tissues protecting the muscle tissue from extensive RRV-induced damage. 20-day-old C57BL/6 mice were infected s.c. with 104 pfu RRV or mock-infected with diluent alone then either treated daily i.p. with PPS at 3 mg/kg in 100 μL PBS or mock-treated with PBS alone. (A) Mice were scored for the development of hind-limb dysfunction and displayed a reduction in RRV-disease severity with PPS treatment. Mock-infected mice were scored zero for the duration of the experiment *p<0.05, **p<0.01 using a Mann-Whitney test. (B) Weight was monitored at 24-hour intervals ***p<0.001 significantly reduced weight loss of RRV-infected PPS-treated compared to RRV-infected mock-treated -using two-way ANOVA with Bonferroni post test. (C) For histological analysis, mice were sacrificed at 7 or 10 days p.i., perfused with 4% PFA, quadriceps and knee joint tissues removed, paraffin-embedded, 5 μm sections generated. Sections were stained with H&E. (D) Quadriceps muscles were removed from RRV-infected ‘pentosan’ and ‘PBS’ treated mice at
day -
FIG. 3 . PPS-treatment does not alter the kinetics of viral replication. - Both infectious virus and viral RNA levels were measured indicating PPS-treatment did not affect viral clearance. Mice were infected s.c. with RRV or mock-infected with diluent alone then either treated daily i.p. with PPS or mock-treated with PBS alone. At
days -
FIG. 4 . PPS-treatment alters the expression of soluble factors in RRV-inflammatory disease. - 20-day-old C57BL/6 mice were infected s.c. with 104 pfu RRV or mock-infected with diluent alone then either treated daily i.p. with PPS at 3 mg/kg in 100 μL PBS or mock-treated with PBS alone (mock-infected mock-treated=Mock control, mock-infected PPS-treated=PPS alone, RRV-infected mock-treated=RRV alone, RRV-infected PPS-treated=RRV+PPS). At
days day 10 p.i., (B) increased levels of chemo-attractants factors atday 1 p.i. and (C) altered the kinetics of M2 cytokine IL-10. Red line indicates the level of mock control. Each data point represents the mean +/− standard error of 5 to 6 mice. (A, B) *p<0.05, ** p<0.01, ***p<0.001 one-way ANOVA with Tukey's post test. (C) *p<0.05, ** p<0.01, ***p<0.001 using two-way ANOVA with Bonferroni post test. Astrices directly on top of bars are compared to mock control levels. -
FIG. 5 . Pentosan polysulfate treatment protects the joints from RRV-induced cartilage damage. - 20-day-old C57BL/6 mice were infected s.c. with 104 pfu RRV or mock-infected with diluent alone then either treated daily i.p. with PPS at 3 mg/kg in 100 μL PBS or mock-treated with PBS alone. For histological analysis, mice were sacrificed at peak disease at 10 days p.i., perfused with 4% PFA, whole legs removed, paraffin-embedded and 5 pm sections generated. Sections were stained with (A) Masson's Trichrome or (B) Safranin 0/fast green and showed an increase in collagen fibres, improvement in the skeletal muscle tissue morphology and protection of the proteoglycan matrix with treatment. Annotations; (B) bone, (C) cartilage, (P) pannus, (M) muscle. Images are representative of at least 5-8 mice per group. (C) The width of cartilage and epiphyseal plate from mice were measured in five areas of per mouse and averaged with each point representing one mouse. Cartilage degradation from the medial femoral condyle (MFC) and the medial tibia plateau (MTP) of at least five mice per group was assessed as outlined in the methods with mock control mice showing a score of 1. Data represent mean ±SEM of 5 mice per group.
-
FIG. 6 . Pentosan polysulfate treatment counteracts the dysregulation of the cartilage matrix components caused by RRV-infection. - PPS treatment significantly reduced the early expression of aggrecan and collagen II and the expression of ADAMTS-5 and TIMP-3 at peak disease. 20-day-old C57BL/6 mice were infected s.c. with 104 pfu RRV or mock-infected with diluent alone then either treated daily i.p. with PPS at 3 mg/kg in 100 μL PBS or mock-treated with PBS alone (mock-infected mock-treated=Mock control, mock-infected PPS-treated=PPS alone, RRV-infected mock- treated=RRV alone, RRV-infected PPS-treated=RRV+PPS). At
days -
FIG. 7 . Pentosan-polysulfate reduces the severity of acute CHIKV- inflammation without affecting the kinetics of viral infection. - 25-day-old C57BL/6 mice were infected s.c. with CHIKV or mock-infected with diluent alone then either treated daily i.p. with PPS at 3 mg/kg in 100 μL PBS or mock-treated with PBS alone. (A) CHIKV-induced footpad swelling was assessed daily by measuring the height and width of the perimetatarsal area of the hind foot. PPS treatment resulted in a significant reduction in swelling. (B) H&E stained histological analysis showed PPS treatment decreased the level of inflammatory infiltrates in CHIKV-infected mouse joints at
peak swelling 3 days p.i.. Both infectious virus and viral RNA levels were measured indicating PPS-treatment did not affect viral clearance. Atdays -
FIG. 8 . PPS-treatment alters soluble factors in CHIKV-inflammatory disease. - 25-day-old C57BL/6 mice were infected with CHIKV or diluent alone then either treated daily i.p. with PPS or mock-treated with PBS alone. (A) Kinetics of IL-10 were altered with PPS-treatment. Grey line indicates the level of mock control. Each data point represents the mean +/− standard error of 5 to 6 mice. *p<0.05, ** p<0.01, ***p<0.001 using two-way ANOVA with Bonferroni post test. Astrices directly on top of bars are compared to mock control levels. (B) Levels pro-inflammatory factors were decreased at peak swelling by
day 3 p.i. with PPS treatment *p<0.05, ** p<0.01, ***p<0.001 one-way ANOVA with Tukey's post test. - The inflammation arising out of the alphavirus infection is in the joints and/or muscles. Alternatively, the inflammation may be confined to the joints or it may be confined to the muscles. In some embodiments, the inflammation will be in both the joints and the muscles.
- Although the present invention is directed towards alphavirus infections generally, in some embodiments, the alphavirus infection will be selected from the group consisting of Ross River virus, chikungunya virus and Barmah Forest virus.
- In particular, the present invention in one embodiment is directed towards Ross River virus.
- In particular, the present invention in another embodiment is directed towards chikungunya virus.
- In particular, the present invention in another embodiment is directed towards Barmah Forest virus.
- Administration of the effective amount of pentosan polysulfate to a subject infected with an alphavirus is parenteral.
- In some embodiments when a joint is suffering from inflammation, administration is intra-articular.
- In other embodiments when a muscle is suffering from inflammation, administration is intra-muscular.
- Generally, a daily dose of pentosan polysulfate will be administered. Such dosing may be into one or more joints, one or more muscles, or into both one or more joints and muscles. Alternatively, a single dose may be administered intravenously to treat inflammation in both joints and muscles.
- It is, however, within the scope of the invention to administer on two more occasions on a daily basis, depending on the severity of the symptoms of the subject.
- Of course it will be recognised that treatment to reduce cartilage damage is undertaken by intra-articular or intravenous administration.
- Based on the animal studies disclosed herein, the amount to be administered is within the range of from 0.1 to 5.0 mg/kg body weight of subject.
- In some embodiments, the amount to be administered is within the range of from 1.0 to 5.0 mg/kg body weight of subject.
- In some embodiments, the amount to be administered is within the range of from 2.0 to 5.0 mg/kg body weight of subject.
- Treatment of subjects is by administration to the subject of pentosan polysulfate. Owing to its solubility and ready availability, preferably the sodium salt of pentosan polysulfate is used. However, other salts such as magnesium and calcium may also be used.
- Commercially, Bene-PharmaChem has supplied their PPS in 1 ml glass ampoules containing 100mg PPS/ml. Because of the ready availability of this sterile injectable product it is preferred to be used in the present invention for treating humans.
- The Bene-PharmaChem product comprises: sodium pentosan polysulfate (PPS) 100 mg, sodium phosphate 2.2mg, sodium hydrogen phosphate 6.8mg, adjusted to pH 6.5 with sodium hydroxide and water for injection, USP, qs. 1mL.
- It is, however, within the scope of this invention to use alternative formulations. For example, standard textbooks in the field of this invention, such as Remington's Practice of Pharmacy teach such alternatives.
- Likewise, for veterinary applications, a product such as Cartrophen Vet® (Biopharm Australia) may be used.
- In order to better understand the nature of this invention, set out below is a description of a series of experiments carried out to demonstrate the effectiveness of PPS in the treatment of subjects infected with an alphavirus and suffering from joint and/or muscle inflammation and cartilage damage.
- Methods
- Virus and Cells. Stocks of the wild-type T48 strain of RRV were generated from the full- length T48 cDNA clone (kindly provided by Dr Richard Kuhn, Purdue University) (20). Stocks of CHIKV Mauritius strain were propagated in BHK-21 cells. All titrations were performed by plaque assay on Vero cells as described previously (21).
- Mice. C57BL/6 wild type (WT) mice were obtained from the Animal Resources Centre (Perth, Australia) and bred in- house. All animal experiments were performed in accordance with the guidelines set out by the Griffith University Animal Ethics Committee. C57BL/6 mice of twenty- to twenty-five day old, were inoculated subcutaneously (s.c.) with 104 pfu virus. Injections of RRV in PBS to a volume of 50 μL was in the thorax as described previously (11), and CHIKV in PBS to a volume of 20 μL in the ventral side of the footpad as described previously (10). Mock-infected mice were inoculated with PBS alone. Mice were weighed and scored for disease signs every 24 h. RRV disease scores were assessed based on animal strength and hind-leg paralysis as outlined previously (22). Swelling of the footpad induced by CHIKV was assessed by measuring the height and width of the perimetatarsal area of the hind foot using Kincrome digital vernier calipers.
- Treatment with pentosan polysulfate (Cartrophen Vet®, Biopharm Australia) or vehicle alone was given intraperitoneally (i.p.) at 3 mg/kg in 100 μL PBS (vehicle), daily for the duration of the experiment, commencing from the day of virus infection. In long-term experiments, PPS treatment was delivered orally by adding to drinking water at a concentration of 100 mg/L which is equivalent to a dose of 25 mg/kg/day based on the daily water consumption of a C57BL/6 mouse as previously reported (23). Using the human to animal conversion outlined by Regan-Shaw et al., (24) 25 mg/kg/day is the equivalent of the human dose of 2 mg/kg which is the recommended dose for Elmiron®.
- Histology. Mice were sacrificed and perfused with 4% paraformaldehyde (PFA). Tissues were collected and fixed in 4% PFA, followed by paraffin embedding. Ankles and knee joints were decalcified prior to embedding. Sagittal sections of five micron thickness were prepared and stained with haematoxylin and eosin (H & E), Masson's trichrome or Safranin O/Fast Green. Cartilage thickness and damage was measured at 200× magnification from the medial femoral condyle (MFC) and the medial tibial plateau (MTP) by averaging five random points of measurement (separated by at least 20 μM of distance) per region per mouse and graphed as the mean ±SEM of 5 mice per group. Epiphyseal thickness was measured from central sagittal sections by averaging five random points of measurement (separated by at least 20 μM of distance) per region per mouse and graphed as the mean ±SEM of 5 mice per group system. Cartilage degradation from the medial femoral condyle (MFC) and the medial tibial plateau (MTP) was assessed according to a modified semi-quantitative scoring system of Glasson et at, (25) where 1=normal cartilage; 2=alteration of the proteoglycan matrix assessed by Safranin O stain; 3=alteration of the proteoglycan matrix and loss of lamina splendens; 4=a score of 2-3 thinning either the transitional or radial cartilage; 5=a score of 2-3 plus thinning of both the transitional and radial layers.
- Multiplex. The level of serum cytokines was determined using multiplex bead arrays kits according to the manufacturer's instructions (Bio-Plex Pro Mouse Cytokine 23-plex kits) (Biorad, Hercules, CA). Data was acquired using a
Luminex 200™ (Biorad) and analysed using the Bio-plex Manager' 6.1 software (Biorad). - Real-Time PCR. Preparation of RNA was performed from cell pellets using TRIzol (Life Technologies, Victoria, Australia) according to the manufacturer's instructions. Quantification of total RNA was measured by
NanoDrop 1000 spectrophotometer (Thermo Scientific, Victoria, Australia). Extracted total RNA (20 ng/μL) was reverse-transcribed using an oligo (dT) primer and reverse transcriptase (Sigma Aldrich, Sydney, Australia) according to the manufacturer's instructions. - Gene expression- SYBR® Green Real-time PCR was performed using 10 ng of template cDNA on a CFX96 TouchTM Real-Time PCR System in 96-well plates, using QuantiTect Primer Assay kits (Qiagen, Hilden, Germany) for HPRT1, or purchased from primers from Sigma-Aldrich with the sequences outlined in table 1.
-
TABLE 1 Primer sequences Gene Forward Reverse TGFβ CAA CGC CAT CTA TGA AAG CCC TGT ATT CCG GAA AAC C TCT CC Aggrecan GCC CAA GAA CAG TAC TGC TAG GTT GGT TGA AAT GGT CCC A Collagen CAG AAC ATC ACC TAC TTC AAC ATC GTT GGA I CAC TGC AA ACC CTG Collagen AGA ACA GCA TCG CCT CTT GCC CCA CTT ACC II ACC TG AGT GT BMP-1/ AGC AGG CTG CAG TTC GAA TGT GTT CCG GGC mTLD TCA GAC AGC ATA GTG CAT ADAMTS-4 CAC TGA CTT CCT GGA GGA AAA GTC GTC GGT CAA TGG TTA T AGA TGG A ADAMTS-5 GAT GAT CAC GAA GAG TCA CAT GAA TGA TGC CAC TAC GA CCA CAT MMP-3 TGG AGC TGA TGC ATA TGA AGC CAC CAA CAT AGC CC CAG GA MMP-9 GGA ACT CAC ACG ACA GAA ACT CAC ACG CCA TCT TCC A GAA GAA TTT TIMP-3 GGC ACT CTG GTC TAC TTT CAG AGG CTT CCG ACT ATT AAG CA TGT GA RRV nsp3 CCG TGG CGG GTA TTA AAC ACT CCC GTC GAC primer* TCA AT AAC AGA *RRV nsp3 Probe ATT AAG AGT GTA GCC ATC C - Viral load quantification-Standard curve was generated using serial dilutions of RRV T48 infectious plasmid DNA as described previously (26). Quantification of viral load was performed using SsoAdvanced Universal Probes Supermix (BioRad) in 12.5 μL reaction volume to detect nsP3 region RNA (table 1) (26).
- All reactions were performed using BioRad CFX96 Touch™ Real-Time PCR Detection System on 96-well plates. Cycler conditions were as follows: (i) PCR initial activation step: 95° C. for 15 min, 1 cycle and (ii) 3-step cycling: 94° C. for 15 sec, follow by 55° C. for 30 sec and 72° C. for 30 sec, 40 cycles. Dissociation curve was acquired using CFX Manager™ software to determine specificity of amplified products. Standard curve was plotted and copy numbers of amplified products were interpolated from standard curve using Prism Graphpad software to determine viral load. The fold change in mRNA expression relative to mock- infected samples for each gene was calculated with the ΔΔCt method. Briefly, ΔΔCt=ΔCt (RRV-infected)−ΔCt (Mock-infected) with ΔCt=Ct (gene of interest)−Ct (housekeeping gene—HPRT). The fold change for each gene was calculated as 2−ΔΔCt.
- Detection of leukocyte infiltrates in quadriceps. Quadriceps muscles were removed and processed as described previously (10). Briefly, tissues were incubated with 3 mg/Ml collagenase IV and 1 mg/mL DNase I in 100 μL RPMI 1640 at 37° C. for 1.5 h then resuspended in 5 mL RPMI and passed through a 40 μm cell strainer. Cells were washed, pelleted and treated with 1× RBC lysis buffer for 5 min, and counted. To determine percentages and numbers of specific leukocyte populations, cells were treated with Fc Block (2.4G2; BD) for 5 min at 4° C.and labelled with fluorochrome-conjugated anti-mouse antibodies, including anti-CD3-FITC (145-2C11, BD), anti-CD19-APC (MB19-1, eBioscience), anti-CD11b-PE (M1/70, BD), anti-Grl-APC (RB6-8C5, eBioscience) and anti-pan-NK/NKT antigen-PE (U5A2-13, BD) in various combinations in the presence of biotinylated anti-CD45 (30-F11, eBioscience), followed by treatment with streptavidin PE-Cy7 at 4° C.for 30 min. Cells were resuspended in 500 μL PBS containing 2% FCS and 1 μg/mL propidium iodine (PI), and analysed by the CyAn ADP flow cytometer (Beckman Coutler) with Kaluza software.
- Statistical analysis. Body mass, plaque assay, multiplex (
FIG. 4C and 8A ), real-time PCR (FIG. 6 ) and joint swelling were analysed using two-way ANOVA with Bonferroni post- test. Real-time PCR (FIG. 1D ), multiplex (FIGS. 4 B and 8B) and histology (FIG. 5C ) were analysed using a one-way ANOVA with a Dunnett's or Tukey's post-test. Flow cytometry data and histology (FIG. 1C ) were analysed using unpaired Students t-test. All data was tested for normality using the D'Agostino-Pearson normality test prior to analysis with these parametric tests. Clinical scores and and real-time PCR for viral load were analysed using the non-parametric Mann-Whitney test. Statistics were performed with GraphPad Prism 5.0 - Results
- Ross River virus infection stimulates the production of proteases ADAMTS-4, MMP-3 and MMP-9 and causes damage to the articular cartilage in joints.
- To determine if RRV infection affects the cartilage of joints, we infected C57BL/6 mice with RRV and isolated joint tissue for histological analysis. At peak disease, extensive joint inflammation was observed along with pannus-like formation and thinning of the articular cartilage in H&E stained sections (
FIG. 1A ). Further analysis of joint sections, stained with Safranin O, revealed considerable disruption of the proteoglycans in the cartilage matrix, as seen by the difference in Safranin O staining intensity (which is directly proportional to the proteoglycan content) between infected and mock tissues (FIG. 1B ) (27). Quantification of cartilage thickness and damage was measured from the MFC and MTP and showed RRV- infection results in an average 20 μm reduction in articular cartilage thickness and cartilage damage characterised by alteration of the proteoglycan matrix and loss of lamina splendens (FIG. 1C ). Furthermore, RRV-infection resulted in an early significant increase (24 hours p.i.) of the enzymes; A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4 (p<0.001), matrix metalloproteinase (MMP)-3 (p<0.05) and MMP-9 (p<0.01) compared to mock infected controls. These are known to cause cartilage damage by degrading aggrecan, collagen, proteogylcans and the extracellular matrix (FIG. 1D i). At peak disease, stimulators of cartilage growth, matrix-transforming growth factor (TGF) β1 (p<0.01) and bone morphogenetic protein (BMP)-1 (p<0.001) were also significantly increased in response to RRV-infection compared to mock-infected controls (FIG. 1D ii). The results suggest that RRV infection results in cartilage degradation and thinning which is associated with arthritic disease symptoms. - Pentosan polysulfate reduces the severity of RRV-induced disease and inflammation
- To assess the potential of PPS as a treatment strategy in alphaviral disease, mice were infected with RRV or mock-infected with PBS alone and then treated i.p. with either PPS at 3 mg/kg or with vehicle daily. PPS treatment resulted in a 65% decrease (p>0.05) in clinical disease score in RRV-infected mice (
FIG. 2A ) and a corresponding protection from disease-associated weight loss (p>0.001) (FIG. 2B ). - To better characterise the reduction in disease, we assessed inflammation and tissue damage in RRV-infected mock and PPS-treated mice. Tissues from RRV-infected mice were collected at the start and end of peak disease (
day FIG. 2C ). Consistent with previous studies, RRV-infected mock-treated mice showed extensive inflammation and myositis in quadriceps muscle atday 7 p.i. (FIG. 2C ,D) and around the ankle joint (FIG. 2C ) (11, 21, 22, 28). In contrast, RRV-infected PPS-treated mice showed markedly reduced numbers of infiltrating cells in both the muscle and the joint tissues (FIG. 2C ). - In order to characterise the effect of PPS treatment on both lymphoid and myeloid infiltrating cells, we analysed the cell populations in the quadriceps muscles at
days day 7 p.i. PPS treatment uniformly reduced the numbers of all CD45+ infiltrating leukocytes including reductions (p<0.05) in the monocytes, T cell and NK cell populations (FIG. 2D ). Byday 10 p.i. PPS treatment showed a decrease in the T cell and NK cell populations (p<0.05) with similar numbers of CD45+ cells and monocytes (FIG. 2D ). Atday 10 p.i. treatment also altered the NK cell to total cell ratio resulting in a reduction in the overall percentage of NK cells in the quadriceps muscle (data not shown). - Reduced disease in treated mice is not due to decreased viral burden.
- Viral titres in the serum of PPS-treated and mock-treated mice were comparable at all days tested, indicative of equivalent systemic replication (
FIG. 3A ). Similarly, RRV titres recovered from quadriceps muscles were comparable (FIG. 3A ). Interestingly, RRV titres in the ankle tissues showed the most variation between PPS-treated and mock-treated mice (FIG. 3A ). Byday 3 p.i. there was a slight reduction in RRV titres for PPS-treated mice; byday 7 p.i titres were comparable; and then byday 10 p.i. RRV titres in PPS-treated mice were elevated compared to mock-treated mice (p<0.05). The results fromday 10 p.i. suggest that PPS-treatment may have an effect on viral clearance within the joint. To assess viral clearance, qPCR to quantify viral RNA was performed on the joint tissues (FIG. 3B ). Although the trend appears to suggest a lower level of specific RRV RNA in the joint tissue of PPS-treated RRV-infected mice in the joint atay 10 p.i., this was not statistically significant compared to mock-treated mice, confirming that PPS-treatment does not affect viral clearance. - Treatment with pentosan polysulfate increases the level of anti-inflammatory IL-10 and decreases pro-inflammatory factors associated with RRV disease.
- A range of pro-inflammatory factors and chemoattractants mediate or contribute to alphaviral disease (29). To elucidate whether PPS-treatment affects the production of soluble immune mediators during RRV infection, sera from PPS-treated and mock-treated mice were analysed using multiplex and compared to mock-infected, PPS-treated or mock-treated mice. As expected, RRV infection resulted in an increase (p<0.05) of pro-inflammatory factors (both cytokines and chemoattractants) at peak disease (
FIG. 4 ). PPS-treatment also altered the levels of the M2 anti-inflammatory cytokine IL-10. IL-10 kinetics corresponded to the kinetics of disease with serum levels increasing over time in RRV-infected, mock-treated mice. PPS-treatment resulted in an early surge of IL-10 in RRV-infected mice, being significantly elevated at bothdays FIG. 4A ). Additionally, PPS treatment significantly reduced the serum levels of IL-1 a, IL-2, IL-6, CCL-2 and MIP-1α at peak disease (p<0.05) (FIG. 4B ). - Pentosan polysulfate treatment protects the joints from cartilage damage associated with RRV infection.
- Recently we showed that RRV could infect osteoblasts and infection results in systemic bone loss including the tibial epiphysis and vertebrae (30-32). To determine the effect of PPS treatment on RRV-induced muscle and joint damage, the tissues of RRV-infected and mock- infected PPS-treated and mock-treated mice were processed for histological analysis using Masson's trichrome and Safranin O/Fast Green staining. Masson's trichrome staining of the tibialis anterior showed PPS treatment protected the morphology of striations within the skeletal muscle with sections of collagen formation characteristic of muscle repair and fibrosis (33). PPS treatment also prevented RRV-induced thinning of the epiphyseal plate, protecting against cartilage loss (
FIG. 5A , C). Safranin O/Fast Green staining of the cartilage in the knee joint revealed that PPS treatment protected the proteoglycan matrix of the articular cartilage, preventing the loss of articular cartilage observed in RRV-infected mock-treated mice as well as maintaining chondrocyte morphology (FIG. 5B , C). - To further characterise the mechanism of PPS treatment we analysed the genes involved in enzyme degradation of cartilage (ADAMTS-4, ADAMTS-5, MMP-3 and MMP-9), stimulation of cartilage protection and synthesis (TGF-β1 and BMP-1) and cartilage matrix proteins (aggrecan, collagen I and collagen II). As shown in
FIG. 1D , RRV-infection caused an increase in ADAMTS-4, which remains elevated atday 3 p.i (p<0.01), but drops by the time of peak disease (day 10 p.i), where there was a surge in ADMATS-5. RRV- infection also resulted in a late rise (at peak disease) of tissue inhibitor of metalloproteinases (TIMP)-3 (p<0.001); known to inhibit both ADAMTS-4 and ADAMTS-5 (FIG. 6A, 6C ). PPS-treatment significantly reduced the levels of ADAMTS-5 and TIMP-3 at peak disease (p<0.01), but not at the early stages of infection. RRV-infection also resulted in an increase of cartilage components aggrecan, collagen I and collagen II, that is largely reduced with PPS-treatment (FIG. 6B ). The genes associated with signalling pathways for cartilage development (TGF-β1 and BMP-1) and the metalloproteinases were unaffected by PPS (FIG. 6 ). - Pentosan polysulfate treatment is a safe long-term treatment strategy for chronic RRV disease.
- To assess PPS-treatment for a long-term treatment in patients with chronic symptoms, mice were RRV-infected and treated orally with PPS or mock-treated in drinking water. Long-term PPS treatment resulted in no adverse clinical signs in the mice for the 3-month duration of the experiment. RRV-infected mock-treated mice showed extended disruption of the cartilage components with a three-fold elevation of aggrecan (p<0.01) (Table 2). PPS-treated mice showed less joint damage and significantly decreased the expression of aggrecan back to base-line levels (p<0.001). The levels of ADAMTS-4 expression were also reduced (p<0.01).
-
TABLE 2 Long-term PPS treatment decreases the expression of aggrecan and ADAMTS-4 in RRV infection. Mock RRV RRV + PPS Gene Fold SEM n Fold SEM n Fold SEM n ADAMT 0.8345 0.203 6 1.2261 0.237 5 0.1689* 0.299 5 ADAMT 1.0854 0.320 6 1.5759 0.373 5 0.8305 0.179 5 MMP- 1.1335 0.349 6 1.6958 0.440 5 2.5028 1.221 5 MMP- 1.0066 0.083 6 1.0858 0.201 5 0.8604 0.247 5 9 3 2 7 Collagen 1.0128 0.117 3 0.8550 0.141 5 0.4014 0.074 5 Collagen 1.0357 0.179 3 0.9584 0.235 5 0.1719 0.055 5 II 3 8 6 BMP-1 1.0597 0.267 3 1.6204 0.295 5 1.4697 0.702 5 TIMP- 1.3321 0.772 6 1.4287 0.318 5 0.6328 0.136 5 - Pentosan polysulfate is a potential treatment for CHIKV-induced inflammation, reducing disease by altering the cytokine response.
- Given the expanding range of the alphavirus-CHIKV, together with the current lack of therapeutic treatment options, we sought to determine the broader application of PPS on alleviating CHIKV-induced disease. PPS treatment decreased the level of joint swelling of CHIKV-infected mice corresponding to a reduction in inflammatory cells infiltrating into the joint (
FIG. 7A , B). Furthermore, as seen in RRV-infection, PPS-treatment did not affect the kinetics of virus infection (FIG. 7C ) and did not increase the viral persistence in the joint tissues, with similar levels of viral RNA detected three weeks p.i. (FIG. 7D ). The reduced disease also correlated to an early surge in anti-inflammatory IL-10 (FIG. 8A ) and reduced the levels of soluble factors CCL-2, IL-6, IL-9 and G-CSF at peak disease (day 3 p.i.) (FIG. 8B ). These collective results, whereby PPS reduces the disease severity of two critical alphaviral diseases suggest that PPS may be a promising broad-range treatment for alphavirus disease manifestations in general. - Discussion
- The mechanisms by which alphaviruses trigger arthritis and myositis are the focus of ongoing studies. Alphavirus-induced disease has many similarities to rheumatoid arthritis (RA) including common inflammatory pathways and the key involvement of macrophages (11,12). The innate immune response is critical in the pathogenesis of alphaviral disease, mediating cell recruitment, viral clearance and inflammation (28, 29, 34). In particular, monocytes and macrophages are the major cellular contributors to disease progression and severity (29). In RA, monocytes play a significant role in disease development and cartilage destruction through the production of pro-inflammatory factors. (35). Despite these clear similarities, the potential of alphavirus infection to damage the articular cartilage in the joint tissues has not been investigated. We now propose that, analogous to RA, RRV-infection leads to an immune poly-arthritis that causes cartilage thinning that contributes to clinical signs associated with alphaviral disease.
- It has long been recognised that the joint tissue is a critical site of viral replication, and we recently identified osteoblasts as a source of infectious virus, being susceptible to RRV infection (31). We showed that RRV-infection results in bone loss by disrupting the receptor activator of nuclear factor κβ ligand (RANKL) and osteoprotegerin (OPG) ratio (31). We now describe thinning of articular cartilage in an RRV disease mouse model correlating to a significant increase in metalloproteinases including ADAMTS-4 and ADAMTS-5. These result in histopathological findings similar to mild onset RA.
- A recent study, using CCR2−/− mice infected with CHIKV, showed that when normal monocyte trafficking is disrupted by this receptor knockout the major inflammatory infiltrates became neutrophil dominant (36). This replaces the usual macrophage dominance of the cellular response observed in alphavirus infections. Comparing the histopathological findings in the feet of CHIKV-infected wild-type (WT) and CCR2−/− mice, Poo et al (36) observed that the neutrophil shift resulted in cartilage damage. We also observe cartilage thinning in immunocompetent C57BL/6 mice (with functional macrophage trafficking) following RRV infection, demonstrating that macrophages may also play a critical role in RRV-induced inflammation including cartilage thinning.
- The use of glycans as novel therapeutics has developed momentum in recent years (37). The interaction of glycans with growth factors, extracellular proteases, protease inhibitors, cytokines/chemokines and adhesive proteins regulate various physiopathologies and diseases including cancer, atherosclerosis and thrombosis (38). In addition many pathogens, including viruses, exploit host glycans to cause infection. The therapeutic potential of this class of molecule to alleviate viral-induced arthritis and inflammatory disease has not been studied and currently remains unknown.
- Pentosan polysulfate is a semisynthetic polysaccharide derivative that chemically and structurally resembles other GAGs, including heparin. In contrast to many other GAGs, PPS is bioavailable in both its injectable and oral forms and produces limited toxic side effects, even when administered in high doses (39). In a clinical setting, PPS has been used as an anti-thrombotic agent for several decades, due to its ability to bind preferentially to the glycocalyx of circulating blood cells (40). In more recent times, PPS has been identified as having anti-inflammatory properties and is currently approved in the United States for the management of patients with interstitial cystitis, having an excellent long-term safety profile (15). Furthermore, injectable forms of PPS are currently used to treat osteoarthritis in veterinary medicine (19).
- Given the promising results of PPS treatment of a range of inflammatory conditions particularly arthritis, and the lack of studies on PPS in treating virus-associated pathologies, we tested the efficacy of PPS to treat alphavirus-induced arthritis. PPS treatment significantly reduced the acute disease signs and the muscle and joint inflammation of both RRV- and CHIKV-induced disease. This corresponded to a reduction in serum levels of pro-inflammatory factors at peak disease. In diabetic kidney nephropathy, disease pathogenesis is dependent on the cellular infiltration of macrophages and pro-inflammatory and chemoattractant factors, similar to those associated with alphavirus-arthritis. These include CCL-2, RANTES and CXCL1, TNF-a (29, 41). Our data are consistent with the treatment observed in diabetic nephropathy, in which PPS reduced the macrophage infiltration and suppressed the induction of pro-inflammatory factors (41).
- At the onset of RRV-disease (day 6-7 p.i), there is a surge in the levels of IL-10 (42). Of interest to the present study is the recent observation demonstrating that a surge in IL-10 resulted in a phenotype switch of monocytes/macrophages (43, 44). This suggests that IL-10 is part of a repair signal that activates specific cellular and molecular cascades to facilitate tissue recovery (43). PPS-treatment altered the kinetics of RRV-induced soluble pro- and anti-inflammatory factors, promoting a swing to anti-inflammatory cytokines with an early induction of IL-10 that enhances myogenesis (43). Furthermore IL-10 inhibits the synthesis of pro-inflammatory soluble factors including, IL-1α, IL-2, IL-6, TNF-α and CCL-2 (45), previously associated with increased severity of alphaviral disease. Overall the early PPS- induced increase of IL-10 may act to reduce inflammation, but also enhance tissue repair, thereby providing a key mechanism by which PPS-treatment reduces the severity of alphaviral disease.
- In addition to the reported action of PPS in reducing pro-inflammatory factors, PPS inhibits both the alternative and classical pathways of complement activation (46). For example, in RRV-pathogenesis, complement activation is essential for the development of severe RRV disease (34). Furthermore, this is specific to RRV-activation of complement via the MBL pathway and is independent of the classical and alternative pathways (28), and therefore it is unlikely that the reduction in RRV disease observed with PPS treatment is due to its effect on complement.
- Treatment with PPS also resulted in protection of the i) epiphysis, 2) articular cartilage and 3) proteoglycan matrix. The anti-inflammatory effect of PPS is due in part to its ability to inhibit IL-6 (47). In our results, serum levels of IL-6 were significantly reduced in PPS treated RRV- infected mice at peak disease. We have shown that the disruption of the RANKL and OPG ratio during RRV-infection occurs in an IL-6 dependent manner such that inhibition of IL-6 protects from RRV-induced bone loss(31). Additionally, studies on CCL-2 have recently demonstrated that inhibition of CCL-2 can both inhibit osteoclast differentiation and protect against CHIKV induced bone loss (30, 48). Therefore, it is likely that PPS protection against RRV-induced bone loss is due to its ability to inhibit both IL-6 and CCL-2. PPS also stimulates hyaluronan synthesis by synovial fibroblasts and proteoglycan synthesis by chondrocytes (49). This may explain the observed protection of both the articular cartilage and the proteoglycan matrix in PPS-treated RRV-infected mice. PPS also promotes the proliferation and chondrogenic differentiation of adult human bone marrow mesenchymal stem cells (50), further explaining the PPS protection of articular cartilage thinning that we see in RRV-infection.
- Although the molecular mechanism of PPS action remains unclear, PPS can repress MMP, including ADAMTS expression and inflammation, as well as NF-κβ activation. It also enhances proteoglycan synthesis, including the production of aggrecan and hyaluronan (49) and has been shown to be efficacious as both a treatment and a prophylactic (51). The results of this study demonstrate that RRV-infection results in cartilage thinning, increasing the levels of ADAMTS-4 and ADMATS-5 (
aggrecanase - In humans, long-term clinical use of PPS is extremely well tolerated and highly efficacious for periods greater than 12 months (53). Alphaviruses can produce chronic musculoskeletal ailments over a prolonged period of time. A long-term therapeutic strategy is therefore required for effective treatment of alphavirus-induced arthritis. We have shown that PPS not only alleviates the acute signs of RRV-induced arthritis but also protects the cartilage over the long-term without compromising host viral clearance. Given that PPS promotes an anti- inflammatory immune state without promoting viral persistence, it is an attractive drug-repurposing candidate for the long-term treatment of RRV-associated inflammation and disease.
- To date, non-steroidal anti-inflammatory drugs are the primary therapeutic means to alleviate the symptoms of alphavirus-associated inflammatory disease. These drugs can cause a variety of undesired side effects and may compromise immunity in treated patients (54). Studies by our group in the past have examined a number of drug candidates for the treatment of alphavirus disease. Bindarit (a CCL-2 inhibitor), while effective in reducing alphavirus induced arthritis and myositis, is currently not a drug that is available for human use (30, 55). Enbrel, while available for human use, was found to suppress the antiviral response and enhance viral replication thereby worsen disease (56). Similarly, methotrexate, a licensed drug for the treatment of RA, increased the onset of RRV-induced musculoskeletal disease and the influx of inflammatory cell infiltrates into the skeletal muscle tissue (57). Here we show PPS treatment significantly reduced both the acute clinical signs and the inflammation in the muscle (myositis) and the joint (arthritis) in alphavirus disease. Additionally PPS has positive and extensive long-term human safety data, and is available as an approved drug by a number of regulatory authorities globally. We therefore conclude that PPS is a promising therapeutic candidate for alphaviral disease; and may also be effective in other infectious inflammatory conditions.
- To this end, the Australian Therapeutic Goods Administration has provided its approval for the evaluation of PPS in four Ross River virus infected subjects. Those subjects were treated with PPS intramuscularly in accordance with the dosage regimen as described herein.
- It will be appreciated by persons skilled in the art that numerous variations and/or modifications may be made to the above-described embodiments, without departing from the broad general scope of the present disclosure. The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive.
-
- 1. Burt F J, Rolph M S, Rulli N E, Mahalingam S, Heise M T. 2012. Chikungunya: a re-emerging virus. Lancet 379:662-671.
- 2. Noel H, Rizzo C. 2014. Spread of chikungunya from the Caribbean to mainland Central and South America: a greater risk of spillover in Europe? Euro surveillance: bulletin Europeen sur les maladies transmissibles =European communicable disease bulletin 19 (28).
- 3. Centers for Disease Control (CDC). 2014. First Chikungunya case acquired in the United States reported in Florida. [cited 2014 27th September]; Available from: http:/www.cdc.gov/media/releases/2014/p0717-chikungunya.html
- 4. Pan American Health Organization (PAHO). 2016. Chikungunya. [cited 2016 23rd May]; Available from: http://www.paho.org/hq/index.php?option=com topics&view=articie&id=343& Itemid=40931
- 5. Herrero L, Nelson M, Bettadapura J, Gahan M E, Mahalingam S. 2011. Applications of animal models of infectious arthritis in drug discovery: a focus on alphaviral disease. Curr Drug Targets 12:1024-1036.
- 6. Manimunda SP, Vijayachari P, Uppoor R, Sugunan A P, Singh S S, Rai S K, Sudeep A B, Muruganandam N, Chaitanya I K, Guruprasad D R. 2010. Clinical progression of chikungunya fever during acute and chronic arthritic stages and the changes in joint morphology as revealed by imaging. Trans R Soc Trop Med Hyg 104:392-399.
- 7. Fraser J R, Cunningham A L, Clarris B J, Aaskov J G, Leach R. 1981. Cytology of synovial effusions in epidemic polyarthritis. Aust N Z J Med 11:168-173.
- 8. Fraser J R, Ratnamohan V M, Dowling J P, Becker G J, Varigos G A. 1983. The exanthem of Ross River virus infection: histology, location of virus antigen and nature of inflammatory infiltrate. J Clin Pathol 36:1256-1263.
- 9. Soden M, Vasudevan H, Roberts B, Coelen R, Hamlin G, Vasudevan S, La Brooy J. 2000. Detection of viral ribonucleic acid and histologic analysis of inflamed synovium in Ross River virus infection. Arthritis Rheum 43:365-369.
- 10. Herrero L, Sheng K-C, Jian P, Herr Z, Herring B, Chow A, Chow Y, Hickey M, Morand E, Ng L, Bucala R, Mahalingam S. 2013. MIF receptor CD74 mediates alphavirus-induced arthritis and myositis murine models of alphavirus infection. Arthritis and Rheumatism 65:2724-2736.
- 11. Herrero LJ, Nelson M, Srikiatkhachorn A, Gu R, Anantapreecha S, Fingerle-Rowson G, Bucala R, Morand E, Santos L L, Mahalingam S. 2011. Critical role for macrophage migration inhibitory factor (MIF) in Ross River virus-induced arthritis and myositis. Proc Natl Acad Sci U S A 108:12048-12053.
- 12. Nakaya H I, Gardner J, Poo Y S, Major L, Pulendran B, Suhrbier A. 2012. Gene profiling of Chikungunya virus arthritis in a mouse model reveals significant overlap with rheumatoid arthritis. Arthritis Rheum 64:3553-3563.
- 13. Bresnihan B. 1999. Pathogenesis of joint damage in rheumatoid arthritis. J Rheumatol 26:717-719.
- 14. Otero M, Goldring M B. 2007. Cells of the synovium in rheumatoid arthritis. Chondrocytes. Arthritis Res Ther 9:220.
- 15. Parsons C L, Mulholland S G. 1987. Successful therapy of interstitial cystitis with pentosanpolysulfate. J Urol 138:513-516.
- 16. Kumagai K, Shirabe S, Miyata N, Murata M, Yamauchi A, Kataoka Y, Niwa M. 2010. Sodium pentosan polysulfate resulted in cartilage improvement in knee osteoarthritis--an open clinical trial. BMC Clin Pharmacol 10:7.
- 17. Ghosh P, Edelman J, March L, Smith M. 2005. Effects of pentosan polysulfate in osteoarthritis of the knee: A randomized, double-blind, placebo-controlled pilot study. Curr Ther Res Clin Exp 66:552-571.
- 18. Ghosh P. 1999. The pathobiology of osteoarthritis and the rationale for the use of pentosan polysulfate for its treatment. Semin Arthritis Rheum 28:211-267.
- 19. Kongtawelert P, Brooks PM, Ghosh P. 1989. Pentosan polysulfate (Cartrophen) prevents the hydrocortisone induced loss of hyaluronic acid and proteoglycans from cartilage of rabbit joints as well as normalizes the keratan sulfate levels in their serum. J Rheumatol 16:1454-1459.
- 20. Kuhn RJ, Niesters HG, Hong Z, Strauss JH. 1991. Infectious RNA transcripts from Ross River virus cDNA clones and the construction and characterization of defined chimeras with Sindbis virus. Virology 182:430-441.
- 21. Lidbury BA, Simeonovic C, Maxwell G E, Marshall I D, Hapel A J. 2000. Macrophage-induced muscle pathology results in morbidity and mortality for Ross River virus-infected mice. J Infect Dis 181:27-34.
- 22. Morrison TE, Whitmore AC, Shabman RS, Lidbury BA, Mahalingam S, Heise MT. 2006. Characterization of Ross River virus tropism and virus-induced inflammation in a mouse model of viral arthritis and myositis. J Virol 80:737-749.
- 23. Bachmanov A A, Reed D R, Beauchamp G K, Tordoff M G. 2002. Food intake, water intake, and drinking spout side preference of 28 mouse strains. Behavior genetics 32:435-443.
- 24. Reagan-Shaw S, Nihal M, Ahmad N. 2008. Dose translation from animal to human studies revisited. FASEB J 22:659-661.
- 25. Glasson S S, Chambers M G, Van Den Berg W B, Little CB. 2010. The OARSI histopathology initiative—recommendations for histological assessments of osteoarthritis in the mouse. Osteoarthritis Cartilage 18 Suppl 3:S17-23.
- 26. Shabman R S, Rogers K M, Heise M T. 2008. Ross River virus envelope glycans contribute to type I interferon production in myeloid dendritic cells. J Virol 82:12374-12383.
- 27. Camplejohn K L, Allard S A. 1988. Limitations of safranin ‘O’ staining in proteoglycan-depleted cartilage demonstrated with monoclonal antibodies. Histochemistry 89:185-189.
- 28. Gunn B M, Morrison T E, Whitmore A C, Blevins L K, Hueston L, Fraser R J, Herrero L J, Ramirez R, Smith P N, Mahalingam S, Heise M T. 2012. Mannose binding lectin is required for alphavirus-induced arthritis/myositis. PLoS Pathog 8:e1002586.
- 29. Lidbury B A, Rulli N E, Suhrbier A, Smith P N, McColl S R, Cunningham A L, Tarkowski A, van Rooijen N, Fraser R J, Mahalingam S. 2008. Macrophage-derived proinflammatory factors contribute to the development of arthritis and myositis after infection with an arthrogenic alphavirus. J Infect Dis 197:1585-1593.
- 30. Chen W, Foo S S, Taylor A, Lulla A, Merits A, Hueston L, Forwood M R, Walsh N C, Sims N A, Herrero L J, Mahalingam S. 2015. Bindarit, an inhibitor of monocyte chemotactic proteins (MCPs) synthesis, protects against bone loss induced by Chikungunya virus infection. J Virol 89:581-593.
- 31. Chen W, Foo S S, Rulli N, Taylor A, Sheng K C, Herrero L J, Herring B L, Lidbury B A, Li R W, Walsh N C, Sims N A, Smith P N, Mahalingam S. 2014. Arthritogenic alphaviral infection perturbs osteoblast function and triggers pathologic bone loss. Proc Natl Acad Sci U S A 111:6040-6045.
- 32. Chen W, Foo S-S, Sims N A, Herrero L J, Walsh N C, Mahalingam S. 2015. Arthritogenic alphaviruses: new insights into arthritis and bone pathology. Trends in Microbiology 23:35-43.
- 33. Mann C J, Perdiguero E, Kharraz Y, Aguilar S, Pessina P, Serrano A L, Munoz-Canoves P. 2011. Aberrant repair and fibrosis development in skeletal muscle. Skelet Muscle 1:21.
- 34. Morrison T E, Fraser R J, Smith P N, Mahalingam S, Heise M T. 2007. Complement contributes to inflammatory tissue destruction in a mouse model of Ross River virus-induced disease. J Virol 81:5132-5143.
- 35. Ma Y, Pope R M. 2005. The role of macrophages in rheumatoid arthritis. Current pharmaceutical design 11:569-580.
- 36. Poo Y S, Nakaya H, Gardner J, Larcher T, Schroder W A, Le T T, Major L D, Suhrbier A. 2014. CCR2 Deficiency Promotes Exacerbated Chronic Erosive Neutrophil-Dominated Chikungunya Virus Arthritis. J Virol 88:6862-6872.
- 37. Bertozzi C R, Freeze H H, Varki A, Esko J D. 2009. Glycans in Biotechnology and the Pharmaceutical Industry. In Varki A, Cummings R D, Esko J D (ed.), Essentials of Glycobiology, 2010/03/20 ed. Cold Spring Harbor (N.Y.).
- 38. Varki A. 2007. Glycan-based interactions involving vertebrate sialic-acid-recognizing proteins. Nature 446:1023-1029.
- 39. Nickel J C, Barkin J, Forrest J, Mosbaugh P G, Hernandez-Graulau J, Kaufman D, Lloyd K, Evans R J, Parsons C L, Atkinson LE. 2005. Randomized, double-blind, dose-ranging study of pentosan polysulfate sodium for interstitial cystitis. Urology 65:654-658.
- 40. Maffrand J P, Herbert J M, Bernat A, Defreyn G, Delebassee D, Savi P, Pinot J J, Sampol J. 1991. Experimental and clinical pharmacology of pentosan polysulfate. Semin Thromb Hemost 17 Suppl 2:186-198.
- 41. Wu J, Guan T J, Zheng S, Grosjean F, Liu W, Xiong H, Gordon R, Vlassara H, Striker G E, Zheng F. 2011. Inhibition of inflammation by pentosan polysulfate impedes the development and progression of severe diabetic nephropathy in aging C57B6 mice. Lab Invest 91:1459-1471.
- 42. Stoermer K A, Burrack A, Oko L, Montgomery S A, Borst L B, Gill R G, Morrison T E. 2012. Genetic ablation of
arginase 1 in macrophages and neutrophils enhances clearance of an arthritogenic alphavirus. J Immuno1189:4047-4059. - 43. Deng B, Wehling-Henricks M, Villalta S A, Wang Y, Tidball J G. 2012. IL-10 triggers changes in macrophage phenotype that promote muscle growth and regeneration. J Immunol 189:3669-3680.
- 44. Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, Plonquet A, Gherardi R K, Chazaud B. 2007. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J Exp Med 204:1057-1069.
- 45. Couper K N, Blount D G, Riley E M. 2008. IL-10: the master regulator of immunity to infection. J Immunol 180:5771-5777.
- 46. Kilgore K S, Naylor K B, Tanhehco E J, Park J L, Booth E A, Washington R A, Lucchesi B R. 1998. The semisynthetic polysaccharide pentosan polysulfate prevents complement-mediated myocardial injury in the rabbit perfused heart. J Pharmacol Exp Ther 285:987-994.
- 47. Smith M M, Ghosh P, Numata Y, Bansal M K. 1994. The effects of orally administered calcium pentosan polysulfate on inflammation and cartilage degradation produced in rabbit joints by intraarticular injection of a hyaluronate-polylysine complex. Arthritis Rheum 37:125-136.
- 48. Morrison N A, Day C J, Nicholson G C. 2014. Dominant negative MCP-1 blocks human osteoclast differentiation. J Cell Biochem 115:303-312.
- 49. Takizawa M, Yatabe T, Okada A, Chijiiwa M, Mochizuki S, Ghosh P, Okada Y. 2008. Calcium pentosan polysulfate directly inhibits enzymatic activity of ADAMTS4 (aggrecanase-1) in osteoarthritic chondrocytes. FEBS Lett 582:2945-2949.
- 50. Ghosh P, Wu J, Shimmon S, Zannettino AC, Gronthos S, Itescu S. 2010. Pentosan polysulfate promotes proliferation and chondrogenic differentiation of adult human bone marrow-derived mesenchymal precursor cells. Arthritis Res Ther 12:R28.
- 51. Kramer C M, Tsang A S, Koenig T, Jeffcott L B, Dart C M, Dart A J. 2014. Survey of the therapeutic approach and efficacy of pentosan polysulfate for the prevention and treatment of equine osteoarthritis in veterinary practice in Australia. Aust Vet J 92:482-487.
- 52. Troeberg L, Fushimi K, Khokha R, Emonard H, Ghosh P, Nagase H. 2008. Calcium pentosan polysulfate is a multifaceted exosite inhibitor of aggrecanases. FASEB J 22:3515-3524.
- 53. Al-Zahrani A A, Gajewski J B. 2011. Long-term efficacy and tolerability of pentosan polysulphate sodium in the treatment of bladder pain syndrome. Canadian Urological Association journal=Journal de l'Association des urologues du Canada 5:113-118.
- 54. Bancos S, Bernard M P, Topham D J, Phipps R P. 2009. Ibuprofen and other widely used non-steroidal anti-inflammatory drugs inhibit antibody production in human cells. Cell Immunol 258:18-28.
- 55. Rulli NE, Guglielmotti A, Mangano G, Rolph MS, Apicella C, Zaid A, Suhrbier A, Mahalingam S. 2009. Amelioration of alphavirus-induced arthritis and myositis in a mouse model by treatment with bindarit, an inhibitor of monocyte chemotactic proteins. Arthritis Rheum 60:2513-2523.
- 56. Zaid A, Sheng K C, Taylor A, Rulli N E, Herrero L J, McNeil P, Mahalingam S. 2013. Exacerbation of Alphaviral Arthritis and Myositis in a Mouse Model after Etanercept Treatment is due to Diminished Levels of Interferon a/b. Virol Mycol 2:122.
- 57. Taylor A, Sheng K C, Herrero L J, Chen W, Rulli N E, Mahalingam S. 2013. Methotrexate treatment causes early onset of disease in a mouse model of Ross River virus-induced inflammatory disease through increased monocyte production. PLoS One 8:e71146.
Claims (20)
1. A method of treating a subject having an alphavirus infection comprising administering parenterally by intra-articular or intra-muscular injection an amount of pentosan polysulfate or a salt thereof effective to reduce alphavirus induced inflammation and/or alphavirus induced cartilage damage in the subject.
2. The method of claim 1 wherein the inflammation is in the joints and/or muscles.
3. The method of claim 2 wherein the inflammation is in the joints.
4. The method of claim 2 wherein the inflammation is in the muscles.
5. The method of claim 2 wherein the inflammation is in the joints and the muscles.
6. The method of claim 1 wherein the alphavirus is selected from the group consisting of Ross River virus, chikungunya virus and Barmah Forest virus.
7. The method of claim 6 wherein the virus is Ross River virus.
8. The method of claim 6 wherein the virus is chikungunya virus.
9. The method of claim 6 wherein the virus is Barmah Forest virus.
10. The method of claim 1 wherein administration is intra-articular.
11. The method of claim 1 wherein administration is intra-muscular.
12. The method of claim 1 wherein the pentosan polysulfate is the sodium salt.
13. The method of claim 1 wherein administration is daily.
14. The method of claim 1 wherein administration is twice daily.
15. The method of claim 13 wherein the amount administered is within the range of from 0.5 to 5.0 mg/kg body weight of subject.
16. The method of claim 15 wherein the amount is within the range of from 1.0 to 5.0 mg/kg body weight of subject.
17. The method of claim 16 wherein the amount is within the range of from 2.0 to 5.0 mg/kg body weight of subject.
18. The method claim 17 wherein the subject is a human.
19-20. (cancelled)
21. A method of treating a subject having an alphavirus infection selected from the group consisting of Ross River virus, chikungunya virus and Barmah Forest virus comprising administering daily parenterally by intra-articular or intra-muscular injection an amount of sodium pentosan polysulfate effective to reduce alphavirus induced inflammation and/or alphavirus induced cartilage damage in the subject.
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/AU2016/050408 WO2017201563A1 (en) | 2016-05-26 | 2016-05-26 | Treatment of alphavirus-induced inflammation |
Related Parent Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/AU2016/050408 A-371-Of-International WO2017201563A1 (en) | 2016-05-26 | 2016-05-26 | Treatment of alphavirus-induced inflammation |
Related Child Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US16/884,628 Continuation US11638718B2 (en) | 2016-05-26 | 2020-05-27 | Treatment of alphavirus-induced inflammation |
Publications (1)
Publication Number | Publication Date |
---|---|
US20190255097A1 true US20190255097A1 (en) | 2019-08-22 |
Family
ID=60410929
Family Applications (3)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US16/304,550 Abandoned US20190255097A1 (en) | 2016-05-26 | 2016-05-26 | Treatment of alphavirus-induced inflammation |
US16/884,628 Active 2036-06-02 US11638718B2 (en) | 2016-05-26 | 2020-05-27 | Treatment of alphavirus-induced inflammation |
US18/130,475 Pending US20230364128A1 (en) | 2016-05-26 | 2023-04-04 | Treatment of alphavirus-induced inflammation |
Family Applications After (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US16/884,628 Active 2036-06-02 US11638718B2 (en) | 2016-05-26 | 2020-05-27 | Treatment of alphavirus-induced inflammation |
US18/130,475 Pending US20230364128A1 (en) | 2016-05-26 | 2023-04-04 | Treatment of alphavirus-induced inflammation |
Country Status (10)
Country | Link |
---|---|
US (3) | US20190255097A1 (en) |
KR (2) | KR20240056805A (en) |
AU (2) | AU2016407635B2 (en) |
BR (1) | BR112018074235A2 (en) |
CA (1) | CA3025018C (en) |
MX (1) | MX2018014499A (en) |
MY (1) | MY190852A (en) |
PH (1) | PH12018502477A1 (en) |
SG (1) | SG11201810109UA (en) |
WO (1) | WO2017201563A1 (en) |
Family Cites Families (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CA1327354C (en) | 1987-03-19 | 1994-03-01 | David Cullis-Hill | Anti-inflamatory compounds and compositions |
AU1052492A (en) * | 1991-01-31 | 1992-08-06 | Farmitalia Carlo Erba S.R.L. | Synergistic composition comprising a fibroblast growth factor and a sulfated polylsaccharide, for use as antiviral agent |
US5872880A (en) * | 1996-08-12 | 1999-02-16 | Ronald S. Maynard | Hybrid-optical multi-axis beam steering apparatus |
US20030181416A1 (en) * | 2002-01-10 | 2003-09-25 | Comper Wayne D. | Antimicrobial charged polymers that exhibit resistance to lysosomal degradation during kidney filtration and renal passage, compositions and method of use thereof |
WO2012027797A1 (en) * | 2010-09-02 | 2012-03-08 | David Charles James | Anti-inflammatory compostion |
US9902765B2 (en) * | 2013-07-19 | 2018-02-27 | Integral Molecular, Inc. | Antibodies against chikungunya virus and uses thereof |
-
2016
- 2016-05-26 CA CA3025018A patent/CA3025018C/en active Active
- 2016-05-26 WO PCT/AU2016/050408 patent/WO2017201563A1/en active Application Filing
- 2016-05-26 KR KR1020247013992A patent/KR20240056805A/en active Application Filing
- 2016-05-26 KR KR1020187037193A patent/KR20190011761A/en active Application Filing
- 2016-05-26 BR BR112018074235-0A patent/BR112018074235A2/en not_active Application Discontinuation
- 2016-05-26 MX MX2018014499A patent/MX2018014499A/en unknown
- 2016-05-26 SG SG11201810109UA patent/SG11201810109UA/en unknown
- 2016-05-26 AU AU2016407635A patent/AU2016407635B2/en active Active
- 2016-05-26 US US16/304,550 patent/US20190255097A1/en not_active Abandoned
- 2016-05-26 MY MYPI2018002104A patent/MY190852A/en unknown
-
2018
- 2018-11-23 PH PH12018502477A patent/PH12018502477A1/en unknown
-
2019
- 2019-05-27 AU AU2019203683A patent/AU2019203683A1/en not_active Abandoned
-
2020
- 2020-05-27 US US16/884,628 patent/US11638718B2/en active Active
-
2023
- 2023-04-04 US US18/130,475 patent/US20230364128A1/en active Pending
Also Published As
Publication number | Publication date |
---|---|
KR20190011761A (en) | 2019-02-07 |
US20210023124A1 (en) | 2021-01-28 |
US20230364128A1 (en) | 2023-11-16 |
KR20240056805A (en) | 2024-04-30 |
US11638718B2 (en) | 2023-05-02 |
PH12018502477A1 (en) | 2019-10-07 |
AU2019203683A1 (en) | 2019-06-20 |
AU2016407635A1 (en) | 2018-11-22 |
MY190852A (en) | 2022-05-12 |
AU2016407635B2 (en) | 2019-03-07 |
CA3025018C (en) | 2023-08-29 |
SG11201810109UA (en) | 2018-12-28 |
MX2018014499A (en) | 2019-05-23 |
CA3025018A1 (en) | 2017-11-30 |
BR112018074235A2 (en) | 2019-03-06 |
WO2017201563A1 (en) | 2017-11-30 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Herrero et al. | Pentosan polysulfate: a novel glycosaminoglycan-like molecule for effective treatment of alphavirus-induced cartilage destruction and inflammatory disease | |
Chen et al. | Specific inhibition of NLRP3 in chikungunya disease reveals a role for inflammasomes in alphavirus-induced inflammation | |
Punzi et al. | Post-traumatic arthritis: overview on pathogenic mechanisms and role of inflammation | |
Cheng et al. | Matrine improves 2, 4, 6-trinitrobenzene sulfonic acid-induced colitis in mice | |
Zhao et al. | TNF‐α stimulates caspase‐3 activation and apoptotic cell death in primary septo‐hippocampal cultures | |
Goldbach-Mansky | Current status of understanding the pathogenesis and management of patients with NOMID/CINCA | |
Bauditz et al. | Treatment with tumour necrosis factor inhibitor oxpentifylline does not improve corticosteroid dependent chronic active Crohn's disease. | |
Herrero et al. | Macrophage migration inhibitory factor receptor CD74 mediates alphavirus‐induced arthritis and myositis in murine models of alphavirus infection | |
Basha | Corona virus drugs–a brief overview of past, present and future | |
Wang et al. | Study on the efficacy and mechanism of triptolide on treating TNF transgenic mice with rheumatoid arthritis | |
KR20070110046A (en) | Compounds for treating inflammatory and demyelinating diseases | |
Fonseca et al. | 7-hydroxyfrullanolide, a sesquiterpene lactone, inhibits pro-inflammatory cytokine production from immune cells and is orally efficacious in animal models of inflammation | |
Li et al. | The P2X7 receptor in osteoarthritis | |
Ramonda et al. | Molecular mechanisms of pain in crystal-induced arthritis | |
Zhang et al. | Recent progress on the treatment of Ebola virus disease with Favipiravir and other related strategies | |
Yan et al. | Autophagy attenuates osteoarthritis in mice by inhibiting chondrocyte pyroptosis and improving subchondral bone remodeling | |
Chen et al. | A natural plant ingredient, menthone, regulates T cell subtypes and lowers pro-inflammatory cytokines of rheumatoid arthritis | |
Jie et al. | Simiao Yong'an decoction ameliorates murine collagen-induced arthritis by modulating neutrophil activities: an in vitro and in vivo study | |
US11638718B2 (en) | Treatment of alphavirus-induced inflammation | |
Wu et al. | Phthalide derivative CD21 regulates the platelet-neutrophil extracellular trap-thrombin axis and protects against ischemic brain injury in rodents | |
Li et al. | Microglia in neuroimmunopharmacology and drug addiction | |
AU2021201198A1 (en) | Treatment of bone marrow pathologies with polysulfated polysaccharides | |
CN110075304B (en) | Pharmaceutical composition for treating osteoarthritis and application thereof | |
Zhang et al. | flg2 as a potential biomarker of acute cerebral ischemic-reperfusion injury | |
Paladino et al. | Resveratrol reverses the effect of TNF-α on inflammatory markers in a model of autoimmune uveitis |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
STPP | Information on status: patent application and granting procedure in general |
Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION |
|
AS | Assignment |
Owner name: GRIFFITH UNIVERSITY, AUSTRALIA Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MAHALINGAM, SURENDRAN;HERRERO, LARA JOSEFINA;SIGNING DATES FROM 20181212 TO 20181214;REEL/FRAME:050799/0980 |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: NON FINAL ACTION MAILED |
|
STCB | Information on status: application discontinuation |
Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION |