WO2013032964A1 - Compositions and methods for preventing and treating biofilms - Google Patents
Compositions and methods for preventing and treating biofilms Download PDFInfo
- Publication number
- WO2013032964A1 WO2013032964A1 PCT/US2012/052421 US2012052421W WO2013032964A1 WO 2013032964 A1 WO2013032964 A1 WO 2013032964A1 US 2012052421 W US2012052421 W US 2012052421W WO 2013032964 A1 WO2013032964 A1 WO 2013032964A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- macrophage
- biofilm
- macrophages
- subject
- infection
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims abstract description 118
- 239000000203 mixture Substances 0.000 title claims description 20
- 210000002540 macrophage Anatomy 0.000 claims abstract description 296
- 208000015181 infectious disease Diseases 0.000 claims abstract description 199
- 239000007943 implant Substances 0.000 claims abstract description 27
- 210000003127 knee Anatomy 0.000 claims abstract description 8
- 210000003423 ankle Anatomy 0.000 claims abstract description 7
- 210000000707 wrist Anatomy 0.000 claims abstract description 7
- 238000011282 treatment Methods 0.000 claims description 60
- 241001465754 Metazoa Species 0.000 claims description 59
- 239000003795 chemical substances by application Substances 0.000 claims description 49
- 210000004027 cell Anatomy 0.000 claims description 48
- -1 spriamycin Chemical compound 0.000 claims description 42
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 41
- 108060008682 Tumor Necrosis Factor Proteins 0.000 claims description 38
- 108010074328 Interferon-gamma Proteins 0.000 claims description 36
- 230000003213 activating effect Effects 0.000 claims description 34
- 102000003390 tumor necrosis factor Human genes 0.000 claims description 34
- 239000003981 vehicle Substances 0.000 claims description 34
- 230000000770 proinflammatory effect Effects 0.000 claims description 33
- 239000003242 anti bacterial agent Substances 0.000 claims description 27
- 230000003115 biocidal effect Effects 0.000 claims description 25
- 229940088710 antibiotic agent Drugs 0.000 claims description 24
- 150000001875 compounds Chemical class 0.000 claims description 24
- 238000002560 therapeutic procedure Methods 0.000 claims description 24
- 108010059426 Anaphylatoxin C5a Receptor Proteins 0.000 claims description 23
- 102000005590 Anaphylatoxin C5a Receptor Human genes 0.000 claims description 23
- 102100037850 Interferon gamma Human genes 0.000 claims description 20
- 102000035195 Peptidases Human genes 0.000 claims description 20
- 108091005804 Peptidases Proteins 0.000 claims description 20
- 230000002458 infectious effect Effects 0.000 claims description 20
- 229960003130 interferon gamma Drugs 0.000 claims description 20
- 230000002265 prevention Effects 0.000 claims description 18
- 239000000018 receptor agonist Substances 0.000 claims description 17
- 229940044601 receptor agonist Drugs 0.000 claims description 17
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 16
- MSFSPUZXLOGKHJ-UHFFFAOYSA-N Muraminsaeure Natural products OC(=O)C(C)OC1C(N)C(O)OC(CO)C1O MSFSPUZXLOGKHJ-UHFFFAOYSA-N 0.000 claims description 15
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 claims description 15
- 108010013639 Peptidoglycan Proteins 0.000 claims description 15
- 239000000411 inducer Substances 0.000 claims description 15
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 15
- ULGZDMOVFRHVEP-RWJQBGPGSA-N Erythromycin Natural products O([C@@H]1[C@@H](C)C(=O)O[C@@H]([C@@]([C@H](O)[C@@H](C)C(=O)[C@H](C)C[C@@](C)(O)[C@H](O[C@H]2[C@@H]([C@H](C[C@@H](C)O2)N(C)C)O)[C@H]1C)(C)O)CC)[C@H]1C[C@@](C)(OC)[C@@H](O)[C@H](C)O1 ULGZDMOVFRHVEP-RWJQBGPGSA-N 0.000 claims description 14
- XIURVHNZVLADCM-IUODEOHRSA-N cefalotin Chemical compound N([C@H]1[C@@H]2N(C1=O)C(=C(CS2)COC(=O)C)C(O)=O)C(=O)CC1=CC=CS1 XIURVHNZVLADCM-IUODEOHRSA-N 0.000 claims description 13
- JQXXHWHPUNPDRT-WLSIYKJHSA-N rifampicin Chemical compound O([C@](C1=O)(C)O/C=C/[C@@H]([C@H]([C@@H](OC(C)=O)[C@H](C)[C@H](O)[C@H](C)[C@@H](O)[C@@H](C)\C=C\C=C(C)/C(=O)NC=2C(O)=C3C([O-])=C4C)C)OC)C4=C1C3=C(O)C=2\C=N\N1CC[NH+](C)CC1 JQXXHWHPUNPDRT-WLSIYKJHSA-N 0.000 claims description 13
- RJQXTJLFIWVMTO-TYNCELHUSA-N Methicillin Chemical compound COC1=CC=CC(OC)=C1C(=O)N[C@@H]1C(=O)N2[C@@H](C(O)=O)C(C)(C)S[C@@H]21 RJQXTJLFIWVMTO-TYNCELHUSA-N 0.000 claims description 12
- 229960003085 meticillin Drugs 0.000 claims description 12
- 210000001616 monocyte Anatomy 0.000 claims description 12
- 229960001225 rifampicin Drugs 0.000 claims description 12
- 241000283690 Bos taurus Species 0.000 claims description 11
- MYSWGUAQZAJSOK-UHFFFAOYSA-N ciprofloxacin Chemical compound C12=CC(N3CCNCC3)=C(F)C=C2C(=O)C(C(=O)O)=CN1C1CC1 MYSWGUAQZAJSOK-UHFFFAOYSA-N 0.000 claims description 11
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 claims description 10
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 claims description 10
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 claims description 10
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 claims description 10
- 229930182555 Penicillin Natural products 0.000 claims description 10
- 208000035475 disorder Diseases 0.000 claims description 10
- AEUTYOVWOVBAKS-UWVGGRQHSA-N ethambutol Chemical compound CC[C@@H](CO)NCCN[C@@H](CC)CO AEUTYOVWOVBAKS-UWVGGRQHSA-N 0.000 claims description 10
- 238000001356 surgical procedure Methods 0.000 claims description 10
- 108010014414 Chemokine CXCL2 Proteins 0.000 claims description 9
- 102000016951 Chemokine CXCL2 Human genes 0.000 claims description 9
- HZZVJAQRINQKSD-UHFFFAOYSA-N Clavulanic acid Natural products OC(=O)C1C(=CCO)OC2CC(=O)N21 HZZVJAQRINQKSD-UHFFFAOYSA-N 0.000 claims description 9
- 108010013198 Daptomycin Proteins 0.000 claims description 9
- 239000004098 Tetracycline Substances 0.000 claims description 9
- LSQZJLSUYDQPKJ-NJBDSQKTSA-N amoxicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=C(O)C=C1 LSQZJLSUYDQPKJ-NJBDSQKTSA-N 0.000 claims description 9
- 230000015572 biosynthetic process Effects 0.000 claims description 9
- ZAIPMKNFIOOWCQ-UEKVPHQBSA-N cephalexin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@@H]3N(C2=O)C(=C(CS3)C)C(O)=O)=CC=CC=C1 ZAIPMKNFIOOWCQ-UEKVPHQBSA-N 0.000 claims description 9
- DOAKLVKFURWEDJ-QCMAZARJSA-N daptomycin Chemical compound C([C@H]1C(=O)O[C@H](C)[C@@H](C(NCC(=O)N[C@@H](CCCN)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@H](C)C(=O)N[C@@H](CC(O)=O)C(=O)NCC(=O)N[C@H](CO)C(=O)N[C@H](C(=O)N1)[C@H](C)CC(O)=O)=O)NC(=O)[C@H](CC(O)=O)NC(=O)[C@@H](CC(N)=O)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)CCCCCCCCC)C(=O)C1=CC=CC=C1N DOAKLVKFURWEDJ-QCMAZARJSA-N 0.000 claims description 9
- 229940124530 sulfonamide Drugs 0.000 claims description 9
- 235000019364 tetracycline Nutrition 0.000 claims description 9
- 150000003522 tetracyclines Chemical class 0.000 claims description 9
- OHKOGUYZJXTSFX-KZFFXBSXSA-N ticarcillin Chemical compound C=1([C@@H](C(O)=O)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)C=CSC=1 OHKOGUYZJXTSFX-KZFFXBSXSA-N 0.000 claims description 9
- GSDSWSVVBLHKDQ-UHFFFAOYSA-N 9-fluoro-3-methyl-10-(4-methylpiperazin-1-yl)-7-oxo-2,3-dihydro-7H-[1,4]oxazino[2,3,4-ij]quinoline-6-carboxylic acid Chemical compound FC1=CC(C(C(C(O)=O)=C2)=O)=C3N2C(C)COC3=C1N1CCN(C)CC1 GSDSWSVVBLHKDQ-UHFFFAOYSA-N 0.000 claims description 8
- 101710155857 C-C motif chemokine 2 Proteins 0.000 claims description 8
- 201000003883 Cystic fibrosis Diseases 0.000 claims description 8
- 102000004889 Interleukin-6 Human genes 0.000 claims description 8
- 108090001005 Interleukin-6 Proteins 0.000 claims description 8
- 241001494479 Pecora Species 0.000 claims description 8
- 229960003022 amoxicillin Drugs 0.000 claims description 8
- 230000000845 anti-microbial effect Effects 0.000 claims description 8
- 229960000603 cefalotin Drugs 0.000 claims description 8
- 229940106164 cephalexin Drugs 0.000 claims description 8
- 230000001684 chronic effect Effects 0.000 claims description 8
- 229940090805 clavulanate Drugs 0.000 claims description 8
- HZZVJAQRINQKSD-PBFISZAISA-N clavulanic acid Chemical compound OC(=O)[C@H]1C(=C/CO)/O[C@@H]2CC(=O)N21 HZZVJAQRINQKSD-PBFISZAISA-N 0.000 claims description 8
- 229960005484 daptomycin Drugs 0.000 claims description 8
- 208000027866 inflammatory disease Diseases 0.000 claims description 8
- LSQZJLSUYDQPKJ-UHFFFAOYSA-N p-Hydroxyampicillin Natural products O=C1N2C(C(O)=O)C(C)(C)SC2C1NC(=O)C(N)C1=CC=C(O)C=C1 LSQZJLSUYDQPKJ-UHFFFAOYSA-N 0.000 claims description 8
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 claims description 8
- 229960004659 ticarcillin Drugs 0.000 claims description 8
- 102000029816 Collagenase Human genes 0.000 claims description 7
- 108060005980 Collagenase Proteins 0.000 claims description 7
- 208000000913 Kidney Calculi Diseases 0.000 claims description 7
- 206010029148 Nephrolithiasis Diseases 0.000 claims description 7
- 206010031252 Osteomyelitis Diseases 0.000 claims description 7
- 206010052428 Wound Diseases 0.000 claims description 7
- 208000027418 Wounds and injury Diseases 0.000 claims description 7
- MQTOSJVFKKJCRP-BICOPXKESA-N azithromycin Chemical compound O([C@@H]1[C@@H](C)C(=O)O[C@@H]([C@@]([C@H](O)[C@@H](C)N(C)C[C@H](C)C[C@@](C)(O)[C@H](O[C@H]2[C@@H]([C@H](C[C@@H](C)O2)N(C)C)O)[C@H]1C)(C)O)CC)[C@H]1C[C@@](C)(OC)[C@@H](O)[C@H](C)O1 MQTOSJVFKKJCRP-BICOPXKESA-N 0.000 claims description 7
- 229960002424 collagenase Drugs 0.000 claims description 7
- 206010014665 endocarditis Diseases 0.000 claims description 7
- 238000007920 subcutaneous administration Methods 0.000 claims description 7
- 201000001320 Atherosclerosis Diseases 0.000 claims description 6
- 241000283707 Capra Species 0.000 claims description 6
- 206010009137 Chronic sinusitis Diseases 0.000 claims description 6
- 241000282326 Felis catus Species 0.000 claims description 6
- 206010024238 Leptospirosis Diseases 0.000 claims description 6
- 206010028851 Necrosis Diseases 0.000 claims description 6
- HJLSLZFTEKNLFI-UHFFFAOYSA-N Tinidazole Chemical compound CCS(=O)(=O)CCN1C(C)=NC=C1[N+]([O-])=O HJLSLZFTEKNLFI-UHFFFAOYSA-N 0.000 claims description 6
- 208000024035 chronic otitis media Diseases 0.000 claims description 6
- 208000027157 chronic rhinosinusitis Diseases 0.000 claims description 6
- 201000003146 cystitis Diseases 0.000 claims description 6
- 230000017074 necrotic cell death Effects 0.000 claims description 6
- NXFQHRVNIOXGAQ-YCRREMRBSA-N nitrofurantoin Chemical compound O1C([N+](=O)[O-])=CC=C1\C=N\N1C(=O)NC(=O)C1 NXFQHRVNIOXGAQ-YCRREMRBSA-N 0.000 claims description 6
- 210000005259 peripheral blood Anatomy 0.000 claims description 6
- 239000011886 peripheral blood Substances 0.000 claims description 6
- 229920001184 polypeptide Polymers 0.000 claims description 6
- 208000015768 polyposis Diseases 0.000 claims description 6
- SKIVFJLNDNKQPD-UHFFFAOYSA-N sulfacetamide Chemical compound CC(=O)NS(=O)(=O)C1=CC=C(N)C=C1 SKIVFJLNDNKQPD-UHFFFAOYSA-N 0.000 claims description 6
- JLKIGFTWXXRPMT-UHFFFAOYSA-N sulphamethoxazole Chemical compound O1C(C)=CC(NS(=O)(=O)C=2C=CC(N)=CC=2)=N1 JLKIGFTWXXRPMT-UHFFFAOYSA-N 0.000 claims description 6
- IEDVJHCEMCRBQM-UHFFFAOYSA-N trimethoprim Chemical compound COC1=C(OC)C(OC)=CC(CC=2C(=NC(N)=NC=2)N)=C1 IEDVJHCEMCRBQM-UHFFFAOYSA-N 0.000 claims description 6
- 208000019206 urinary tract infection Diseases 0.000 claims description 6
- VCOPTHOUUNAYKQ-WBTCAYNUSA-N (3s)-3,6-diamino-n-[[(2s,5s,8e,11s,15s)-15-amino-11-[(6r)-2-amino-1,4,5,6-tetrahydropyrimidin-6-yl]-8-[(carbamoylamino)methylidene]-2-(hydroxymethyl)-3,6,9,12,16-pentaoxo-1,4,7,10,13-pentazacyclohexadec-5-yl]methyl]hexanamide;(3s)-3,6-diamino-n-[[(2s,5s,8 Chemical compound N1C(=O)\C(=C/NC(N)=O)NC(=O)[C@H](CNC(=O)C[C@@H](N)CCCN)NC(=O)[C@H](C)NC(=O)[C@@H](N)CNC(=O)[C@@H]1[C@@H]1NC(N)=NCC1.N1C(=O)\C(=C/NC(N)=O)NC(=O)[C@H](CNC(=O)C[C@@H](N)CCCN)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CNC(=O)[C@@H]1[C@@H]1NC(N)=NCC1 VCOPTHOUUNAYKQ-WBTCAYNUSA-N 0.000 claims description 5
- FFTVPQUHLQBXQZ-KVUCHLLUSA-N (4s,4as,5ar,12ar)-4,7-bis(dimethylamino)-1,10,11,12a-tetrahydroxy-3,12-dioxo-4a,5,5a,6-tetrahydro-4h-tetracene-2-carboxamide Chemical compound C1C2=C(N(C)C)C=CC(O)=C2C(O)=C2[C@@H]1C[C@H]1[C@H](N(C)C)C(=O)C(C(N)=O)=C(O)[C@@]1(O)C2=O FFTVPQUHLQBXQZ-KVUCHLLUSA-N 0.000 claims description 5
- WDLWHQDACQUCJR-ZAMMOSSLSA-N (6r,7r)-7-[[(2r)-2-azaniumyl-2-(4-hydroxyphenyl)acetyl]amino]-8-oxo-3-[(e)-prop-1-enyl]-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylate Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@@H]3N(C2=O)C(=C(CS3)/C=C/C)C(O)=O)=CC=C(O)C=C1 WDLWHQDACQUCJR-ZAMMOSSLSA-N 0.000 claims description 5
- MINDHVHHQZYEEK-UHFFFAOYSA-N (E)-(2S,3R,4R,5S)-5-[(2S,3S,4S,5S)-2,3-epoxy-5-hydroxy-4-methylhexyl]tetrahydro-3,4-dihydroxy-(beta)-methyl-2H-pyran-2-crotonic acid ester with 9-hydroxynonanoic acid Natural products CC(O)C(C)C1OC1CC1C(O)C(O)C(CC(C)=CC(=O)OCCCCCCCCC(O)=O)OC1 MINDHVHHQZYEEK-UHFFFAOYSA-N 0.000 claims description 5
- FUBFWTUFPGFHOJ-UHFFFAOYSA-N 2-nitrofuran Chemical class [O-][N+](=O)C1=CC=CO1 FUBFWTUFPGFHOJ-UHFFFAOYSA-N 0.000 claims description 5
- 102100026802 72 kDa type IV collagenase Human genes 0.000 claims description 5
- 101710151806 72 kDa type IV collagenase Proteins 0.000 claims description 5
- 108010065839 Capreomycin Proteins 0.000 claims description 5
- 229930186147 Cephalosporin Natural products 0.000 claims description 5
- 108010078777 Colistin Proteins 0.000 claims description 5
- DYDCUQKUCUHJBH-UWTATZPHSA-N D-Cycloserine Chemical compound N[C@@H]1CONC1=O DYDCUQKUCUHJBH-UWTATZPHSA-N 0.000 claims description 5
- MQJKPEGWNLWLTK-UHFFFAOYSA-N Dapsone Chemical compound C1=CC(N)=CC=C1S(=O)(=O)C1=CC=C(N)C=C1 MQJKPEGWNLWLTK-UHFFFAOYSA-N 0.000 claims description 5
- FMTDIUIBLCQGJB-UHFFFAOYSA-N Demethylchlortetracyclin Natural products C1C2C(O)C3=C(Cl)C=CC(O)=C3C(=O)C2=C(O)C2(O)C1C(N(C)C)C(O)=C(C(N)=O)C2=O FMTDIUIBLCQGJB-UHFFFAOYSA-N 0.000 claims description 5
- UIOFUWFRIANQPC-JKIFEVAISA-N Floxacillin Chemical compound N([C@@H]1C(N2[C@H](C(C)(C)S[C@@H]21)C(O)=O)=O)C(=O)C1=C(C)ON=C1C1=C(F)C=CC=C1Cl UIOFUWFRIANQPC-JKIFEVAISA-N 0.000 claims description 5
- IECPWNUMDGFDKC-UHFFFAOYSA-N Fusicsaeure Natural products C12C(O)CC3C(=C(CCC=C(C)C)C(O)=O)C(OC(C)=O)CC3(C)C1(C)CCC1C2(C)CCC(O)C1C IECPWNUMDGFDKC-UHFFFAOYSA-N 0.000 claims description 5
- CEAZRRDELHUEMR-URQXQFDESA-N Gentamicin Chemical compound O1[C@H](C(C)NC)CC[C@@H](N)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](NC)[C@@](C)(O)CO2)O)[C@H](N)C[C@@H]1N CEAZRRDELHUEMR-URQXQFDESA-N 0.000 claims description 5
- 108010015899 Glycopeptides Proteins 0.000 claims description 5
- 102000002068 Glycopeptides Human genes 0.000 claims description 5
- AIJTTZAVMXIJGM-UHFFFAOYSA-N Grepafloxacin Chemical compound C1CNC(C)CN1C(C(=C1C)F)=CC2=C1C(=O)C(C(O)=O)=CN2C1CC1 AIJTTZAVMXIJGM-UHFFFAOYSA-N 0.000 claims description 5
- 108010065805 Interleukin-12 Proteins 0.000 claims description 5
- 102000013462 Interleukin-12 Human genes 0.000 claims description 5
- JUZNIMUFDBIJCM-ANEDZVCMSA-N Invanz Chemical compound O=C([C@H]1NC[C@H](C1)SC=1[C@H](C)[C@@H]2[C@H](C(N2C=1C(O)=O)=O)[C@H](O)C)NC1=CC=CC(C(O)=O)=C1 JUZNIMUFDBIJCM-ANEDZVCMSA-N 0.000 claims description 5
- 108010028921 Lipopeptides Proteins 0.000 claims description 5
- 102100027998 Macrophage metalloelastase Human genes 0.000 claims description 5
- 101710187853 Macrophage metalloelastase Proteins 0.000 claims description 5
- TYMRLRRVMHJFTF-UHFFFAOYSA-N Mafenide Chemical compound NCC1=CC=C(S(N)(=O)=O)C=C1 TYMRLRRVMHJFTF-UHFFFAOYSA-N 0.000 claims description 5
- 102100030417 Matrilysin Human genes 0.000 claims description 5
- 108090000855 Matrilysin Proteins 0.000 claims description 5
- 102000000380 Matrix Metalloproteinase 1 Human genes 0.000 claims description 5
- 108010016113 Matrix Metalloproteinase 1 Proteins 0.000 claims description 5
- 108010015302 Matrix metalloproteinase-9 Proteins 0.000 claims description 5
- 102100030412 Matrix metalloproteinase-9 Human genes 0.000 claims description 5
- UOZODPSAJZTQNH-UHFFFAOYSA-N Paromomycin II Natural products NC1C(O)C(O)C(CN)OC1OC1C(O)C(OC2C(C(N)CC(N)C2O)OC2C(C(O)C(O)C(CO)O2)N)OC1CO UOZODPSAJZTQNH-UHFFFAOYSA-N 0.000 claims description 5
- NHUHCSRWZMLRLA-UHFFFAOYSA-N Sulfisoxazole Chemical compound CC1=NOC(NS(=O)(=O)C=2C=CC(N)=CC=2)=C1C NHUHCSRWZMLRLA-UHFFFAOYSA-N 0.000 claims description 5
- 108010053950 Teicoplanin Proteins 0.000 claims description 5
- 108010059993 Vancomycin Proteins 0.000 claims description 5
- LKCWBDHBTVXHDL-RMDFUYIESA-N amikacin Chemical compound O([C@@H]1[C@@H](N)C[C@H]([C@@H]([C@H]1O)O[C@@H]1[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O1)O)NC(=O)[C@@H](O)CCN)[C@H]1O[C@H](CN)[C@@H](O)[C@H](O)[C@H]1O LKCWBDHBTVXHDL-RMDFUYIESA-N 0.000 claims description 5
- 229940126575 aminoglycoside Drugs 0.000 claims description 5
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 claims description 5
- 230000001355 anti-mycobacterial effect Effects 0.000 claims description 5
- 229940034014 antimycobacterial agent Drugs 0.000 claims description 5
- WZPBZJONDBGPKJ-VEHQQRBSSA-N aztreonam Chemical compound O=C1N(S([O-])(=O)=O)[C@@H](C)[C@@H]1NC(=O)C(=N/OC(C)(C)C(O)=O)\C1=CSC([NH3+])=N1 WZPBZJONDBGPKJ-VEHQQRBSSA-N 0.000 claims description 5
- 229940041011 carbapenems Drugs 0.000 claims description 5
- FPPNZSSZRUTDAP-UWFZAAFLSA-N carbenicillin Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)C(C(O)=O)C1=CC=CC=C1 FPPNZSSZRUTDAP-UWFZAAFLSA-N 0.000 claims description 5
- 229960005361 cefaclor Drugs 0.000 claims description 5
- QYIYFLOTGYLRGG-GPCCPHFNSA-N cefaclor Chemical compound C1([C@H](C(=O)N[C@@H]2C(N3C(=C(Cl)CS[C@@H]32)C(O)=O)=O)N)=CC=CC=C1 QYIYFLOTGYLRGG-GPCCPHFNSA-N 0.000 claims description 5
- 229960003719 cefdinir Drugs 0.000 claims description 5
- RTXOFQZKPXMALH-GHXIOONMSA-N cefdinir Chemical compound S1C(N)=NC(C(=N\O)\C(=O)N[C@@H]2C(N3C(=C(C=C)CS[C@@H]32)C(O)=O)=O)=C1 RTXOFQZKPXMALH-GHXIOONMSA-N 0.000 claims description 5
- OKBVVJOGVLARMR-QSWIMTSFSA-N cefixime Chemical compound S1C(N)=NC(C(=N\OCC(O)=O)\C(=O)N[C@@H]2C(N3C(=C(C=C)CS[C@@H]32)C(O)=O)=O)=C1 OKBVVJOGVLARMR-QSWIMTSFSA-N 0.000 claims description 5
- 229960004682 cefoperazone Drugs 0.000 claims description 5
- GCFBRXLSHGKWDP-XCGNWRKASA-N cefoperazone Chemical compound O=C1C(=O)N(CC)CCN1C(=O)N[C@H](C=1C=CC(O)=CC=1)C(=O)N[C@@H]1C(=O)N2C(C(O)=O)=C(CSC=3N(N=NN=3)C)CS[C@@H]21 GCFBRXLSHGKWDP-XCGNWRKASA-N 0.000 claims description 5
- AZZMGZXNTDTSME-JUZDKLSSSA-M cefotaxime sodium Chemical compound [Na+].N([C@@H]1C(N2C(=C(COC(C)=O)CS[C@@H]21)C([O-])=O)=O)C(=O)\C(=N/OC)C1=CSC(N)=N1 AZZMGZXNTDTSME-JUZDKLSSSA-M 0.000 claims description 5
- ZCCUWMICIWSJIX-NQJJCJBVSA-N ceftaroline fosamil Chemical compound S([C@@H]1[C@@H](C(N1C=1C([O-])=O)=O)NC(=O)\C(=N/OCC)C=2N=C(NP(O)(O)=O)SN=2)CC=1SC(SC=1)=NC=1C1=CC=[N+](C)C=C1 ZCCUWMICIWSJIX-NQJJCJBVSA-N 0.000 claims description 5
- NMVPEQXCMGEDNH-TZVUEUGBSA-N ceftazidime pentahydrate Chemical compound O.O.O.O.O.S([C@@H]1[C@@H](C(N1C=1C([O-])=O)=O)NC(=O)\C(=N/OC(C)(C)C(O)=O)C=2N=C(N)SC=2)CC=1C[N+]1=CC=CC=C1 NMVPEQXCMGEDNH-TZVUEUGBSA-N 0.000 claims description 5
- UNJFKXSSGBWRBZ-BJCIPQKHSA-N ceftibuten Chemical compound S1C(N)=NC(C(=C\CC(O)=O)\C(=O)N[C@@H]2C(N3C(=CCS[C@@H]32)C(O)=O)=O)=C1 UNJFKXSSGBWRBZ-BJCIPQKHSA-N 0.000 claims description 5
- VOAZJEPQLGBXGO-SDAWRPRTSA-N ceftobiprole Chemical compound S1C(N)=NC(C(=N\O)\C(=O)N[C@@H]2C(N3C(=C(\C=C/4C(N([C@H]5CNCC5)CC\4)=O)CS[C@@H]32)C(O)=O)=O)=N1 VOAZJEPQLGBXGO-SDAWRPRTSA-N 0.000 claims description 5
- 229940124587 cephalosporin Drugs 0.000 claims description 5
- 150000001780 cephalosporins Chemical class 0.000 claims description 5
- 239000002975 chemoattractant Substances 0.000 claims description 5
- DHSUYTOATWAVLW-WFVMDLQDSA-N cilastatin Chemical compound CC1(C)C[C@@H]1C(=O)N\C(=C/CCCCSC[C@H](N)C(O)=O)C(O)=O DHSUYTOATWAVLW-WFVMDLQDSA-N 0.000 claims description 5
- AGOYDEPGAOXOCK-KCBOHYOISA-N clarithromycin Chemical compound O([C@@H]1[C@@H](C)C(=O)O[C@@H]([C@@]([C@H](O)[C@@H](C)C(=O)[C@H](C)C[C@](C)([C@H](O[C@H]2[C@@H]([C@H](C[C@@H](C)O2)N(C)C)O)[C@H]1C)OC)(C)O)CC)[C@H]1C[C@@](C)(OC)[C@@H](O)[C@H](C)O1 AGOYDEPGAOXOCK-KCBOHYOISA-N 0.000 claims description 5
- KDLRVYVGXIQJDK-AWPVFWJPSA-N clindamycin Chemical compound CN1C[C@H](CCC)C[C@H]1C(=O)N[C@H]([C@H](C)Cl)[C@@H]1[C@H](O)[C@H](O)[C@@H](O)[C@@H](SC)O1 KDLRVYVGXIQJDK-AWPVFWJPSA-N 0.000 claims description 5
- 229960004287 clofazimine Drugs 0.000 claims description 5
- WDQPAMHFFCXSNU-BGABXYSRSA-N clofazimine Chemical compound C12=CC=CC=C2N=C2C=C(NC=3C=CC(Cl)=CC=3)C(=N/C(C)C)/C=C2N1C1=CC=C(Cl)C=C1 WDQPAMHFFCXSNU-BGABXYSRSA-N 0.000 claims description 5
- LQOLIRLGBULYKD-JKIFEVAISA-N cloxacillin Chemical compound N([C@@H]1C(N2[C@H](C(C)(C)S[C@@H]21)C(O)=O)=O)C(=O)C1=C(C)ON=C1C1=CC=CC=C1Cl LQOLIRLGBULYKD-JKIFEVAISA-N 0.000 claims description 5
- YFAGHNZHGGCZAX-JKIFEVAISA-N dicloxacillin Chemical compound N([C@@H]1C(N2[C@H](C(C)(C)S[C@@H]21)C(O)=O)=O)C(=O)C1=C(C)ON=C1C1=C(Cl)C=CC=C1Cl YFAGHNZHGGCZAX-JKIFEVAISA-N 0.000 claims description 5
- AVAACINZEOAHHE-VFZPANTDSA-N doripenem Chemical compound C=1([C@H](C)[C@@H]2[C@H](C(N2C=1C(O)=O)=O)[C@H](O)C)S[C@@H]1CN[C@H](CNS(N)(=O)=O)C1 AVAACINZEOAHHE-VFZPANTDSA-N 0.000 claims description 5
- 229960003276 erythromycin Drugs 0.000 claims description 5
- NSYZCCDSJNWWJL-YXOIYICCSA-N erythromycin ethylsuccinate Chemical compound O1[C@H](C)C[C@H](N(C)C)[C@@H](OC(=O)CCC(=O)OCC)[C@@H]1O[C@H]1[C@@](O)(C)C[C@@H](C)C(=O)[C@H](C)[C@@H](O)[C@](C)(O)[C@@H](CC)OC(=O)[C@H](C)[C@@H](O[C@@H]2O[C@@H](C)[C@H](O)[C@](C)(OC)C2)[C@@H]1C NSYZCCDSJNWWJL-YXOIYICCSA-N 0.000 claims description 5
- AEOCXXJPGCBFJA-UHFFFAOYSA-N ethionamide Chemical compound CCC1=CC(C(N)=S)=CC=N1 AEOCXXJPGCBFJA-UHFFFAOYSA-N 0.000 claims description 5
- YMDXZJFXQJVXBF-STHAYSLISA-N fosfomycin Chemical compound C[C@@H]1O[C@@H]1P(O)(O)=O YMDXZJFXQJVXBF-STHAYSLISA-N 0.000 claims description 5
- 229960001625 furazolidone Drugs 0.000 claims description 5
- PLHJDBGFXBMTGZ-WEVVVXLNSA-N furazolidone Chemical compound O1C([N+](=O)[O-])=CC=C1\C=N\N1C(=O)OCC1 PLHJDBGFXBMTGZ-WEVVVXLNSA-N 0.000 claims description 5
- 229940117681 interleukin-12 Drugs 0.000 claims description 5
- 229940100601 interleukin-6 Drugs 0.000 claims description 5
- 210000002510 keratinocyte Anatomy 0.000 claims description 5
- OJMMVQQUTAEWLP-KIDUDLJLSA-N lincomycin Chemical compound CN1C[C@H](CCC)C[C@H]1C(=O)N[C@H]([C@@H](C)O)[C@@H]1[C@H](O)[C@H](O)[C@@H](O)[C@@H](SC)O1 OJMMVQQUTAEWLP-KIDUDLJLSA-N 0.000 claims description 5
- 229940041028 lincosamides Drugs 0.000 claims description 5
- ZEKZLJVOYLTDKK-UHFFFAOYSA-N lomefloxacin Chemical compound FC1=C2N(CC)C=C(C(O)=O)C(=O)C2=CC(F)=C1N1CCNC(C)C1 ZEKZLJVOYLTDKK-UHFFFAOYSA-N 0.000 claims description 5
- 229960001977 loracarbef Drugs 0.000 claims description 5
- JAPHQRWPEGVNBT-UTUOFQBUSA-N loracarbef Chemical compound C1([C@H](C(=O)N[C@@H]2C(N3C(=C(Cl)CC[C@@H]32)C([O-])=O)=O)[NH3+])=CC=CC=C1 JAPHQRWPEGVNBT-UTUOFQBUSA-N 0.000 claims description 5
- 239000003120 macrolide antibiotic agent Substances 0.000 claims description 5
- 229940041033 macrolides Drugs 0.000 claims description 5
- DMJNNHOOLUXYBV-PQTSNVLCSA-N meropenem Chemical compound C=1([C@H](C)[C@@H]2[C@H](C(N2C=1C(O)=O)=O)[C@H](O)C)S[C@@H]1CN[C@H](C(=O)N(C)C)C1 DMJNNHOOLUXYBV-PQTSNVLCSA-N 0.000 claims description 5
- YPBATNHYBCGSSN-VWPFQQQWSA-N mezlocillin Chemical compound N([C@@H](C(=O)N[C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C=1C=CC=CC=1)C(=O)N1CCN(S(C)(=O)=O)C1=O YPBATNHYBCGSSN-VWPFQQQWSA-N 0.000 claims description 5
- 239000004005 microsphere Substances 0.000 claims description 5
- 229940041009 monobactams Drugs 0.000 claims description 5
- DDHVILIIHBIMQU-YJGQQKNPSA-L mupirocin calcium hydrate Chemical compound O.O.[Ca+2].C[C@H](O)[C@H](C)[C@@H]1O[C@H]1C[C@@H]1[C@@H](O)[C@@H](O)[C@H](C\C(C)=C\C(=O)OCCCCCCCCC([O-])=O)OC1.C[C@H](O)[C@H](C)[C@@H]1O[C@H]1C[C@@H]1[C@@H](O)[C@@H](O)[C@H](C\C(C)=C\C(=O)OCCCCCCCCC([O-])=O)OC1 DDHVILIIHBIMQU-YJGQQKNPSA-L 0.000 claims description 5
- GPXLMGHLHQJAGZ-JTDSTZFVSA-N nafcillin Chemical compound C1=CC=CC2=C(C(=O)N[C@@H]3C(N4[C@H](C(C)(C)S[C@@H]43)C(O)=O)=O)C(OCC)=CC=C21 GPXLMGHLHQJAGZ-JTDSTZFVSA-N 0.000 claims description 5
- MHWLWQUZZRMNGJ-UHFFFAOYSA-N nalidixic acid Chemical compound C1=C(C)N=C2N(CC)C=C(C(O)=O)C(=O)C2=C1 MHWLWQUZZRMNGJ-UHFFFAOYSA-N 0.000 claims description 5
- 229960000808 netilmicin Drugs 0.000 claims description 5
- OGJPXUAPXNRGGI-UHFFFAOYSA-N norfloxacin Chemical compound C1=C2N(CC)C=C(C(O)=O)C(=O)C2=CC(F)=C1N1CCNCC1 OGJPXUAPXNRGGI-UHFFFAOYSA-N 0.000 claims description 5
- UWYHMGVUTGAWSP-JKIFEVAISA-N oxacillin Chemical compound N([C@@H]1C(N2[C@H](C(C)(C)S[C@@H]21)C(O)=O)=O)C(=O)C1=C(C)ON=C1C1=CC=CC=C1 UWYHMGVUTGAWSP-JKIFEVAISA-N 0.000 claims description 5
- UOZODPSAJZTQNH-LSWIJEOBSA-N paromomycin Chemical compound N[C@@H]1[C@@H](O)[C@H](O)[C@H](CN)O[C@@H]1O[C@H]1[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](N)C[C@@H](N)[C@@H]2O)O[C@@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)N)O[C@@H]1CO UOZODPSAJZTQNH-LSWIJEOBSA-N 0.000 claims description 5
- 229940049954 penicillin Drugs 0.000 claims description 5
- 150000002960 penicillins Chemical class 0.000 claims description 5
- BPLBGHOLXOTWMN-MBNYWOFBSA-N phenoxymethylpenicillin Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)COC1=CC=CC=C1 BPLBGHOLXOTWMN-MBNYWOFBSA-N 0.000 claims description 5
- WCMIIGXFCMNQDS-IDYPWDAWSA-M piperacillin sodium Chemical compound [Na+].O=C1C(=O)N(CC)CCN1C(=O)N[C@H](C=1C=CC=CC=1)C(=O)N[C@@H]1C(=O)N2[C@@H](C([O-])=O)C(C)(C)S[C@@H]21 WCMIIGXFCMNQDS-IDYPWDAWSA-M 0.000 claims description 5
- 150000007660 quinolones Chemical class 0.000 claims description 5
- NZCRJKRKKOLAOJ-XRCRFVBUSA-N rifaximin Chemical compound OC1=C(C(O)=C2C)C3=C4N=C5C=C(C)C=CN5C4=C1NC(=O)\C(C)=C/C=C/[C@H](C)[C@H](O)[C@@H](C)[C@@H](O)[C@@H](C)[C@H](OC(C)=O)[C@H](C)[C@@H](OC)\C=C\O[C@@]1(C)OC2=C3C1=O NZCRJKRKKOLAOJ-XRCRFVBUSA-N 0.000 claims description 5
- DZZWHBIBMUVIIW-DTORHVGOSA-N sparfloxacin Chemical compound C1[C@@H](C)N[C@@H](C)CN1C1=C(F)C(N)=C2C(=O)C(C(O)=O)=CN(C3CC3)C2=C1F DZZWHBIBMUVIIW-DTORHVGOSA-N 0.000 claims description 5
- 229960000268 spectinomycin Drugs 0.000 claims description 5
- UNFWWIHTNXNPBV-WXKVUWSESA-N spectinomycin Chemical compound O([C@@H]1[C@@H](NC)[C@@H](O)[C@H]([C@@H]([C@H]1O1)O)NC)[C@]2(O)[C@H]1O[C@H](C)CC2=O UNFWWIHTNXNPBV-WXKVUWSESA-N 0.000 claims description 5
- VACCAVUAMIDAGB-UHFFFAOYSA-N sulfamethizole Chemical compound S1C(C)=NN=C1NS(=O)(=O)C1=CC=C(N)C=C1 VACCAVUAMIDAGB-UHFFFAOYSA-N 0.000 claims description 5
- 150000003456 sulfonamides Chemical class 0.000 claims description 5
- 108010089019 telavancin Proteins 0.000 claims description 5
- LJVAJPDWBABPEJ-PNUFFHFMSA-N telithromycin Chemical compound O([C@@H]1[C@@H](C)C(=O)[C@@H](C)C(=O)O[C@@H]([C@]2(OC(=O)N(CCCCN3C=C(N=C3)C=3C=NC=CC=3)[C@@H]2[C@@H](C)C(=O)[C@H](C)C[C@@]1(C)OC)C)CC)[C@@H]1O[C@H](C)C[C@H](N(C)C)[C@H]1O LJVAJPDWBABPEJ-PNUFFHFMSA-N 0.000 claims description 5
- 229940040944 tetracyclines Drugs 0.000 claims description 5
- 229960000707 tobramycin Drugs 0.000 claims description 5
- MYPYJXKWCTUITO-UHFFFAOYSA-N vancomycin Natural products O1C(C(=C2)Cl)=CC=C2C(O)C(C(NC(C2=CC(O)=CC(O)=C2C=2C(O)=CC=C3C=2)C(O)=O)=O)NC(=O)C3NC(=O)C2NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(CC(C)C)NC)C(O)C(C=C3Cl)=CC=C3OC3=CC2=CC1=C3OC1OC(CO)C(O)C(O)C1OC1CC(C)(N)C(O)C(C)O1 MYPYJXKWCTUITO-UHFFFAOYSA-N 0.000 claims description 5
- KMEGBUCIGMEPME-LQYKFRDPSA-N (2s,5r,6r)-6-[[(2r)-2-amino-2-phenylacetyl]amino]-3,3-dimethyl-7-oxo-4-thia-1-azabicyclo[3.2.0]heptane-2-carboxylic acid;(1r,4s)-3,3-dimethyl-2,2,6-trioxo-2$l^{6}-thiabicyclo[3.2.0]heptane-4-carboxylic acid Chemical compound O=S1(=O)C(C)(C)[C@H](C(O)=O)C2C(=O)C[C@H]21.C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 KMEGBUCIGMEPME-LQYKFRDPSA-N 0.000 claims description 4
- NNRXCKZMQLFUPL-WBMZRJHASA-N (3r,4s,5s,6r,7r,9r,11r,12r,13s,14r)-6-[(2s,3r,4s,6r)-4-(dimethylamino)-3-hydroxy-6-methyloxan-2-yl]oxy-14-ethyl-7,12,13-trihydroxy-4-[(2r,4r,5s,6s)-5-hydroxy-4-methoxy-4,6-dimethyloxan-2-yl]oxy-3,5,7,9,11,13-hexamethyl-oxacyclotetradecane-2,10-dione;(2r,3 Chemical compound OC(=O)[C@H](O)[C@@H](O)[C@@H]([C@H](O)CO)O[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O.O([C@@H]1[C@@H](C)C(=O)O[C@@H]([C@@]([C@H](O)[C@@H](C)C(=O)[C@H](C)C[C@@](C)(O)[C@H](O[C@H]2[C@@H]([C@H](C[C@@H](C)O2)N(C)C)O)[C@H]1C)(C)O)CC)[C@H]1C[C@@](C)(OC)[C@@H](O)[C@H](C)O1 NNRXCKZMQLFUPL-WBMZRJHASA-N 0.000 claims description 4
- SGKRLCUYIXIAHR-AKNGSSGZSA-N (4s,4ar,5s,5ar,6r,12ar)-4-(dimethylamino)-1,5,10,11,12a-pentahydroxy-6-methyl-3,12-dioxo-4a,5,5a,6-tetrahydro-4h-tetracene-2-carboxamide Chemical compound C1=CC=C2[C@H](C)[C@@H]([C@H](O)[C@@H]3[C@](C(O)=C(C(N)=O)C(=O)[C@H]3N(C)C)(O)C3=O)C3=C(O)C2=C1O SGKRLCUYIXIAHR-AKNGSSGZSA-N 0.000 claims description 4
- GUXHBMASAHGULD-SEYHBJAFSA-N (4s,4as,5as,6s,12ar)-7-chloro-4-(dimethylamino)-1,6,10,11,12a-pentahydroxy-3,12-dioxo-4a,5,5a,6-tetrahydro-4h-tetracene-2-carboxamide Chemical compound C1([C@H]2O)=C(Cl)C=CC(O)=C1C(O)=C1[C@@H]2C[C@H]2[C@H](N(C)C)C(=O)C(C(N)=O)=C(O)[C@@]2(O)C1=O GUXHBMASAHGULD-SEYHBJAFSA-N 0.000 claims description 4
- MMRINLZOZVAPDZ-LSGRDSQZSA-N (6r,7r)-7-[[(2z)-2-(2-amino-1,3-thiazol-4-yl)-2-methoxyiminoacetyl]amino]-3-[(1-methylpyrrolidin-1-ium-1-yl)methyl]-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid;chloride Chemical compound Cl.S([C@@H]1[C@@H](C(N1C=1C([O-])=O)=O)NC(=O)\C(=N/OC)C=2N=C(N)SC=2)CC=1C[N+]1(C)CCCC1 MMRINLZOZVAPDZ-LSGRDSQZSA-N 0.000 claims description 4
- RXZBMPWDPOLZGW-XMRMVWPWSA-N (E)-roxithromycin Chemical compound O([C@@H]1[C@@H](C)C(=O)O[C@@H]([C@@]([C@H](O)[C@@H](C)C(=N/OCOCCOC)/[C@H](C)C[C@@](C)(O)[C@H](O[C@H]2[C@@H]([C@H](C[C@@H](C)O2)N(C)C)O)[C@H]1C)(C)O)CC)[C@H]1C[C@@](C)(OC)[C@@H](O)[C@H](C)O1 RXZBMPWDPOLZGW-XMRMVWPWSA-N 0.000 claims description 4
- QKDHBVNJCZBTMR-LLVKDONJSA-N (R)-temafloxacin Chemical compound C1CN[C@H](C)CN1C(C(=C1)F)=CC2=C1C(=O)C(C(O)=O)=CN2C1=CC=C(F)C=C1F QKDHBVNJCZBTMR-LLVKDONJSA-N 0.000 claims description 4
- XUBOMFCQGDBHNK-JTQLQIEISA-N (S)-gatifloxacin Chemical compound FC1=CC(C(C(C(O)=O)=CN2C3CC3)=O)=C2C(OC)=C1N1CCN[C@@H](C)C1 XUBOMFCQGDBHNK-JTQLQIEISA-N 0.000 claims description 4
- WZRJTRPJURQBRM-UHFFFAOYSA-N 4-amino-n-(5-methyl-1,2-oxazol-3-yl)benzenesulfonamide;5-[(3,4,5-trimethoxyphenyl)methyl]pyrimidine-2,4-diamine Chemical compound O1C(C)=CC(NS(=O)(=O)C=2C=CC(N)=CC=2)=N1.COC1=C(OC)C(OC)=CC(CC=2C(=NC(N)=NC=2)N)=C1 WZRJTRPJURQBRM-UHFFFAOYSA-N 0.000 claims description 4
- WZPBZJONDBGPKJ-UHFFFAOYSA-N Antibiotic SQ 26917 Natural products O=C1N(S(O)(=O)=O)C(C)C1NC(=O)C(=NOC(C)(C)C(O)=O)C1=CSC(N)=N1 WZPBZJONDBGPKJ-UHFFFAOYSA-N 0.000 claims description 4
- 108010001478 Bacitracin Proteins 0.000 claims description 4
- GNWUOVJNSFPWDD-XMZRARIVSA-M Cefoxitin sodium Chemical compound [Na+].N([C@]1(OC)C(N2C(=C(COC(N)=O)CS[C@@H]21)C([O-])=O)=O)C(=O)CC1=CC=CS1 GNWUOVJNSFPWDD-XMZRARIVSA-M 0.000 claims description 4
- DYDCUQKUCUHJBH-UHFFFAOYSA-N D-Cycloserine Natural products NC1CONC1=O DYDCUQKUCUHJBH-UHFFFAOYSA-N 0.000 claims description 4
- YAVZHCFFUATPRK-YZPBMOCRSA-N Erythromycin stearate Chemical compound CCCCCCCCCCCCCCCCCC(O)=O.O([C@@H]1[C@@H](C)C(=O)O[C@@H]([C@@]([C@H](O)[C@@H](C)C(=O)[C@H](C)C[C@@](C)(O)[C@H](O[C@H]2[C@@H]([C@H](C[C@@H](C)O2)N(C)C)O)[C@H]1C)(C)O)CC)[C@H]1C[C@@](C)(OC)[C@@H](O)[C@H](C)O1 YAVZHCFFUATPRK-YZPBMOCRSA-N 0.000 claims description 4
- 229930182566 Gentamicin Natural products 0.000 claims description 4
- GSDSWSVVBLHKDQ-JTQLQIEISA-N Levofloxacin Chemical compound C([C@@H](N1C2=C(C(C(C(O)=O)=C1)=O)C=C1F)C)OC2=C1N1CCN(C)CC1 GSDSWSVVBLHKDQ-JTQLQIEISA-N 0.000 claims description 4
- OJMMVQQUTAEWLP-UHFFFAOYSA-N Lincomycin Natural products CN1CC(CCC)CC1C(=O)NC(C(C)O)C1C(O)C(O)C(O)C(SC)O1 OJMMVQQUTAEWLP-UHFFFAOYSA-N 0.000 claims description 4
- 229930193140 Neomycin Natural products 0.000 claims description 4
- 229930195708 Penicillin V Natural products 0.000 claims description 4
- 108010093965 Polymyxin B Proteins 0.000 claims description 4
- WKDDRNSBRWANNC-UHFFFAOYSA-N Thienamycin Natural products C1C(SCCN)=C(C(O)=O)N2C(=O)C(C(O)C)C21 WKDDRNSBRWANNC-UHFFFAOYSA-N 0.000 claims description 4
- 239000002253 acid Substances 0.000 claims description 4
- 150000007513 acids Chemical class 0.000 claims description 4
- 229960004821 amikacin Drugs 0.000 claims description 4
- 229960000723 ampicillin Drugs 0.000 claims description 4
- 229960004099 azithromycin Drugs 0.000 claims description 4
- 229960003623 azlocillin Drugs 0.000 claims description 4
- JTWOMNBEOCYFNV-NFFDBFGFSA-N azlocillin Chemical compound N([C@@H](C(=O)N[C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C=1C=CC=CC=1)C(=O)N1CCNC1=O JTWOMNBEOCYFNV-NFFDBFGFSA-N 0.000 claims description 4
- 229960003644 aztreonam Drugs 0.000 claims description 4
- 229960003071 bacitracin Drugs 0.000 claims description 4
- 229930184125 bacitracin Natural products 0.000 claims description 4
- CLKOFPXJLQSYAH-ABRJDSQDSA-N bacitracin A Chemical compound C1SC([C@@H](N)[C@@H](C)CC)=N[C@@H]1C(=O)N[C@@H](CC(C)C)C(=O)N[C@H](CCC(O)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H]1C(=O)N[C@H](CCCN)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@H](CC=2C=CC=CC=2)C(=O)N[C@@H](CC=2N=CNC=2)C(=O)N[C@H](CC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)NCCCC1 CLKOFPXJLQSYAH-ABRJDSQDSA-N 0.000 claims description 4
- 229960004602 capreomycin Drugs 0.000 claims description 4
- 229960003669 carbenicillin Drugs 0.000 claims description 4
- 229960004841 cefadroxil Drugs 0.000 claims description 4
- NBFNMSULHIODTC-CYJZLJNKSA-N cefadroxil monohydrate Chemical compound O.C1([C@@H](N)C(=O)N[C@H]2[C@@H]3N(C2=O)C(=C(CS3)C)C(O)=O)=CC=C(O)C=C1 NBFNMSULHIODTC-CYJZLJNKSA-N 0.000 claims description 4
- OLVCFLKTBJRLHI-AXAPSJFSSA-N cefamandole Chemical compound CN1N=NN=C1SCC1=C(C(O)=O)N2C(=O)[C@@H](NC(=O)[C@H](O)C=3C=CC=CC=3)[C@H]2SC1 OLVCFLKTBJRLHI-AXAPSJFSSA-N 0.000 claims description 4
- 229960003012 cefamandole Drugs 0.000 claims description 4
- 229960001139 cefazolin Drugs 0.000 claims description 4
- MLYYVTUWGNIJIB-BXKDBHETSA-N cefazolin Chemical compound S1C(C)=NN=C1SCC1=C(C(O)=O)N2C(=O)[C@@H](NC(=O)CN3N=NN=C3)[C@H]2SC1 MLYYVTUWGNIJIB-BXKDBHETSA-N 0.000 claims description 4
- 229960004069 cefditoren Drugs 0.000 claims description 4
- 229960002100 cefepime Drugs 0.000 claims description 4
- 229960004041 cefetamet Drugs 0.000 claims description 4
- MQLRYUCJDNBWMV-GHXIOONMSA-N cefetamet Chemical compound N([C@@H]1C(N2C(=C(C)CS[C@@H]21)C(O)=O)=O)C(=O)\C(=N/OC)C1=CSC(N)=N1 MQLRYUCJDNBWMV-GHXIOONMSA-N 0.000 claims description 4
- 229960002129 cefixime Drugs 0.000 claims description 4
- SNBUBQHDYVFSQF-HIFRSBDPSA-N cefmetazole Chemical compound S([C@@H]1[C@@](C(N1C=1C(O)=O)=O)(NC(=O)CSCC#N)OC)CC=1CSC1=NN=NN1C SNBUBQHDYVFSQF-HIFRSBDPSA-N 0.000 claims description 4
- 229960003585 cefmetazole Drugs 0.000 claims description 4
- 229960004489 cefonicid Drugs 0.000 claims description 4
- DYAIAHUQIPBDIP-AXAPSJFSSA-N cefonicid Chemical compound S([C@@H]1[C@@H](C(N1C=1C(O)=O)=O)NC(=O)[C@H](O)C=2C=CC=CC=2)CC=1CSC1=NN=NN1CS(O)(=O)=O DYAIAHUQIPBDIP-AXAPSJFSSA-N 0.000 claims description 4
- 229960004261 cefotaxime Drugs 0.000 claims description 4
- SRZNHPXWXCNNDU-RHBCBLIFSA-N cefotetan Chemical compound N([C@]1(OC)C(N2C(=C(CSC=3N(N=NN=3)C)CS[C@@H]21)C(O)=O)=O)C(=O)C1SC(=C(C(N)=O)C(O)=O)S1 SRZNHPXWXCNNDU-RHBCBLIFSA-N 0.000 claims description 4
- 229960005495 cefotetan Drugs 0.000 claims description 4
- 229960002682 cefoxitin Drugs 0.000 claims description 4
- WYUSVOMTXWRGEK-HBWVYFAYSA-N cefpodoxime Chemical compound N([C@H]1[C@@H]2N(C1=O)C(=C(CS2)COC)C(O)=O)C(=O)C(=N/OC)\C1=CSC(N)=N1 WYUSVOMTXWRGEK-HBWVYFAYSA-N 0.000 claims description 4
- 229960005090 cefpodoxime Drugs 0.000 claims description 4
- 229960002580 cefprozil Drugs 0.000 claims description 4
- 229960003202 cefsulodin Drugs 0.000 claims description 4
- SYLKGLMBLAAGSC-QLVMHMETSA-N cefsulodin Chemical compound C1=CC(C(=O)N)=CC=[N+]1CC1=C(C([O-])=O)N2C(=O)[C@@H](NC(=O)[C@@H](C=3C=CC=CC=3)S(O)(=O)=O)[C@H]2SC1 SYLKGLMBLAAGSC-QLVMHMETSA-N 0.000 claims description 4
- 229940036735 ceftaroline Drugs 0.000 claims description 4
- 229960000484 ceftazidime Drugs 0.000 claims description 4
- 229960004086 ceftibuten Drugs 0.000 claims description 4
- 229960001991 ceftizoxime Drugs 0.000 claims description 4
- NNULBSISHYWZJU-LLKWHZGFSA-N ceftizoxime Chemical compound N([C@@H]1C(N2C(=CCS[C@@H]21)C(O)=O)=O)C(=O)\C(=N/OC)C1=CSC(N)=N1 NNULBSISHYWZJU-LLKWHZGFSA-N 0.000 claims description 4
- 229950004259 ceftobiprole Drugs 0.000 claims description 4
- 229960004755 ceftriaxone Drugs 0.000 claims description 4
- VAAUVRVFOQPIGI-SPQHTLEESA-N ceftriaxone Chemical compound S([C@@H]1[C@@H](C(N1C=1C(O)=O)=O)NC(=O)\C(=N/OC)C=2N=C(N)SC=2)CC=1CSC1=NC(=O)C(=O)NN1C VAAUVRVFOQPIGI-SPQHTLEESA-N 0.000 claims description 4
- 229960001668 cefuroxime Drugs 0.000 claims description 4
- JFPVXVDWJQMJEE-IZRZKJBUSA-N cefuroxime Chemical compound N([C@@H]1C(N2C(=C(COC(N)=O)CS[C@@H]21)C(O)=O)=O)C(=O)\C(=N/OC)C1=CC=CO1 JFPVXVDWJQMJEE-IZRZKJBUSA-N 0.000 claims description 4
- DDTDNCYHLGRFBM-YZEKDTGTSA-N chembl2367892 Chemical compound CC(=O)N[C@H]1[C@@H](O)[C@H](O)[C@H](CO)O[C@H]1O[C@@H]([C@H]1C(N[C@@H](C2=CC(O)=CC(O[C@@H]3[C@H]([C@H](O)[C@H](O)[C@@H](CO)O3)O)=C2C=2C(O)=CC=C(C=2)[C@@H](NC(=O)[C@@H]2NC(=O)[C@@H]3C=4C=C(O)C=C(C=4)OC=4C(O)=CC=C(C=4)[C@@H](N)C(=O)N[C@H](CC=4C=C(Cl)C(O5)=CC=4)C(=O)N3)C(=O)N1)C(O)=O)=O)C(C=C1Cl)=CC=C1OC1=C(O[C@H]3[C@H]([C@@H](O)[C@H](O)[C@H](CO)O3)NC(C)=O)C5=CC2=C1 DDTDNCYHLGRFBM-YZEKDTGTSA-N 0.000 claims description 4
- WIIZWVCIJKGZOK-RKDXNWHRSA-N chloramphenicol Chemical compound ClC(Cl)C(=O)N[C@H](CO)[C@H](O)C1=CC=C([N+]([O-])=O)C=C1 WIIZWVCIJKGZOK-RKDXNWHRSA-N 0.000 claims description 4
- 229960004912 cilastatin Drugs 0.000 claims description 4
- 229960004621 cinoxacin Drugs 0.000 claims description 4
- VDUWPHTZYNWKRN-UHFFFAOYSA-N cinoxacin Chemical compound C1=C2N(CC)N=C(C(O)=O)C(=O)C2=CC2=C1OCO2 VDUWPHTZYNWKRN-UHFFFAOYSA-N 0.000 claims description 4
- 229960003405 ciprofloxacin Drugs 0.000 claims description 4
- 229960002626 clarithromycin Drugs 0.000 claims description 4
- 229960002227 clindamycin Drugs 0.000 claims description 4
- 229960003326 cloxacillin Drugs 0.000 claims description 4
- 229940047766 co-trimoxazole Drugs 0.000 claims description 4
- 229960003346 colistin Drugs 0.000 claims description 4
- 238000012258 culturing Methods 0.000 claims description 4
- 229960003077 cycloserine Drugs 0.000 claims description 4
- 229960000860 dapsone Drugs 0.000 claims description 4
- 229960002398 demeclocycline Drugs 0.000 claims description 4
- 239000004053 dental implant Substances 0.000 claims description 4
- 229960001585 dicloxacillin Drugs 0.000 claims description 4
- 229960000895 doripenem Drugs 0.000 claims description 4
- 229960003722 doxycycline Drugs 0.000 claims description 4
- 239000002158 endotoxin Substances 0.000 claims description 4
- IDYZIJYBMGIQMJ-UHFFFAOYSA-N enoxacin Chemical compound N1=C2N(CC)C=C(C(O)=O)C(=O)C2=CC(F)=C1N1CCNCC1 IDYZIJYBMGIQMJ-UHFFFAOYSA-N 0.000 claims description 4
- 229960002770 ertapenem Drugs 0.000 claims description 4
- 229960003203 erythromycin estolate Drugs 0.000 claims description 4
- AWMFUEJKWXESNL-JZBHMOKNSA-N erythromycin estolate Chemical compound CCCCCCCCCCCCOS(O)(=O)=O.O([C@@H]1[C@@H](C)C(=O)O[C@@H]([C@@]([C@H](O)[C@@H](C)C(=O)[C@H](C)C[C@@](C)(O)[C@H](O[C@H]2[C@@H]([C@H](C[C@@H](C)O2)N(C)C)OC(=O)CC)[C@H]1C)(C)O)CC)[C@H]1C[C@@](C)(OC)[C@@H](O)[C@H](C)O1 AWMFUEJKWXESNL-JZBHMOKNSA-N 0.000 claims description 4
- 229960000741 erythromycin ethylsuccinate Drugs 0.000 claims description 4
- 229960004213 erythromycin lactobionate Drugs 0.000 claims description 4
- 229960004142 erythromycin stearate Drugs 0.000 claims description 4
- 229960000285 ethambutol Drugs 0.000 claims description 4
- 229960002001 ethionamide Drugs 0.000 claims description 4
- 229960003306 fleroxacin Drugs 0.000 claims description 4
- XBJBPGROQZJDOJ-UHFFFAOYSA-N fleroxacin Chemical compound C1CN(C)CCN1C1=C(F)C=C2C(=O)C(C(O)=O)=CN(CCF)C2=C1F XBJBPGROQZJDOJ-UHFFFAOYSA-N 0.000 claims description 4
- 229960004273 floxacillin Drugs 0.000 claims description 4
- 229960000308 fosfomycin Drugs 0.000 claims description 4
- 229960004675 fusidic acid Drugs 0.000 claims description 4
- IECPWNUMDGFDKC-MZJAQBGESA-N fusidic acid Chemical compound O[C@@H]([C@@H]12)C[C@H]3\C(=C(/CCC=C(C)C)C(O)=O)[C@@H](OC(C)=O)C[C@]3(C)[C@@]2(C)CC[C@@H]2[C@]1(C)CC[C@@H](O)[C@H]2C IECPWNUMDGFDKC-MZJAQBGESA-N 0.000 claims description 4
- 229960003923 gatifloxacin Drugs 0.000 claims description 4
- 229960002518 gentamicin Drugs 0.000 claims description 4
- 229960000642 grepafloxacin Drugs 0.000 claims description 4
- 229960002182 imipenem Drugs 0.000 claims description 4
- ZSKVGTPCRGIANV-ZXFLCMHBSA-N imipenem Chemical compound C1C(SCC\N=C\N)=C(C(O)=O)N2C(=O)[C@H]([C@H](O)C)[C@H]21 ZSKVGTPCRGIANV-ZXFLCMHBSA-N 0.000 claims description 4
- QRXWMOHMRWLFEY-UHFFFAOYSA-N isoniazide Chemical compound NNC(=O)C1=CC=NC=C1 QRXWMOHMRWLFEY-UHFFFAOYSA-N 0.000 claims description 4
- 229960000318 kanamycin Drugs 0.000 claims description 4
- 229930027917 kanamycin Natural products 0.000 claims description 4
- SBUJHOSQTJFQJX-NOAMYHISSA-N kanamycin Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N SBUJHOSQTJFQJX-NOAMYHISSA-N 0.000 claims description 4
- 229930182823 kanamycin A Natural products 0.000 claims description 4
- 229960005287 lincomycin Drugs 0.000 claims description 4
- TYZROVQLWOKYKF-ZDUSSCGKSA-N linezolid Chemical compound O=C1O[C@@H](CNC(=O)C)CN1C(C=C1F)=CC=C1N1CCOCC1 TYZROVQLWOKYKF-ZDUSSCGKSA-N 0.000 claims description 4
- 229920006008 lipopolysaccharide Polymers 0.000 claims description 4
- 239000002502 liposome Substances 0.000 claims description 4
- 229960002422 lomefloxacin Drugs 0.000 claims description 4
- 229960003640 mafenide Drugs 0.000 claims description 4
- 229960002260 meropenem Drugs 0.000 claims description 4
- VAOCPAMSLUNLGC-UHFFFAOYSA-N metronidazole Chemical compound CC1=NC=C([N+]([O-])=O)N1CCO VAOCPAMSLUNLGC-UHFFFAOYSA-N 0.000 claims description 4
- 229960000198 mezlocillin Drugs 0.000 claims description 4
- 239000000693 micelle Substances 0.000 claims description 4
- 239000011859 microparticle Substances 0.000 claims description 4
- 229960004023 minocycline Drugs 0.000 claims description 4
- 229960003128 mupirocin Drugs 0.000 claims description 4
- 229930187697 mupirocin Natural products 0.000 claims description 4
- JORAUNFTUVJTNG-BSTBCYLQSA-N n-[(2s)-4-amino-1-[[(2s,3r)-1-[[(2s)-4-amino-1-oxo-1-[[(3s,6s,9s,12s,15r,18s,21s)-6,9,18-tris(2-aminoethyl)-3-[(1r)-1-hydroxyethyl]-12,15-bis(2-methylpropyl)-2,5,8,11,14,17,20-heptaoxo-1,4,7,10,13,16,19-heptazacyclotricos-21-yl]amino]butan-2-yl]amino]-3-h Chemical compound CC(C)CCCCC(=O)N[C@@H](CCN)C(=O)N[C@H]([C@@H](C)O)CN[C@@H](CCN)C(=O)N[C@H]1CCNC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCN)NC(=O)[C@H](CCN)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@H](CCN)NC1=O.CCC(C)CCCCC(=O)N[C@@H](CCN)C(=O)N[C@H]([C@@H](C)O)CN[C@@H](CCN)C(=O)N[C@H]1CCNC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCN)NC(=O)[C@H](CCN)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@H](CCN)NC1=O JORAUNFTUVJTNG-BSTBCYLQSA-N 0.000 claims description 4
- 229960000515 nafcillin Drugs 0.000 claims description 4
- 229960000210 nalidixic acid Drugs 0.000 claims description 4
- 239000002105 nanoparticle Substances 0.000 claims description 4
- 239000002077 nanosphere Substances 0.000 claims description 4
- 229960004927 neomycin Drugs 0.000 claims description 4
- ZBGPYVZLYBDXKO-HILBYHGXSA-N netilmycin Chemical compound O([C@@H]1[C@@H](N)C[C@H]([C@@H]([C@H]1O)O[C@@H]1[C@]([C@H](NC)[C@@H](O)CO1)(C)O)NCC)[C@H]1OC(CN)=CC[C@H]1N ZBGPYVZLYBDXKO-HILBYHGXSA-N 0.000 claims description 4
- 229960000564 nitrofurantoin Drugs 0.000 claims description 4
- 229960001180 norfloxacin Drugs 0.000 claims description 4
- 229960001699 ofloxacin Drugs 0.000 claims description 4
- 229960001019 oxacillin Drugs 0.000 claims description 4
- 229960001914 paromomycin Drugs 0.000 claims description 4
- 229940056360 penicillin g Drugs 0.000 claims description 4
- 229940056367 penicillin v Drugs 0.000 claims description 4
- 229960002292 piperacillin Drugs 0.000 claims description 4
- CSOMAHTTWTVBFL-OFBLZTNGSA-N platensimycin Chemical compound C([C@]1([C@@H]2[C@@H]3C[C@@H]4C[C@@]2(C=CC1=O)C[C@@]4(O3)C)C)CC(=O)NC1=C(O)C=CC(C(O)=O)=C1O CSOMAHTTWTVBFL-OFBLZTNGSA-N 0.000 claims description 4
- CSOMAHTTWTVBFL-UHFFFAOYSA-N platensimycin Natural products O1C2(C)CC3(C=CC4=O)CC2CC1C3C4(C)CCC(=O)NC1=C(O)C=CC(C(O)=O)=C1O CSOMAHTTWTVBFL-UHFFFAOYSA-N 0.000 claims description 4
- 229920000024 polymyxin B Polymers 0.000 claims description 4
- XDJYMJULXQKGMM-UHFFFAOYSA-N polymyxin E1 Natural products CCC(C)CCCCC(=O)NC(CCN)C(=O)NC(C(C)O)C(=O)NC(CCN)C(=O)NC1CCNC(=O)C(C(C)O)NC(=O)C(CCN)NC(=O)C(CCN)NC(=O)C(CC(C)C)NC(=O)C(CC(C)C)NC(=O)C(CCN)NC1=O XDJYMJULXQKGMM-UHFFFAOYSA-N 0.000 claims description 4
- KNIWPHSUTGNZST-UHFFFAOYSA-N polymyxin E2 Natural products CC(C)CCCCC(=O)NC(CCN)C(=O)NC(C(C)O)C(=O)NC(CCN)C(=O)NC1CCNC(=O)C(C(C)O)NC(=O)C(CCN)NC(=O)C(CCN)NC(=O)C(CC(C)C)NC(=O)C(CC(C)C)NC(=O)C(CCN)NC1=O KNIWPHSUTGNZST-UHFFFAOYSA-N 0.000 claims description 4
- 229960005266 polymyxin b Drugs 0.000 claims description 4
- 229960003040 rifaximin Drugs 0.000 claims description 4
- 229960005224 roxithromycin Drugs 0.000 claims description 4
- 150000003839 salts Chemical class 0.000 claims description 4
- 229960003600 silver sulfadiazine Drugs 0.000 claims description 4
- 229960004954 sparfloxacin Drugs 0.000 claims description 4
- 229960005322 streptomycin Drugs 0.000 claims description 4
- 229960002673 sulfacetamide Drugs 0.000 claims description 4
- 229960000654 sulfafurazole Drugs 0.000 claims description 4
- 229960005158 sulfamethizole Drugs 0.000 claims description 4
- 229960005404 sulfamethoxazole Drugs 0.000 claims description 4
- FDDDEECHVMSUSB-UHFFFAOYSA-N sulfanilamide Chemical compound NC1=CC=C(S(N)(=O)=O)C=C1 FDDDEECHVMSUSB-UHFFFAOYSA-N 0.000 claims description 4
- NCEXYHBECQHGNR-UHFFFAOYSA-N sulfasalazine Natural products C1=C(O)C(C(=O)O)=CC(N=NC=2C=CC(=CC=2)S(=O)(=O)NC=2N=CC=CC=2)=C1 NCEXYHBECQHGNR-UHFFFAOYSA-N 0.000 claims description 4
- 229960003865 tazobactam Drugs 0.000 claims description 4
- LPQZKKCYTLCDGQ-WEDXCCLWSA-N tazobactam Chemical compound C([C@]1(C)S([C@H]2N(C(C2)=O)[C@H]1C(O)=O)(=O)=O)N1C=CN=N1 LPQZKKCYTLCDGQ-WEDXCCLWSA-N 0.000 claims description 4
- 229960001608 teicoplanin Drugs 0.000 claims description 4
- 229960005240 telavancin Drugs 0.000 claims description 4
- ONUMZHGUFYIKPM-MXNFEBESSA-N telavancin Chemical compound O1[C@@H](C)[C@@H](O)[C@](NCCNCCCCCCCCCC)(C)C[C@@H]1O[C@H]1[C@H](OC=2C3=CC=4[C@H](C(N[C@H]5C(=O)N[C@H](C(N[C@@H](C6=CC(O)=C(CNCP(O)(O)=O)C(O)=C6C=6C(O)=CC=C5C=6)C(O)=O)=O)[C@H](O)C5=CC=C(C(=C5)Cl)O3)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](NC(=O)[C@@H](CC(C)C)NC)[C@H](O)C3=CC=C(C(=C3)Cl)OC=2C=4)O[C@H](CO)[C@@H](O)[C@@H]1O ONUMZHGUFYIKPM-MXNFEBESSA-N 0.000 claims description 4
- 229960003250 telithromycin Drugs 0.000 claims description 4
- 229960004576 temafloxacin Drugs 0.000 claims description 4
- 229960002180 tetracycline Drugs 0.000 claims description 4
- 229930101283 tetracycline Natural products 0.000 claims description 4
- OTVAEFIXJLOWRX-NXEZZACHSA-N thiamphenicol Chemical compound CS(=O)(=O)C1=CC=C([C@@H](O)[C@@H](CO)NC(=O)C(Cl)Cl)C=C1 OTVAEFIXJLOWRX-NXEZZACHSA-N 0.000 claims description 4
- 229960003053 thiamphenicol Drugs 0.000 claims description 4
- 229960005053 tinidazole Drugs 0.000 claims description 4
- NLVFBUXFDBBNBW-PBSUHMDJSA-N tobramycin Chemical compound N[C@@H]1C[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N NLVFBUXFDBBNBW-PBSUHMDJSA-N 0.000 claims description 4
- 229960001082 trimethoprim Drugs 0.000 claims description 4
- 229960000497 trovafloxacin Drugs 0.000 claims description 4
- WVPSKSLAZQPAKQ-CDMJZVDBSA-N trovafloxacin Chemical compound C([C@H]1[C@@H]([C@H]1C1)N)N1C(C(=CC=1C(=O)C(C(O)=O)=C2)F)=NC=1N2C1=CC=C(F)C=C1F WVPSKSLAZQPAKQ-CDMJZVDBSA-N 0.000 claims description 4
- 229960003165 vancomycin Drugs 0.000 claims description 4
- ZXBDZLHAHGPXIG-VTXLJDRKSA-N (3r,4s,5s,6r,7r,9r,11r,12r,13s,14r)-6-[(2s,3r,4s,6r)-4-(dimethylamino)-3-hydroxy-6-methyloxan-2-yl]oxy-14-ethyl-7,12,13-trihydroxy-4-[(2r,4r,5s,6s)-5-hydroxy-4-methoxy-4,6-dimethyloxan-2-yl]oxy-3,5,7,9,11,13-hexamethyl-oxacyclotetradecane-2,10-dione;(2r,3 Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)[C@@H](O)C(O)=O.O([C@@H]1[C@@H](C)C(=O)O[C@@H]([C@@]([C@H](O)[C@@H](C)C(=O)[C@H](C)C[C@@](C)(O)[C@H](O[C@H]2[C@@H]([C@H](C[C@@H](C)O2)N(C)C)O)[C@H]1C)(C)O)CC)[C@H]1C[C@@](C)(OC)[C@@H](O)[C@H](C)O1 ZXBDZLHAHGPXIG-VTXLJDRKSA-N 0.000 claims description 3
- UETNIIAIRMUTSM-UHFFFAOYSA-N Jacareubin Natural products CC1(C)OC2=CC3Oc4c(O)c(O)ccc4C(=O)C3C(=C2C=C1)O UETNIIAIRMUTSM-UHFFFAOYSA-N 0.000 claims description 3
- 239000004100 Oxytetracycline Substances 0.000 claims description 3
- ZWBTYMGEBZUQTK-PVLSIAFMSA-N [(7S,9E,11S,12R,13S,14R,15R,16R,17S,18S,19E,21Z)-2,15,17,32-tetrahydroxy-11-methoxy-3,7,12,14,16,18,22-heptamethyl-1'-(2-methylpropyl)-6,23-dioxospiro[8,33-dioxa-24,27,29-triazapentacyclo[23.6.1.14,7.05,31.026,30]tritriaconta-1(32),2,4,9,19,21,24,26,30-nonaene-28,4'-piperidine]-13-yl] acetate Chemical compound CO[C@H]1\C=C\O[C@@]2(C)Oc3c(C2=O)c2c4NC5(CCN(CC(C)C)CC5)N=c4c(=NC(=O)\C(C)=C/C=C/[C@H](C)[C@H](O)[C@@H](C)[C@@H](O)[C@@H](C)[C@H](OC(C)=O)[C@@H]1C)c(O)c2c(O)c3C ZWBTYMGEBZUQTK-PVLSIAFMSA-N 0.000 claims description 3
- 229960005091 chloramphenicol Drugs 0.000 claims description 3
- 229960002615 dalfopristin Drugs 0.000 claims description 3
- 108700028430 dalfopristin Proteins 0.000 claims description 3
- SUYRLXYYZQTJHF-VMBLUXKRSA-N dalfopristin Chemical compound O=C([C@@H]1N(C2=O)CC[C@H]1S(=O)(=O)CCN(CC)CC)O[C@H](C(C)C)[C@H](C)\C=C\C(=O)NC\C=C\C(\C)=C\[C@@H](O)CC(=O)CC1=NC2=CO1 SUYRLXYYZQTJHF-VMBLUXKRSA-N 0.000 claims description 3
- 229960002549 enoxacin Drugs 0.000 claims description 3
- 229960003350 isoniazid Drugs 0.000 claims description 3
- 229960003376 levofloxacin Drugs 0.000 claims description 3
- 229960003907 linezolid Drugs 0.000 claims description 3
- 229960000282 metronidazole Drugs 0.000 claims description 3
- 229960000625 oxytetracycline Drugs 0.000 claims description 3
- 235000019366 oxytetracycline Nutrition 0.000 claims description 3
- 229960005442 quinupristin Drugs 0.000 claims description 3
- WTHRRGMBUAHGNI-LCYNINFDSA-N quinupristin Chemical compound N([C@@H]1C(=O)N[C@@H](C(N2CCC[C@H]2C(=O)N(C)[C@@H](CC=2C=CC(=CC=2)N(C)C)C(=O)N2C[C@@H](CS[C@H]3C4CCN(CC4)C3)C(=O)C[C@H]2C(=O)N[C@H](C(=O)O[C@@H]1C)C=1C=CC=CC=1)=O)CC)C(=O)C1=NC=CC=C1O WTHRRGMBUAHGNI-LCYNINFDSA-N 0.000 claims description 3
- 108700028429 quinupristin Proteins 0.000 claims description 3
- 229960000885 rifabutin Drugs 0.000 claims description 3
- 229960001940 sulfasalazine Drugs 0.000 claims description 3
- NCEXYHBECQHGNR-QZQOTICOSA-N sulfasalazine Chemical compound C1=C(O)C(C(=O)O)=CC(\N=N\C=2C=CC(=CC=2)S(=O)(=O)NC=2N=CC=CC=2)=C1 NCEXYHBECQHGNR-QZQOTICOSA-N 0.000 claims description 3
- IWVCMVBTMGNXQD-UHFFFAOYSA-N terramycin dehydrate Natural products C1=CC=C2C(O)(C)C3C(O)C4C(N(C)C)C(O)=C(C(N)=O)C(=O)C4(O)C(O)=C3C(=O)C2=C1O IWVCMVBTMGNXQD-UHFFFAOYSA-N 0.000 claims description 3
- 101100504320 Caenorhabditis elegans mcp-1 gene Proteins 0.000 claims description 2
- 102000000018 Chemokine CCL2 Human genes 0.000 claims 1
- KMIPKYQIOVAHOP-YLGJWRNMSA-N cefditoren Chemical compound S([C@@H]1[C@@H](C(N1C=1C(O)=O)=O)NC(=O)\C(=N/OC)C=2N=C(N)SC=2)CC=1\C=C/C=1SC=NC=1C KMIPKYQIOVAHOP-YLGJWRNMSA-N 0.000 claims 1
- IWVCMVBTMGNXQD-PXOLEDIWSA-N oxytetracycline Chemical compound C1=CC=C2[C@](O)(C)[C@H]3[C@H](O)[C@H]4[C@H](N(C)C)C(O)=C(C(N)=O)C(=O)[C@@]4(O)C(O)=C3C(=O)C2=C1O IWVCMVBTMGNXQD-PXOLEDIWSA-N 0.000 claims 1
- UEJSSZHHYBHCEL-UHFFFAOYSA-N silver(1+) sulfadiazinate Chemical compound [Ag+].C1=CC(N)=CC=C1S(=O)(=O)[N-]C1=NC=CC=N1 UEJSSZHHYBHCEL-UHFFFAOYSA-N 0.000 claims 1
- MYPYJXKWCTUITO-LYRMYLQWSA-O vancomycin(1+) Chemical compound O([C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1OC1=C2C=C3C=C1OC1=CC=C(C=C1Cl)[C@@H](O)[C@H](C(N[C@@H](CC(N)=O)C(=O)N[C@H]3C(=O)N[C@H]1C(=O)N[C@H](C(N[C@@H](C3=CC(O)=CC(O)=C3C=3C(O)=CC=C1C=3)C([O-])=O)=O)[C@H](O)C1=CC=C(C(=C1)Cl)O2)=O)NC(=O)[C@@H](CC(C)C)[NH2+]C)[C@H]1C[C@](C)([NH3+])[C@H](O)[C@H](C)O1 MYPYJXKWCTUITO-LYRMYLQWSA-O 0.000 claims 1
- 230000012010 growth Effects 0.000 abstract description 21
- 239000000463 material Substances 0.000 abstract description 9
- 210000001624 hip Anatomy 0.000 abstract description 7
- 241000191967 Staphylococcus aureus Species 0.000 abstract description 3
- 210000001513 elbow Anatomy 0.000 abstract description 3
- 210000002832 shoulder Anatomy 0.000 abstract description 3
- 210000001519 tissue Anatomy 0.000 description 52
- 230000001580 bacterial effect Effects 0.000 description 46
- 241000894006 Bacteria Species 0.000 description 37
- 210000000440 neutrophil Anatomy 0.000 description 31
- 241000699670 Mus sp. Species 0.000 description 24
- 108700033771 Tyr(65)-Phe(67)-Pro(69,71)-Ala(73)- (65-74) complement C5a Proteins 0.000 description 24
- 210000002865 immune cell Anatomy 0.000 description 19
- 238000001727 in vivo Methods 0.000 description 19
- 102000008070 Interferon-gamma Human genes 0.000 description 16
- 238000002474 experimental method Methods 0.000 description 16
- 102000004127 Cytokines Human genes 0.000 description 15
- 108090000695 Cytokines Proteins 0.000 description 15
- 230000032770 biofilm formation Effects 0.000 description 13
- 230000014509 gene expression Effects 0.000 description 13
- 102100031506 Complement C5 Human genes 0.000 description 12
- 230000000694 effects Effects 0.000 description 12
- 238000011740 C57BL/6 mouse Methods 0.000 description 11
- 125000003275 alpha amino acid group Chemical group 0.000 description 11
- 238000013459 approach Methods 0.000 description 11
- 238000000338 in vitro Methods 0.000 description 11
- 238000002347 injection Methods 0.000 description 11
- 239000007924 injection Substances 0.000 description 11
- 230000000069 prophylactic effect Effects 0.000 description 11
- 102000010168 Myeloid Differentiation Factor 88 Human genes 0.000 description 10
- 108010077432 Myeloid Differentiation Factor 88 Proteins 0.000 description 10
- 230000008901 benefit Effects 0.000 description 10
- 230000003176 fibrotic effect Effects 0.000 description 10
- 230000001965 increasing effect Effects 0.000 description 10
- 230000001225 therapeutic effect Effects 0.000 description 10
- 102100021943 C-C motif chemokine 2 Human genes 0.000 description 8
- 102100036170 C-X-C motif chemokine 9 Human genes 0.000 description 8
- 230000003110 anti-inflammatory effect Effects 0.000 description 8
- 230000002829 reductive effect Effects 0.000 description 8
- 102100021723 Arginase-1 Human genes 0.000 description 7
- 101710129000 Arginase-1 Proteins 0.000 description 7
- 230000004888 barrier function Effects 0.000 description 7
- 230000001332 colony forming effect Effects 0.000 description 7
- 230000001276 controlling effect Effects 0.000 description 7
- 230000036541 health Effects 0.000 description 7
- 210000000987 immune system Anatomy 0.000 description 7
- 230000000813 microbial effect Effects 0.000 description 7
- 230000002688 persistence Effects 0.000 description 7
- 108090000623 proteins and genes Proteins 0.000 description 7
- 102000004169 proteins and genes Human genes 0.000 description 7
- 230000004044 response Effects 0.000 description 7
- 238000003501 co-culture Methods 0.000 description 6
- 201000010099 disease Diseases 0.000 description 6
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 6
- 238000003125 immunofluorescent labeling Methods 0.000 description 6
- 230000002757 inflammatory effect Effects 0.000 description 6
- 230000007774 longterm Effects 0.000 description 6
- 239000011159 matrix material Substances 0.000 description 6
- 239000002096 quantum dot Substances 0.000 description 6
- 230000009467 reduction Effects 0.000 description 6
- 238000005070 sampling Methods 0.000 description 6
- 108010058068 tyrosyl-seryl-phenylalanyl-lysyl-aspartyl-methionyl-prolyl-N-methyleucyl-alanyl-arginine Proteins 0.000 description 6
- 102100032367 C-C motif chemokine 5 Human genes 0.000 description 5
- 102000013691 Interleukin-17 Human genes 0.000 description 5
- 108050003558 Interleukin-17 Proteins 0.000 description 5
- 230000004913 activation Effects 0.000 description 5
- 239000000556 agonist Substances 0.000 description 5
- 230000000844 anti-bacterial effect Effects 0.000 description 5
- 230000027455 binding Effects 0.000 description 5
- 210000004369 blood Anatomy 0.000 description 5
- 239000008280 blood Substances 0.000 description 5
- 239000000872 buffer Substances 0.000 description 5
- 244000309464 bull Species 0.000 description 5
- 239000002775 capsule Substances 0.000 description 5
- 230000028993 immune response Effects 0.000 description 5
- 230000002147 killing effect Effects 0.000 description 5
- 244000052769 pathogen Species 0.000 description 5
- 230000028327 secretion Effects 0.000 description 5
- 230000003068 static effect Effects 0.000 description 5
- SEEPANYCNGTZFQ-UHFFFAOYSA-N sulfadiazine Chemical compound C1=CC(N)=CC=C1S(=O)(=O)NC1=NC=CC=N1 SEEPANYCNGTZFQ-UHFFFAOYSA-N 0.000 description 5
- 238000012546 transfer Methods 0.000 description 5
- 230000002485 urinary effect Effects 0.000 description 5
- 101710085500 C-X-C motif chemokine 9 Proteins 0.000 description 4
- JFPVXVDWJQMJEE-QMTHXVAHSA-N Cefuroxime Chemical compound N([C@@H]1C(N2C(=C(COC(N)=O)CS[C@@H]21)C(O)=O)=O)C(=O)C(=NOC)C1=CC=CO1 JFPVXVDWJQMJEE-QMTHXVAHSA-N 0.000 description 4
- 208000002064 Dental Plaque Diseases 0.000 description 4
- 101000797762 Homo sapiens C-C motif chemokine 5 Proteins 0.000 description 4
- 101000947172 Homo sapiens C-X-C motif chemokine 9 Proteins 0.000 description 4
- 101000867983 Homo sapiens C5a anaphylatoxin chemotactic receptor 1 Proteins 0.000 description 4
- 102000003814 Interleukin-10 Human genes 0.000 description 4
- 108090000174 Interleukin-10 Proteins 0.000 description 4
- 208000037942 Methicillin-resistant Staphylococcus aureus infection Diseases 0.000 description 4
- 241000699666 Mus <mouse, genus> Species 0.000 description 4
- 108010003894 Protein-Lysine 6-Oxidase Proteins 0.000 description 4
- 102100026858 Protein-lysine 6-oxidase Human genes 0.000 description 4
- 102100040247 Tumor necrosis factor Human genes 0.000 description 4
- 230000009286 beneficial effect Effects 0.000 description 4
- AFZFFLVORLEPPO-UVYJNCLZSA-N cefditoren pivoxil Chemical compound S([C@@H]1[C@@H](C(N1C=1C(=O)OCOC(=O)C(C)(C)C)=O)NC(=O)\C(=N/OC)C=2N=C(N)SC=2)CC=1\C=C/C=1SC=NC=1C AFZFFLVORLEPPO-UVYJNCLZSA-N 0.000 description 4
- 238000002659 cell therapy Methods 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- 230000018109 developmental process Effects 0.000 description 4
- 230000001747 exhibiting effect Effects 0.000 description 4
- 239000011521 glass Substances 0.000 description 4
- 238000001990 intravenous administration Methods 0.000 description 4
- 239000011325 microbead Substances 0.000 description 4
- 238000010172 mouse model Methods 0.000 description 4
- 230000007112 pro inflammatory response Effects 0.000 description 4
- 230000002206 pro-fibrotic effect Effects 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- MYPYJXKWCTUITO-LYRMYLQWSA-N vancomycin Chemical compound O([C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1OC1=C2C=C3C=C1OC1=CC=C(C=C1Cl)[C@@H](O)[C@H](C(N[C@@H](CC(N)=O)C(=O)N[C@H]3C(=O)N[C@H]1C(=O)N[C@H](C(N[C@@H](C3=CC(O)=CC(O)=C3C=3C(O)=CC=C1C=3)C(O)=O)=O)[C@H](O)C1=CC=C(C(=C1)Cl)O2)=O)NC(=O)[C@@H](CC(C)C)NC)[C@H]1C[C@](C)(N)[C@H](O)[C@H](C)O1 MYPYJXKWCTUITO-LYRMYLQWSA-N 0.000 description 4
- OWFJMIVZYSDULZ-PXOLEDIWSA-N (4s,4ar,5s,5ar,6s,12ar)-4-(dimethylamino)-1,5,6,10,11,12a-hexahydroxy-6-methyl-3,12-dioxo-4,4a,5,5a-tetrahydrotetracene-2-carboxamide Chemical compound C1=CC=C2[C@](O)(C)[C@H]3[C@H](O)[C@H]4[C@H](N(C)C)C(=O)C(C(N)=O)=C(O)[C@@]4(O)C(=O)C3=C(O)C2=C1O OWFJMIVZYSDULZ-PXOLEDIWSA-N 0.000 description 3
- 206010002091 Anaesthesia Diseases 0.000 description 3
- 102100039398 C-X-C motif chemokine 2 Human genes 0.000 description 3
- 102100032957 C5a anaphylatoxin chemotactic receptor 1 Human genes 0.000 description 3
- 102000004190 Enzymes Human genes 0.000 description 3
- 108090000790 Enzymes Proteins 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 101000889128 Homo sapiens C-X-C motif chemokine 2 Proteins 0.000 description 3
- 102000014158 Interleukin-12 Subunit p40 Human genes 0.000 description 3
- 108010011429 Interleukin-12 Subunit p40 Proteins 0.000 description 3
- 208000037581 Persistent Infection Diseases 0.000 description 3
- 230000001464 adherent effect Effects 0.000 description 3
- 150000001413 amino acids Chemical class 0.000 description 3
- 230000037005 anaesthesia Effects 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 230000002238 attenuated effect Effects 0.000 description 3
- 230000003190 augmentative effect Effects 0.000 description 3
- 210000004979 bone marrow derived macrophage Anatomy 0.000 description 3
- 238000004624 confocal microscopy Methods 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 229940088598 enzyme Drugs 0.000 description 3
- 238000000684 flow cytometry Methods 0.000 description 3
- 238000003780 insertion Methods 0.000 description 3
- 230000037431 insertion Effects 0.000 description 3
- 244000005700 microbiome Species 0.000 description 3
- 230000000399 orthopedic effect Effects 0.000 description 3
- 230000000242 pagocytic effect Effects 0.000 description 3
- 230000001717 pathogenic effect Effects 0.000 description 3
- 230000037361 pathway Effects 0.000 description 3
- 210000001539 phagocyte Anatomy 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 230000000284 resting effect Effects 0.000 description 3
- 239000000523 sample Substances 0.000 description 3
- 238000000527 sonication Methods 0.000 description 3
- 238000001228 spectrum Methods 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- 238000006467 substitution reaction Methods 0.000 description 3
- 239000006228 supernatant Substances 0.000 description 3
- 230000008685 targeting Effects 0.000 description 3
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 2
- 206010011409 Cross infection Diseases 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- 102100030131 Interferon regulatory factor 5 Human genes 0.000 description 2
- 206010033078 Otitis media Diseases 0.000 description 2
- 206010034133 Pathogen resistance Diseases 0.000 description 2
- 241000589517 Pseudomonas aeruginosa Species 0.000 description 2
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 2
- 239000000427 antigen Substances 0.000 description 2
- 102000036639 antigens Human genes 0.000 description 2
- 108091007433 antigens Proteins 0.000 description 2
- 210000000544 articulatio talocruralis Anatomy 0.000 description 2
- AGSPXMVUFBBBMO-UHFFFAOYSA-N beta-aminopropionitrile Chemical compound NCCC#N AGSPXMVUFBBBMO-UHFFFAOYSA-N 0.000 description 2
- 210000001185 bone marrow Anatomy 0.000 description 2
- 210000004899 c-terminal region Anatomy 0.000 description 2
- PPKJUHVNTMYXOD-PZGPJMECSA-N c49ws9n75l Chemical compound O=C([C@@H]1N(C2=O)CC[C@H]1S(=O)(=O)CCN(CC)CC)O[C@H](C(C)C)[C@H](C)\C=C\C(=O)NC\C=C\C(\C)=C\[C@@H](O)CC(=O)CC1=NC2=CO1.N([C@@H]1C(=O)N[C@@H](C(N2CCC[C@H]2C(=O)N(C)[C@@H](CC=2C=CC(=CC=2)N(C)C)C(=O)N2C[C@@H](CS[C@H]3C4CCN(CC4)C3)C(=O)C[C@H]2C(=O)N[C@H](C(=O)O[C@@H]1C)C=1C=CC=CC=1)=O)CC)C(=O)C1=NC=CC=C1O PPKJUHVNTMYXOD-PZGPJMECSA-N 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- 230000001010 compromised effect Effects 0.000 description 2
- 238000010494 dissociation reaction Methods 0.000 description 2
- 230000005593 dissociations Effects 0.000 description 2
- 210000002310 elbow joint Anatomy 0.000 description 2
- 229940072686 floxin Drugs 0.000 description 2
- 239000012634 fragment Substances 0.000 description 2
- 150000004676 glycans Chemical class 0.000 description 2
- 210000004394 hip joint Anatomy 0.000 description 2
- 238000003384 imaging method Methods 0.000 description 2
- 230000002519 immonomodulatory effect Effects 0.000 description 2
- 230000003259 immunoinhibitory effect Effects 0.000 description 2
- 239000012678 infectious agent Substances 0.000 description 2
- 201000007119 infective endocarditis Diseases 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 230000002401 inhibitory effect Effects 0.000 description 2
- 238000001361 intraarterial administration Methods 0.000 description 2
- 238000011835 investigation Methods 0.000 description 2
- 210000000629 knee joint Anatomy 0.000 description 2
- 230000005923 long-lasting effect Effects 0.000 description 2
- 230000002132 lysosomal effect Effects 0.000 description 2
- 238000007726 management method Methods 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 239000003550 marker Substances 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 230000001404 mediated effect Effects 0.000 description 2
- 239000002609 medium Substances 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 239000003068 molecular probe Substances 0.000 description 2
- 230000000869 mutational effect Effects 0.000 description 2
- ZESIAEVDVPWEKB-ORCFLVBFSA-N n-[(2s)-4-amino-1-[[(2s,3r)-1-[[(2s)-4-amino-1-oxo-1-[[(3s,6s,9s,12s,15r,18s,21s)-6,9,18-tris(2-aminoethyl)-3-[(1r)-1-hydroxyethyl]-12,15-bis(2-methylpropyl)-2,5,8,11,14,17,20-heptaoxo-1,4,7,10,13,16,19-heptazacyclotricos-21-yl]amino]butan-2-yl]amino]-3-h Chemical compound OS(O)(=O)=O.OS(O)(=O)=O.CC(C)CCCCC(=O)N[C@@H](CCN)C(=O)N[C@H]([C@@H](C)O)CN[C@@H](CCN)C(=O)N[C@H]1CCNC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCN)NC(=O)[C@H](CCN)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@H](CCN)NC1=O.CCC(C)CCCCC(=O)N[C@@H](CCN)C(=O)N[C@H]([C@@H](C)O)CN[C@@H](CCN)C(=O)N[C@H]1CCNC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCN)NC(=O)[C@H](CCN)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@H](CCN)NC1=O ZESIAEVDVPWEKB-ORCFLVBFSA-N 0.000 description 2
- 238000012758 nuclear staining Methods 0.000 description 2
- 229940099980 ocuflox Drugs 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 229920001282 polysaccharide Polymers 0.000 description 2
- 239000005017 polysaccharide Substances 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 230000008672 reprogramming Effects 0.000 description 2
- 210000000323 shoulder joint Anatomy 0.000 description 2
- 210000003491 skin Anatomy 0.000 description 2
- 244000005714 skin microbiome Species 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 230000004936 stimulating effect Effects 0.000 description 2
- 230000000638 stimulation Effects 0.000 description 2
- 238000011269 treatment regimen Methods 0.000 description 2
- YFDSDPIBEUFTMI-UHFFFAOYSA-N tribromoethanol Chemical compound OCC(Br)(Br)Br YFDSDPIBEUFTMI-UHFFFAOYSA-N 0.000 description 2
- 229950004616 tribromoethanol Drugs 0.000 description 2
- 238000002255 vaccination Methods 0.000 description 2
- 229960005486 vaccine Drugs 0.000 description 2
- 210000003857 wrist joint Anatomy 0.000 description 2
- BJNLLBUOHPVGFT-CAYRISATSA-N (1S,2R,19R,22R,34S,37R,40R,52S)-2-[(2R,3R,4R,5S,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-22-amino-5,15-dichloro-64-[(2S,3R,4R,5S,6R)-3-(decanoylamino)-4,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-26,31,44,49-tetrahydroxy-21,35,38,54,56,59-hexaoxo-47-[(3S,4S,5S,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-7,13,28-trioxa-20,36,39,53,55,58-hexazaundecacyclo[38.14.2.23,6.214,17.219,34.18,12.123,27.129,33.141,45.010,37.046,51]hexahexaconta-3,5,8,10,12(64),14,16,23(61),24,26,29(60),30,32,41(57),42,44,46(51),47,49,62,65-henicosaene-52-carboxylic acid Chemical compound CCCCCCCCCC(=O)N[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1Oc1c2Oc3ccc(C[C@H]4NC(=O)[C@H](N)c5ccc(O)c(Oc6cc(O)cc(c6)[C@H](NC4=O)C(=O)N[C@@H]4c(c2)cc1Oc1ccc(cc1Cl)[C@@H](O[C@@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1NC(C)=O)[C@@H]1NC(=O)[C@H](NC4=O)c2ccc(O)c(c2)-c2c(OC4O[C@H](CO)[C@@H](O)[C@H](O)[C@@H]4O)cc(O)cc2[C@H](NC1=O)C(O)=O)c5)cc3Cl BJNLLBUOHPVGFT-CAYRISATSA-N 0.000 description 1
- OIXVKQDWLFHVGR-GQTDVWSESA-N (2r,3s,4r,5r,6r)-5-amino-2-(aminomethyl)-6-[(1r,2r,3s,4r,6s)-4,6-diamino-2-[(2s,3r,4s,5r)-4-[(3r,4r,5s,6s)-3-amino-6-(aminomethyl)-4,5-dihydroxyoxan-2-yl]oxy-3-hydroxy-5-(hydroxymethyl)oxolan-2-yl]oxy-3-hydroxycyclohexyl]oxyoxane-3,4-diol;sulfuric acid Chemical compound OS(O)(=O)=O.N[C@@H]1[C@@H](O)[C@H](O)[C@H](CN)OC1O[C@H]1[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](N)C[C@@H](N)[C@@H]2O)O[C@@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CN)O2)N)O[C@@H]1CO OIXVKQDWLFHVGR-GQTDVWSESA-N 0.000 description 1
- ASROOJRFUZQUBC-MUHZOXKTSA-N (2s)-2-[[2-[[(2s)-2-[[(2s)-5-amino-2-[[(2s)-2-[[(2s)-2-[[(2s)-6-amino-2-[[(2s)-2-[[(2s)-2-[[(2s,3s)-2-amino-3-methylpentanoyl]amino]-3-hydroxypropanoyl]amino]-3-(1h-imidazol-5-yl)propanoyl]amino]hexanoyl]amino]-3-carboxypropanoyl]amino]-4-methylsulfanylbu Chemical compound NC(N)=NCCC[C@@H](C(O)=O)NC(=O)CNC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCSC)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCCCN)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@@H](N)[C@@H](C)CC)CC1=CNC=N1 ASROOJRFUZQUBC-MUHZOXKTSA-N 0.000 description 1
- NZKFUBQRAWPZJP-BXKLGIMVSA-N (2s,3r,4s,5s,6r)-4-amino-2-[(1s,2s,3r,4s,6r)-4,6-diamino-3-[(2r,3r,5s,6r)-3-amino-6-(aminomethyl)-5-hydroxyoxan-2-yl]oxy-2-hydroxycyclohexyl]oxy-6-(hydroxymethyl)oxane-3,5-diol;sulfuric acid Chemical compound OS(O)(=O)=O.OS(O)(=O)=O.OS(O)(=O)=O.OS(O)(=O)=O.OS(O)(=O)=O.N[C@@H]1C[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N.N[C@@H]1C[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N NZKFUBQRAWPZJP-BXKLGIMVSA-N 0.000 description 1
- NWXMGUDVXFXRIG-WESIUVDSSA-N (4s,4as,5as,6s,12ar)-4-(dimethylamino)-1,6,10,11,12a-pentahydroxy-6-methyl-3,12-dioxo-4,4a,5,5a-tetrahydrotetracene-2-carboxamide Chemical compound C1=CC=C2[C@](O)(C)[C@H]3C[C@H]4[C@H](N(C)C)C(=O)C(C(N)=O)=C(O)[C@@]4(O)C(=O)C3=C(O)C2=C1O NWXMGUDVXFXRIG-WESIUVDSSA-N 0.000 description 1
- OAPVUSSHCBRCOL-KBHRXELFSA-N (4s,4as,5as,6s,12ar)-7-chloro-4-(dimethylamino)-1,6,10,11,12a-pentahydroxy-3,12-dioxo-4a,5,5a,6-tetrahydro-4h-tetracene-2-carboxamide;hydrochloride Chemical compound Cl.C1([C@H]2O)=C(Cl)C=CC(O)=C1C(O)=C1[C@@H]2C[C@H]2[C@H](N(C)C)C(=O)C(C(N)=O)=C(O)[C@@]2(O)C1=O OAPVUSSHCBRCOL-KBHRXELFSA-N 0.000 description 1
- QKDHBVNJCZBTMR-UHFFFAOYSA-N 1-(2,4-difluorophenyl)-6-fluoro-7-(3-methylpiperazin-1-yl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid Chemical compound C1CNC(C)CN1C(C(=C1)F)=CC2=C1C(=O)C(C(O)=O)=CN2C1=CC=C(F)C=C1F QKDHBVNJCZBTMR-UHFFFAOYSA-N 0.000 description 1
- ACTOXUHEUCPTEW-CEUOBAOPSA-N 2-[(4r,5s,6s,7r,9r,10r,11e,13e,16r)-6-[(2s,3r,4r,5s,6r)-5-[(2s,4r,5s,6s)-4,5-dihydroxy-4,6-dimethyloxan-2-yl]oxy-4-(dimethylamino)-3-hydroxy-6-methyloxan-2-yl]oxy-10-[(2r,5s,6r)-5-(dimethylamino)-6-methyloxan-2-yl]oxy-4-hydroxy-5-methoxy-9,16-dimethyl-2-o Chemical compound O([C@H]1/C=C/C=C/C[C@@H](C)OC(=O)C[C@@H](O)[C@@H]([C@H]([C@@H](CC=O)C[C@H]1C)O[C@H]1[C@@H]([C@H]([C@H](O[C@@H]2O[C@@H](C)[C@H](O)[C@](C)(O)C2)[C@@H](C)O1)N(C)C)O)OC)[C@H]1CC[C@H](N(C)C)[C@@H](C)O1 ACTOXUHEUCPTEW-CEUOBAOPSA-N 0.000 description 1
- NWXMGUDVXFXRIG-UHFFFAOYSA-N 2-carbamoyl-4-(dimethylazaniumyl)-6,10,11,12a-tetrahydroxy-6-methyl-3,12-dioxo-4,4a,5,5a-tetrahydrotetracen-1-olate Chemical compound C1=CC=C2C(O)(C)C3CC4C(N(C)C)C(=O)C(C(N)=O)=C(O)C4(O)C(=O)C3=C(O)C2=C1O NWXMGUDVXFXRIG-UHFFFAOYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- RZVAJINKPMORJF-UHFFFAOYSA-N Acetaminophen Chemical compound CC(=O)NC1=CC=C(O)C=C1 RZVAJINKPMORJF-UHFFFAOYSA-N 0.000 description 1
- 208000035143 Bacterial infection Diseases 0.000 description 1
- 206010061695 Biliary tract infection Diseases 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 102100025248 C-X-C motif chemokine 10 Human genes 0.000 description 1
- 241000222122 Candida albicans Species 0.000 description 1
- 208000032840 Catheter-Related Infections Diseases 0.000 description 1
- RTXOFQZKPXMALH-PRHODGIISA-N Cefzon Chemical compound S1C(N)=NC(C(=NO)C(=O)N[C@@H]2C(N3C(=C(C=C)CS[C@@H]32)C(O)=O)=O)=C1 RTXOFQZKPXMALH-PRHODGIISA-N 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
- 208000035473 Communicable disease Diseases 0.000 description 1
- 101150031350 Cxcl2 gene Proteins 0.000 description 1
- 229920001651 Cyanoacrylate Polymers 0.000 description 1
- 206010064687 Device related infection Diseases 0.000 description 1
- 206010012735 Diarrhoea Diseases 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 206010014666 Endocarditis bacterial Diseases 0.000 description 1
- 241000194032 Enterococcus faecalis Species 0.000 description 1
- 241000194031 Enterococcus faecium Species 0.000 description 1
- 241000283074 Equus asinus Species 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 102100027286 Fanconi anemia group C protein Human genes 0.000 description 1
- 241000192125 Firmicutes Species 0.000 description 1
- 238000012413 Fluorescence activated cell sorting analysis Methods 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 102100034221 Growth-regulated alpha protein Human genes 0.000 description 1
- 241000606768 Haemophilus influenzae Species 0.000 description 1
- 101000897480 Homo sapiens C-C motif chemokine 2 Proteins 0.000 description 1
- 101000858088 Homo sapiens C-X-C motif chemokine 10 Proteins 0.000 description 1
- 101001069921 Homo sapiens Growth-regulated alpha protein Proteins 0.000 description 1
- 101001011442 Homo sapiens Interferon regulatory factor 5 Proteins 0.000 description 1
- 101710157897 Interferon regulatory factor 5 Proteins 0.000 description 1
- 102000000589 Interleukin-1 Human genes 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- 102000003812 Interleukin-15 Human genes 0.000 description 1
- 108090000172 Interleukin-15 Proteins 0.000 description 1
- 108010002335 Interleukin-9 Proteins 0.000 description 1
- 102000000585 Interleukin-9 Human genes 0.000 description 1
- PIWKPBJCKXDKJR-UHFFFAOYSA-N Isoflurane Chemical compound FC(F)OC(Cl)C(F)(F)F PIWKPBJCKXDKJR-UHFFFAOYSA-N 0.000 description 1
- 208000007764 Legionnaires' Disease Diseases 0.000 description 1
- 241000589902 Leptospira Species 0.000 description 1
- 241000589929 Leptospira interrogans Species 0.000 description 1
- 102000002274 Matrix Metalloproteinases Human genes 0.000 description 1
- 108010000684 Matrix Metalloproteinases Proteins 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 101001065556 Mus musculus Lymphocyte antigen 6G Proteins 0.000 description 1
- 206010028885 Necrotising fasciitis Diseases 0.000 description 1
- CTQNGGLPUBDAKN-UHFFFAOYSA-N O-Xylene Chemical compound CC1=CC=CC=C1C CTQNGGLPUBDAKN-UHFFFAOYSA-N 0.000 description 1
- 206010057249 Phagocytosis Diseases 0.000 description 1
- 206010035664 Pneumonia Diseases 0.000 description 1
- 206010035718 Pneumonia legionella Diseases 0.000 description 1
- 101100070542 Podospora anserina het-s gene Proteins 0.000 description 1
- 239000004793 Polystyrene Substances 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 241000589540 Pseudomonas fluorescens Species 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 208000035415 Reinfection Diseases 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 239000004809 Teflon Substances 0.000 description 1
- 229920006362 Teflon® Polymers 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 229940049505 achromycin v Drugs 0.000 description 1
- 230000004721 adaptive immunity Effects 0.000 description 1
- 150000001298 alcohols Chemical class 0.000 description 1
- 229940090588 amoxil Drugs 0.000 description 1
- 239000012491 analyte Substances 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000001745 anti-biotin effect Effects 0.000 description 1
- 238000009175 antibody therapy Methods 0.000 description 1
- VLAXZGHHBIJLAD-UHFFFAOYSA-N arsphenamine Chemical compound [Cl-].[Cl-].C1=C(O)C([NH3+])=CC([As]=[As]C=2C=C([NH3+])C(O)=CC=2)=C1 VLAXZGHHBIJLAD-UHFFFAOYSA-N 0.000 description 1
- 238000011882 arthroplasty Methods 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 229940098164 augmentin Drugs 0.000 description 1
- 229940062316 avelox Drugs 0.000 description 1
- 229940073066 azactam Drugs 0.000 description 1
- 208000009361 bacterial endocarditis Diseases 0.000 description 1
- 208000022362 bacterial infectious disease Diseases 0.000 description 1
- 244000052616 bacterial pathogen Species 0.000 description 1
- 229940098166 bactrim Drugs 0.000 description 1
- 229940028420 bactroban Drugs 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 229940087430 biaxin Drugs 0.000 description 1
- 229960002685 biotin Drugs 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 229940057194 bleph-10 Drugs 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 210000002798 bone marrow cell Anatomy 0.000 description 1
- FATUQANACHZLRT-KMRXSBRUSA-L calcium glucoheptonate Chemical compound [Ca+2].OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C(O)C([O-])=O.OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C(O)C([O-])=O FATUQANACHZLRT-KMRXSBRUSA-L 0.000 description 1
- 229940095731 candida albicans Drugs 0.000 description 1
- 229940063703 capastat Drugs 0.000 description 1
- 239000011545 carbonate/bicarbonate buffer Substances 0.000 description 1
- 229940097644 cedax Drugs 0.000 description 1
- ICZOIXFFVKYXOM-YCLOEFEOSA-M cefamandole nafate Chemical compound [Na+].CN1N=NN=C1SCC1=C(C([O-])=O)N2C(=O)[C@@H](NC(=O)[C@H](OC=O)C=3C=CC=CC=3)[C@H]2SC1 ICZOIXFFVKYXOM-YCLOEFEOSA-M 0.000 description 1
- FLKYBGKDCCEQQM-WYUVZMMLSA-M cefazolin sodium Chemical compound [Na+].S1C(C)=NN=C1SCC1=C(C([O-])=O)N2C(=O)[C@@H](NC(=O)CN3N=NN=C3)[C@H]2SC1 FLKYBGKDCCEQQM-WYUVZMMLSA-M 0.000 description 1
- LTINZAODLRIQIX-FBXRGJNPSA-N cefpodoxime proxetil Chemical compound N([C@H]1[C@@H]2N(C1=O)C(=C(CS2)COC)C(=O)OC(C)OC(=O)OC(C)C)C(=O)C(=N/OC)\C1=CSC(N)=N1 LTINZAODLRIQIX-FBXRGJNPSA-N 0.000 description 1
- 229940099237 cefzil Drugs 0.000 description 1
- 239000006143 cell culture medium Substances 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 210000002421 cell wall Anatomy 0.000 description 1
- AZFBLLCNOQPJGJ-DVAZEERGSA-N chembl1332607 Chemical compound O([C@](C1=O)(C)O\C=C/[C@@H]([C@H]([C@@H](OC(C)=O)[C@H](C)[C@H](O)[C@H](C)[C@@H](O)[C@@H](C)/C=C\C=C(C)/C(=O)N2)C)OC)C(C(=C3O)C)=C1C(C1=N4)=C3C(O)=C2C1=NC14CCN(CC(C)C)CC1 AZFBLLCNOQPJGJ-DVAZEERGSA-N 0.000 description 1
- 230000014564 chemokine production Effects 0.000 description 1
- 208000003167 cholangitis Diseases 0.000 description 1
- 231100000749 chronicity Toxicity 0.000 description 1
- 229940088516 cipro Drugs 0.000 description 1
- 229940088530 claforan Drugs 0.000 description 1
- 210000003690 classically activated macrophage Anatomy 0.000 description 1
- 229940063193 cleocin Drugs 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 238000010226 confocal imaging Methods 0.000 description 1
- 239000000356 contaminant Substances 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 238000012937 correction Methods 0.000 description 1
- 229940021392 cubicin Drugs 0.000 description 1
- 238000012136 culture method Methods 0.000 description 1
- 238000011461 current therapy Methods 0.000 description 1
- 230000016396 cytokine production Effects 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 231100000517 death Toxicity 0.000 description 1
- 238000001804 debridement Methods 0.000 description 1
- 229940088965 declomycin Drugs 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 208000002925 dental caries Diseases 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- WLOHNSSYAXHWNR-DWIOZXRMSA-N dirithromycin Chemical compound O([C@@H]1[C@@H](C)C(=O)O[C@@H]([C@@]([C@@H]2O[C@H](COCCOC)N[C@H]([C@@H]2C)[C@H](C)C[C@@](C)(O)[C@H](O[C@H]2[C@@H]([C@H](C[C@@H](C)O2)N(C)C)O)[C@H]1C)(C)O)CC)[C@H]1C[C@@](C)(OC)[C@@H](O)[C@H](C)O1 WLOHNSSYAXHWNR-DWIOZXRMSA-N 0.000 description 1
- 231100000676 disease causative agent Toxicity 0.000 description 1
- 229940111539 doribax Drugs 0.000 description 1
- HALQELOKLVRWRI-VDBOFHIQSA-N doxycycline hyclate Chemical compound O.[Cl-].[Cl-].CCO.O=C1C2=C(O)C=CC=C2[C@H](C)[C@@H]2C1=C(O)[C@]1(O)C(=O)C(C(N)=O)=C(O)[C@@H]([NH+](C)C)[C@@H]1[C@H]2O.O=C1C2=C(O)C=CC=C2[C@H](C)[C@@H]2C1=C(O)[C@]1(O)C(=O)C(C(N)=O)=C(O)[C@@H]([NH+](C)C)[C@@H]1[C@H]2O HALQELOKLVRWRI-VDBOFHIQSA-N 0.000 description 1
- 239000003814 drug Substances 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 229940099739 duricef Drugs 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- JJJFUHOGVZWXNQ-UHFFFAOYSA-N enbucrilate Chemical compound CCCCOC(=O)C(=C)C#N JJJFUHOGVZWXNQ-UHFFFAOYSA-N 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 229940032049 enterococcus faecalis Drugs 0.000 description 1
- 230000017188 evasion or tolerance of host immune response Effects 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 229940089936 fortaz Drugs 0.000 description 1
- 229940048400 fucidin Drugs 0.000 description 1
- 230000006870 function Effects 0.000 description 1
- 229940072360 garamycin Drugs 0.000 description 1
- XUBOMFCQGDBHNK-UHFFFAOYSA-N gatifloxacin Chemical compound FC1=CC(C(C(C(O)=O)=CN2C3CC3)=O)=C2C(OC)=C1N1CCNC(C)C1 XUBOMFCQGDBHNK-UHFFFAOYSA-N 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 230000003862 health status Effects 0.000 description 1
- 239000002874 hemostatic agent Substances 0.000 description 1
- 239000011539 homogenization buffer Substances 0.000 description 1
- 230000005745 host immune response Effects 0.000 description 1
- 102000056190 human C5AR1 Human genes 0.000 description 1
- 230000005934 immune activation Effects 0.000 description 1
- 230000007124 immune defense Effects 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000008076 immune mechanism Effects 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000011503 in vivo imaging Methods 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 208000022760 infectious otitis media Diseases 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 230000004941 influx Effects 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 239000000543 intermediate Substances 0.000 description 1
- 229940054114 invanz Drugs 0.000 description 1
- 230000009545 invasion Effects 0.000 description 1
- 229960002725 isoflurane Drugs 0.000 description 1
- 229940090589 keflex Drugs 0.000 description 1
- 229940083668 ketek Drugs 0.000 description 1
- 238000011813 knockout mouse model Methods 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 229940033683 lincocin Drugs 0.000 description 1
- 210000003712 lysosome Anatomy 0.000 description 1
- 230000001868 lysosomic effect Effects 0.000 description 1
- 229940110128 macrobid Drugs 0.000 description 1
- 229940090037 macrodantin Drugs 0.000 description 1
- 238000007885 magnetic separation Methods 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 229940103196 maxaquin Drugs 0.000 description 1
- 229940021422 maxipime Drugs 0.000 description 1
- 229940087515 mefoxin Drugs 0.000 description 1
- 229940032713 merrem Drugs 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 230000003641 microbiacidal effect Effects 0.000 description 1
- 244000000010 microbial pathogen Species 0.000 description 1
- 229940110254 minocin Drugs 0.000 description 1
- 229940112659 monurol Drugs 0.000 description 1
- FABPRXSRWADJSP-MEDUHNTESA-N moxifloxacin Chemical compound COC1=C(N2C[C@H]3NCCC[C@H]3C2)C(F)=CC(C(C(C(O)=O)=C2)=O)=C1N2C1CC1 FABPRXSRWADJSP-MEDUHNTESA-N 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 229940052202 myambutol Drugs 0.000 description 1
- XJODGRWDFZVTKW-ZCFIWIBFSA-N n-methylleucine Chemical compound CN[C@@H](C(O)=O)CC(C)C XJODGRWDFZVTKW-ZCFIWIBFSA-N 0.000 description 1
- 201000007970 necrotizing fasciitis Diseases 0.000 description 1
- AGFWIZQEWFGATK-UNZHCMSXSA-N netilmicin sulfate Chemical compound OS(O)(=O)=O.OS(O)(=O)=O.OS(O)(=O)=O.OS(O)(=O)=O.OS(O)(=O)=O.O([C@@H]1[C@@H](N)C[C@H]([C@@H]([C@H]1O)O[C@@H]1[C@@H]([C@@H](NC)[C@@](C)(O)CO1)O)NCC)[C@H]1OC(CN)=CC[C@H]1N.O([C@@H]1[C@@H](N)C[C@H]([C@@H]([C@H]1O)O[C@@H]1[C@@H]([C@@H](NC)[C@@](C)(O)CO1)O)NCC)[C@H]1OC(CN)=CC[C@H]1N AGFWIZQEWFGATK-UNZHCMSXSA-N 0.000 description 1
- 230000037311 normal skin Effects 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 229940064764 noroxin Drugs 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 229940031908 omnicef Drugs 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 238000004806 packaging method and process Methods 0.000 description 1
- 230000036407 pain Effects 0.000 description 1
- PXDJXZJSCPSGGI-UHFFFAOYSA-N palmityl palmitate Chemical compound CCCCCCCCCCCCCCCCOC(=O)CCCCCCCCCCCCCCC PXDJXZJSCPSGGI-UHFFFAOYSA-N 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 201000001245 periodontitis Diseases 0.000 description 1
- 230000002085 persistent effect Effects 0.000 description 1
- 229940066843 pfizerpen Drugs 0.000 description 1
- 230000008782 phagocytosis Effects 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 229940099765 pipracil Drugs 0.000 description 1
- 230000004260 plant-type cell wall biogenesis Effects 0.000 description 1
- 230000010287 polarization Effects 0.000 description 1
- 229920002223 polystyrene Polymers 0.000 description 1
- 230000008092 positive effect Effects 0.000 description 1
- 229940087661 priftin Drugs 0.000 description 1
- 229940027836 primaxin Drugs 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- ABBQGOCHXSPKHJ-WUKNDPDISA-N prontosil Chemical compound NC1=CC(N)=CC=C1\N=N\C1=CC=C(S(N)(=O)=O)C=C1 ABBQGOCHXSPKHJ-WUKNDPDISA-N 0.000 description 1
- 230000000644 propagated effect Effects 0.000 description 1
- 238000011321 prophylaxis Methods 0.000 description 1
- 201000007094 prostatitis Diseases 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- 230000017854 proteolysis Effects 0.000 description 1
- 229940024999 proteolytic enzymes for treatment of wounds and ulcers Drugs 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 108010071077 quinupristin-dalfopristin Proteins 0.000 description 1
- 229940052337 quinupristin/dalfopristin Drugs 0.000 description 1
- 230000007115 recruitment Effects 0.000 description 1
- 230000000306 recurrent effect Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 229940063639 rifadin Drugs 0.000 description 1
- WDZCUPBHRAEYDL-GZAUEHORSA-N rifapentine Chemical compound O([C@](C1=O)(C)O/C=C/[C@@H]([C@H]([C@@H](OC(C)=O)[C@H](C)[C@H](O)[C@H](C)[C@@H](O)[C@@H](C)\C=C\C=C(C)/C(=O)NC=2C(O)=C3C(O)=C4C)C)OC)C4=C1C3=C(O)C=2\C=N\N(CC1)CCN1C1CCCC1 WDZCUPBHRAEYDL-GZAUEHORSA-N 0.000 description 1
- 229940081561 rocephin Drugs 0.000 description 1
- 238000003118 sandwich ELISA Methods 0.000 description 1
- 229940048278 septra Drugs 0.000 description 1
- 238000013207 serial dilution Methods 0.000 description 1
- 229940099992 seromycin Drugs 0.000 description 1
- 229940099261 silvadene Drugs 0.000 description 1
- ORVLUIMCZUPAPB-LBTQIPEASA-M sodium (4S,4aS,5aS,6S,12aR)-4-(dimethylamino)-1,6,10,11,12a-pentahydroxy-6-methyl-3,12-dioxo-4,4a,5,5a-tetrahydrotetracene-2-carboxamide dioxido(oxo)phosphanium phosphenic acid Chemical compound [Na+].O[P+]([O-])=O.O[P+]([O-])=O.O[P+]([O-])=O.O[P+]([O-])=O.O[P+]([O-])=O.[O-][P+]([O-])=O.CN(C)[C@H]1[C@@H]2C[C@H]3C(=C(O)c4c(O)cccc4[C@@]3(C)O)C(=O)[C@]2(O)C(O)=C(C(N)=O)C1=O.CN(C)[C@H]1[C@@H]2C[C@H]3C(=C(O)c4c(O)cccc4[C@@]3(C)O)C(=O)[C@]2(O)C(O)=C(C(N)=O)C1=O.CN(C)[C@H]1[C@@H]2C[C@H]3C(=C(O)c4c(O)cccc4[C@@]3(C)O)C(=O)[C@]2(O)C(O)=C(C(N)=O)C1=O.CN(C)[C@H]1[C@@H]2C[C@H]3C(=C(O)c4c(O)cccc4[C@@]3(C)O)C(=O)[C@]2(O)C(O)=C(C(N)=O)C1=O.CN(C)[C@H]1[C@@H]2C[C@H]3C(=C(O)c4c(O)cccc4[C@@]3(C)O)C(=O)[C@]2(O)C(O)=C(C(N)=O)C1=O ORVLUIMCZUPAPB-LBTQIPEASA-M 0.000 description 1
- TUPFOYXHAYOHIB-YCAIQWGJSA-M sodium;(2s,5r,6r)-6-[[(2r)-2-[(4-ethyl-2,3-dioxopiperazine-1-carbonyl)amino]-2-phenylacetyl]amino]-3,3-dimethyl-7-oxo-4-thia-1-azabicyclo[3.2.0]heptane-2-carboxylate;(2s,3s,5r)-3-methyl-4,4,7-trioxo-3-(triazol-1-ylmethyl)-4$l^{6}-thia-1-azabicyclo[3.2.0]h Chemical compound [Na+].C([C@]1(C)S([C@H]2N(C(C2)=O)[C@H]1C(O)=O)(=O)=O)N1C=CN=N1.O=C1C(=O)N(CC)CCN1C(=O)N[C@H](C=1C=CC=CC=1)C(=O)N[C@@H]1C(=O)N2[C@@H](C([O-])=O)C(C)(C)S[C@@H]21 TUPFOYXHAYOHIB-YCAIQWGJSA-M 0.000 description 1
- HJHVQCXHVMGZNC-JCJNLNMISA-M sodium;(2z)-2-[(3r,4s,5s,8s,9s,10s,11r,13r,14s,16s)-16-acetyloxy-3,11-dihydroxy-4,8,10,14-tetramethyl-2,3,4,5,6,7,9,11,12,13,15,16-dodecahydro-1h-cyclopenta[a]phenanthren-17-ylidene]-6-methylhept-5-enoate Chemical compound [Na+].O[C@@H]([C@@H]12)C[C@H]3\C(=C(/CCC=C(C)C)C([O-])=O)[C@@H](OC(C)=O)C[C@]3(C)[C@@]2(C)CC[C@@H]2[C@]1(C)CC[C@@H](O)[C@H]2C HJHVQCXHVMGZNC-JCJNLNMISA-M 0.000 description 1
- 125000006850 spacer group Chemical group 0.000 description 1
- 229940010329 spectracef Drugs 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 229940091629 sulfamylon Drugs 0.000 description 1
- 229950006904 sulfisoxazole acetyl Drugs 0.000 description 1
- OPYGFNJSCUDTBT-PMLPCWDUSA-N sultamicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(=O)OCOC(=O)[C@H]2C(S(=O)(=O)[C@H]3N2C(C3)=O)(C)C)(C)C)=CC=CC=C1 OPYGFNJSCUDTBT-PMLPCWDUSA-N 0.000 description 1
- 229940072226 suprax Drugs 0.000 description 1
- 238000011477 surgical intervention Methods 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 230000008961 swelling Effects 0.000 description 1
- 229940020707 synercid Drugs 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 229940049565 targocid Drugs 0.000 description 1
- 229940036731 teflaro Drugs 0.000 description 1
- GSSIWSIRBWAZHG-ACOPVEIWSA-N telavancin hydrochloride Chemical compound Cl.O1[C@@H](C)[C@@H](O)[C@](NCCNCCCCCCCCCC)(C)C[C@@H]1O[C@H]1[C@H](OC=2C3=CC=4[C@H](C(N[C@H]5C(=O)N[C@H](C(N[C@@H](C6=CC(O)=C(CNCP(O)(O)=O)C(O)=C6C=6C(O)=CC=C5C=6)C(O)=O)=O)[C@H](O)C5=CC=C(C(=C5)Cl)O3)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](NC(=O)[C@@H](CC(C)C)NC)[C@H](O)C3=CC=C(C(=C3)Cl)OC=2C=4)O[C@H](CO)[C@@H](O)[C@@H]1O GSSIWSIRBWAZHG-ACOPVEIWSA-N 0.000 description 1
- BVCKFLJARNKCSS-DWPRYXJFSA-N temocillin Chemical compound N([C@]1(OC)C(N2[C@H](C(C)(C)S[C@@H]21)C(O)=O)=O)C(=O)C(C(O)=O)C=1C=CSC=1 BVCKFLJARNKCSS-DWPRYXJFSA-N 0.000 description 1
- 229940061354 tequin Drugs 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- KTAVBOYXMBQFGR-MAODNAKNSA-J tetrasodium;(6r,7r)-7-[[(2z)-2-(2-amino-1,3-thiazol-4-yl)-2-methoxyimino-1-oxidoethylidene]amino]-3-[(2-methyl-5,6-dioxo-1h-1,2,4-triazin-3-yl)sulfanylmethyl]-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylate;heptahydrate Chemical compound O.O.O.O.O.O.O.[Na+].[Na+].[Na+].[Na+].S([C@@H]1[C@@H](C(N1C=1C([O-])=O)=O)NC(=O)\C(=N/OC)C=2N=C(N)SC=2)CC=1CSC1=NC(=O)C([O-])=NN1C.S([C@@H]1[C@@H](C(N1C=1C([O-])=O)=O)NC(=O)\C(=N/OC)C=2N=C(N)SC=2)CC=1CSC1=NC(=O)C([O-])=NN1C KTAVBOYXMBQFGR-MAODNAKNSA-J 0.000 description 1
- 229940027257 timentin Drugs 0.000 description 1
- 229940063418 tindamax Drugs 0.000 description 1
- 239000003106 tissue adhesive Substances 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 229940079342 trecator Drugs 0.000 description 1
- DYNZICQDCVYXFW-AHZSKCOESA-N trovafloxacin mesylate Chemical compound CS(O)(=O)=O.C([C@H]1[C@@H]([C@H]1C1)N)N1C(C(=CC=1C(=O)C(C(O)=O)=C2)F)=NC=1N2C1=CC=C(F)C=C1F DYNZICQDCVYXFW-AHZSKCOESA-N 0.000 description 1
- 229940055820 trovan Drugs 0.000 description 1
- 229940020930 unasyn Drugs 0.000 description 1
- 229940072335 vancocin Drugs 0.000 description 1
- 229940054969 vantin Drugs 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 229940020733 vibativ Drugs 0.000 description 1
- 229940063678 vibramycin Drugs 0.000 description 1
- 230000001018 virulence Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 229940064406 xifaxan Drugs 0.000 description 1
- 239000008096 xylene Substances 0.000 description 1
- 229940049589 zagam Drugs 0.000 description 1
- 229940072251 zithromax Drugs 0.000 description 1
- 229940104666 zosyn Drugs 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/43—Enzymes; Proenzymes; Derivatives thereof
- A61K38/46—Hydrolases (3)
- A61K38/48—Hydrolases (3) acting on peptide bonds (3.4)
- A61K38/4886—Metalloendopeptidases (3.4.24), e.g. collagenase
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4614—Monocytes; Macrophages
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/462—Cellular immunotherapy characterized by the effect or the function of the cells
- A61K39/4622—Antigen presenting cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4648—Bacterial antigens
- A61K39/464821—Staphylococcus
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/38—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
Definitions
- Microorganisms such as bacteria, can form biofilms, where organisms are encased in a complex three-dimensional matrix consisting of extracellular polysaccharides, DNA, and proteins. This structure protects the microbes from a variety of physical and chemical challenges, making biofilm treatment difficult. In addition, upon biofilm formation, some bacteria assume a metabolically dormant state, which protects them from antibiotic killing that requires active cell wall and/or protein synthesis.
- Biofilm infections often lead to significant morbidity due to their chronicity and recalcitrance to antibiotics.
- Biofilms cause over 80% of infections and approximately 65% of nosocomial infections caused by microorganisms in the developed world involve biofilms.
- Biofilms are responsible for catheter infections that cause about 10,000 U.S. deaths and more than 11 billion US dollars in losses in hospital costs annually. Twenty percent of urinary catheters inserted into 5 million U.S. patients have developed biofilm infections, resulting in medical expenses of 1.6 billion U.S. dollars each year.
- biofilms of a single potentially pathogenic bacterium Pseudomonas aeruginosa
- Pseudomonas aeruginosa are the cause of 70,000 cases of cystic fibrosis worldwide.
- U.S. dollars are spent in medical care and productivity losses in the United States due to 25 to 80 million cases annually of biofilm-related food-borne diarrhea and 8,000 to 18,000 cases of biofilm-related
- Bacterial biofilm infections alter host immune responses to favor anti-inflammatory and pro-fibrotic pathways, which contribute to biofilm persistence.
- a variety of microbes e.g., bacteria, have been identified as being associated with, or causative agents of, biofilm.
- a number of these infectious microbes are major health scourges, including methicillin- resistant Staphylococcus aureus (MRSA), a common etiologic agent of biofilms that often cause chronic and recurrent infections when associated with indwelling medical devices, such as implants and catheters.
- MRSA methicillin- resistant Staphylococcus aureus
- the persistent biofilms caused by these microbes are associated with a variety of diseases and medical conditions, including but not limited to, dental caries, periodontitis, infection complications associated with cystic fibrosis, pneumonia, infective endocarditis, muscle skeletal infections, necrotizing fasciitis, osteomyelitis, meloidosis, infectious kidney stones, bacterial endocarditis, airway infections, otitis media, biliary tract infections, chronic bacterial prostatitis, and infections associated with medical devices ⁇ e.g., intravenous catheters, urinary catheters, artificial joints, contact lenses).
- diseases and medical conditions including but not limited to, dental caries, periodontitis, infection complications associated with cystic fibrosis, pneumonia, infective endocarditis, muscle skeletal infections, necrotizing fasciitis, osteomyelitis, meloidosis, infectious kidney stones, bacterial endocarditis, airway infections, otitis media, biliary tract infections, chronic bacterial prostatitis, and
- antibiotics are commonly used to control microbes, e.g., bacteria, that escape the biofilm matrix in order to prevent their colonization of other tissue sites.
- Such uses of antibiotics impose selection pressures on microbes such as bacteria and increase the chance of antibiotic-resistant strains arising.
- a relatively recent development regarding antibiotic treatment involves an administration schedule characterized by the delivery of intermittent low dosages of antibiotics to treat biofilm infection.
- the schedule is designed to better attack persister cells, which are biofilm-associated microbes that survive initial, high-dosage treatments.
- Persister cells have been offered as an explanation for the capacity of many biofilm infections to not only survive initial antibiotic treatments, but to propagate biofilm infection over time. This capacity of biofilm infections has been suggested as a basis for the chronic infections associated with biofilm.
- the intermittent, low-dose approach to biofilm infection treatment may help to treat persister cells associated with biofilm, this approach does not advance methods to prevent biofilm infection and it does not reduce the risk of developing resistant strains because it continues to apply selective pressure in the form of antibiotics.
- biofilm infections associated with artificial devices such as implants or catheters
- current therapy involves a cycle of implant or catheter removal and replacement, which is inconvenient, ineffective, and typically undesirable.
- Management of implant- or device-associated biofilm infections involves a staged replacement of the hardware, either as a single-step exchange, whereby the entire implant is replaced in a single procedure or, more commonly, as a two-stage exchange.
- patients receive extended antibiotic regimens in addition to surgical management, which generally consists of device removal and replacement with an antibiotic-impregnated temporary spacer, followed by insertion of a new prosthesis after a two- to eight- week period. This is a long and debilitating process associated with significant morbidity and economic impact for patients.
- compositions and methods for preventing and treating biofilms that are efficacious and cost-effective without incurring significant risk of toxicity or other complications arising from prophylactic or therapeutic treatment, while minimizing the need for surgical intervention.
- the materials and methods (and uses) disclosed herein provide cells, such as autologous cells, that can be manipulated ex vivo and/or in vivo to become activated macrophages suitable for use in preventing or treating biofilm infections.
- the disclosure provides materials and methods of wide applicability, i.e., any biofilm associated with a native surface or "foreign" device such as an infectious microbe ⁇ e.g., bacterium) is amenable to prevention or treatment with the disclosed materials and methods.
- the disclosed methods provide the benefit of phrophylactic and therapeutic approaches that will not contribute to the collective evolutionary problem of effectively selecting for infectious superbugs resistant to most treatments, such as methicillin-resistant S. aureus (i.e., MRSA), because the methods take advantage of bacterial killing attributes inherent to the host's immune system.
- biofilms can skew the immune response to favor anti-inflammatory and pro-fibrotic pathways, which contribute to biofilm persistence.
- macrophages are a prominent infiltrate in S. aureus biofilm infections, their penetration into the biofilm itself is impeded by a robust fibrotic response surrounding the infection.
- biofilm-associated macrophages are polarized towards an alternatively activated M2 phenotype that exhibits anti-inflammatory and pro-fibrotic properties that limit bacterial clearance.
- the programming of macrophages towards a microbicidal Ml response is diverted towards a M2 phenotype prior to physical contact with the biofilm.
- a macrophage-activating peptide such as a C5a receptor agonist, a C5a analog, or a C5a fragment
- EP67 YSFKDMP(MeL)aR; SEQ ID NO:l; uppercase letters designate the L stereoisomeric form and lower case the D stereoisomeric form of the amino acids; (MeL) corresponds to N-methyl leucine)
- EP67 was derived from the C-terminal region of human complement component, C5a (Taylor SM, et al., Curr Med Chem 2001;8:675-684.).
- EP67 possesses potent immune-enhancing properties (Morgan et al., Vaccine, 28(2): 463-469 (2009)).
- the disclosure further provides a C5a analog comprising 1, 2, 3, 4, or 5 amino acid substitutions in the amino acid sequence set out in SEQ ID NO:l wherein the peptide having the substituted amino acid sequence retains binding activity of the peptide set out in SEQ ID NO:l.
- the C-terminus of C5a comprises amino acids 65-74 of C5a (i.e. , ISHKDMQLGR ("C5a 65 _74") (SEQ ID NO:4).
- the disclosure further provides a C5a analog comprising 1, 2, 3, 4, or 5 amino acid substitutions in the amino acid set out in SEQ ID NO: 4 wherein the peptide having the substituted amino acid sequence retains binding activity of the peptide set out in SEQ ID NO:4.
- the disclosure further provides a C5a analog comprising 1, 2, 3, 4, or 5 amino acid substitutions in the amino acid sequence set out in SEQ ID NO:2 wherein the peptide having the substituted amino acid sequence retains binding activity of the peptide set out in SEQ ID NO:2. Additional disclosure of C5a analogs is provided in USSN 61/359,444 and International (PCT) Patent Application No.
- One or more of the macrophage-activating compounds disclosed herein e.g., a C5a receptor agonist, a C5a analog or a C5a fragment, is useful in methods of activating macrophages prior to, concurrent with, or after administration of macrophage to prevent or treat a biofilm infection.
- the macrophage-activating compound contacts the macrophage ex vivo prior to administration of the macrophages.
- the macrophage-activating compound is coadministered with the macrophages or the macrophage-activating compound is administered after the administration of the macrophages, where administration beyond the fibrotic barrier and in the vicinity of the biofilm places the macrophage-activating compound(s) and macrophages in sufficiently close proximity to result in contact and macrophage activation in vivo.
- Targeting macrophage pro-inflammatory activity with exogenous Ml -activated macrophages inhibited S. aureus biofilm formation and provided long-lasting protection from catheter colonization and infection in surrounding tissues.
- biofilm prevention and treatment exploit natural host immune mechanisms for therapeutic benefit.
- Targeting the host response rather than the pathogen itself offers certain advantages by largely avoiding selective pressures for the evolution of microbial resistance.
- stimulating adaptive immunity through vaccination has remained effective in the face of microbial resistance over decades of clinical use, although not all pathogens have been amenable to vaccination strategies, most notably S. aureus.
- innate immune defenses are geared to rapidly recognize an infinite pathogen repertoire, indicates that immunomodulation will afford broad- spectrum protection against a range of microbial pathogens, enabling prophylactic use in high-risk groups and early treatment prior to the identification of causative infectious agents.
- Ml macrophages for biofilm prevention and treatment disclosed herein overcomes immune evasion by the causative microbes by triggering a pro-inflammatory milieu.
- Early administration of Ml-activated macrophages significantly attenuated biofilm formation in a mouse model of MRSA catheter- associated infection.
- pro-inflammatory mediators were significantly elevated in biofilm-infected tissues from macrophage-treated animals, revealing effective reprogramming of the biofilm environment to a pro-inflammatory milieu.
- Treatment of established biofilm infections with activated macrophages also significantly reduced catheter-associated biofilm burdens compared to antibiotic treatment.
- the disclosure provides a convenient and effective way to prophylactically and/or therapeutically reduce the incidence and/or severity of microbial, e.g., bacterial, biofilm infections generally, and more particularly, of microbial, e.g., bacterial, biofilm infections associated with catheters (e.g., urinary, intravenous, an intraarterial catheters) and other artificial implants such as artificial joint components (e.g., artificial components of hip, knee, ankle, shoulder, elbow and wrist joints).
- catheters e.g., urinary, intravenous, an intraarterial catheters
- other artificial implants such as artificial joint components (e.g., artificial components of hip, knee, ankle, shoulder, elbow and wrist joints).
- compositions comprising an isolated activated macrophage for the prevention or treatment of a biofilm.
- the composition further comprises a macrophage- activating peptide selected from the group consisting of EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2) and C5a 65 - 7 4 (SEQ ID NO:4).
- Variants and derivatives of the EP67 peptide, the EP54 peptide, or the C5a 6 5- 74 peptide that retain the capacity to beneficially affect endogenous macrophages, as disclosed herein, are also contemplated, and in particular those variants and derivatives of the EP67 peptide, EP54 peptide, and C5a 6 5- 7 4 peptide that differ by 1, 2, 3, 4, or 5 residues from the relevant wild-type sequence, i.e., SEQ ID NO:l for EP67, SEQ ID NO:2 for EP54, and SEQ ID NO:4 for C5a 65 _ 74 are within the scope of the compositions, and methods, of the disclosure.
- kits comprising an isolated macrophage and a macrophage- activating agent.
- a kit comprising a macrophage- activating agent.
- the macrophage- activating agent is selected from the group consisting of a C5a receptor agonist, including but not limited to macrophage-activating peptides such as EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), C5a 65 - 74 (SEQ ID NO:4) and analogs and variants thereof, interleukin- ⁇ as well as combinations of interferon- ⁇ and tumor necrosis factor (TNF), and interferon- ⁇ and an inducer of TNF.
- a C5a receptor agonist including but not limited to macrophage-activating peptides such as EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), C5a 65 - 74 (SEQ ID NO:4) and analogs and variants thereof, interleukin- ⁇ as well as combinations of interferon- ⁇ and tumor necrosis
- kits according to the disclosure include variants and derivatives of the EP67 peptide, the EP54 peptide, or the C5a 6 5- 74 peptide that retain the capacity to beneficially affect endogenous macrophages, as disclosed herein, and in particular, those variants and derivatives that differ by 1, 2, 3, 4, or 5 residues from the relevant wild-type sequence, i.e., SEQ ID NO:l for EP67, SEQ ID NO:2 for EP54, and SEQ ID NO:4 for C5a 6 5- 74 are within the scope of the kits of the disclosure.
- Another aspect of the disclosure is a method of providing biofilm therapy to a subject-in-need comprising: (a) isolating peripheral blood monocytes from a subject; (b) culturing the monocytes under conditions compatible with formation of macrophages; and (c) administering a prophylactically or therapeutically effective amount of the macrophages to a subject-in-need, wherein the macrophages are activated by contact with a macrophage- activating agent prior to, simultaneously with, or after administration of the macrophages.
- the macrophages are activated by contact with a macrophage- activating agent prior to administration of the macrophages.
- a related aspect is drawn to the use of an activated macrophage in the treatment of an established biofilm infection in a subject-in-need.
- the subject-in-need is at risk of developing a biofilm infection (i.e., the subject-in-need does not have an established biofilm infection), while in other embodiments, the subject-in-need comprises a biofilm infection (i.e., the subject-in-need does have an established biofilm infection).
- the subject-in-need is a human patient or a veterinary animal, such as a veterinary animal selected from the group consisting of a dog, a cat, a horse, a bovine (a bull or cow), a sheep, a goat and a pig.
- a veterinary animal selected from the group consisting of a dog, a cat, a horse, a bovine (a bull or cow), a sheep, a goat and a pig.
- the subject is the subject-in- need (i.e., the isolated cells are autologous cells).
- the dose or doses of macrophages, e.g., activated macrophages, for administration according to the methods and uses of the disclosure are determined on an individual basis using factors known in the art, e.g., general health, immunocompetency, age, weight, and the like.
- the disclosure contemplates dosages within the range of about 10 5 to 10 8 macrophages, e.g., 10 6
- Dosage scheduling is expected to range from about one to five doses of macrophages, with each dose separated from other doses by about one to five days. In some embodiments, multiple doses are administered to different sites around the expected or real periphery of a potential or existing biofilm infection.
- An exemplary dosage schedule for prophylactic methods and uses is about three daily doses of 10 5 to 108 macrophages, which is compatible with the average hospital stay for receiving a surgical implant, which places the patient at risk of a device-associated biofilm infection.
- the macrophage-activating agent is selected from the group consisting of a C5a receptor agonist, including but not limited to macrophage-activating peptides such as EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), C5a 65 - 7 4 (SEQ ID NO:4), and analogs and variants thereof, interleukin- ⁇ as well as combinations of interferon- ⁇ and tumor necrosis factor (TNF), and interferon- ⁇ and an inducer of TNF.
- An exemplary inducer of TNF is a TNF inducer selected from the group consisting of peptidoglycan and lipopolysacchande.
- An exemplary C5a receptor agonist is selected from the group consisting of EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2) and C5a 65 - 7 4 (SEQ ID NO:4).
- the macrophage- activating agent is a combination selected from the group consisting of interferon- ⁇ and tumor necrosis factor (TNF) or interferon- ⁇ and an inducer of TNF.
- TNF tumor necrosis factor
- An exemplary inducer of TNF is selected from the group consisting of peptidoglycan and lipopolysaccharide.
- Yet another stimulatory agent is the above-noted macrophage- activating peptide EP67, a C5a receptor/CD88 agonist that specifically induces pro-inflammatory properties of CD88 + macrophages without any effects on CD88 + neutrophils.
- a pro-inflammatory compound such as interleukin-6, interleukin-12, monocyte chemoattractant protein- 1 (MCP-1), monokine- induced by IFN- ⁇ (MIG), interferon-regulatory protein- 10 (IP- 10), macrophage inflammatory protein-2 (MIP-2), and keratinocyte chemoattractant (KC).
- M-CSF macrophage colony-stimulating factor
- GM-CSF granulocyte-macrophage colony- stimulating factor
- the subject- in-need undergoes surgery to receive a surgical implant, such as an artificial joint component, a cranial plate (natural or artificial), a dental implant, or a subcutaneous pacemaker.
- a surgical implant such as an artificial joint component, a cranial plate (natural or artificial), a dental implant, or a subcutaneous pacemaker.
- exemplary artificial joint components are an artificial hip component, an artificial knee component, an artificial ankle component, an artificial elbow component, an artificial shoulder component, and an artificial wrist component.
- the subject-in-need is scheduled to receive, or has received, a catheter.
- exemplary artificial joint components are an artificial hip component, an artificial knee component, an artificial ankle component, an artificial elbow component, an artificial shoulder component, and an artificial wrist component.
- embodiments of the method or use further comprise delivery of a proteolytic enzyme, such as MMP-1, MMP-2, MMP-7, MMP-9, MMP-12 or collagenase to the subject-in-need, for example to prevent biofilm infection.
- a proteolytic enzyme such as MMP-1, MMP-2, MMP-7, MMP-9, MMP-12, and collagenase, in the treatment of an existing biofilm infection in a subject-in-need.
- the proteolytic enzyme is expected to degrade the fibrotic capsule often associated with biofilms, enhancing the access of activated
- the proteolytic enzyme is packaged in a delivery vehicle such as a microsphere, microparticle, nanosphere, nanoparticle, liposome, micelle, or gel.
- delivery vehicles may be formulated to provide for the controlled, e.g., slow, release of their contents, as would be known in the art.
- the delivery vehicle is selected from the group consisting of a microsphere, a microparticle, a nanosphere, a nanoparticle, a liposome, a micelle and a gel, wherein the delivery vehicle comprises a macrophage and a macrophage- activating agent, or an activated macrophage, or a macrophage, a macrophage-activating agent and a proteolytic enzyme, or an activated macrophage and a proteolytic enzyme.
- Delivery vehicles suitable for the disclosed methods and uses include vehicles that are targeted to a biofilm, such as by exhibiting a binding partner to a marker displayed on the infectious organism, and vehicles that are targeted to the vicinity of a biofilm infection, for example by exhibiting a binding partner to a component of the fibrotic matrix frequently associated with biofilms.
- An exemplary proteolytic enzyme is collagenase, which may be packaged in any of the foregoing vehicles, such as a microsphere.
- the method and use further comprise the delivery of at least one molecule that interferes with the formation of the fibrotic capsule surrounding biofilm infections, such as slow-release delivery vehicles (any of the above-described vehicles) containing at least one proteolytic enzyme (e.g., collagenase or an MMP) and, optionally, at least one compound that interferes with the formation of a fibrotic capsule, such as a Lysyl Oxidase (i.e., LOX) inhibitor (e.g., BAPN).
- slow-release delivery vehicles any of the above-described vehicles
- at least one proteolytic enzyme e.g., collagenase or an MMP
- LOX Lysyl Oxidase
- BAPN Lysyl Oxidase
- the disclosure comprehends any delivery vehicle known in the art for use in packaging and delivering activated macrophages, or macrophages and at least one macrophage-activating agent, or activated macrophages and at least one proteolytic enzyme, or macrophages, at least one macrophage-activating agent and at least one proteolytic enzyme.
- Any of these formulations may further comprise a compound that interferes with the formation of the fibrotic capsule, such as BAPN or another Lysyl Oxidase (LOX) inhibitor.
- Embodiments of the use of activated macrophages further comprise using a prophylactically or therapeutically effective amount of an EP67 peptide (SEQ ID NO:l) in the treatment of a biofilm infection in a subject-in-need.
- an EP67 peptide SEQ ID NO:l
- the subject-in-need exhibits an inflammatory disorder.
- the subject-in-need exhibits a disorder or condition selected from the group consisting of atherosclerosis, infectious complications of cystic fibrosis, endocarditis, chronic sinusitis, nasal polyposis, chronic otitis media, leptospirosis, kidney stones, osteomyelitis, necrosis of the jaw, a urinary tract infection, a bladder infection, a chronic wound, an artificial component of a joint, a temporary catheter and a permanent catheter.
- a disorder or condition selected from the group consisting of atherosclerosis, infectious complications of cystic fibrosis, endocarditis, chronic sinusitis, nasal polyposis, chronic otitis media, leptospirosis, kidney stones, osteomyelitis, necrosis of the jaw, a urinary tract infection, a bladder infection, a chronic wound, an artificial component of a joint, a temporary catheter and a permanent catheter.
- Embodiments of the methods and uses for preventing or treating biofilm infections include methods and uses in which the subject-in-need exhibits an inflammatory disorder.
- An inflammatory disorder as used herein means any inflammatory disorder of humans or veterinary animals that is known in the art.
- Exemplary methods and uses comprise a subject- in-need that exhibits a disorder or condition selected from the group consisting of atherosclerosis, infectious complications of cystic fibrosis, endocarditis, chronic sinusitis, nasal polyposis, chronic otitis media, leptospirosis, kidney stones, osteomyelitis, necrosis of the jaw, a urinary tract infection, a bladder infection, a chronic wound, an artificial component of a joint, a temporary catheter and a permanent catheter.
- Embodiments of the methods and uses for preventing or treating biofilm infections further comprise administering a prophylactically or therapeutically effective amount of an antibiotic.
- the antibiotic is a member of a class of antibiotics selected from the group consisting of aminoglycosides, ansamycins, carbacephems, carbapenems, cephalosporins, glycopeptides, lincosamides, lipopeptides, macrolides, erythromycins, monobactams, nitrofurans, penicillins, penicillin combinations, antimicrobial polypeptides, quinolones, sulfonamides, tetracyclines and antimycobacterials.
- the antibiotic is selected from the group consisting of aminoglycosides, e.g., , amikacin (amikin), gentamicin (garamycin), kanamycin (kantrex), neomycin (neofradin), roxithromycin, netilmicin (netromycin), tobramycin (nebcin), paromomycin (humatin); ansamycins, e.g., geldamycin, herimycin; Carbacephems, e.g., loracarbef (lorabid); carbapenems, e.g., ertapenem (invanz), doripenem (doribax), imipenem or cilastatin (primaxin), meropenem (merrem); cephalosporins, e.g., cefadroxil (duricef), cefazolin (ancef), cefalotin or cefalothin (keflin
- macrolides e.g., azithromycin (zithromax, sumamed, xithrone), clarithromycin (biaxin, dynabac); erythromycin (erythocin, erythroped), erythromycin estolate, erythromycin ethyl succinate, erythromycin glucoheptonate, erythromycin lactobionate, erythromycin stearate, roximthromycin, trolandomycin (tao), telithromycin (ketek), spectinomycin (trobicin), spriamycin (rovamycine); monobactams, e.g., aztreonam (azactam); nitrofurans, e.g., furazolidone (furoxone), nitrofurantoi (macrodantin, macrobid); penicillins, e.g., amoxicillin (novamox, amoxil), ampicillin (principen), azlocillin, carben
- antimicrobial polypeptides e.g., bacitracin, colistin (colymycin S), polymyxin B; quinolones, e.g., ciprofloxacin (cipro, ciproxin, ciprobay), enoxacin (penetrex), gatifloxacin (tequin), levofloxacin (levaquin), lomefloxacin (maxaquin), moxifloxicin (avelox), ofloxacin (floxin, ocuflox), nalidixic acid (negram), norfloxacin (noroxin), ofloxicin (floxin, ocuflox), trovafloxacin (trovan), grepafloxacin (raxar), sparfloxacin (zagam), temafloxacin (omniflox); sulfonamides, e.g., mafenide (sulfamylon), sufonamidochry
- sulfamethizole thiosulfil forte
- sulfamethoxazole gantanol
- sulfanilamide sulfasalazine
- sulfisoxazole gantrisin
- trimethoprim-sulfamethoxazole bactrim, Septra
- Tetracyclines e.g., demeclocycline (declomycin), doxycycline (vibramycin), minocycline (minocin), oxytetracycline (terramycin), tetracycline (sumycin, achromycin V, steclin); and antimycobacterials, e.g., clofazimine (lamprene), dapsone (avlosulfon), capreomycin
- capastat cycloserine (seromycin), ethambutol (myambutol), ethionamide (trecator), isoniazid (INH), rifampicin or rifampin (rifadin, fimactane), rifabutin (mycobutin), rifapenine (priftin), streptomycin, arshenamine (salvarsan), chloramphenicol (Chloromycetin), fosfomycin (monurol), fusidic acid (fucidin), linezolid (zyvox), metronidazole (flagyl);
- mupirocin (bactroban), platensimycin, quinupristin/dalfopristin (synercid), rifaximin
- antibiotics include but not limited to their salts, acids, and bases.
- a composition comprising an isolated activated macrophage for the prevention or treatment of a biofilm.
- composition according to paragraph 1 further comprising a macrophage- activating peptide selected from the group consisting of EP67, EP54 and C5a 6 5-74.
- a kit comprising an isolated macrophage and a macrophage- activating agent.
- a kit comprising a macrophage-activating agent.
- the macrophage-activating agent is selected from the group consisting of a C5a receptor agonist, interleukin- ⁇ , interferon- ⁇ and tumor necrosis factor (TNF), and interferon- ⁇ and an inducer of TNF.
- kits according to paragraph 5 wherein the C5a receptor agonist is selected from the group consisting of EP67, EP54 and C5a 6 5-74.
- a method of providing biofilm therapy to a subject-in-need comprising:
- the macrophage- activating agent is selected from the group consisting of a C5a receptor agonist, interleukin- ⁇ , interferon- ⁇ and tumor necrosis factor (TNF), and interferon- ⁇ and an inducer of TNF.
- C5a receptor agonist is selected from the group consisting of EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2) and C5a 65 - 74 (SEQ ID NO:4).
- a pro -inflammatory compound selected from the group consisting of interleukin-6, interleukin-12, monocyte chemoattractant protein- 1 (MCP- 1), monokine-induced by IFN- ⁇ (MIG), interferon-regulatory protein- 10 (IP- 10), macrophage inflammatory protein-2 (MIP-2), keratinocyte chemoattractant (KC), macrophage colony- stimulating factor (M-CSF) and granulocyte-macrophage colony- stimulating factor (GM- CSF).
- MCP-1 monocyte chemoattractant protein- 1
- MIG monokine-induced by IFN- ⁇
- IP- 10 interferon-regulatory protein- 10
- MIP-2 macrophage inflammatory protein-2
- KC keratinocyte chemoattractant
- M-CSF macrophage colony- stimulating factor
- GM- CSF granulocyte-macrophage colony- stimulating factor
- the surgical implant is selected from the group consisting of an artificial joint component, a cranial plate (natural or artificial), a dental implant and a subcutaneous pacemaker.
- the artificial joint component is selected from the group consisting of an artificial hip component, an artificial knee component, an artificial ankle component, an artificial elbow component, an artificial shoulder component, and an artificial wrist component.
- proteolytic enzyme is selected from the group consisting of MMP-1, MMP-2, MMP-7, MMP-9, MMP-12 and collagenase.
- [0061] 28 The method according to paragraph 7, 8, or 27 further comprising a delivery vehicle selected from the group consisting of a microsphere, a microparticle, a nanosphere, a nanoparticle, a liposome, a micelle and a gel, wherein the delivery vehicle comprises a macrophage and a macrophage-activating agent, or an activated macrophage, or a macrophage, a macrophage-activating agent and a proteolytic enzyme, or an activated macrophage and a proteolytic enzyme.
- a delivery vehicle selected from the group consisting of a microsphere, a microparticle, a nanosphere, a nanoparticle, a liposome, a micelle and a gel, wherein the delivery vehicle comprises a macrophage and a macrophage-activating agent, or an activated macrophage, or a macrophage, a macrophage-activating agent and a proteolytic enzyme, or an activated macrophage and a proteolytic enzyme.
- the antibiotic is a member of a class of antibiotics selected from the group consisting of aminoglycosides, ansamycins, carbacephems, carbapenems, cephalosporins, glycopeptides, lincosamides, lipopeptides, macrolides, erythromycins, monobactams, nitrofurans, penicillins, penicillin combinations, antimicrobial polypeptides, quinolones, sulfonamides, tetracyclines and antimycobacterials.
- the antibiotic is a member of a class of antibiotics selected from the group consisting of aminoglycosides, ansamycins, carbacephems, carbapenems, cephalosporins, glycopeptides, lincosamides, lipopeptides, macrolides, erythromycins, monobactams, nitrofurans, penicillins, penicillin combinations, antimicrobial polypeptides, quinolones, sul
- the antibiotic is selected from the group consisting of amikacin, gentamicin, kanamycin, neomycin, roxithromycin, netilmicin, tobramycin, paromomycin, geldamycin, herimycin, loracarbef, ertapenem, doripenem, imipenem or cilastatin, meropenem, cefadroxil, cefazolin, cefalotin or cefalothin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefonicid, cefixime, cefdinir, cefditoren, cefotaxime, cefetamet, cefoperazone, cefpodoxime, ceftazidime, ceftibuten, ceftriaxone, cefepime, ceftaroline or fasamil, ceftobiprole
- spriamycin Aztreonam, furazolidone, nitrofurantoi, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin G, penicillin V, piperacillin, ticarcillin, amoxicillin and clavulanate, ampicillin and sulbactam, piperaccillin and tazobactam, ticarcillin and clavulanate; bacitracin, colistin, polymyxin B, ciprofloxacin, enoxacin, gatifloxacin, levofloxacin, lomefloxacin
- C57BL/6 mice were infected with 10 colony forming units (cfu) of methicillin-resistant S. aureus (MRSA) USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection.
- Animals were initially treated with 10 6 neutrophils or Ml -activated macrophages derived from C57BL/6 mice at 12, 24 and 48 hours post-infection. Mice were sacrificed at day 3 following MRSA infection, whereupon catheters (A) and tissues surrounding the catheters (B) were removed to quantitate bacterial burdens. Results are expressed as the number of cfu per ml for catheters or cfu per mg tissue, to correct for differences in tissue sampling size. Significant differences are denoted by asterisks (*, p ⁇
- Results are presented from individual animals combined from at least two independent experiments.
- FIG. 1 Long-term effect of activated macrophages on biofilm-causing bacteria. Growth of S. aureus on a catheter surface, measured on day 3 and again on day 14 as the log io of the number of S. aureus colony forming units (cfu) per ml. Activated macrophages were administered at 12, 24 and 48 hours post-infection and no additional cells were administered beyond 48 hours following infection. Control mice received injections of vehicle (PBS) alone. Results showed that activated macrophages were capable of long-term attenuation of S. aureus biofilm growth in vivo.
- PBS vehicle
- FIG. 3 Reduction of established S. aureus biofilm burden associated with catheters.
- S. aureus biofilms were established using a mouse s.c. catheter model with therapies initiated at day 7 following biofilm growth. A robust biofilm is formed within this time interval.
- Mice received either vehicle (PBS) or 10 6 activated macrophages ( ⁇ ) within the catheter lumen at days 7 and 9 post-infection, whereas antibiotic-treated mice received
- PBS vehicle
- ⁇ activated macrophages
- Ml macrophage polarization increases S. aureus biofilm phagocytosis and killing.
- Biofilms without the addition of immune cells were used as untreated controls. Arrows indicate phagocytic cells and significant differences are denoted by asterisks (**, p ⁇ 0.01; ***, /> ⁇ 0.001).
- FIG. 5 S. aureus biofilms decrease macrophage cytokine secretion. Bone marrow-derived macrophages were incubated with 6-day-old biofilms or planktonic bacteria for 2 hours in glass chamber slides, whereupon viable macrophages were purified by FACS and then incubated at 37°C for 24 hours without any further stimulation after which supernatants were collected to quantitate TNF-a, IL-10, and IL-1RA by ELISA. Significant differences in secreted cytokine levels between biofilm and planktonic exposed macrophages are denoted by asterisks (*, p ⁇ 0.05; **, p ⁇ 0.01; ⁇ 0.001).
- FIG. 6 Activated macrophages but not neutrophils impair MRSA biofilm formation in vivo.
- C57BL/6 mice were infected with 10 3 colony forming units (cfu) of methicillin-resistant S. aureus (MRSA) USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection.
- Animals were treated with 10 6 neutrophils, non- activated macrophage, or Ml-activated macrophages derived from C57BL/6 mice at 12, 24 and 48 hours post-infection. Mice were sacrificed at day 3 following MRSA infection, whereupon catheters (A) and tissues surrounding the catheters (B) were removed to quantitate bacterial burdens.
- MRSA methicillin-resistant S. aureus
- Results are expressed as the number of cfu per ml for catheters or cfu per mg tissue, to correct for differences in tissue sampling size. Significant differences are denoted by asterisks (*, p ⁇ 0.05). Results are presented from individual animals combined from at least two independent experiments.
- FIG. 7 The ability of Ml-polarized macrophages to impair MRSA biofilm development is mediated by MyD88-dependent signals.
- C57BL/6 mice were infected with 10 colony forming units (cfu) of methicillin-resistant S. aureus (MRSA) USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection.
- Animals were initially treated with 10 6 activated macrophages derived from C57BL/6 or MyD88 knockout (KO) mice at 12, 24 and 48 hours post-infection. Mice were sacrificed at day 3 following MRSA infection, whereupon catheters (A) and tissue surrounding the catheters (B) were removed to quantitate bacterial burdens.
- MRSA methicillin-resistant S. aureus
- Results are expressed as the number of cfu per ml for catheters or cfu per mg tissue, to correct for differences in tissue sampling size. Results are presented from individual animals combined from at least two independent experiments. Significant differences are denoted by asterisks (*, p ⁇ 0.05).
- Figure 8 Activated macrophage therapy alters the local inflammatory milieu in MRSA biofilm associated tissue. Animals were sacrificed at day 3 post-infection
- FIG. 9 Ml-activated macrophages attenuate MRSA biofilm catheter persistence in an established infection.
- C57BL/6 mice were infected with 10 3 colony forming units (cfu) of methicillin-resistant S. aureus (MRSA) USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection.
- animals received vehicle control (PBS), 10 6 activated macrophages, or antibiotic combination (rifampicin 25 ⁇ g/ml + daptomycin 5 ⁇ g/ml).
- Catheters (A) and surrounding host tissues (B) were removed on day 10 to quantitate bacterial burdens. Significant differences between groups are denoted by asterisks (*, p ⁇ 0.05).
- FIG. 12 The macrophage-activating peptide EP67 attenuates S. aureus biofilm growth in vivo.
- EP67- or vehicle-treated mice were infected with 10 3 CFU of MRSA USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection. Animals were sacrificed at days 3 (A & B) or 14 (C & D) following S. aureus infection, whereupon catheters (A and C) or tissues surrounding the catheters (B and D) were removed to quantitate bacterial burdens. Data are expressed as the number of CFU per ml, or CFU per mg host tissue, for normalization. Results are presented from individual animals pooled from at least two independent experiments with bars representing the mean of each group. Significant differences in bacterial burdens between vehicle and EP67-treated mice are denoted by asterisks (*, p ⁇ 0.05).
- EP67 augments inflammatory mediator expression in tissues surrounding MRSA biofilm infections.
- Tissues surrounding MRSA biofilms from EP67- or vehicle-treated mice were homogenized at day 3 post-infection to quantitate the effects of EP67 treatment on IL-12p40 (A), CCL5 (B), IL-17 (C), IL-la (D) and IFN- ⁇ (E) expression by MILLIPLEX analysis.
- Results were normalized to the amount of total protein recovered to correct for differences in tissue sampling size. Results are presented from individual animals combined from two independent experiments. Significant differences between EP67- vs. vehicle-treated catheter-associated tissues are denoted by asterisks (*, p ⁇ 0.05).
- the disclosure provides compositions and methods that are useful in preventing the formation of biofilm infections and in treating existing biofilm infections.
- methods of administering cells such as macrophages, including previously activated macrophages, macrophages being activated simultaneously with administration, or macrophages activated in vivo post-administration, to prevent and/or treat biofilms.
- peripheral blood monocytes are collected from a patient prior to surgery, such as one week prior to surgery.
- Macrophages are prepared from this sample and are activated using various activating agents, including but not limited to, peptidoglycan ⁇ e.g., S. aureus- derived peptidoglycan) and interferon- ⁇ .
- activated macrophages are then administered to the patient at the site of implantation to prevent biofilm formation.
- activated macrophages are administered to a patient who already has an established biofilm to treat the biofilm infection. It is envisioned that one or multiple doses of activated
- macrophages could be administered to treat an established biofilm.
- evidence documenting that activated macrophages reduce bacterial burdens on infected devices supports embodiments where activated macrophages are used in combination with antibiotics.
- Another aspect of treating biofilms is interfering with the extensive fibrotic matrices that form, which represents a barrier to effective treatment. To overcome this barrier, it is envisioned that proteolytic enzymes or drugs are co-administered with activated macrophages.
- one or more compounds or factors that are capable of activating macrophages in vivo is/are co-administered or administered pre- or post- administration of activated macrophages to further enhance macrophage activation.
- Biofilm or "biofilm infection” refers to an organized community of bacteria contained within a matrix or “slime,” typically composed of bacterial polysaccharides, DNA, and proteins, within which is found at least one species of infectious microbe (e.g. , bacterium) and which typically exhibits a complex heterogeneous composition of microbes.
- Biofilms may exist within the body on natural surfaces (e.g. , the lining of bones and the heart) or inanimate objects inserted for therapeutic treatment (e.g., indwelling devices or artificial joints).
- a "subject” is a human being or a veterinary animal, including but not limited to, a dog, a cat, a horse, a bovine (a cow or a bull), a sheep, a goat and a pig, as well as a zoo animal and a game animal.
- Subject-in-need means a subject, such as a human patient or veterinary animal including but not limited to a dog, a cat, a horse, a bovine (a cow or a bull), a sheep, a goat and a pig, that is in need of prevention for a possible biofilm infection or is in need of treatment for an existing biofilm infection.
- Isolating in the context of isolating a cell, is given its ordinary and accustomed meaning in the art of separating that cell from at least one other component with which the cell is normally associated in the body of a subject.
- Peripheral blood monocyte is given its ordinary and accustomed meaning in the art of a white blood cell in peripheral blood that has a single nucleus and can take in (ingest or phagocytose) foreign material.
- a monocyte like a macrophage, is a phagocyte. Under the proper conditions, a monocyte can differentiate into a macrophage.
- “Culturing” is given its ordinary and accustomed meaning of incubating a cell in the presence of nutrients and under conditions permissive for continued viability and/or growth.
- “Contacting” means removing any barrier to a coming together, a locating in the immediate vicinity of, or a touching, such as between a cell and a compound(s) such as an activating agent.
- a "macrophage-activating agent” is one or a combination of compounds that induce an activated state or phenotype in a macrophage.
- exemplary macrophage-activating agents include macrophage-activating peptides such as EP67 (SEQ ID NO: l), EP54 (SEQ ID NO:2), C5a 6 5-74 (SEQ ID NO:4), and analogs and variations thereof, interleukin- ⁇ as well as combinations of compounds selected from the group consisting of interferon- ⁇ and tumor necrosis factor (TNF), and interferon- ⁇ and an inducer of TNF.
- a "macrophage” is a phagocyte having a single nucleus that ingests or
- Macrophages are derived from monocytes by differentiation.
- an "activated macrophage” is a macrophage that exhibits an activated phenotype relative to a resting macrophage.
- the activated macrophage becomes larger, more motile, adherent, expresses more MHCII proteins on its surface, contains more lysosomes and lysosomal enzymes, and secretes a variety of substances including interleukin-1 and tumor necrosis factor, relative to a resting macrophage.
- an activated macrophage has increased phagocytic activity and increased bacterial killing via reactive oxygen intermediates and lysosomal enzymes.
- an activated macrophage as used herein is an "Ml -activated macrophage" or a "classically activated macrophage.”
- a “prophylactically effective amount” is an amount effective in prevention of a disease, disorder or condition and is determined on an individual basis with consideration given to well-known factors such as weight, gender, general health, and the like.
- a “therapeutically effective amount” is an amount effective in treatment of a disease, disorder or condition and is determined on an individual basis with consideration given to well-known factors such as weight, gender, general health, and the like.
- At risk of developing a biofilm infection means a subject exhibiting a greater risk of developing a biofilm infection than the average subject, such as a subject with a compromised immune system, a subject with a documented history of recurrent bacterial infections, or a subject harboring a surgical implant such as an artificial component of a joint (e.g., hip, knee, ankle, shoulder, elbow, wrist) or an implanted pacemaker.
- a surgical implant such as an artificial component of a joint (e.g., hip, knee, ankle, shoulder, elbow, wrist) or an implanted pacemaker.
- a "veterinary animal” means any animal conventionally receiving treatment from a veterinarian and having an immune system, including but not limited to a dog, a cat, a horse, a bovine (a cow or a bull), a sheep, a goat a pig, a zoo animal, and a game animal.
- An "immunomodulatory cytokine” is a cytokine exhibiting the capacity to modulate an immune response under at least one set of conditions.
- exemplary cytokines are proinflammatory cytokines, such as interleukin-6, interleukin-12, monocyte chemoattractant protein- 1 (MCP-1), monokine-induced by IFN- ⁇ (MIG), interferon-regulatory protein- 10 (IP- 10), macrophage inflammatory protein-2 (MIP-2), and keratinocyte chemoattractant (KC).
- M-CSF macrophage colony-stimulating factor
- GM-CSF granulocyte-macrophage colony-stimulating factor
- a "surgical implant” is an artificial implant or device placed within the body of a subject and includes, but is not limited to, an artificial joint component (an artificial hip component, an artificial knee component, an artificial ankle component, an artificial elbow component, an artificial shoulder component, and an artificial wrist component), a cranial plate (natural or artificial ), a dental implant, and a subcutaneous pacemaker.
- Catheter is given its ordinary and accustomed meaning of a hollow, typically flexible, tube for insertion into a body cavity, duct, or vessel to allow the passage of fluids or to distend a passageway.
- a "proteolytic enzyme” is an enzyme that catalyzes the proteolysis of at least one protein, polypeptide or peptide, and includes, but is not limited to, MMP-1, MMP-2, MMP-7, MMP-9, MMP-12 and collagenase.
- An "EP67 peptide” is a peptide functioning as a C5a receptor agonist and exhibits the amino acid sequence set forth in SEQ ID NO: 1, or a variant or derivative of that sequence.
- An "EP54 peptide” is a peptide functioning as a C5a receptor agonist and exhibits the amino acid sequence set forth in SEQ ID NO: 2, or a variant or derivative of that sequence.
- An "C5a65_74” is a peptide functioning as a C5a receptor agonist and exhibits the amino acid sequence set forth in SEQ ID NO:4, or a variant or derivative of that sequence.
- Inflammatory disorder is a disorder characterized by an inflammatory response causing redness, swelling, heat and/or pain. Inflammatory disorders include, but are not limited to, atherosclerosis, infectious complications of cystic fibrosis, endocarditis, chronic sinusitis, nasal polyposis, chronic otitis media, leptospirosis, kidney stones, osteomyelitis, necrosis of the jaw, a urinary tract infection, a bladder infection, and a chronic wound.
- compositions and methods for preventing and treating biofilm infection comprise administering a prophylactically effective amount of an immune cell, such as an activated macrophage.
- the method involves administering a therapeutically effective amount of autologous activated macrophages.
- a blood sample is obtained from a patient prior to implant placement, e.g., approximately one week prior to the medical device placement procedure.
- the blood is sent to a laboratory where immune stimulation would take place, which would allow for quality control and
- cells Once cells are propagated, they are delivered to the recipient for local treatment at the implant site, making the cells immediately available to respond to any potential bacterial contaminant introduced during the surgical procedure (most commonly via skin microflora).
- Macrophage-based immune-cell therapy is not only effective in treating/controlling microbe, e.g., bacterial, burdens associated with biofilms, it has the added advantage of doing so by utilizing the host's own innate immune cells.
- the methods of the disclosure eliminate or minimize selection pressures imposed directly on the microbe, e.g., bacteria, and decrease the likelihood of developing resistant strains.
- minimal side effects would be expected because autologous cells that won't survive long-term (i.e., no longer than 7-14 days) are being introduced.
- Biofilms have been shown to skew the immune response to favor anti-inflammatory and pro-fibrotic pathways, which likely contributes to biofilm persistence.
- antimicrobial activity has been augmented through the local administration of classically activated Ml macrophages, optionally supplemented by administration of a macrophage-activating peptide such as EP67, EP54, C5a 6 5_74, or their analogs or variants, provided such peptides retain the capacity to invoke macrophage pro-inflammatory responses.
- a macrophage-activating peptide such as EP67, EP54, C5a 6 5_74, or their analogs or variants, provided such peptides retain the capacity to invoke macrophage pro-inflammatory responses.
- Ml- activated macrophages limited biofilm formation and treatment of established biofilm infections with activated macrophages also significantly reduced catheter-associated biofilm burdens compared to antibiotic treatment.
- pro-inflammatory mediators were significantly elevated in tissues surrounding biofilm infections following Ml -activated macrophage treatment, which may account
- Ml-activated macrophage therapy provides a new strategy for preventing and treating biofilms, optionally supplemented with macrophage-activating agents such as macrophage- activating peptides like EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), or C5a 65 - 7 4 (SEQ ID NO:4), or their analogs or variants.
- macrophage-activating agents such as macrophage- activating peptides like EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), or C5a 65 - 7 4 (SEQ ID NO:4), or their analogs or variants.
- the activated macrophage therapies disclosed herein are suitable for combination with antibiotics, particularly for surgical-implant patients who are unable or unwilling to undergo additional surgery to manage the infection and thereby maintain the implanted device.
- antibiotics particularly for surgical-implant patients who are unable or unwilling to undergo additional surgery to manage the infection and thereby maintain the implanted device.
- the significance of this approach is even more pronounced against the backdrop of the rapidly increasing elderly population, which grows progressively less immunoresponsive and will be the primary recipients of more permanent artificial implants, such as hips and knees.
- S. aureus e.g., MRS A
- MRS A MRS A
- MRS A MRS A
- the therapeutic potential of activated macrophages is supported by results demonstrating that early treatment with proinflammatory Ml-activated macrophages significantly limited S. aureus biofilm growth in vivo and provided long-term protection from biofilm colonization.
- activated macrophages were also effective at reducing S. aureus burdens in established biofilms by 100-fold. Without wishing to be bound by theory, this reduction in device- associated bacterial burdens may result from dispersal of organisms from the biofilm which, in turn, would restore the metabolic activity of the bacteria.
- Ml- activated macrophages will display synergy with antibiotics to maximize biofilm clearance, because these organisms are no longer foreign body-associated and, thus, more amenable to antibiotic action.
- Monocytes can be easily collected from patient's blood by elutriation, expanded, induced to differentiate into mature macrophages, and activated in vitro for introduction into the surgical site for infection prophylaxis.
- a relatively wide range of activated macrophage dosages are suitable for use in the prophylactic and therapeutic methods and uses according to the disclosure.
- Exemplary macrophage dosages are 10 5 , 10 6 , 10 7 , and 10 8 activated macrophages per dose.
- the methods and uses of the disclosure are amenable to a variety of dosing schedules, which can readily be determined by those of skill in the art.
- the activated macrophage can be delivered in one to five doses, over one to five days.
- administered doses may be placed in a region approximately surrounding the expected or actual location of a potential or existing biofilm.
- the disclosure provided herein establishes a previously unappreciated role for Ml- activated macrophages in biofilm containment and bacterial clearance.
- biofilms such as MRSA biofilms have the capacity to thwart this response by skewing macrophages away from a pro-inflammatory Ml to an anti-inflammatory M2 phenotype, which could ensure biofilm persistence in an immunocompetent host.
- activated macrophage transfer therapy would allow macrophages to be "on site" to deal with possible contamination of the implant from normal bacterial flora on the skin.
- compositions and methods of the disclosure are contemplated as suitable for preventing or treating biofilm infections caused by, or associated with, any known infectious organism, such as microbes, including gram-positive and gram-negative bacteria, fungi, and yeast.
- infectious microbes causing or associated with biofilm infections include gram- positive bacteria such as Staphylococcus aureus, Enterococcus faecalis and Enterococcus faecium and gram-negative bacteria such as Escherichia coli, Pseudomonas aeruginosa, Pseudomonas fluorescens, and Haemophilus influenza,.
- exemplary microbes causing or associated with biofilm infections include Leptospira species such as Leptospira interrogans, and the yeast, Candida albicans. These exemplary organisms are representative of the hundreds and hundreds of microbes capable of causing or being associated with biofilm infections.
- dental plaque is known to involve a biofilm infection associated with the teeth of humans and veterinary animals. Investigations have shown that biofilms associated with dental plaque contain over 500 species of microorganisms. The disclosure contemplates the prevention or treatment of biofilm infections caused by or associated with each of these organisms, and other microbes capable of causing or being associated with biofilm infection.
- biofilm- associated microbes of dental plaque are provided to illustrate the number and diversity of microbes associated with biofilm infections amenable to prevention or treatment with the disclosed compositions and methods, even if the administration of cells such as activated macrophages to prevent or treat biofilm infections associated with dental plaque is not contemplated as a preferred embodiment of the disclosed subject matter.
- biofilm infection A variety of diseases, disorders and conditions are known to be associated with biofilm infection, and the compositions and methods disclosed herein are contemplated as suitable for preventing or treating any of these biofilm infections.
- exemplary diseases associated with biofilm infection include, but are not limited to, atherosclerosis, infectious complications of cystic fibrosis, endocarditis, chronic sinusitis, nasal polyposis, chronic otitis media or middle ear infections, leptospirosis, kidney stones, osteomyelitis, necrosis of the jaw, urinary tract infections, and bladder infections.
- Exemplary disorders include chronic wounds, and exemplary conditions include subjects, e.g., human patients, that have or are receiving a prosthetic device such as an artificial component of a joint (e.g., a component of a hip, knee, ankle, elbow, shoulder, or wrist joint), or a temporary or permanent catheter (e.g., urinary, intravenous or intraarterial catheter).
- a prosthetic device such as an artificial component of a joint (e.g., a component of a hip, knee, ankle, elbow, shoulder, or wrist joint), or a temporary or permanent catheter (e.g., urinary, intravenous or intraarterial catheter).
- a prosthetic device such as an artificial component of a joint (e.g., a component of a hip, knee, ankle, elbow, shoulder, or wrist joint), or a temporary or permanent catheter (e.g., urinary, intravenous or intraarterial catheter).
- typically non-pathogenic microbes e.g., benign members of the normal skin
- infectious diseases of veterinary animals are associated with biofilm infection and are comprehended as suitable for prevention or treatment using the disclosed compositions and methods.
- the disclosed compositions and methods are contemplated as suitable for preventing or treating any known biofilm infection.
- Activating agents suitable for use in activating a macrophage include any compound, or combination of compounds, known to activate macrophages.
- compounds effective in activating macrophages only in combination with at least one other compound are referred to herein as activating agents.
- Exemplary activating agents include macrophage- activating peptides such as EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), C5a 6 5-74 (SEQ ID NO:4) and their analogs and variants, interleukin- ⁇ as well as
- TNF tumor necrosis factor
- exemplary inducers of TNF are peptidoglycan and
- compositions further comprise delivering to the patient in need an effective amount of a pro -inflammatory compound, such as interleukin-6, interleukin-12, monocyte chemoattractant protein-1 (MCP-1), monokine-induced by IFN- ⁇ (MIG), interferon-regulatory protein-10 (IP-10), macrophage inflammatory protein-2 (MIP- 2), and keratinocyte chemoattractant (KC).
- a pro -inflammatory compound such as interleukin-6, interleukin-12, monocyte chemoattractant protein-1 (MCP-1), monokine-induced by IFN- ⁇ (MIG), interferon-regulatory protein-10 (IP-10), macrophage inflammatory protein-2 (MIP- 2), and keratinocyte chemoattractant (KC).
- macrophage colony- stimulating factor M-CSF
- GM-CSF granulocyte-macrophage colony-stimulating factor
- a cell e.g., a macrophage, is exposed to one or more activating agents ex vivo or in vivo, and prior to, concurrent with, or following administration of the cell to the subject, such as a human patient or veterinary animal.
- a cell to be administered to a subject is an activated macrophage or a macrophage.
- a subject such as a human patient or veterinary animal
- an activated macrophage or a macrophage results in a
- a syngeneic cell is utilized, such as an autologous cell.
- Autologous cells are administered in order to minimize complicating influences from the host immune system and cell rejection.
- Example 1 provides the materials and methods used in the experiments disclosed herein.
- Example 2 established that activated macrophages are bactericidal towards biofilm-associated S. aureus in vitro.
- Example 3 showed that biofilm infections alter the gene expression profile of administered macrophage.
- Example 4 demonstrated that activated macrophages prevented the formation of a biofilm infection (i.e., MRS A infection) in vivo.
- Example 5 established that activated macrophages reduced the bacterial burden of biofilm infections, while establishing a pro-inflammatory microenvironment at the biofilm site.
- Example 6 showed that administering a C5aR agonist, e.g., EP67, successfully prevented or inhibited formation of a biofilm infection, i.e., a MRS A biofilm infection. Without wishing to be bound by theory, this effect is believed to be due to the C5aR agonist reprogramming endogenous macrophages from an M2- to a pro-inflammatory Ml-activation state, which would complement or enhance activated-macrophage therapy.
- Example 7 established that activated macrophages eliminated or reduced the bacterial burden of a catheter-associated biofilm infection in vitro (i.e., MRS A infection).
- Antibiotics e.g., EP67
- compositions, methods and uses according to the disclosure are generally drawn to macrophages that are or can be activated for use in preventing or treating biofilm infections.
- the macrophages can be combined with macrophage- activating agents, including macrophage-activating peptides such as EP67, EP54 and C5a 6 5_74.
- the macrophages can be combined, alone or with a macrophage-activating agent, with one or more antibiotics.
- the macrophages, with or without a macrophage- activating agent can be administered separately or jointly with one or more antibiotics in the prophylactic or therapeutic methods and uses according to the disclosure. Any known antibiotic is expected to be suitable for these methods and uses.
- Suitable classes of antibiotics include , but are not limited to, aminoglycosides, ansamycins, carbacephems, carbapenems, cephalosporins, glycopeptides, lincosamides, lipopeptides, macrolides, erythromycins, monobactams, nitrofurans, penicillins, penicillin combinations, antimicrobial polypeptides, quinolones, sulfonamides, tetracyclines and antimycobacterials.
- the antibiotic is selected from the group consisting of amikacin, gentamicin, kanamycin, neomycin, roxithromycin, netilmicin, tobramycin, paromomycin, geldamycin, herimycin, loracarbef, ertapenem, doripenem, imipenem or cilastatin, meropenem, cefadroxil, cefazolin, cefalotin or cefalothin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefonicid, cefixime, cefdinir, cefditoren, cefotaxime, cefetamet, cefoperazone, cefpodoxime, ceftazidime, ceftibuten, ceftriaxone, cefepime, ceftaroline or fasamil, ceftobipro
- spriamycin Aztreonam, furazolidone, nitrofurantoi, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin G, penicillin V, piperacillin, ticarcillin, amoxicillin and clavulanate, ampicillin and sulbactam, piperaccillin and tazobactam, ticarcillin and clavulanate, ampicillin and sulbactam, piperaccillin and tazobactam, ticarcillin and clavulanate, ampicillin and sulbactam, piperaccillin and tazobactam, ticarcillin and clavulanate, ampicillin and sulbactam, piperaccillin and tazobactam, ticarcillin and clav
- sufonamidochrysoidine sulfacetamide, sufadiazine, silver sulfadiazine, sulfamethizole, sulfamethoxazole, sulfanilamide, sulfasalazine, sulfisoxazole, trimethoprim- sulfamethoxazole, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol, ethionamide, isoniazid, rifampicin or rifampin, rifabutin, rifapenine, streptomycin, arshenamine, chloramphenicol, fosfomycin, fusidic acid, linezolid, metronidazole, mupirocin, platensimycin, qui
- mice Male C57BL/6 mice (6-8 weeks old) were obtained from Charles River Laboratories (Frederick, MD) and MyD88 KO animals were purchased from the Centre de La Recherche Scientifique and have been backcrossed with C57BL/6 mice for over 10 generations (36-38). Mice were housed in restricted-access rooms equipped with ventilated microisolator cages and maintained at 21 °C under a 12-hour light: 12-hour dark cycle with ad libitum access to water (HydropacTM; Lab Products, Seaford, DE) and Teklad rodent chow (Harlan, Indianapolis, IN).
- S. aureus strains For in vitro biofilms, the S. aureus MRSA strain USA300 LAC was used and, for confocal microscopy studies, was transformed with the plasmid pCMl l to express GFP driven by the sarA PI promoter (USA300 LAC-GFP) and plasmid expression was maintained with erm selection (10 ⁇ g/ml) (15, 39).
- the bioluminescent MRSA USA300 LAC::lux strain was used as previously described (15, 40, 41).
- USA300 LAC or USA300LAC-GFP were prepared in macrophage medium, supplemented with 10 mg/ml erm for the GFP strain. The following day, plasma coating buffer was removed and chambers were inoculated with bacteria, diluted in mammalian cell culture medium (RPMI-1640 supplemented with 10% fetal bovine serum) to an OD600 of 0.050 in 2 ml, and incubated at 37°C under static aerobic conditions for a period of up to 6 days.
- mammalian cell culture medium RPMI-1640 supplemented with 10% fetal bovine serum
- BMDM and neutrophils were labeled with either 5 ⁇ CellTracker Orange (CTO) or CellTracker Blue (CTB) (both from Molecular Probes, San Diego, CA) depending on the experimental setup.
- CTO CellTracker Orange
- CTB CellTracker Blue
- aureus-dedved peptidoglycan i.e., PGN
- PGN aureus-dedved peptidoglycan
- a Zeiss 510 META laser scanning microscope Carl Zeiss, Oberkochen, Germany
- Neutrophil or macrophage-biofilm co- cultures were harvested at 24 hours after immune cell addition by mechanical dissociation followed by sonication, whereupon bacterial enumeration was performed by serial dilution on TSA plates supplemented with 5% sheep blood (Hemostat Laboratories, Dixon, CA).
- 200,000 cells/well were plated in a 96-well polystyrene microtiter plate (BD Biosciences) and incubated for 24 hours at 37°C with 5% C0 2 . After 24 hours, supernatants were collected to compare the production of inflammatory mediators associated with biofilm- versus planktonic-exposed macrophages utilizing sandwich ELISA kits to quantitate TNF-a, IL-10 (OptiEIA; BD Biosciences, Franklin Lakes, NJ), and IL-1RA (DuoSet; R & D Systems, Minneapolis, MN) per the manufacturers' protocols.
- sandwich ELISA kits to quantitate TNF-a, IL-10 (OptiEIA; BD Biosciences, Franklin Lakes, NJ), and IL-1RA (DuoSet; R & D Systems, Minneapolis, MN) per the manufacturers' protocols.
- mice were infected with 10 colony forming units (cfu) of MRSA USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection and on days 7 and 9 post-infection, animals received vehicle control (PBS), 10 6 activated macrophages, or antibiotic treatment consisting of rifampicin (25 ⁇ g/ml) and daptomycin (5 ⁇ g/ml).
- PBS vehicle control
- rifampicin 25 ⁇ g/ml
- daptomycin 5 ⁇ g/ml
- mice received vehicle or 10 activated macrophages loaded with Quantum dots (Molecular Probe) at 24 hours post-infection for the prophylactic paradigm or at 7 days post-infection for established biofilm paradigms.
- Quantum dots Molecular Probe
- the longevity of the administered macrophages was monitored daily in the same cohort of mice using an In vivo Imaging System (IVIS Spectrum; Caliper Life Sciences, Hopkinton, MA) under isoflurane anesthesia until 5 days after administration, at which point animals were sacrificed to determine absolute bacterial burdens associated with catheters and surrounding tissues.
- IVIS Spectrum Caliper Life Sciences, Hopkinton, MA
- antibiotic treatment consisting of rifampicin (25 ⁇ g/ml) and daptomycin (5 ⁇ g/ml) given twice daily starting 12 hours post-infection and continuing until 48 hours post-infection, was used to compare experimental treatments with the standard of care typically administered in the clinic, which does not exhibit any beneficial effects on biofilm clearance.
- EP67 Peptide-Activated Macrophage Combined Treatment To demonstrate that macrophage-activating agents, such as macrophage-activating peptides, were suitable for combination with activated macrophage therapy in preventing or treating biofilm infection, the effect of administering a macrophage-activating peptide alone was assessed. Animals were initially treated locally at the site of S. aureus infection with EP67 or a biologically inactive scrambled derivative (sEP67) having the sequence set forth in SEQ ID NO:3. 200 ⁇ g of either peptide was injected into the catheter at the time of infection (time 0) followed by 800 ⁇ g peptide divided into four different injection sites surrounding the catheter 24 and 48 hours post-infection.
- time 0 time of infection
- 800 ⁇ g peptide divided into four different injection sites surrounding the catheter 24 and 48 hours post-infection.
- MILLIPLEX multi-analyte bead array.
- MILLIPLEX Millipore, Billerica, MA
- inflammatory mediators IL-la, IL- ⁇ , TNF-a, IFN- ⁇ , IL-6, IL-9, IL-10, IL-12p40, IL-12p70, IL-15, IL-17, CXCL1, CXCL2, CXCL9, CXCL10, CCL2, CCL3, CCL4, and CCL5. Results were analyzed using a Bio-Plex workstation (Bio-Rad, Hercules, CA) and normalized to the amount of total protein recovered to correct for differences in tissue sampling size. [0131] Flow cytometry. Tissues surrounding biofilm-infected catheters were collected and processed for flow cytometry as previously described (15). Cells were stained with the following antibodies: F4/80-PE-Cy7 and Ly6G-AF700 (all from BD Biosciences).
- Macrophages were identified as F4/80 + and Ly6G " , while neutrophils were identified as F4/80 " and Ly6G + .
- Activated macrophages exhibit S. aureus biofllm bactericidal activity in vitro.
- MRSA biofilms are capable of attenuating traditional pro-inflammatory responses, explaining, in part, why these infections persist in an immunocompetent host (15).
- macrophages were stimulated with S. aureus-derived peptidoglycan (PGN) and interferon- gamma (IFN- ⁇ ) for 6 hours prior to their addition to MRSA biofilms or planktonic cultures as a control.
- PPN S. aureus-derived peptidoglycan
- IFN- ⁇ interferon- gamma
- activated macrophages were capable of phagocytosing biofilm- associated bacteria, which resulted in significant reductions in bacterial burdens following a 24 hour co-culture period (Fig. 4B).
- neutrophils were isolated from murine bone marrow and co-cultured with biofilms. Unlike macrophages, neutrophils were not pre-activated prior to biofilm addition, because this would lead to rapid degranulation and reduced cell viability. Interestingly, neutrophils were able to phagocytose MRSA biofilms, yet this did not translate into reduced bacterial numbers (Fig. 4B), revealing disconnect between the two processes. This may result from additional virulence determinants released by S. aureus during biofilm growth, because the organism is known to produce numerous factors that interfere with neutrophil function/survival (20-22).
- biofilm formation did afford some protection against phagocytic uptake compared to planktonic growth conditions, because both macrophages and neutrophils actively phagocytosed planktonic S. aureus but were less capable of internalizing biofilm-associated bacteria (Fig. 4A).
- Biofilms alter macrophage activation profiles. S. aureus biofilms skew
- macrophage gene expression towards an alternatively activated (M2) phenotype compared to planktonic bacteria 15, 23.
- M2 alternatively activated phenotype compared to planktonic bacteria
- macrophages were exposed to S. aureus biofilms or planktonic bacteria for 2 hours, separated from bacteria by FACS and incubated at 37°C for 24 hours, at which point supernatants were collected to determine cytokine secretion profiles.
- Macrophages co- cultured with biofilms demonstrated a decreased cytokine secretion profile with reductions in TNF-a, IL-10 and IL-1RA expression compared to macrophages exposed to planktonic bacteria (Fig. 5).
- Example 4 Introduction of activated macrophages prevents MRSA biofilm formation in vivo.
- MRSA biofilms attenuated the expression of numerous pro-inflammatory mediators compared to a sterile foreign body (15, 23).
- the introduction of Ml-activated macrophages led to increases in CXCL9, CCL5, and IFN- ⁇ expression within biofilm-infected tissues (Fig. 9), revealing successful re-direction of the macrophages towards a pro-inflammatory milieu.
- the pro-inflammatory activity of activated macrophages is expected to play an important role in limiting biofilm growth.
- Ml -activated macrophage administration attenuated arginase-1 expression within the surrounding tissue, indicating that macrophages were able to modify the local tissue environment to favor a pro-inflammatory response (Fig. 11).
- the introduction of Ml -activated macrophages into established biofilms augmented CXCL9, CXCL2, IL-17, and IL-6 expression within established biofilm-infected tissues (Fig. 9D-G), again revealing the successful re-direction of macrophages towards a pro-inflammatory milieu.
- Ml-activated macrophages were labeled with near-infrared Quantum Dots (QDots) and injected at either the time of infection or on day 7 in the early- and established-biofilm models, respectively.
- QDots near-infrared Quantum Dots
- Animals were subjected to IVIS imaging immediately following macrophage transfer to confirm macrophage signals immediately following injection, and subsequently reimaged every 24 hours (Fig. 12A).
- Qdot-labeled Ml macrophages were still visible at 5 days post-treatment, at which point animals were sacrificed in order to recover tissues for FACs analysis. Data indicated a slight increase in F4/80 + /IRF5 + macrophages in Qdot-labeled Ml macrophage-treated animals compared to vehicle-treated animals (Fig. 12B).
- EP67 impacts MRSA biofilm establishment in vivo and stimulates local proinflammatory responses.
- a supplementary approach to the introduction of exogenous Ml- activated macrophages is the combined administration of Ml-activated macrophages and a macrophage- activating compound, such as a macrophage- activating peptide like EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), or C5a 65 -74 (SEQ ID NO:4) or their analogs or variants as described herein.
- a macrophage- activating peptide like EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), or C5a 65 -74 (SEQ ID NO:4) or their analogs or variants as described herein.
- Ml-activated macrophages and macrophage- activating compound such as a macrophage- activating peptide as disclosed herein, when administered in combination, are effective in the prevention and treatment of biofilms.
- biofilms including MRSA biofilms, have the capacity to thwart this response by skewing macrophages away from a pro-inflammatory Ml phenotype to an anti-inflammatory M2 phenotype to ensure biofilm persistence in an immunocompetent host.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical & Material Sciences (AREA)
- Cell Biology (AREA)
- Immunology (AREA)
- Medicinal Chemistry (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Engineering & Computer Science (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Gastroenterology & Hepatology (AREA)
- Zoology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Disclosed are materials, uses and methods for preventing and treating biofilm infection, such as a device-associated biofilm infection. Exemplary devices include catheters and artificial implants, such as artificial hips, knees, ankles, shoulders, elbows, wrists, and the like. The methods comprise administration of a prophylactically or therapeutically effective amount of activated macrophages to reduce the spread of infection, to reduce the growth of microbes associated with a biofilm, such as Staphylococcus aureus (e.g., MRS A), or to kill such microbes, thereby effectively preventing or treating a biofilm infection.
Description
COMPOSITIONS AND METHODS FOR PREVENTING AND TREATING BIOFILMS
CROSS-REFERENCE TO RELATED APPLICATION
[0001] This application claims the priority benefit of provisional U.S. Patent Application Number 61/527,773 filed August 26, 2011, which is incorporated herein by reference in its entirety.
GOVERNMENT SUPPORT
[0002] This invention was made with government support under Grant No. P01 A1083211 awarded by the National Institutes of Health (NIH/NIAID). The government has certain rights in the invention.
BACKGROUND
[0003] Microorganisms, such as bacteria, can form biofilms, where organisms are encased in a complex three-dimensional matrix consisting of extracellular polysaccharides, DNA, and proteins. This structure protects the microbes from a variety of physical and chemical challenges, making biofilm treatment difficult. In addition, upon biofilm formation, some bacteria assume a metabolically dormant state, which protects them from antibiotic killing that requires active cell wall and/or protein synthesis.
[0004] Biofilm infections often lead to significant morbidity due to their chronicity and recalcitrance to antibiotics. Biofilms cause over 80% of infections and approximately 65% of nosocomial infections caused by microorganisms in the developed world involve biofilms.
[0005] One patient population particularly vulnerable to biofilm infection is the group of patients harboring foreign devices, such as implants (e.g., artificial joints, subcutaneous pacemakers) or indwelling catheters. Biofilms are responsible for catheter infections that cause about 10,000 U.S. deaths and more than 11 billion US dollars in losses in hospital costs annually. Twenty percent of urinary catheters inserted into 5 million U.S. patients have developed biofilm infections, resulting in medical expenses of 1.6 billion U.S. dollars each year.
[0006] More specifically with respect to patients harboring foreign devices, e.g. , implants or catheters, the frequency of device-related biofilms continues to rise, with current infection rates totaling 1-2% of primary arthroplasties, 3-5% of all revisions, about 10-50% of short- term urinary catheterizations, and the majority of all patients with long-term catheterization. These numbers do not take into account the propensity of bacterial biofilms to colonize other
medical devices as well as natural body surfaces. It is estimated that the total cost of biofilm infections, in treatment, lost productivity and fouled equipment, is about 500 billion US dollars annually.
[0007] In addition to the significant cost associated with biofilm infections, devastating health consequences can result from such infections. As but one example, biofilms of a single potentially pathogenic bacterium, Pseudomonas aeruginosa, are the cause of 70,000 cases of cystic fibrosis worldwide. Between 5 to 7 billion U.S. dollars are spent in medical care and productivity losses in the United States due to 25 to 80 million cases annually of biofilm-related food-borne diarrhea and 8,000 to 18,000 cases of biofilm-related
Legionnaire's disease.
[0008] Bacterial biofilm infections alter host immune responses to favor anti-inflammatory and pro-fibrotic pathways, which contribute to biofilm persistence. A variety of microbes, e.g., bacteria, have been identified as being associated with, or causative agents of, biofilm. A number of these infectious microbes are major health scourges, including methicillin- resistant Staphylococcus aureus (MRSA), a common etiologic agent of biofilms that often cause chronic and recurrent infections when associated with indwelling medical devices, such as implants and catheters. The persistent biofilms caused by these microbes, moreover, are associated with a variety of diseases and medical conditions, including but not limited to, dental caries, periodontitis, infection complications associated with cystic fibrosis, pneumonia, infective endocarditis, muscle skeletal infections, necrotizing fasciitis, osteomyelitis, meloidosis, infectious kidney stones, bacterial endocarditis, airway infections, otitis media, biliary tract infections, chronic bacterial prostatitis, and infections associated with medical devices {e.g., intravenous catheters, urinary catheters, artificial joints, contact lenses).
[0009] Current therapeutic options are ineffective in treating and controlling microbial burden, e.g., bacterial burden, in biofilms. For example, conventional antibiotics are ineffective in treating or controlling bacterial burden in biofilms. The reasons for the ineffectiveness of antibiotics and other chemical-based approaches to biofilm control are not fully understood, but it is believed that the biofilm matrix or "slime" presents a barrier to antibiotics that is exacerbated by a host-derived fibrous capsule typically found associated with chronic biofilm infections. In addition, it is believed that the altered physiological state of microbes associated with a biofilm affect their susceptibility to many antibiotics.
Regardless of the reasons, however, empirical evidence demonstrates that antibiotic treatment of biofilm infection is insufficient to address the problem. Nevertheless, antibiotics are
commonly used to control microbes, e.g., bacteria, that escape the biofilm matrix in order to prevent their colonization of other tissue sites. Such uses of antibiotics impose selection pressures on microbes such as bacteria and increase the chance of antibiotic-resistant strains arising.
[0010] A relatively recent development regarding antibiotic treatment involves an administration schedule characterized by the delivery of intermittent low dosages of antibiotics to treat biofilm infection. The schedule is designed to better attack persister cells, which are biofilm-associated microbes that survive initial, high-dosage treatments. Persister cells have been offered as an explanation for the capacity of many biofilm infections to not only survive initial antibiotic treatments, but to propagate biofilm infection over time. This capacity of biofilm infections has been suggested as a basis for the chronic infections associated with biofilm. Although the intermittent, low-dose approach to biofilm infection treatment may help to treat persister cells associated with biofilm, this approach does not advance methods to prevent biofilm infection and it does not reduce the risk of developing resistant strains because it continues to apply selective pressure in the form of antibiotics.
[0011] For biofilm infections associated with artificial devices such as implants or catheters, current therapy involves a cycle of implant or catheter removal and replacement, which is inconvenient, ineffective, and typically undesirable. Management of implant- or device-associated biofilm infections involves a staged replacement of the hardware, either as a single-step exchange, whereby the entire implant is replaced in a single procedure or, more commonly, as a two-stage exchange. In the latter case, patients receive extended antibiotic regimens in addition to surgical management, which generally consists of device removal and replacement with an antibiotic-impregnated temporary spacer, followed by insertion of a new prosthesis after a two- to eight- week period. This is a long and debilitating process associated with significant morbidity and economic impact for patients. The difficulties of biofilm treatment are further underscored in the context of more permanent implants such as artificial joints (e.g. , hips, knees, shoulders, ankles, elbows, and wrists), procedures that are particularly common in the elderly, who grow increasingly less immune-responsive over time.
[0012] Accordingly, a need exists in the art for compositions and methods for preventing and treating biofilms that are efficacious and cost-effective without incurring significant risk of toxicity or other complications arising from prophylactic or therapeutic treatment, while minimizing the need for surgical intervention.
SUMMARY
[0013] The materials and methods (and uses) disclosed herein provide cells, such as autologous cells, that can be manipulated ex vivo and/or in vivo to become activated macrophages suitable for use in preventing or treating biofilm infections. In providing an immune-based approach to the prevention and treatment of biofilm, the disclosure provides materials and methods of wide applicability, i.e., any biofilm associated with a native surface or "foreign" device such as an infectious microbe {e.g., bacterium) is amenable to prevention or treatment with the disclosed materials and methods. In addition, the disclosed methods provide the benefit of phrophylactic and therapeutic approaches that will not contribute to the collective evolutionary problem of effectively selecting for infectious superbugs resistant to most treatments, such as methicillin-resistant S. aureus (i.e., MRSA), because the methods take advantage of bacterial killing attributes inherent to the host's immune system.
[0014] In developing the disclosed materials and methods/uses, it was recognized that biofilms can skew the immune response to favor anti-inflammatory and pro-fibrotic pathways, which contribute to biofilm persistence. As a specific example, although macrophages are a prominent infiltrate in S. aureus biofilm infections, their penetration into the biofilm itself is impeded by a robust fibrotic response surrounding the infection. In addition, biofilm-associated macrophages are polarized towards an alternatively activated M2 phenotype that exhibits anti-inflammatory and pro-fibrotic properties that limit bacterial clearance. By extension, the programming of macrophages towards a microbicidal Ml response is diverted towards a M2 phenotype prior to physical contact with the biofilm. These findings led to methods of treating biofilms by exogenous administration of Ml- activated macrophages directly into sites of biofilm infection. As disclosed herein, the administration overcame the local immunoinhibitory environment and bypassed the fibrotic barrier associated with biofilm infection.
[0015] Also contemplated are variations of the above-described methods/uses to prevent and/or treat biofilms further comprising administration of an effective amount of a macrophage-activating peptide, such as a C5a receptor agonist, a C5a analog, or a C5a fragment, wherein each of these peptides facilitates bacterial clearance by inducing a proinflammatory milieu. One exemplary C5a receptor agonist is EP67 (YSFKDMP(MeL)aR; SEQ ID NO:l; uppercase letters designate the L stereoisomeric form and lower case the D stereoisomeric form of the amino acids; (MeL) corresponds to N-methyl leucine)), which is a response- selective agonist of the human C5a receptor (C5aR) that preferentially elicits proinflammatory mediator production from C5aR-bearing macrophages without affecting C5aR-
bearing neutrophils. EP67 was derived from the C-terminal region of human complement component, C5a (Taylor SM, et al., Curr Med Chem 2001;8:675-684.). EP67 possesses potent immune-enhancing properties (Morgan et al., Vaccine, 28(2): 463-469 (2009)). The disclosure further provides a C5a analog comprising 1, 2, 3, 4, or 5 amino acid substitutions in the amino acid sequence set out in SEQ ID NO:l wherein the peptide having the substituted amino acid sequence retains binding activity of the peptide set out in SEQ ID NO:l.
[0016] The C-terminus of C5a comprises amino acids 65-74 of C5a (i.e. , ISHKDMQLGR ("C5a65_74") (SEQ ID NO:4). The disclosure further provides a C5a analog comprising 1, 2, 3, 4, or 5 amino acid substitutions in the amino acid set out in SEQ ID NO: 4 wherein the peptide having the substituted amino acid sequence retains binding activity of the peptide set out in SEQ ID NO:4.
[0017] The C-terminal analog of C5a, EP54 (YSFKPMPLaR; SEQ ID NO:2) is a C5a receptor agonist that has been described previously (Duryee et al. (2009) Vaccine 27:2981- 2988; Hegde et al. (2008) Int. Immunopharmacol. 8:819-827; and Ulrich et al. (2000) J. Immunol. 164:5492-5498). However, these studies described an EP54 construct wherein EP54 was conjugated to a specific antigen or hapten. The disclosure further provides a C5a analog comprising 1, 2, 3, 4, or 5 amino acid substitutions in the amino acid sequence set out in SEQ ID NO:2 wherein the peptide having the substituted amino acid sequence retains binding activity of the peptide set out in SEQ ID NO:2. Additional disclosure of C5a analogs is provided in USSN 61/359,444 and International (PCT) Patent Application No.
PCT/US2011/042344 , each of which is incorporated herein by reference.
[0018] One or more of the macrophage-activating compounds disclosed herein, e.g., a C5a receptor agonist, a C5a analog or a C5a fragment, is useful in methods of activating macrophages prior to, concurrent with, or after administration of macrophage to prevent or treat a biofilm infection. In a typical embodiment, the macrophage-activating compound contacts the macrophage ex vivo prior to administration of the macrophages. Given that, in some embodiments, administration will place the macrophages beyond the fibrotic barrier in the vicinity of the biofilm, it is contemplated that the macrophage-activating compound is coadministered with the macrophages or the macrophage-activating compound is administered after the administration of the macrophages, where administration beyond the fibrotic barrier and in the vicinity of the biofilm places the macrophage-activating compound(s) and macrophages in sufficiently close proximity to result in contact and macrophage activation in vivo.
[0019] Targeting macrophage pro-inflammatory activity with exogenous Ml -activated macrophages inhibited S. aureus biofilm formation and provided long-lasting protection from catheter colonization and infection in surrounding tissues. This protection was not provided by neutrophils, consistent with the failure of these cells to become a prominent infiltrate during S. aureus biofilm infection in some settings. Collectively, these results indicate that immune cell-based therapy using activated macrophages overcomes the current difficulties associated with biofilm treatment and control.
[0020] The approaches to biofilm prevention and treatment disclosed herein exploit natural host immune mechanisms for therapeutic benefit. Targeting the host response rather than the pathogen itself offers certain advantages by largely avoiding selective pressures for the evolution of microbial resistance. Indeed, stimulating adaptive immunity through vaccination has remained effective in the face of microbial resistance over decades of clinical use, although not all pathogens have been amenable to vaccination strategies, most notably S. aureus. Furthermore, the fact that innate immune defenses are geared to rapidly recognize an infinite pathogen repertoire, indicates that immunomodulation will afford broad- spectrum protection against a range of microbial pathogens, enabling prophylactic use in high-risk groups and early treatment prior to the identification of causative infectious agents.
[0021] The local administration of classically activated Ml macrophages for biofilm prevention and treatment disclosed herein overcomes immune evasion by the causative microbes by triggering a pro-inflammatory milieu. Early administration of Ml-activated macrophages significantly attenuated biofilm formation in a mouse model of MRSA catheter- associated infection. Several pro-inflammatory mediators were significantly elevated in biofilm-infected tissues from macrophage-treated animals, revealing effective reprogramming of the biofilm environment to a pro-inflammatory milieu. Treatment of established biofilm infections with activated macrophages also significantly reduced catheter-associated biofilm burdens compared to antibiotic treatment. A requirement for macrophage pro-inflammatory activity was demonstrated by the fact that transfer of MyD 88 -deficient macrophages had minimal impact on biofilm growth. Likewise, neutrophil administration had no effect on biofilm formation. Collectively, these results demonstrate that targeting macrophage proinflammatory activity can overcome the local immunoinhibitory environment created during biofilm infections and represents a promising therapeutic strategy to the prevention and treatment of biofilm infections.
[0022] The disclosure provides a convenient and effective way to prophylactically and/or therapeutically reduce the incidence and/or severity of microbial, e.g., bacterial, biofilm
infections generally, and more particularly, of microbial, e.g., bacterial, biofilm infections associated with catheters (e.g., urinary, intravenous, an intraarterial catheters) and other artificial implants such as artificial joint components (e.g., artificial components of hip, knee, ankle, shoulder, elbow and wrist joints).
[0023] One aspect of the disclosure is a composition comprising an isolated activated macrophage for the prevention or treatment of a biofilm. In some embodiments, the composition further comprises a macrophage- activating peptide selected from the group consisting of EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2) and C5a65-74 (SEQ ID NO:4). Variants and derivatives of the EP67 peptide, the EP54 peptide, or the C5a65-74 peptide that retain the capacity to beneficially affect endogenous macrophages, as disclosed herein, are also contemplated, and in particular those variants and derivatives of the EP67 peptide, EP54 peptide, and C5a65-74 peptide that differ by 1, 2, 3, 4, or 5 residues from the relevant wild-type sequence, i.e., SEQ ID NO:l for EP67, SEQ ID NO:2 for EP54, and SEQ ID NO:4 for C5a65_ 74 are within the scope of the compositions, and methods, of the disclosure.
[0024] A related aspect of the disclosure provides a kit comprising an isolated macrophage and a macrophage- activating agent. Also provided is a kit comprising a macrophage- activating agent. In some embodiments of the kit, the macrophage- activating agent is selected from the group consisting of a C5a receptor agonist, including but not limited to macrophage-activating peptides such as EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), C5a65- 74 (SEQ ID NO:4) and analogs and variants thereof, interleukin-ΐβ as well as combinations of interferon-γ and tumor necrosis factor (TNF), and interferon-γ and an inducer of TNF. An exemplary kit comprises a C5a receptor agonist that is selected from the group consisting of EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2) and C5a65-74 (SEQ ID NO:4). As with the compositions according to the disclosure, kits according to the disclosure include variants and derivatives of the EP67 peptide, the EP54 peptide, or the C5a65-74 peptide that retain the capacity to beneficially affect endogenous macrophages, as disclosed herein, and in particular, those variants and derivatives that differ by 1, 2, 3, 4, or 5 residues from the relevant wild-type sequence, i.e., SEQ ID NO:l for EP67, SEQ ID NO:2 for EP54, and SEQ ID NO:4 for C5a65-74 are within the scope of the kits of the disclosure.
[0025] Another aspect of the disclosure is a method of providing biofilm therapy to a subject-in-need comprising: (a) isolating peripheral blood monocytes from a subject; (b) culturing the monocytes under conditions compatible with formation of macrophages; and (c) administering a prophylactically or therapeutically effective amount of the macrophages to a subject-in-need, wherein the macrophages are activated by contact with a macrophage-
activating agent prior to, simultaneously with, or after administration of the macrophages. In some embodiments of the method, the macrophages are activated by contact with a macrophage- activating agent prior to administration of the macrophages. A related aspect is drawn to the use of an activated macrophage in the treatment of an established biofilm infection in a subject-in-need. In some embodiments of the method and of the use, the subject-in-need is at risk of developing a biofilm infection (i.e., the subject-in-need does not have an established biofilm infection), while in other embodiments, the subject-in-need comprises a biofilm infection (i.e., the subject-in-need does have an established biofilm infection).
[0026] Also contemplated by the disclosure are embodiments wherein the subject-in-need is a human patient or a veterinary animal, such as a veterinary animal selected from the group consisting of a dog, a cat, a horse, a bovine (a bull or cow), a sheep, a goat and a pig.
Embodiments of the method and use are contemplated in which the subject is the subject-in- need (i.e., the isolated cells are autologous cells). In some embodiments, the dose or doses of macrophages, e.g., activated macrophages, for administration according to the methods and uses of the disclosure are determined on an individual basis using factors known in the art, e.g., general health, immunocompetency, age, weight, and the like. In general, the disclosure contemplates dosages within the range of about 105 to 108 macrophages, e.g., 106
macrophage cells. Consistent with relative infectious microbe burden, lower dosages and/or a reduced schedule of administration of macrophages would be expected in prophylactic methods and uses compared to treatment methods and uses. Dosage scheduling is expected to range from about one to five doses of macrophages, with each dose separated from other doses by about one to five days. In some embodiments, multiple doses are administered to different sites around the expected or real periphery of a potential or existing biofilm infection. An exemplary dosage schedule for prophylactic methods and uses is about three daily doses of 10 5 to 108 macrophages, which is compatible with the average hospital stay for receiving a surgical implant, which places the patient at risk of a device-associated biofilm infection.
[0027] In some embodiments, the macrophage-activating agent is selected from the group consisting of a C5a receptor agonist, including but not limited to macrophage-activating peptides such as EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), C5a65-74 (SEQ ID NO:4), and analogs and variants thereof, interleukin-ΐβ as well as combinations of interferon-γ and tumor necrosis factor (TNF), and interferon-γ and an inducer of TNF. An exemplary inducer of TNF is a TNF inducer selected from the group consisting of peptidoglycan and
lipopolysacchande. An exemplary C5a receptor agonist is selected from the group consisting of EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2) and C5a65-74 (SEQ ID NO:4). In some embodiments of the method and use, the macrophage- activating agent is a combination selected from the group consisting of interferon-γ and tumor necrosis factor (TNF) or interferon-γ and an inducer of TNF. An exemplary inducer of TNF is selected from the group consisting of peptidoglycan and lipopolysaccharide. Yet another stimulatory agent is the above-noted macrophage- activating peptide EP67, a C5a receptor/CD88 agonist that specifically induces pro-inflammatory properties of CD88+ macrophages without any effects on CD88+ neutrophils. Yet other embodiments of the method and use further comprise delivering to the subject- in-need an effective amount of a pro-inflammatory compound, such as interleukin-6, interleukin-12, monocyte chemoattractant protein- 1 (MCP-1), monokine- induced by IFN-γ (MIG), interferon-regulatory protein- 10 (IP- 10), macrophage inflammatory protein-2 (MIP-2), and keratinocyte chemoattractant (KC). Also contemplated are macrophage colony-stimulating factor (M-CSF) and granulocyte-macrophage colony- stimulating factor (GM-CSF) to encourage macrophage and neutrophil expansion at the site of biofilm infection, whereupon pro-inflammatory cytokines would be present to activate these cells to trigger their bactericidal activity.
[0028] It is also contemplated that, in some embodiments of both the method and use, the subject- in-need undergoes surgery to receive a surgical implant, such as an artificial joint component, a cranial plate (natural or artificial), a dental implant, or a subcutaneous pacemaker. Exemplary artificial joint components are an artificial hip component, an artificial knee component, an artificial ankle component, an artificial elbow component, an artificial shoulder component, and an artificial wrist component. In some embodiments of each of the disclosed method and use, the subject-in-need is scheduled to receive, or has received, a catheter. In some implementations of the method and of the use, the activating agent and the macrophage are co-administered to the subject-in-need. Yet other
embodiments of the method or use further comprise delivery of a proteolytic enzyme, such as MMP-1, MMP-2, MMP-7, MMP-9, MMP-12 or collagenase to the subject-in-need, for example to prevent biofilm infection. Additional embodiments of the method or use further comprise use of a proteolytic enzyme, such as MMP-1, MMP-2, MMP-7, MMP-9, MMP-12, and collagenase, in the treatment of an existing biofilm infection in a subject-in-need.
Without wishing to be bound by theory, the proteolytic enzyme is expected to degrade the fibrotic capsule often associated with biofilms, enhancing the access of activated
macrophages to the biofilm.
[0029] In some embodiments of both the methods and uses according to the disclosure, the proteolytic enzyme is packaged in a delivery vehicle such as a microsphere, microparticle, nanosphere, nanoparticle, liposome, micelle, or gel. These delivery vehicles may be formulated to provide for the controlled, e.g., slow, release of their contents, as would be known in the art. In various embodiments, the delivery vehicle is selected from the group consisting of a microsphere, a microparticle, a nanosphere, a nanoparticle, a liposome, a micelle and a gel, wherein the delivery vehicle comprises a macrophage and a macrophage- activating agent, or an activated macrophage, or a macrophage, a macrophage-activating agent and a proteolytic enzyme, or an activated macrophage and a proteolytic enzyme.
Delivery vehicles suitable for the disclosed methods and uses include vehicles that are targeted to a biofilm, such as by exhibiting a binding partner to a marker displayed on the infectious organism, and vehicles that are targeted to the vicinity of a biofilm infection, for example by exhibiting a binding partner to a component of the fibrotic matrix frequently associated with biofilms. An exemplary proteolytic enzyme is collagenase, which may be packaged in any of the foregoing vehicles, such as a microsphere. In some embodiments, the method and use further comprise the delivery of at least one molecule that interferes with the formation of the fibrotic capsule surrounding biofilm infections, such as slow-release delivery vehicles (any of the above-described vehicles) containing at least one proteolytic enzyme (e.g., collagenase or an MMP) and, optionally, at least one compound that interferes with the formation of a fibrotic capsule, such as a Lysyl Oxidase (i.e., LOX) inhibitor (e.g., BAPN). In general, the disclosure comprehends any delivery vehicle known in the art for use in packaging and delivering activated macrophages, or macrophages and at least one macrophage-activating agent, or activated macrophages and at least one proteolytic enzyme, or macrophages, at least one macrophage-activating agent and at least one proteolytic enzyme. Any of these formulations may further comprise a compound that interferes with the formation of the fibrotic capsule, such as BAPN or another Lysyl Oxidase (LOX) inhibitor.
[0030] Embodiments of the use of activated macrophages further comprise using a prophylactically or therapeutically effective amount of an EP67 peptide (SEQ ID NO:l) in the treatment of a biofilm infection in a subject-in-need. In some embodiments of each of the method and the use, the subject-in-need exhibits an inflammatory disorder. In some embodiments of each of the method and thus use, the subject-in-need exhibits a disorder or condition selected from the group consisting of atherosclerosis, infectious complications of cystic fibrosis, endocarditis, chronic sinusitis, nasal polyposis, chronic otitis media,
leptospirosis, kidney stones, osteomyelitis, necrosis of the jaw, a urinary tract infection, a bladder infection, a chronic wound, an artificial component of a joint, a temporary catheter and a permanent catheter.
[0031] Embodiments of the methods and uses for preventing or treating biofilm infections include methods and uses in which the subject-in-need exhibits an inflammatory disorder. An inflammatory disorder as used herein means any inflammatory disorder of humans or veterinary animals that is known in the art. Exemplary methods and uses comprise a subject- in-need that exhibits a disorder or condition selected from the group consisting of atherosclerosis, infectious complications of cystic fibrosis, endocarditis, chronic sinusitis, nasal polyposis, chronic otitis media, leptospirosis, kidney stones, osteomyelitis, necrosis of the jaw, a urinary tract infection, a bladder infection, a chronic wound, an artificial component of a joint, a temporary catheter and a permanent catheter.
[0032] Embodiments of the methods and uses for preventing or treating biofilm infections further comprise administering a prophylactically or therapeutically effective amount of an antibiotic. In some embodiments, the antibiotic is a member of a class of antibiotics selected from the group consisting of aminoglycosides, ansamycins, carbacephems, carbapenems, cephalosporins, glycopeptides, lincosamides, lipopeptides, macrolides, erythromycins, monobactams, nitrofurans, penicillins, penicillin combinations, antimicrobial polypeptides, quinolones, sulfonamides, tetracyclines and antimycobacterials. In some embodiments, the antibiotic is selected from the group consisting of aminoglycosides, e.g., , amikacin (amikin), gentamicin (garamycin), kanamycin (kantrex), neomycin (neofradin), roxithromycin, netilmicin (netromycin), tobramycin (nebcin), paromomycin (humatin); ansamycins, e.g., geldamycin, herimycin; Carbacephems, e.g., loracarbef (lorabid); carbapenems, e.g., ertapenem (invanz), doripenem (doribax), imipenem or cilastatin (primaxin), meropenem (merrem); cephalosporins, e.g., cefadroxil (duricef), cefazolin (ancef), cefalotin or cefalothin (keflin), cefalexin (keflex), cefaclor (distaclor), cefamandole (mandol), cefoxitin (mefoxin), cefprozil (cefzil), cefuroxime (cetin, zinnat), cefonicid, cefixime (suprax), cefdinir (omnicef, cefdiel), cefditoren (spectracef, cefobid), cefotaxime (claforan), cefetamet, cefoperazone, cefpodoxime (vantin), ceftazidime (fortaz), ceftibuten (cedax), ceftriaxone (rocephin), cefepime (maxipime), ceftaroline or fasamil (teflaro), ceftobiprole (zeftera), cephalothin, cefmetazole, cefotetan, ceftizoxime, cefsulodin, cephalexin, cinoxacin; glycopeptides, e.g., teicoplanin (targocid), vancomycin (vancocin), telavancin (vibativ); lincosamides, e.g., clindamycin (cleocin), lincomycin (lincocin); lipopeptides, e.g., daptomycin (cubicin);
macrolides, e.g., azithromycin (zithromax, sumamed, xithrone), clarithromycin (biaxin,
dynabac); erythromycin (erythocin, erythroped), erythromycin estolate, erythromycin ethyl succinate, erythromycin glucoheptonate, erythromycin lactobionate, erythromycin stearate, roximthromycin, trolandomycin (tao), telithromycin (ketek), spectinomycin (trobicin), spriamycin (rovamycine); monobactams, e.g., aztreonam (azactam); nitrofurans, e.g., furazolidone (furoxone), nitrofurantoi (macrodantin, macrobid); penicillins, e.g., amoxicillin (novamox, amoxil), ampicillin (principen), azlocillin, carbenicillin (geocillin), cloxacillin (tegopen), dicloxacillin (dynapen), flucloxacillin (floxapen), mezlocillin (mezlin), methicillin (staphcillin), nafcillin (unipen), oxacillin (prostaphlin), penicillin G (pentids, pfizerpen, negaban), penicillin V (veetids), piperacillin (pipracil), ticarcillin (ticar); penicillin combinations, e.g., amoxicillin and clavulanate (augmentin), ampicillin and sulbactam (unasyn), piperaccillin and tazobactam (zosyn), ticarcillin and clavulanate (timentin);
antimicrobial polypeptides, e.g., bacitracin, colistin (colymycin S), polymyxin B; quinolones, e.g., ciprofloxacin (cipro, ciproxin, ciprobay), enoxacin (penetrex), gatifloxacin (tequin), levofloxacin (levaquin), lomefloxacin (maxaquin), moxifloxicin (avelox), ofloxacin (floxin, ocuflox), nalidixic acid (negram), norfloxacin (noroxin), ofloxicin (floxin, ocuflox), trovafloxacin (trovan), grepafloxacin (raxar), sparfloxacin (zagam), temafloxacin (omniflox); sulfonamides, e.g., mafenide (sulfamylon), sufonamidochrysoidine (prontosil), sulfacetamide (sulamyd, bleph-10), sufadiazine (micro-sulfon), silver sulfadiazine (silvadene),
sulfamethizole (thiosulfil forte), sulfamethoxazole (gantanol), sulfanilamide, sulfasalazine (azulfidine), sulfisoxazole (gantrisin), trimethoprim-sulfamethoxazole (bactrim, Septra); Tetracyclines, e.g., demeclocycline (declomycin), doxycycline (vibramycin), minocycline (minocin), oxytetracycline (terramycin), tetracycline (sumycin, achromycin V, steclin); and antimycobacterials, e.g., clofazimine (lamprene), dapsone (avlosulfon), capreomycin
(capastat), cycloserine (seromycin), ethambutol (myambutol), ethionamide (trecator), isoniazid (INH), rifampicin or rifampin (rifadin, fimactane), rifabutin (mycobutin), rifapenine (priftin), streptomycin, arshenamine (salvarsan), chloramphenicol (Chloromycetin), fosfomycin (monurol), fusidic acid (fucidin), linezolid (zyvox), metronidazole (flagyl);
mupirocin (bactroban), platensimycin, quinupristin/dalfopristin (synercid), rifaximin
(xifaxan), thiamphenicol, tigercycline (tigacyl) tinidazole (tindamax fasigyn); and other compounds, e.g., trimethoprim (proloprim, trimpex); fleroxacin, nitrofurantoin, and coamoxiclavuanate. Also contemplated are various forms of each of these antibiotics, including but not limited to their salts, acids, and bases.
[0033] Particular aspects of the disclosure are described in the following enumerated paragraphs.
[0034] 1. A composition comprising an isolated activated macrophage for the prevention or treatment of a biofilm.
[0035] 2. The composition according to paragraph 1 further comprising a macrophage- activating peptide selected from the group consisting of EP67, EP54 and C5a65-74.
[0036] 3. A kit comprising an isolated macrophage and a macrophage- activating agent.
[0037] 4. A kit comprising a macrophage-activating agent.
[0038] 5. The kit according to paragraph 3 or 4 wherein the macrophage-activating agent is selected from the group consisting of a C5a receptor agonist, interleukin-ΐβ, interferon-γ and tumor necrosis factor (TNF), and interferon-γ and an inducer of TNF.
[0039] 6. The kit according to paragraph 5 wherein the C5a receptor agonist is selected from the group consisting of EP67, EP54 and C5a65-74.
[0040] 7. A method of providing biofilm therapy to a subject-in-need comprising:
(a) isolating peripheral blood monocytes from a subject;
(b) culturing the monocytes under conditions compatible with formation of
macrophages; and
(c) administering a prophylactically or therapeutically effective amount of the macrophages to a subject-in-need, wherein the macrophages are activated by contact with a macrophage-activating agent prior to, simultaneously with, or after administration of the macrophages.
[0041] 8. The method according to paragraph 7 wherein the macrophages are activated by contact with a macrophage-activating agent prior to administration of the macrophages.
[0042] 9. A use of macrophages in the prevention or treatment of a biofilm infection in a subject-in-need, wherein the macrophages are activated by contact with a macrophage- activating agent prior to, simultaneously with, or after administration of the macrophages.
[0043] 10. The method according to paragraph 7 or 8 wherein the subject-in-need is at risk of developing a biofilm infection.
[0044] 11. The method according to paragraph 7 or 8 wherein the subject-in-need comprises an established biofilm infection.
[0045] 12. The method according to paragraph 7 or 8 wherein the subject-in-need is a human patient.
[0046] 13. The method according to paragraph 7 or 8 wherein the subject-in-need is a veterinary animal selected from the group consisting of a dog, a cat, a horse, a bovine (a cow or a bull), a sheep, a goat and a pig.
[0047] 14. The method according to paragraph 7 or 8 wherein the subject is the subject- in-need.
[0048] 15. The method according to paragraph 7 or 8 wherein the macrophage- activating agent is selected from the group consisting of a C5a receptor agonist, interleukin-ΐβ, interferon-γ and tumor necrosis factor (TNF), and interferon-γ and an inducer of TNF.
[0049] 16. The method according to paragraph 15 wherein the inducer of TNF is selected from the group consisting of peptidoglycan and lipopolysaccharide.
[0050] 17. The method according to paragraph 15 wherein the C5a receptor agonist is selected from the group consisting of EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2) and C5a65- 74 (SEQ ID NO:4).
[0051] 18. The method according to paragraph 7 or 8 wherein the macrophages are administered at a dose of 10 5 to 108 cells.
[0052] 19. The method according to paragraph 18 wherein one to five doses of macrophages are administered.
[0053] 20. The method according to paragraph 15 further comprising delivering to the subject-in-need an effective amount of a pro -inflammatory compound selected from the group consisting of interleukin-6, interleukin-12, monocyte chemoattractant protein- 1 (MCP- 1), monokine-induced by IFN-γ (MIG), interferon-regulatory protein- 10 (IP- 10), macrophage inflammatory protein-2 (MIP-2), keratinocyte chemoattractant (KC), macrophage colony- stimulating factor (M-CSF) and granulocyte-macrophage colony- stimulating factor (GM- CSF).
[0054] 21. The method according to paragraph 7 or 8 wherein the subject-in-need undergoes surgery to receive a surgical implant.
[0055] 22. The method according to paragraph 21 wherein the surgical implant is selected from the group consisting of an artificial joint component, a cranial plate (natural or artificial), a dental implant and a subcutaneous pacemaker.
[0056] 23. The method according to paragraph 22 wherein the artificial joint component is selected from the group consisting of an artificial hip component, an artificial knee
component, an artificial ankle component, an artificial elbow component, an artificial shoulder component, and an artificial wrist component.
[0057] 24. The method according to paragraph 7 or 8 wherein the subject-in-need is scheduled to receive, or has received, a catheter.
[0058] 25. The method according to paragraph 7 wherein the macrophage-activating agent and the macrophage are co-administered to the subject-in-need.
[0059] 26. The method according to paragraph 7 or 8 further comprising delivery of a proteolytic enzyme to the subject-in-need.
[0060] 27. The method according to paragraph 26 wherein the proteolytic enzyme is selected from the group consisting of MMP-1, MMP-2, MMP-7, MMP-9, MMP-12 and collagenase.
[0061] 28. The method according to paragraph 7, 8, or 27 further comprising a delivery vehicle selected from the group consisting of a microsphere, a microparticle, a nanosphere, a nanoparticle, a liposome, a micelle and a gel, wherein the delivery vehicle comprises a macrophage and a macrophage-activating agent, or an activated macrophage, or a macrophage, a macrophage-activating agent and a proteolytic enzyme, or an activated macrophage and a proteolytic enzyme.
[0062] 29. The method according to paragraph 7 or 8 wherein the subject-in-need exhibits an inflammatory disorder.
[0063] 30. The method according to paragraph 7 or 8 wherein the subject-in-need exhibits a disorder or condition selected from the group consisting of atherosclerosis, infectious complications of cystic fibrosis, endocarditis, chronic sinusitis, nasal polyposis, chronic otitis media, leptospirosis, kidney stones, osteomyelitis, necrosis of the jaw, a urinary tract infection, a bladder infection, a chronic wound, an artificial component of a joint, a temporary catheter and a permanent catheter.
[0064] 31. The method according to paragraph 7 or 8 further comprising administering a prophylactically or therapeutically effective amount of an antibiotic.
[0065] 32. The method according to paragraph 31 wherein the antibiotic is a member of a class of antibiotics selected from the group consisting of aminoglycosides, ansamycins, carbacephems, carbapenems, cephalosporins, glycopeptides, lincosamides, lipopeptides, macrolides, erythromycins, monobactams, nitrofurans, penicillins, penicillin combinations, antimicrobial polypeptides, quinolones, sulfonamides, tetracyclines and antimycobacterials.
[0066] 33. The method according to paragraph 32 wherein the antibiotic is selected from the group consisting of amikacin, gentamicin, kanamycin, neomycin, roxithromycin, netilmicin, tobramycin, paromomycin, geldamycin, herimycin, loracarbef, ertapenem, doripenem, imipenem or cilastatin, meropenem, cefadroxil, cefazolin, cefalotin or cefalothin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefonicid, cefixime, cefdinir, cefditoren, cefotaxime, cefetamet, cefoperazone, cefpodoxime, ceftazidime, ceftibuten, ceftriaxone, cefepime, ceftaroline or fasamil, ceftobiprole, cephalothin, cefmetazole, cefotetan, ceftizoxime, cefsulodin, cephalexin, cinoxacin; teicoplanin, vancomycin, telavancin; clindamycin, lincomycin, daptomycin, azithromycin, clarithromycin, erythromycin, erythromycin estolate, erythromycin ethyl succinate, erythromycin
glucoheptonate, erythromycin lactobionate, erythromycin stearate, roximthromycin, trolandomycin, telithromycin, spectinomycin, spriamycin, Aztreonam, furazolidone, nitrofurantoi, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin G, penicillin V, piperacillin, ticarcillin, amoxicillin and clavulanate, ampicillin and sulbactam, piperaccillin and tazobactam, ticarcillin and clavulanate; bacitracin, colistin, polymyxin B, ciprofloxacin, enoxacin, gatifloxacin, levofloxacin, lomefloxacin, moxifloxicin, ofloxacin, nalidixic acid, norfloxacin, ofloxicin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sufonamidochrysoidine, sulfacetamide, sufadiazine, silver sulfadiazine, sulfamethizole, sulfamethoxazole, sulfanilamide, sulfasalazine, sulfisoxazole, trimethoprim- sulfamethoxazole, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol, ethionamide, isoniazid, rifampicin or rifampin, rifabutin, rifapenine, streptomycin, arshenamine, chloramphenicol, fosfomycin, fusidic acid, linezolid, metronidazole, mupirocin, platensimycin, quinupristin or dalfopristin, rifaximin, thiamphenicol, tigercycline, tinidazole, trimethoprim, fleroxacin, nitrofurantoin, coamoxiclavuanate, and their salts, acids, and bases.
[0067] Other features and advantages of the present disclosure will become apparent from the following detailed description, including the drawing. It should be understood, however, that the detailed description and the specific examples, while indicating preferred
embodiments, are provided for illustration only, because various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from the detailed description.
BRIEF DESCRIPTION OF THE DRAWING
[0068] Figure 1. Effect of activated macrophages on biofilm-causing bacteria.
C57BL/6 mice were infected with 10 colony forming units (cfu) of methicillin-resistant S. aureus (MRSA) USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection. Animals were initially treated with 106 neutrophils or Ml -activated macrophages derived from C57BL/6 mice at 12, 24 and 48 hours post-infection. Mice were sacrificed at day 3 following MRSA infection, whereupon catheters (A) and tissues surrounding the catheters (B) were removed to quantitate bacterial burdens. Results are expressed as the number of cfu per ml for catheters or cfu per mg tissue, to correct for differences in tissue sampling size. Significant differences are denoted by asterisks (*, p <
0.05). Results are presented from individual animals combined from at least two independent experiments.
[0069] Figure 2. Long-term effect of activated macrophages on biofilm-causing bacteria. Growth of S. aureus on a catheter surface, measured on day 3 and again on day 14 as the log io of the number of S. aureus colony forming units (cfu) per ml. Activated macrophages were administered at 12, 24 and 48 hours post-infection and no additional cells were administered beyond 48 hours following infection. Control mice received injections of vehicle (PBS) alone. Results showed that activated macrophages were capable of long-term attenuation of S. aureus biofilm growth in vivo.
[0070] Figure 3. Reduction of established S. aureus biofilm burden associated with catheters. S. aureus biofilms were established using a mouse s.c. catheter model with therapies initiated at day 7 following biofilm growth. A robust biofilm is formed within this time interval. Mice received either vehicle (PBS) or 106 activated macrophages (ΜΦ) within the catheter lumen at days 7 and 9 post-infection, whereas antibiotic-treated mice received
1. p. injections of rifampin and daptomycin at the same intervals (25 and 5 mg/kg,
respectively, which is a standard therapeutic cocktail for S. aureus infection). Mice were sacrificed at day 10 post-infection, whereupon catheter-associated biofilm burdens were determined by sonication and quantitative culture. Significant differences between mice receiving activated macrophage therapy versus vehicle control were observed. Each circle represents data from an individual animal. Left panel: Results from study 1, which compared activated macrophage therapy, antibiotic therapy, and vehicle alone as a control. Right panel: Results from a later study confirming the positive effect of activated macrophage therapy relative to vehicle alone as a control. (Additional studies had confirmed the relative ineffectiveness of antibody therapy.) The results establish that macrophage transfer therapy is effective at reducing established S. aureus biofilm burdens on foreign devices.
[0071] Figure 4. Ml macrophage polarization increases S. aureus biofilm phagocytosis and killing. A) Neutrophils, non-activated, and Ml -activated (lOng/ml IFN-γ + l( g/ml PGN) macrophages from C57BL/6 mice, as well as Ml-activated macrophages from MyD88 knockout (KO) animals were labeled with CellTracker Orange (orange-red) or CellTracker Blue (blue) and co-cultured with mature MRSA biofilms or planktonic bacteria (green) for 2 hours and imaged to observe their phagocytic ability. B) After 24 hours following immune cell addition, biofilms were mechanically disrupted by triturating to evaluate the ability of the various immune cell populations to attenuate biofilm growth.
Biofilms without the addition of immune cells were used as untreated controls. Arrows indicate phagocytic cells and significant differences are denoted by asterisks (**, p < 0.01; ***, /> < 0.001).
[0072] Figure 5. S. aureus biofilms decrease macrophage cytokine secretion. Bone marrow-derived macrophages were incubated with 6-day-old biofilms or planktonic bacteria for 2 hours in glass chamber slides, whereupon viable macrophages were purified by FACS and then incubated at 37°C for 24 hours without any further stimulation after which supernatants were collected to quantitate TNF-a, IL-10, and IL-1RA by ELISA. Significant differences in secreted cytokine levels between biofilm and planktonic exposed macrophages are denoted by asterisks (*, p < 0.05; **, p < 0.01; < 0.001).
[0073] Figure 6. Activated macrophages but not neutrophils impair MRSA biofilm formation in vivo. C57BL/6 mice were infected with 103 colony forming units (cfu) of methicillin-resistant S. aureus (MRSA) USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection. Animals were treated with 106 neutrophils, non- activated macrophage, or Ml-activated macrophages derived from C57BL/6 mice at 12, 24 and 48 hours post-infection. Mice were sacrificed at day 3 following MRSA infection, whereupon catheters (A) and tissues surrounding the catheters (B) were removed to quantitate bacterial burdens. Results are expressed as the number of cfu per ml for catheters or cfu per mg tissue, to correct for differences in tissue sampling size. Significant differences are denoted by asterisks (*, p < 0.05). Results are presented from individual animals combined from at least two independent experiments.
[0074] Figure 7. The ability of Ml-polarized macrophages to impair MRSA biofilm development is mediated by MyD88-dependent signals. C57BL/6 mice were infected with 10 colony forming units (cfu) of methicillin-resistant S. aureus (MRSA) USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection. Animals were initially treated with 106 activated macrophages derived from C57BL/6 or MyD88 knockout
(KO) mice at 12, 24 and 48 hours post-infection. Mice were sacrificed at day 3 following MRSA infection, whereupon catheters (A) and tissue surrounding the catheters (B) were removed to quantitate bacterial burdens. Results are expressed as the number of cfu per ml for catheters or cfu per mg tissue, to correct for differences in tissue sampling size. Results are presented from individual animals combined from at least two independent experiments. Significant differences are denoted by asterisks (*, p < 0.05).
[0075] Figure 8. Activated macrophage therapy alters the local inflammatory milieu in MRSA biofilm associated tissue. Animals were sacrificed at day 3 post-infection
(prophylactic) or day 10 (established) following methicillin-resistant S. aureus (MRSA) exposure. Tissues surrounding MRSA biofilms from vehicle- and Ml-activated macrophage treated mice were homogenized to quantitate CXCL9 (A and D), CCL5 (B), IFN-γ (C), CXCL2 (E), IL-17 (F) and IL-6 (G) expression by MILLIPLEX analysis. Results were normalized to the amount of total protein recovered to correct for differences in tissue sampling size. Significant differences between groups are denoted by asterisks (*, p < 0.05).
[0076] Figure 9. Ml-activated macrophages attenuate MRSA biofilm catheter persistence in an established infection. C57BL/6 mice were infected with 103 colony forming units (cfu) of methicillin-resistant S. aureus (MRSA) USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection. On days 7 and 9 post-infection, animals received vehicle control (PBS), 106 activated macrophages, or antibiotic combination (rifampicin 25 μg/ml + daptomycin 5 μg/ml). Catheters (A) and surrounding host tissues (B) were removed on day 10 to quantitate bacterial burdens. Significant differences between groups are denoted by asterisks (*, p < 0.05).
[0077] Figure 10. Ml-activated macrophage therapy attenuates arginase-1
expression in biofilms. Vehicle- (A & C) or Ml macrophage-treated (B & D) animals from the established biofilm paradigm were sacrificed at day 10 following methicillin-resistant S. aureus (MRSA) exposure, whereupon tissues surrounding infected catheters were subjected to immunofluorescence staining with Iba-1 (red) to identify macrophages, arginase-1 (green), and nuclear staining with DAPI (blue). Asterisks represent the original location of the catheter, which is non-adherent to glass slides. (E & F) Quantitation of arginase-1 and Iba-1 fluorescence surrounding S. aureus biofilms of vehicle- or Ml macrophage-treated animals from the established biofilm paradigm. Significant differences are indicated with asterisks (*, p < 0.05; **, p < 0.01).
[0078] Figure 11. Qdot labeling of Ml-activated macrophages. (A) Mice received one dose of 10 Ml-activated macrophages at the time of S. aureus challenge or at day 7 following infection, representing prophylactic and established therapies, respectively. The same cohort of animals was subjected to IVIS imaging daily to visualize macrophage persistence (red dots). Results are representative of 10 individual animals per group. (B) F4/80+ macrophages were recovered at day 5 post- infection, whereupon Ml (IRF-5) or M2 (arginase-1) markers were assessed by FACS to determine if exogenously introduced Qdot- labeled macrophages retained their Ml-activated state.
[0079] Figure 12. The macrophage-activating peptide EP67 attenuates S. aureus biofilm growth in vivo. EP67- or vehicle-treated mice were infected with 103 CFU of MRSA USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection. Animals were sacrificed at days 3 (A & B) or 14 (C & D) following S. aureus infection, whereupon catheters (A and C) or tissues surrounding the catheters (B and D) were removed to quantitate bacterial burdens. Data are expressed as the number of CFU per ml, or CFU per mg host tissue, for normalization. Results are presented from individual animals pooled from at least two independent experiments with bars representing the mean of each group. Significant differences in bacterial burdens between vehicle and EP67-treated mice are denoted by asterisks (*, p < 0.05).
[0080] Figure 13. EP67 augments inflammatory mediator expression in tissues surrounding MRSA biofilm infections. Tissues surrounding MRSA biofilms from EP67- or vehicle-treated mice were homogenized at day 3 post-infection to quantitate the effects of EP67 treatment on IL-12p40 (A), CCL5 (B), IL-17 (C), IL-la (D) and IFN-γ (E) expression by MILLIPLEX analysis. Results were normalized to the amount of total protein recovered to correct for differences in tissue sampling size. Results are presented from individual animals combined from two independent experiments. Significant differences between EP67- vs. vehicle-treated catheter-associated tissues are denoted by asterisks (*, p < 0.05).
[0081] Figure 14. EP67 augments macrophage infiltration into MRSA biofilms. (A &
B) Mice were sacrificed at day 3 post-infection, whereupon tissues surrounding infected catheters were subjected to immunofluorescence staining with Iba-1 (red) to identify macrophages and nuclear staining with DAPI (blue); asterisks represent the original location of the catheter, which is non-adherent to glass slides. (C & D) Macrophage infiltrates in tissues surrounding infected catheters from vehicle- or EP67-treated animals were quantitated by FACS to identify macrophage (F4/80+) infiltrates. Results are expressed as the percentage of cells after correction for isotype control staining (*, p < 0.05; ***, p < 0.001).
DETAILED DESCRIPTION
[0082] The disclosure provides compositions and methods that are useful in preventing the formation of biofilm infections and in treating existing biofilm infections. Disclosed herein are methods of administering cells such as macrophages, including previously activated macrophages, macrophages being activated simultaneously with administration, or macrophages activated in vivo post-administration, to prevent and/or treat biofilms. In one embodiment, peripheral blood monocytes are collected from a patient prior to surgery, such as one week prior to surgery. Macrophages are prepared from this sample and are activated using various activating agents, including but not limited to, peptidoglycan {e.g., S. aureus- derived peptidoglycan) and interferon-γ. The activated macrophages are then administered to the patient at the site of implantation to prevent biofilm formation. In another embodiment, activated macrophages are administered to a patient who already has an established biofilm to treat the biofilm infection. It is envisioned that one or multiple doses of activated
macrophages could be administered to treat an established biofilm. In addition, evidence documenting that activated macrophages reduce bacterial burdens on infected devices, supports embodiments where activated macrophages are used in combination with antibiotics. Another aspect of treating biofilms is interfering with the extensive fibrotic matrices that form, which represents a barrier to effective treatment. To overcome this barrier, it is envisioned that proteolytic enzymes or drugs are co-administered with activated macrophages. In another embodiment, one or more compounds or factors that are capable of activating macrophages in vivo is/are co-administered or administered pre- or post- administration of activated macrophages to further enhance macrophage activation.
[0083] The use of immune cell-based therapy using activated macrophages overcomes at least one of the aforementioned disadvantages of preventing or treating biofilms. The data disclosed herein demonstrate that macrophages are superior to chemicals, such as antibiotics, and to such physical processes as wound debridement, for effective clearance of biofilm- associated bacteria, such as biofilm-associated bacteria from infected medical devices, as well as the surrounding tissue. Importantly, macrophage therapy provides long-lasting protection. In one embodiment, only three activated macrophage cell treatments were required for sustained bacterial containment (out to two weeks post-infection). Multiple cell injections would be feasible from a clinical perspective, given the fact that patients undergoing orthopedic joint surgeries are hospitalized for a minimum of two-three days. Consequently, given the growing prevalence of nosicomial infections and biofilm
development, the increased use of procedures that can give rise to biofilm development, the
growing elderly population who will be receiving artificial implants and who exhibit progressively compromised immune responses, and the difficulties of treating and/or controlling bacteria within a biofilm, increasing demands will be placed on the use of macrophage-based immune cell therapy as a way of treating biofilms in both young and old.
[0084] The following definitions will be helpful in understanding the detailed description of the disclosure.
[0085] "Biofilm" or "biofilm infection" refers to an organized community of bacteria contained within a matrix or "slime," typically composed of bacterial polysaccharides, DNA, and proteins, within which is found at least one species of infectious microbe (e.g. , bacterium) and which typically exhibits a complex heterogeneous composition of microbes. Biofilms may exist within the body on natural surfaces (e.g. , the lining of bones and the heart) or inanimate objects inserted for therapeutic treatment (e.g., indwelling devices or artificial joints).
[0086] A "subject" is a human being or a veterinary animal, including but not limited to, a dog, a cat, a horse, a bovine (a cow or a bull), a sheep, a goat and a pig, as well as a zoo animal and a game animal.
[0087] "Subject-in-need" means a subject, such as a human patient or veterinary animal including but not limited to a dog, a cat, a horse, a bovine (a cow or a bull), a sheep, a goat and a pig, that is in need of prevention for a possible biofilm infection or is in need of treatment for an existing biofilm infection.
[0088] "Isolating," in the context of isolating a cell, is given its ordinary and accustomed meaning in the art of separating that cell from at least one other component with which the cell is normally associated in the body of a subject.
[0089] "Peripheral blood monocyte" is given its ordinary and accustomed meaning in the art of a white blood cell in peripheral blood that has a single nucleus and can take in (ingest or phagocytose) foreign material. A monocyte, like a macrophage, is a phagocyte. Under the proper conditions, a monocyte can differentiate into a macrophage.
[0090] "Culturing" is given its ordinary and accustomed meaning of incubating a cell in the presence of nutrients and under conditions permissive for continued viability and/or growth.
[0091] "Contacting" means removing any barrier to a coming together, a locating in the immediate vicinity of, or a touching, such as between a cell and a compound(s) such as an activating agent.
[0092] A "macrophage-activating agent" is one or a combination of compounds that induce an activated state or phenotype in a macrophage. Exemplary macrophage-activating agents include macrophage-activating peptides such as EP67 (SEQ ID NO: l), EP54 (SEQ ID NO:2), C5a65-74 (SEQ ID NO:4), and analogs and variations thereof, interleukin-ΐβ as well as combinations of compounds selected from the group consisting of interferon-γ and tumor necrosis factor (TNF), and interferon-γ and an inducer of TNF.
[0093] A "macrophage" is a phagocyte having a single nucleus that ingests or
phagocytoses foreign material, such as infectious microbes, including infectious bacteria. Macrophages are derived from monocytes by differentiation.
[0094] An "activated macrophage" is a macrophage that exhibits an activated phenotype relative to a resting macrophage. The activated macrophage becomes larger, more motile, adherent, expresses more MHCII proteins on its surface, contains more lysosomes and lysosomal enzymes, and secretes a variety of substances including interleukin-1 and tumor necrosis factor, relative to a resting macrophage. Again relative to a resting macrophage, an activated macrophage has increased phagocytic activity and increased bacterial killing via reactive oxygen intermediates and lysosomal enzymes. Unless otherwise noted, an activated macrophage as used herein is an "Ml -activated macrophage" or a "classically activated macrophage."
[0095] A "prophylactically effective amount" is an amount effective in prevention of a disease, disorder or condition and is determined on an individual basis with consideration given to well-known factors such as weight, gender, general health, and the like.
[0096] A "therapeutically effective amount" is an amount effective in treatment of a disease, disorder or condition and is determined on an individual basis with consideration given to well-known factors such as weight, gender, general health, and the like.
[0097] "At risk of developing a biofilm infection" means a subject exhibiting a greater risk of developing a biofilm infection than the average subject, such as a subject with a compromised immune system, a subject with a documented history of recurrent bacterial infections, or a subject harboring a surgical implant such as an artificial component of a joint (e.g., hip, knee, ankle, shoulder, elbow, wrist) or an implanted pacemaker.
[0098] A "veterinary animal" means any animal conventionally receiving treatment from a veterinarian and having an immune system,, including but not limited to a dog, a cat, a horse, a bovine (a cow or a bull), a sheep, a goat a pig, a zoo animal, and a game animal.
[0099] An "immunomodulatory cytokine" is a cytokine exhibiting the capacity to modulate an immune response under at least one set of conditions. Exemplary cytokines are proinflammatory cytokines, such as interleukin-6, interleukin-12, monocyte chemoattractant protein- 1 (MCP-1), monokine-induced by IFN-γ (MIG), interferon-regulatory protein- 10 (IP- 10), macrophage inflammatory protein-2 (MIP-2), and keratinocyte chemoattractant (KC). In addition, macrophage colony-stimulating factor (M-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) are contemplated as these molecules encourage macrophage and neutrophil expansion at the site of biofilm infection, whereupon proinflammatory cytokines would be present to activate these cells to trigger their bactericidal activity.
[0100] A "surgical implant" is an artificial implant or device placed within the body of a subject and includes, but is not limited to, an artificial joint component (an artificial hip component, an artificial knee component, an artificial ankle component, an artificial elbow component, an artificial shoulder component, and an artificial wrist component), a cranial plate (natural or artificial ), a dental implant, and a subcutaneous pacemaker.
[0101] "Catheter" is given its ordinary and accustomed meaning of a hollow, typically flexible, tube for insertion into a body cavity, duct, or vessel to allow the passage of fluids or to distend a passageway.
[0102] A "proteolytic enzyme" is an enzyme that catalyzes the proteolysis of at least one protein, polypeptide or peptide, and includes, but is not limited to, MMP-1, MMP-2, MMP-7, MMP-9, MMP-12 and collagenase.
[0103] An "EP67 peptide" is a peptide functioning as a C5a receptor agonist and exhibits the amino acid sequence set forth in SEQ ID NO: 1, or a variant or derivative of that sequence. An "EP54 peptide" is a peptide functioning as a C5a receptor agonist and exhibits the amino acid sequence set forth in SEQ ID NO: 2, or a variant or derivative of that sequence. An "C5a65_74" is a peptide functioning as a C5a receptor agonist and exhibits the amino acid sequence set forth in SEQ ID NO:4, or a variant or derivative of that sequence.
[0104] "Inflammatory disorder" is a disorder characterized by an inflammatory response causing redness, swelling, heat and/or pain. Inflammatory disorders include, but are not limited to, atherosclerosis, infectious complications of cystic fibrosis, endocarditis, chronic
sinusitis, nasal polyposis, chronic otitis media, leptospirosis, kidney stones, osteomyelitis, necrosis of the jaw, a urinary tract infection, a bladder infection, and a chronic wound.
[0105] Terms used in this disclosure that are not expressly defined herein are given their ordinary and accustomed meanings in the art.
[0106] With these terms defined, disclosure of the compositions and methods for preventing and treating biofilm infection will be straightforward. Methods for prophylactic immune-cell therapy comprise administering a prophylactically effective amount of an immune cell, such as an activated macrophage. In some embodiments, the method involves administering a therapeutically effective amount of autologous activated macrophages.
These cells must be obtained and, in some embodiments, activated. For example, a blood sample is obtained from a patient prior to implant placement, e.g., approximately one week prior to the medical device placement procedure. The blood is sent to a laboratory where immune stimulation would take place, which would allow for quality control and
standardization. Once cells are propagated, they are delivered to the recipient for local treatment at the implant site, making the cells immediately available to respond to any potential bacterial contaminant introduced during the surgical procedure (most commonly via skin microflora).
[0107] Macrophage-based immune-cell therapy is not only effective in treating/controlling microbe, e.g., bacterial, burdens associated with biofilms, it has the added advantage of doing so by utilizing the host's own innate immune cells. In relying on a subject's own innate immune cells, the methods of the disclosure eliminate or minimize selection pressures imposed directly on the microbe, e.g., bacteria, and decrease the likelihood of developing resistant strains. In addition, minimal side effects would be expected because autologous cells that won't survive long-term (i.e., no longer than 7-14 days) are being introduced.
[0108] Biofilms have been shown to skew the immune response to favor anti-inflammatory and pro-fibrotic pathways, which likely contributes to biofilm persistence. To overcome this immune deviation and provide a treatment strategy for biofilm infections, antimicrobial activity has been augmented through the local administration of classically activated Ml macrophages, optionally supplemented by administration of a macrophage-activating peptide such as EP67, EP54, C5a65_74, or their analogs or variants, provided such peptides retain the capacity to invoke macrophage pro-inflammatory responses. Early administration of Ml- activated macrophages limited biofilm formation and treatment of established biofilm infections with activated macrophages also significantly reduced catheter-associated biofilm
burdens compared to antibiotic treatment. Several pro-inflammatory mediators were significantly elevated in tissues surrounding biofilm infections following Ml -activated macrophage treatment, which may account for the observed antimicrobial effects.
Furthermore, a role for macrophage pro-inflammatory activity was demonstrated by showing that transfer of neutrophils or macrophages from MyD88 knockout mice had minimal impact on biofilm growth in vivo. Based on this evidence, a therapeutic strategy has been developed to prevent and/or treat biofilm infections, including S. aureus (e.g., MRS A) device- (e.g., implant-) or catheter-associated biofilm infections, by exploiting macrophage activation.
[0109] These macrophage -based approaches boost pro-inflammatory activity, which is useful in preventing biofilm infection and in treating existing biofilm infections. These approaches are expected to be useful when administered to patients undergoing orthopedic surgery or other device-related implant procedures to prevent nosocomial infections, particularly for individuals who are at high-risk for developing infectious complications. Ml-activated macrophage therapy provides a new strategy for preventing and treating biofilms, optionally supplemented with macrophage-activating agents such as macrophage- activating peptides like EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), or C5a65-74 (SEQ ID NO:4), or their analogs or variants. In addition, the activated macrophage therapies disclosed herein are suitable for combination with antibiotics, particularly for surgical-implant patients who are unable or unwilling to undergo additional surgery to manage the infection and thereby maintain the implanted device. The significance of this approach is even more pronounced against the backdrop of the rapidly increasing elderly population, which grows progressively less immunoresponsive and will be the primary recipients of more permanent artificial implants, such as hips and knees.
[0110] S. aureus (e.g., MRS A) is a frequent etiological agent of biofilm infections on indwelling devices and orthopedic implants. The therapeutic potential of activated macrophages is supported by results demonstrating that early treatment with proinflammatory Ml-activated macrophages significantly limited S. aureus biofilm growth in vivo and provided long-term protection from biofilm colonization. Likewise, activated macrophages were also effective at reducing S. aureus burdens in established biofilms by 100-fold. Without wishing to be bound by theory, this reduction in device- associated bacterial burdens may result from dispersal of organisms from the biofilm which, in turn, would restore the metabolic activity of the bacteria. By extension, it is expected that Ml- activated macrophages will display synergy with antibiotics to maximize biofilm clearance,
because these organisms are no longer foreign body-associated and, thus, more amenable to antibiotic action.
[0111] There are several advantages associated with the activated macrophage approach to preventing or treating biofilm infections. Monocytes can be easily collected from patient's blood by elutriation, expanded, induced to differentiate into mature macrophages, and activated in vitro for introduction into the surgical site for infection prophylaxis. A relatively wide range of activated macrophage dosages are suitable for use in the prophylactic and therapeutic methods and uses according to the disclosure. Exemplary macrophage dosages are 105, 106, 107, and 108 activated macrophages per dose. Further, the methods and uses of the disclosure are amenable to a variety of dosing schedules, which can readily be determined by those of skill in the art. By way of example, the activated macrophage can be delivered in one to five doses, over one to five days. For multiple administrations, it is contemplated that administered doses may be placed in a region approximately surrounding the expected or actual location of a potential or existing biofilm.
[0112] Modulating macrophage effector responses represents a promising therapeutic avenue for the prevention and/or treatment of device-associated biofilm infections. Although initial efforts have been focused on demonstrating the therapeutic methods on MRSA biofilms, additional advantages are that activated macrophages possess potent anti-microbial activity and are capable of eliminating a wide spectrum of infectious pathogens based on their immunological characteristics. Stated in the alternative, the disclosed methods rely on elements of the immune response for efficacy. Any infectious agent will be perceived as foreign by the host's immune system, including all microbes such as bacteria, including all infectious gram-positive and gram-negative bacteria. This is far superior to antibiotics, which are typically only active against specific bacterial classes and are ineffective at clearing biofilm infections. Also of significance is that bacteria are less likely to develop mutations to escape immune-directed therapies, because activated macrophages are a primary source of microbial killing in the immune system and most pathogens have not evolved resistance to the effects of the immune system.
[0113] The disclosure provided herein establishes a previously unappreciated role for Ml- activated macrophages in biofilm containment and bacterial clearance. By extension, it is not unexpected that biofilms such as MRSA biofilms have the capacity to thwart this response by skewing macrophages away from a pro-inflammatory Ml to an anti-inflammatory M2 phenotype, which could ensure biofilm persistence in an immunocompetent host. The implementation of activated macrophage transfer therapy would allow macrophages to be "on
site" to deal with possible contamination of the implant from normal bacterial flora on the skin. While conventional antibiotics are ineffective in treating/controlling biofilms, they are commonly used to control bacteria that escape the biofilm matrix thereby preventing their colonization of other tissue sites. Such uses of antibiotics impose mutational pressures on the bacteria and increase the risk of selecting antibiotic -resistant strains. Macrophage-based immune cell therapy not only appears to be effective in treating/controlling microbial, e.g., bacterial, burdens associated with biofilms, it has the added advantage of doing so by utilizing the hosts own innate immune cells, thus eliminating or minimizing mutational pressures imposed directly on the microbes, such as bacteria, and decreasing the likelihood of promoting the growth of resistant strains.
Organisms Associated with Biofilm Infections
[0114] Investigations over the past 20-30 years have revealed that many, if not most, infections of humans and veterinary animals, including pets and domesticated farm animals, are caused by organisms capable of forming biofilm infections. Accordingly, the
compositions and methods of the disclosure are contemplated as suitable for preventing or treating biofilm infections caused by, or associated with, any known infectious organism, such as microbes, including gram-positive and gram-negative bacteria, fungi, and yeast. Exemplary infectious microbes causing or associated with biofilm infections include gram- positive bacteria such as Staphylococcus aureus, Enterococcus faecalis and Enterococcus faecium and gram-negative bacteria such as Escherichia coli, Pseudomonas aeruginosa, Pseudomonas fluorescens, and Haemophilus influenza,. Other exemplary microbes causing or associated with biofilm infections include Leptospira species such as Leptospira interrogans, and the yeast, Candida albicans. These exemplary organisms are representative of the hundreds and hundreds of microbes capable of causing or being associated with biofilm infections. For example, dental plaque is known to involve a biofilm infection associated with the teeth of humans and veterinary animals. Investigations have shown that biofilms associated with dental plaque contain over 500 species of microorganisms. The disclosure contemplates the prevention or treatment of biofilm infections caused by or associated with each of these organisms, and other microbes capable of causing or being associated with biofilm infection. The biofilm- associated microbes of dental plaque are provided to illustrate the number and diversity of microbes associated with biofilm infections amenable to prevention or treatment with the disclosed compositions and methods, even if the administration of cells such as activated macrophages to prevent or treat biofilm infections
associated with dental plaque is not contemplated as a preferred embodiment of the disclosed subject matter.
Diseases, Disorders, and Conditions Associated with Biofilm Infection
[0115] A variety of diseases, disorders and conditions are known to be associated with biofilm infection, and the compositions and methods disclosed herein are contemplated as suitable for preventing or treating any of these biofilm infections. Exemplary diseases associated with biofilm infection include, but are not limited to, atherosclerosis, infectious complications of cystic fibrosis, endocarditis, chronic sinusitis, nasal polyposis, chronic otitis media or middle ear infections, leptospirosis, kidney stones, osteomyelitis, necrosis of the jaw, urinary tract infections, and bladder infections. Exemplary disorders include chronic wounds, and exemplary conditions include subjects, e.g., human patients, that have or are receiving a prosthetic device such as an artificial component of a joint (e.g., a component of a hip, knee, ankle, elbow, shoulder, or wrist joint), or a temporary or permanent catheter (e.g., urinary, intravenous or intraarterial catheter). With respect to prosthetic implants and catheters, moreover, it is known that typically non-pathogenic microbes, e.g., benign members of the normal skin flora, can form biofilm infections in association with these artificial surfaces, and the prevention or treatment of such biofilm infections is also contemplated. In addition, infectious diseases of veterinary animals are associated with biofilm infection and are comprehended as suitable for prevention or treatment using the disclosed compositions and methods. Given the prevalence and wide variety of biofilm infections, the disclosed compositions and methods are contemplated as suitable for preventing or treating any known biofilm infection.
Activating Agents
[0116] Activating agents suitable for use in activating a macrophage include any compound, or combination of compounds, known to activate macrophages. For ease of discourse, compounds effective in activating macrophages only in combination with at least one other compound are referred to herein as activating agents. Exemplary activating agents include macrophage- activating peptides such as EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), C5a65-74 (SEQ ID NO:4) and their analogs and variants, interleukin-ΐβ as well as
combinations of compounds such as interferon-γ and tumor necrosis factor (TNF), interferon- γ and an inducer of TNF. Exemplary inducers of TNF are peptidoglycan and
lipopolysaccharide. Yet other embodiments of the method further comprise delivering to the patient in need an effective amount of a pro -inflammatory compound, such as interleukin-6, interleukin-12, monocyte chemoattractant protein-1 (MCP-1), monokine-induced by IFN-γ
(MIG), interferon-regulatory protein-10 (IP-10), macrophage inflammatory protein-2 (MIP- 2), and keratinocyte chemoattractant (KC). In addition, macrophage colony- stimulating factor (M-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) are contemplated as useful in the methods and uses according to the disclosure to encourage macrophage and neutrophil expansion at the site of biofilm infection, whereupon proinflammatory cytokines would be present to activate these cells to trigger their bactericidal activity. A cell, e.g., a macrophage, is exposed to one or more activating agents ex vivo or in vivo, and prior to, concurrent with, or following administration of the cell to the subject, such as a human patient or veterinary animal.
Administered Cells
[0117] A cell to be administered to a subject, such as a human patient or veterinary animal, is an activated macrophage or a macrophage. Each of these cell types results in a
prophylactically and/or therapeutically effective active macrophage in vivo, at the site or in the region where a biofilm infection is to be prevented and/or treated. Relative to the recipient human patient or veterinary animal, a syngeneic cell is utilized, such as an autologous cell. Autologous cells are administered in order to minimize complicating influences from the host immune system and cell rejection.
[0118] The following examples illustrate embodiments of the disclosure. Example 1 provides the materials and methods used in the experiments disclosed herein. Example 2 established that activated macrophages are bactericidal towards biofilm-associated S. aureus in vitro. Example 3 showed that biofilm infections alter the gene expression profile of administered macrophage. Example 4 demonstrated that activated macrophages prevented the formation of a biofilm infection (i.e., MRS A infection) in vivo. Example 5 established that activated macrophages reduced the bacterial burden of biofilm infections, while establishing a pro-inflammatory microenvironment at the biofilm site. Example 6 showed that administering a C5aR agonist, e.g., EP67, successfully prevented or inhibited formation of a biofilm infection, i.e., a MRS A biofilm infection. Without wishing to be bound by theory, this effect is believed to be due to the C5aR agonist reprogramming endogenous macrophages from an M2- to a pro-inflammatory Ml-activation state, which would complement or enhance activated-macrophage therapy. Example 7 established that activated macrophages eliminated or reduced the bacterial burden of a catheter-associated biofilm infection in vitro (i.e., MRS A infection).
Antibiotics
[0119] The compositions, methods and uses according to the disclosure are generally drawn to macrophages that are or can be activated for use in preventing or treating biofilm infections. As noted herein, the macrophages can be combined with macrophage- activating agents, including macrophage-activating peptides such as EP67, EP54 and C5a65_74. In addition, the macrophages can be combined, alone or with a macrophage-activating agent, with one or more antibiotics. Further, the macrophages, with or without a macrophage- activating agent, can be administered separately or jointly with one or more antibiotics in the prophylactic or therapeutic methods and uses according to the disclosure. Any known antibiotic is expected to be suitable for these methods and uses. Suitable classes of antibiotics include , but are not limited to, aminoglycosides, ansamycins, carbacephems, carbapenems, cephalosporins, glycopeptides, lincosamides, lipopeptides, macrolides, erythromycins, monobactams, nitrofurans, penicillins, penicillin combinations, antimicrobial polypeptides, quinolones, sulfonamides, tetracyclines and antimycobacterials.
[0120] In some embodiments, the antibiotic is selected from the group consisting of amikacin, gentamicin, kanamycin, neomycin, roxithromycin, netilmicin, tobramycin, paromomycin, geldamycin, herimycin, loracarbef, ertapenem, doripenem, imipenem or cilastatin, meropenem, cefadroxil, cefazolin, cefalotin or cefalothin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefonicid, cefixime, cefdinir, cefditoren, cefotaxime, cefetamet, cefoperazone, cefpodoxime, ceftazidime, ceftibuten, ceftriaxone, cefepime, ceftaroline or fasamil, ceftobiprole, cephalothin, cefmetazole, cefotetan, ceftizoxime, cefsulodin, cephalexin, cinoxacin; teicoplanin, vancomycin, telavancin;
clindamycin, lincomycin, daptomycin, azithromycin, clarithromycin, erythromycin, erythromycin estolate, erythromycin ethyl succinate, erythromycin glucoheptonate, erythromycin lactobionate, erythromycin stearate, roximthromycin, trolandomycin, telithromycin, spectinomycin, spriamycin, Aztreonam, furazolidone, nitrofurantoi, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin G, penicillin V, piperacillin, ticarcillin, amoxicillin and clavulanate, ampicillin and sulbactam, piperaccillin and tazobactam, ticarcillin and clavulanate; bacitracin, colistin, polymyxin B, ciprofloxacin, enoxacin, gatifloxacin, levofloxacin, lomefloxacin, moxifloxicin, ofloxacin, nalidixic acid, norfloxacin, ofloxicin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide,
sufonamidochrysoidine, sulfacetamide, sufadiazine, silver sulfadiazine, sulfamethizole, sulfamethoxazole, sulfanilamide, sulfasalazine, sulfisoxazole, trimethoprim-
sulfamethoxazole, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol, ethionamide, isoniazid, rifampicin or rifampin, rifabutin, rifapenine, streptomycin, arshenamine, chloramphenicol, fosfomycin, fusidic acid, linezolid, metronidazole, mupirocin, platensimycin, quinupristin or dalfopristin, rifaximin, thiamphenicol, tigercycline, tinidazole, trimethoprim, fleroxacin, nitrofurantoin, coamoxiclavuanate, and their salts, acids, and bases.
Example 1
Materials and Methods
[0121] Animals. Male C57BL/6 mice (6-8 weeks old) were obtained from Charles River Laboratories (Frederick, MD) and MyD88 KO animals were purchased from the Centre de La Recherche Scientifique and have been backcrossed with C57BL/6 mice for over 10 generations (36-38). Mice were housed in restricted-access rooms equipped with ventilated microisolator cages and maintained at 21 °C under a 12-hour light: 12-hour dark cycle with ad libitum access to water (Hydropac™; Lab Products, Seaford, DE) and Teklad rodent chow (Harlan, Indianapolis, IN).
[0122] S. aureus strains. For in vitro biofilms, the S. aureus MRSA strain USA300 LAC was used and, for confocal microscopy studies, was transformed with the plasmid pCMl l to express GFP driven by the sarA PI promoter (USA300 LAC-GFP) and plasmid expression was maintained with erm selection (10 μg/ml) (15, 39). For in vivo biofilm infections, the bioluminescent MRSA USA300 LAC::lux strain was used as previously described (15, 40, 41).
[0123] In vitro S. aureus biofilms. Static biofilms were generated as previously described (15). Briefly, sterile two-well glass chamber slides (Nunc, Rochester, NY) were treated with 20% human plasma in sterile carbonate-bicarbonate buffer (Sigma- Aldrich, St. Louis, MO) overnight at 4°C to facilitate bacterial attachment (40). Overnight cultures of MRSA
USA300 LAC or USA300LAC-GFP were prepared in macrophage medium, supplemented with 10 mg/ml erm for the GFP strain. The following day, plasma coating buffer was removed and chambers were inoculated with bacteria, diluted in mammalian cell culture medium (RPMI-1640 supplemented with 10% fetal bovine serum) to an OD600 of 0.050 in 2 ml, and incubated at 37°C under static aerobic conditions for a period of up to 6 days.
Medium was carefully replenished every 24 hours to prevent disruption of the biofilm structure.
[0124] Macrophage or neutrophil co-culture with S. aureus biofilms in vitro. MRSA USA300 LAC-GFP static biofilms and bone marrow-derived macrophages (BMDMs) from either WT or MyD88 KO mice were prepared as previously described (42, 43). Neutrophils were recovered from the bone marrow utilizing a three-layer Percoll gradient of 78%, 69%, and 52% Percoll (Amersham Pharmacia Biotech, Uppsala, Sweden) diluted in PBS (100% Percoll = 9 parts Percoll and 1 part 10X PBS), and centrifuged (1500 g, 30 minutes, room temperature without braking) as previously described (44). Neutrophils were further purified using a Miltenyi Anti-Ly6G MicroBead Kit (Miltenyi Biotec, Cambridge, MA) according to the manufacturer's instructions. Briefly, 50 μΐ Anti-Ly-6G-Biotin microbeads were added to 10 total bone marrow cells in 200 μΐ MACs buffer, and samples were vortexed and incubated for 10 minutes at 4°C. Subsequently, 150 μΐ MACs buffer and 100 μΐ of anti-biotin MicroBeads per 10° total cells were added and cells were incubated for 15 minutes at 4°C. After washing, labeled cells were then positively selected by magnetic separation using MS MACs columns and a MACs separator (Miltenyi Biotec). The column was washed three times with 500 μΐ of MACs buffer. After the column was removed from the MACs separator, the positive fraction containing Ly-6G neutrophils was eluted and collected in 1 ml of MACs buffer. BMDM and neutrophils were labeled with either 5 μΜ CellTracker Orange (CTO) or CellTracker Blue (CTB) (both from Molecular Probes, San Diego, CA) depending on the experimental setup. Co-cultures of mature S. aureus biofilms (six-day-old biofilms) with 10 neutrophils, non-activated macrophages, or activated macrophages (pre-treated with 10 ng/ml IFN-γ and 10 μg/ml S. aureus-dedved peptidoglycan (i.e., PGN) for 6 hours) were established at 37°C under static aerobic conditions and imaged by confocal microscopy using a Zeiss 510 META laser scanning microscope (Carl Zeiss, Oberkochen, Germany) at 2 and 24 hours following the addition of immune cells. Neutrophil or macrophage-biofilm co- cultures were harvested at 24 hours after immune cell addition by mechanical dissociation followed by sonication, whereupon bacterial enumeration was performed by serial dilution on TSA plates supplemented with 5% sheep blood (Hemostat Laboratories, Dixon, CA).
[0125] To determine the effect of biofilm exposure on macrophage activation, as measured by cytokine secretion, a separate set of experiments was performed. Co-cultures of mature S. aureus biofilms with 10 non-activated CTB-labeled macrophages were established at 37°C under static aerobic conditions for 2 hours following the addition of immune cells, as described above. To separate macrophages from the biofilm, FACs purification was performed using a BD FACSAria (BD Biosciences, San Diego, CA) after mechanical dissociation of the macrophage biofilm co-culture. Macrophages were distinguished from
bacteria based on their size and CTB staining. Following macrophage recovery by FACS, 200,000 cells/well were plated in a 96-well polystyrene microtiter plate (BD Biosciences) and incubated for 24 hours at 37°C with 5% C02. After 24 hours, supernatants were collected to compare the production of inflammatory mediators associated with biofilm- versus planktonic-exposed macrophages utilizing sandwich ELISA kits to quantitate TNF-a, IL-10 (OptiEIA; BD Biosciences, Franklin Lakes, NJ), and IL-1RA (DuoSet; R & D Systems, Minneapolis, MN) per the manufacturers' protocols.
[0126] Mouse model ofS. aureus catheter-associated biofilm infection. S. aureus biofilm infections were performed as previously described (15, 40, 41). Briefly, a small s.c. incision was made in the mouse flank under avertin anesthesia and a blunt probe was used to create a pocket for insertion of a sterile 14-gauge teflon intravenous catheter 1 cm in length (Exel International, St Petersburg, FL). The incision was sealed using Vetbond Tissue Adhesive (3M, St. Paul, MN) and 103 cfu MRSA USA300 LAC::lux in 20 μΐ of sterile PBS was slowly injected through the skin into the catheter lumen to initiate infection. The health status of each mouse was regularly monitored throughout the course of infection and any moribund animals were immediately euthanized.
[0127] Immune cell administration into biofilm infections in vivo. Mice received injections of 106 neutrophils or activated WT or MyD88 KO macrophages (10 ng/ml IFN-γ + 10 μg/ml S. aureus-denved PGN) beginning at 12 hours, with repeat administration at 24 and 48 hours post-infection. Cells were introduced at four distinct sites surrounding infected catheters to ensure maximal immune cell access to the developing biofilm. A separate cohort of animals received activated macrophages or vehicle until 48-hours post-infection, whereupon no additional cells were administered to determine the longevity of early macrophage treatment on inhibiting biofilm growth until day 14 after infection. In some experiments, the numbers of neutrophils injected was increased by 1-log (i.e., to 10 ) to confirm that these cells did not exhibit any beneficial effects on biofilm clearance. For established biofilm experiments, mice were infected with 10 colony forming units (cfu) of MRSA USA300 LAC in the lumen of surgically implanted catheters to establish biofilm infection and on days 7 and 9 post-infection, animals received vehicle control (PBS), 106 activated macrophages, or antibiotic treatment consisting of rifampicin (25 μg/ml) and daptomycin (5 μg/ml). In a separate experiment, animals received vehicle or 10 activated macrophages loaded with Quantum dots (Molecular Probe) at 24 hours post-infection for the prophylactic paradigm or at 7 days post-infection for established biofilm paradigms. The longevity of the administered macrophages was monitored daily in the same cohort of mice
using an In vivo Imaging System (IVIS Spectrum; Caliper Life Sciences, Hopkinton, MA) under isoflurane anesthesia until 5 days after administration, at which point animals were sacrificed to determine absolute bacterial burdens associated with catheters and surrounding tissues. In some experiments, antibiotic treatment, consisting of rifampicin (25 μg/ml) and daptomycin (5 μg/ml) given twice daily starting 12 hours post-infection and continuing until 48 hours post-infection, was used to compare experimental treatments with the standard of care typically administered in the clinic, which does not exhibit any beneficial effects on biofilm clearance.
[0128] EP67 Peptide-Activated Macrophage Combined Treatment. To demonstrate that macrophage-activating agents, such as macrophage-activating peptides, were suitable for combination with activated macrophage therapy in preventing or treating biofilm infection, the effect of administering a macrophage-activating peptide alone was assessed. Animals were initially treated locally at the site of S. aureus infection with EP67 or a biologically inactive scrambled derivative (sEP67) having the sequence set forth in SEQ ID NO:3. 200 μg of either peptide was injected into the catheter at the time of infection (time 0) followed by 800 μg peptide divided into four different injection sites surrounding the catheter 24 and 48 hours post-infection.
[0129] Recovery of Catheter and Associated Tissues for S.aureus enumeration. At day 3 or 14 following infection, animals were sacrificed and catheters removed and placed in 1 ml of PBS for sonication to dissociate bacteria from the catheter surface. Tissues surrounding infected catheters were also collected, weighed, minced, and disrupted in 500 μΐ
homogenization buffer [PBS supplemented with 100 μΐ RNasin and a protease inhibitor tablet (Roche Diagnostics, Indianapolis, IN)] using a Bullet Blender (Next Advance Inc., Averill Park, NY). Bacterial titers associated with catheters and surrounding tissues were
enumerated using TSA plates supplemented with 5% sheep blood and were expressed as Logio cfu/ml for catheters or Logio cfu/g wet tissue weight.
[0130] MILLIPLEX multi-analyte bead array. To evaluate the ability of Ml -activated macrophages to alter the inflammatory milieu associated with MRS A biofilm infections, a custom-designed mouse microbead array was utilized according to the manufacturer' s instructions (MILLIPLEX; Millipore, Billerica, MA), which detects the following
inflammatory mediators: IL-la, IL-Ιβ, TNF-a, IFN-γ, IL-6, IL-9, IL-10, IL-12p40, IL-12p70, IL-15, IL-17, CXCL1, CXCL2, CXCL9, CXCL10, CCL2, CCL3, CCL4, and CCL5. Results were analyzed using a Bio-Plex workstation (Bio-Rad, Hercules, CA) and normalized to the amount of total protein recovered to correct for differences in tissue sampling size.
[0131] Flow cytometry. Tissues surrounding biofilm-infected catheters were collected and processed for flow cytometry as previously described (15). Cells were stained with the following antibodies: F4/80-PE-Cy7 and Ly6G-AF700 (all from BD Biosciences).
Macrophages were identified as F4/80+ and Ly6G", while neutrophils were identified as F4/80" and Ly6G+.
[0132] Immunofluorescence staining and confocal microscopy. Tissues surrounding infected catheters were fixed in 10% formalin and embedded in paraffin, whereupon 10 μπι thick sections were deparaffinized in xylene and a graded series of alcohols followed by antigen retrieval, as previously described (15). Sections were processed for
immunofluorescence staining using primary antibodies specific for Iba-1 (macrophage- specific marker; Biocare Medical, Concord, CA) and arginase-1 (Santa Cruz Biotechnology, San Diego, CA), followed by donkey anti-rabbit-FITC or -biotinylated secondary antibodies (Jackson ImmunoResearch Laboratories, West Grove, PA), and a streptavidin-594 conjugate for the latter (Invitrogen, Carlsbad, CA). Confocal imaging was performed using a Zeiss 510 META laser scanning microscope with staining specificity confirmed by tissues incubated with secondary antibodies only. Quantitation of arginase-1 or Iba-1 fluorescence was calculated from at least 10 random fields of view using Axio Vision software 4.8 (Carl Zeiss, Oberkochen, Germany).
[0133] Statistical Analysis. Significant differences between experimental groups were determined using an unpaired two-tailed student's i-test in GraphPad Prism 4 (La Jolla, CA). For all analyses, a p-value of less than 0.05 was considered statistically significant.
[0134] Ethics Statement. This study was conducted in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was approved by the Institutional Animal Care and Use Committee of the University of Nebraska Medical Center (Approval ID: 09-049-00-EP). All surgery was performed under avertin anesthesia, and every effort was made to minimize suffering.
Example 2
[0135] Activated macrophages exhibit S. aureus biofllm bactericidal activity in vitro.
MRSA biofilms are capable of attenuating traditional pro-inflammatory responses, explaining, in part, why these infections persist in an immunocompetent host (15). To determine whether macrophages that were preprogrammed towards a pro-inflammatory Ml phenotype were capable of overcoming the immune inhibitory biofilm environment,
macrophages were stimulated with S. aureus-derived peptidoglycan (PGN) and interferon- gamma (IFN-γ) for 6 hours prior to their addition to MRSA biofilms or planktonic cultures as a control. Importantly, activated macrophages were capable of phagocytosing biofilm- associated bacteria, which resulted in significant reductions in bacterial burdens following a 24 hour co-culture period (Fig. 4B). The ability of activated macrophages to phagocytose biofilm-associated bacteria and reduce bacterial burdens required MyD88-dependent signals, because MyD88 KO macrophages treated with PGN + IFN-γ had no impact on biofilm growth (Fig. 4B).
[0136] To compare the efficacy of macrophages versus neutrophils in regulating MRSA biofilm formation, neutrophils were isolated from murine bone marrow and co-cultured with biofilms. Unlike macrophages, neutrophils were not pre-activated prior to biofilm addition, because this would lead to rapid degranulation and reduced cell viability. Interestingly, neutrophils were able to phagocytose MRSA biofilms, yet this did not translate into reduced bacterial numbers (Fig. 4B), revealing disconnect between the two processes. This may result from additional virulence determinants released by S. aureus during biofilm growth, because the organism is known to produce numerous factors that interfere with neutrophil function/survival (20-22). On a comparative basis, biofilm formation did afford some protection against phagocytic uptake compared to planktonic growth conditions, because both macrophages and neutrophils actively phagocytosed planktonic S. aureus but were less capable of internalizing biofilm-associated bacteria (Fig. 4A).
Example 3
[0137] Biofilms alter macrophage activation profiles. S. aureus biofilms skew
macrophage gene expression towards an alternatively activated (M2) phenotype compared to planktonic bacteria (15, 23). To determine whether biofilms influence macrophage cytokine secretion profiles, macrophages were exposed to S. aureus biofilms or planktonic bacteria for 2 hours, separated from bacteria by FACS and incubated at 37°C for 24 hours, at which point supernatants were collected to determine cytokine secretion profiles. Macrophages co- cultured with biofilms demonstrated a decreased cytokine secretion profile with reductions in TNF-a, IL-10 and IL-1RA expression compared to macrophages exposed to planktonic bacteria (Fig. 5). These results further establish that biofilm growth dampens the classic Ml macrophage response (24).
Example 4
[0138] Introduction of activated macrophages prevents MRSA biofilm formation in vivo.
Based on the in vitro studies demonstrating the ability of Ml -polarized macrophages to phagocytose biofilm-associated S. aureus and reduce bacterial burdens, experiments to determine whether this would translate in vivo to overcome the immune attenuation observed during biofilm growth were designed and undertaken. These experiments utilized a mouse model of MRSA catheter-associated biofilm infection that limits macrophage invasion into biofilms and skews cells towards an alternatively activated M2 phenotype (15). Initially employed was a paradigm where activated cells were administered beginning at 12 hours following MRSA infection, with repeat injections occurring at 24 and 48 hours after bacterial exposure. Importantly, the introduction of activated macrophages directly into the biofilm infection site resulted in significant reductions in bacterial burdens on both infected catheters and in surrounding tissues at day 3 post-infection (Fig. 6A & B). More importantly, this early intervention with activated macrophages led to long-term inhibition of biofilm formation, since catheters and surrounding tissues showed little/no evidence of biofilm growth at day 14 without any additional macrophage treatment (Fig. 7). Similar to the in vitro studies, MyD 88 -dependent mechanisms played an important role in Ml macrophage- mediated biofilm clearance, because macrophages from MyD88 KO mice did not
demonstrate any efficacy in controlling biofilm burdens on either infected catheters or surrounding tissues (Fig. 8). In addition, neutrophils had no impact on biofilm formation when injected at 106 cells per administration (Fig. 6), and even when the number of injected cells was increased to 10 per administration, which confirmed the in vitro findings and the fact that neutrophils do not significantly infiltrate catheter-associated MRSA biofilms.
[0139] MRSA biofilms attenuated the expression of numerous pro-inflammatory mediators compared to a sterile foreign body (15, 23). The introduction of Ml-activated macrophages led to increases in CXCL9, CCL5, and IFN-γ expression within biofilm-infected tissues (Fig. 9), revealing successful re-direction of the macrophages towards a pro-inflammatory milieu. The pro-inflammatory activity of activated macrophages is expected to play an important role in limiting biofilm growth.
Example 5
[0140] Introduction of Ml-activated macrophages for the treatment of established MRSA biofilm infections. Based on the efficacy of the early Ml macrophage treatment paradigm, it was next determined whether this would extend to attenuate bacterial growth in established MRSA biofilm infections. A strategy similar to the early treatment regimen for macrophage administration was employed, except that activated cells were initially given at
day 7 following S. aureus infection, with a repeat injection occurring at day 9. Similar to the early treatment paradigm, the introduction of activated macrophages directly into the biofilm infection site led to significant reductions in bacterial burdens on infected catheters at day 10 post-infection, although no effect was seen in surrounding tissues (Fig. 10A and B, respectively). In contrast to macrophage delivery, antibiotic treatment had no effect on biofilm formation (Fig. 10). Interestingly, Ml -activated macrophage administration attenuated arginase-1 expression within the surrounding tissue, indicating that macrophages were able to modify the local tissue environment to favor a pro-inflammatory response (Fig. 11). Likewise, the introduction of Ml -activated macrophages into established biofilms augmented CXCL9, CXCL2, IL-17, and IL-6 expression within established biofilm-infected tissues (Fig. 9D-G), again revealing the successful re-direction of macrophages towards a pro-inflammatory milieu. To establish the longevity of Ml -activated macrophages after injection into the site of biofilm infection, 10 Ml-activated macrophages were labeled with near-infrared Quantum Dots (QDots) and injected at either the time of infection or on day 7 in the early- and established-biofilm models, respectively. Animals were subjected to IVIS imaging immediately following macrophage transfer to confirm macrophage signals immediately following injection, and subsequently reimaged every 24 hours (Fig. 12A). Qdot-labeled Ml macrophages were still visible at 5 days post-treatment, at which point animals were sacrificed in order to recover tissues for FACs analysis. Data indicated a slight increase in F4/80+/IRF5+ macrophages in Qdot-labeled Ml macrophage-treated animals compared to vehicle-treated animals (Fig. 12B).
Example 6
[0141] EP67 impacts MRSA biofilm establishment in vivo and stimulates local proinflammatory responses. A supplementary approach to the introduction of exogenous Ml- activated macrophages is the combined administration of Ml-activated macrophages and a macrophage- activating compound, such as a macrophage- activating peptide like EP67 (SEQ ID NO:l), EP54 (SEQ ID NO:2), or C5a65-74 (SEQ ID NO:4) or their analogs or variants as described herein. To establish that a macrophage-activating compound could have a beneficial effect, an experiment was designed to determine whether the administration of the C5aR agonist EP67 would re-program the endogenous macrophage infiltrate associated with MRSA biofilms in vivo from an anti-inflammatory M2 to a pro-inflammatory Ml phenotype to facilitate bacterial clearance. The in vivo efficacy of EP67 against MRSA was examined using a mouse model of catheter-associated biofilm infection, as previously described (17, 22, 23). Animals were initially treated with EP67 at the time of infection followed by a series
of injections at 24 and 48 hours, two at sites adjacent to the external catheter wall and two into each open end of the catheter. Bacterial burdens associated with biofilm-infected catheters as well as surrounding tissues were significantly decreased following EP67 treatment compared to animals receiving an inactive scrambled sequence of EP67 (sEP67) or vehicle control (Fig. 13). Importantly, early peptide treatment was key to preventing MRSA biofilm establishment, since minimal bacterial growth was detected at day 14 following infection, even though the last dosing interval of EP67 occurred at 48 hours (Fig. 13C and D).
[0142] To determine whether EP67 could restore the biofilm environment to a proinflammatory state, cytokine and chemokine expression were evaluated in biofilm-infected tissues. Several inflammatory mediators that are predominantly expressed by activated macrophages, such as IL-12p40 and RANTES, were significantly increased in EP67-treated animals compared to vehicle-treated animals (Fig. 14A and B). Similarly, IL-17, IL-la, and IFN-γ may also have been elevated, although the observed increases did not reach statistical significance (Fig. 14C, D and E). To further investigate mechanisms of EP67 immune activation during biofilm infections, the degree of macrophage influx into tissues surrounding MRSA biofilms was examined by immunofluorescence staining and flow cytometry.
Immunofluorescence staining revealed that macrophage accumulation into EP67-treated biofilms was significantly increased at day 3 post-infection compared to vehicle-treated animals (Fig. 15A and B). Importantly, while only a few macrophages were recruited to the biofilm surface in vehicle-treated mice, EP67 administration dramatically increased the numbers of macrophages that migrated into the biofilm (Fig. 15A). The ability of EP67 to augment macrophage infiltrates in MRSA biofilms was confirmed by FACS (Fig. 15C and D). Collectively, these findings demonstrate that EP67 induces a pro-inflammatory milieu by augmenting macrophage recruitment and cytokine/chemokine production, which effectively counteracts the anti-inflammatory environment elicited by MRSA biofilms. Experiments were also designed and conducted to determine whether EP67 treatment could impact established biofilms; however, the peptide did not exert any beneficial effects in this setting, suggesting its optimal use as a prophylactic modality.
Example 7
Activated Macrophage Effect on Biofilms
[0143] The local injection of macrophages pre-activated in vitro with S. aareas-derived PGN and IFN-γ into the area surrounding a catheter-associated S. aureus biofilm infection eliminated/reduced bacterial burden within the biofilm as well as the healthy tissue in the vicinity of the biofilm. This was demonstrated by the following experiment.
[0144] Briefly, an S. aureus biofilm (USA 300 LAC strain) was established on a hollow catheter (1 cm in length) inserted subcutaneously into C57BL/6 mice by introducing 1,000 colony forming units (CFUs) of S. aureus into the catheter lumen. At 12, 24 and 48 hours following infection, 106 macrophages were introduced directly into the catheter lumen. Samples of the biofilm and surrounding tissue were obtained at days 3 and 14 post-infection to quantitate the number of viable bacteria by standard culture methods. Mice treated with activated macrophages exhibited a significant decrease in bacterial burdens within the biofilm and surrounding tissue relative to mice treated with neutrophils or PBS vehicle.
[0145] Collectively, the studies disclosed in Examples 1-7 have established that Ml- activated macrophages are effective in biofilm containment and bacterial clearance.
Additionally, data disclosed herein supports the position that Ml-activated macrophages and macrophage- activating compound such as a macrophage- activating peptide as disclosed herein, when administered in combination, are effective in the prevention and treatment of biofilms. By extension, it is not unexpected that biofilms, including MRSA biofilms, have the capacity to thwart this response by skewing macrophages away from a pro-inflammatory Ml phenotype to an anti-inflammatory M2 phenotype to ensure biofilm persistence in an immunocompetent host.
REFERENCES
[0146] 1. Donlan, R.M., and Costerton, J.W. 2002. Biofilms: survival mechanisms of clinically relevant microorganisms. Clin Microbiol Rev 15:167-193
[0147] 2. Fitzpatrick, F., Humphreys, H., and O'Gara, J.P. 2005. The genetics of staphylococcal biofilm formation— will a greater understanding of pathogenesis lead to better management of device-related infection? Clin Microbiol Infect 11:967-973.
[0148] 3. Otto, M. 2008. Staphylococcal biofilms. Curr Top Microbiol Immunol 322:207-228.
[0149] 4. Fong, I.W. 2009. Device-Related Infections. In Emerging Issues and
Controversies in Infectious Disease. I.W. Fong, editor. New York, NY: Springer. 261-297.
[0150] 5. Del Pozo, J.L., and Patel, R. 2009. Clinical practice. Infection associated with prosthetic joints. N Engl J Med. 361:787-794.
[0151] 6. Sia, I.G., Berbari, E.F., and Karchmer, A.W. 2005. Prosthetic joint infections. Infect Dis Clin N Am 19:885-914.
[0152] 7. Zimmerli, W., Trampuz, A., and Ochsner, P.E. 2004. Prosthetic -joint infections. N Engl J Med. 351:1645-1654.
[0153] 8. Anderl, J.N., Zahller, J., Roe, F., and Stewart, P.S. 2003. Role of nutrient limitation and stationary-phase existence in Klebsiella pneumoniae biofilm resistance to ampicillin and ciprofloxacin. Antimicrob Agents Chemother 47:1251-1256.
[0154] 9. Ceri, H., Olson, M.E., Stremick, C, Read, R.R., Morck, D., and Buret, A. 1999. The Calgary Biofilm Device: new technology for rapid determination of antibiotic susceptibilities of bacterial biofilms. J Clin Microbiol 37:1771-1776.
[0155] 10. Agarwal, S., and Busse, P.J. 2010. Innate and adaptive immunosenescence. Ann Allergy Asthma Immunol 104:183-193.
[0156] 11. Scherer, A., and McLean, A. 2002. Mathematical models of vaccination. Br. Med. Bull. 62:187-199.
[0157] 12. Proctor, R.A. 2012. Challenges for a universal Staphylococcus aureus vaccine. Clin Infect Dis. 54: 1179-1186.
[0158] 13. Otto, M. 2006. Bacterial evasion of antimicrobial peptides by biofilm formation. Curr Top Microbiol Immunol 306:251-258.
[0159] 14. Kristian, S.A., T. A. Birkenstock, U. Sauder, D. Mack, F. Gotz, and R.
Landmann. 2008. Biofilm formation induces C3a release and protects Staphylococcus epidermidis from IgG and complement deposition and from neutrophil-dependent killing. J Infect Dis 197:1028-1035.
[0160] 15. Thurlow, L.R., Hanke, M.L., Fritz, T., Angle, A., Williams, S.H.,
Engebretsen, I.L., Bayles, K.W., Horswill, A.R., and Kielian, T. 2011. Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo. J Immunol 186:6585-6596.
[0161] 16. Bernthal, N.M., Pribaz, J.R., Stavrakis, A., Billi, F., Cho, J.S., Ramos, R.I., Francis, K.P., Iwakura, Y., and Miller, L.S. 2011 Protective role of IL-Ιβ against post- arthroplasty Staphylococcus aureus infection. J Orthop Res. 29:1621-1626.
[0162] 17. Taylor, S.M., Sherman, S.A., Kirnarsky, L., and Sanderson, S.D. 2001.
Development of response-selective agonists of human C5a anaphylatoxin: conformational, biological, and therapeutic considerations. Curr Med Chem. 8:675-684.
[0163] 18. Morgan, E.L., Morgan, B.N., Stein, E.A., Vitrs, E.L., Thoman, M.L.,
Sanderson, S.D., and Phillips, J.A. 2009. Enhancement of in vivo and in vitro immune functions by a conformational^ biased, response- selective agonist of human C5a:
implications for a novel adjuvant in vaccine design. Vaccine. 28:463-469.
[0164] 19. Hanke, M.L., and Kielian, T. 2012. Deciphering mechanisms of
staphylococcal biofilm evasion of host immunity. Front. Cell. Inf. Microbio. 2:doi:
10.3389/fcimb.2012.00062.
[0165] 20. Urban, C.F., Lourido, S., and Zychlinsky, A. 2006. How do microbes evade neutrophil killing? Cell Microbiol. 8:1687-1696.
[0166] 21. Graves, S.F., Kobayashi, S.D., and DeLeo, F.R. 2010. Community-associated methicillin-resistant Staphylococcus aureus immune evasion and virulence. J Mol Med (Bed). 88:109-114.
[0167] 22. Foster, T.J. 2009. Colonization and infection of the human host by
staphylococci: adhesion, survival and immune evasion. Vet Dermatol. 20:456-470.
[0168] 23. Hanke, M.L., Angle, A., and Kielian, T. 2012. MyD88-Dependent Signaling Influences Fibrosis and Alternative Macrophage Activation during Staphylococcus aureus Biofilm Infection. PLoS One In Press.
[0169] 24. Benoit, M., Desnues, B., and Mege, J.L. 2008. Macrophage polarization in bacterial infections. J Immunol 181:3733-3739.
[0170] 25. Gristina, A.G. 1987. Biomaterial-centered infection: microbial adhesion versus tissue integration. Science 237:1588-1595.
[0171] 26. Nathan, C. 2006. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol 6:173-182.
[0172] 27. Nauseef, W.M. 2007. How human neutrophils kill and degrade microbes: an integrated view. Immunol Rev 219:88-102.
[0173] 28. Brinkmann, V., Reichard, U., Goosmann, C, Fauler, B., Uhlemann, Y., Weiss, D.S., Weinrauch, Y., and Zychlinsky, A. 2004. Neutrophil extracellular traps kill bacteria. Science 303:1532-1535.
[0174] 29. Short, A.J., Paczkowski, N.J., Vogen, S.M., Sanderson, S.D., and Taylor, S.M. 1999. Response-selective C5a agonists: differential effects on neutropenia and hypotension in the rat. Br J Pharmacol 128:511-514.
[0175] 30. Morgan, E.L., Thoman, M.L., Sanderson, S.D., and Phillips, J.A. 2010. A novel adjuvant for vaccine development in the aged. Vaccine. 28:8275-8279.
[0176] 31. Buret, A., Ward, K.H., Olson, M.E., and Costerton, J.W. 1991. An in vivo model to study the pathobiology of infectious biofilms on biomaterial surfaces. J Biomed Mater Res 25:865-874.
[0177] 32. Duch, J.M., and Yee, J. 2001. Successful use of recombinant tissue
plasminogen activator in a patient with relapsing peritonitis. Am J Kidney Dis 37:149-153.
[0178] 33. Pickering, S.J., Fleming, S.J., Bowley, J.A., Sissons, P., Oppenheim, B.A., Burnie, J., Ralston, A.J., and Ackrill, P. 1989. Urokinase: a treatment for relapsing peritonitis due to coagulase-negative staphylococci. Nephrol Dial Transplant 4:62-65.
[0179] 34. Xu, K.D., McFeters, G.A., and Stewart, P.S. 2000. Biofilm resistance to antimicrobial agents. Microbiology 146:547-549.
[0180] 35. Singh, R., Ray, P., Das, A., and Sharma, M. 2010. Penetration of antibiotics through Staphylococcus aureus and Staphylococcus epidermidis biofilms. J Antimicrob Chemother 65:1955-1958.
[0181] 36. Fremond, CM., Yeremeev, V., Nicolle, D.M., Jacobs, M., Quesniaux, V.F., and Ryffel, B. 2004. Fatal Mycobacterium tuberculosis infection despite adaptive immune response in the absence of MyD88. /. Clin. Invest. 114:1790-1799.
[0182] 37. Kawai, T., Adachi, O., Ogawa, T., Takeda, K., and Akira, S. 1999.
Unresponsiveness of MyD 88 -deficient mice to endotoxin. Immunity 11:115-122.
[0183] 38. Adachi, O., Kawai, T., Takeda, K., Matsumoto, M., Tsutsui, H., Sakagami, M., Nakanishi, K., and Akira, S. 1998. Targeted disruption of the MyD88 gene results in loss of IL-1- and IL-18-mediated function. Immunity 9: 143-150.
[0184] 39. Lauderdale, K.J., Malone, C.L., Boles, B.R., Morcuende, J., and Horswill, A.R. 2010. Biofilm dispersal of community-associated methicillin-resistant Staphylococcus aureus on orthopedic implant material. J Orthop Res 28:55-61.
[0185] 40. Cassat, J.E., Lee, C.Y., and Smeltzer, M.S. 2007. Investigation of biofilm formation in clinical isolates of Staphylococcus aureus. Methods Mol Biol 391:127-144.
[0186] 41. Rupp, M.E., Ulphani, J.S., Fey, P.D., Bartscht, K., and Mack, D. 1999.
Characterization of the importance of polysaccharide intercellular adhesin/hemagglutinin of Staphylococcus epidermidis in the pathogenesis of biomaterial-based infection in a mouse foreign body infection model. Infect Immun 67:2627-2632.
[0187] 42. Longbrake, E.E., Lai, W., Ankeny, D.P., and Popovich, P.G. 2007.
Characterization and modeling of monocyte-derived macrophages after spinal cord injury. J Neurochem 102:1083-1094.
[0188] 43. Kigerl, K.A., Gensel, J.C., Ankeny, D.P., Alexander, J.K., Donnelly, D.J., and Popovich, P.G. 2009. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci 29:13435-13444.
[0189] 44. Boxio, R., Bossenmeyer-Pourie, C, Steinckwich, N., Dournon, C, and Ntisse, O. 2004. Mouse bone marrow contains large numbers of functionally competent neutrophils. JLeukoc Biol 75:604-611.
[0190] Each of the references cited herein is incorporated by reference in its entirety or as relevant apparent from the context of its usage.
[0191] From the disclosure herein it will be appreciated that, although specific
embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention.
Claims
1. A composition comprising an isolated activated macrophage for the prevention or treatment of a biofilm.
2. A kit comprising an isolated macrophage and a macrophage-activating agent.
3. A kit comprising a macrophage-activating agent.
4. The kit according to claim 2 or 3 wherein the macrophage-activating agent is selected from the group consisting of a C5a receptor agonist, interleukin-ΐβ, interferon-γ and tumor necrosis factor (TNF), and interferon-γ and an inducer of TNF.
5. A method of providing biofilm therapy to a subject-in-need comprising:
(a) isolating peripheral blood monocytes from a subject;
(b) culturing the monocytes under conditions compatible with formation of
macrophages; and
(c) administering a prophylactically or therapeutically effective amount of the macrophages to a subject-in-need, wherein the macrophages are activated by contact with a macrophage-activating agent prior to, simultaneously with, or after administration of the macrophages.
6. The method according to claim 5 wherein the macrophages are activated by contact with a macrophage-activating agent prior to administration of the macrophages.
7. A use of macrophages in the prevention or treatment of a biofilm infection in a subject-in-need, wherein the macrophages are activated by contact with a macrophage- activating agent prior to, simultaneously with, or after administration of the macrophages.
8. The method according to claim 5 or 6 wherein the subject-in-need is at risk of developing a biofilm infection.
9. The method according to claim 5 or 6 wherein the subject-in-need comprises an established biofilm infection.
10. The method according to claim 5 or 6 wherein the subject-in-need is a human patient.
11. The method according to claim 5 or 6 wherein the subject-in-need is a veterinary animal selected from the group consisting of a dog, a cat, a horse, a bovine, a sheep, a goat and a pig.
12. The method according to claim 5 or 6 wherein the subject is the subject-in- need.
13. The method according to claim 5 or 6 wherein the macrophage-activating agent is selected from the group consisting of a C5a receptor agonist, interleukin-ΐβ, interferon-γ and tumor necrosis factor (TNF), and interferon-γ and an inducer of TNF.
14. The method according to claim 13 wherein the inducer of TNF is selected from the group consisting of peptidoglycan and lipopolysaccharide.
15. The method according to claim 5 or 6 wherein the macrophages are administered at a dose of 10 5 to 108 cells.
16. The method according to claim 15 wherein one to five doses of macrophages are administered.
17. The method according to claim 13 further comprising delivering to the subject-in-need an effective amount of a pro-inflammatory compound selected from the group consisting of interleukin-6, interleukin-12, monocyte chemoattractant protein- 1 (MCP- 1), monokine-induced by IFN-γ (MIG), interferon-regulatory protein-10 (IP-10), macrophage inflammatory protein-2 (MIP-2), keratinocyte chemoattractant (KC), macrophage colony- stimulating factor (M-CSF) and granulocyte-macrophage colony- stimulating factor (GM- CSF).
18. The method according to claim 5 or 6 wherein the subject-in-need undergoes surgery to receive a surgical implant.
19. The method according to claim 18 wherein the surgical implant is selected from the group consisting of an artificial joint component, a cranial plate, a dental implant and a subcutaneous pacemaker.
20. The method according to claim 19 wherein the artificial joint component is selected from the group consisting of an artificial hip component, an artificial knee component, an artificial ankle component, an artificial elbow component, an artificial shoulder component, and an artificial wrist component.
21. The method according to claim 5 or 6 wherein the subject- in-need is scheduled to receive, or has received, a catheter.
22. The method according to claim 5 wherein the macrophage-activating agent and the macrophage are co-administered to the subject-in-need.
23. The method according to claim 5 or 6 further comprising delivery of a proteolytic enzyme to the subject-in-need.
24. The method according to claim 23 wherein the proteolytic enzyme is selected from the group consisting of MMP-1, MMP-2, MMP-7, MMP-9, MMP-12 and collagenase.
25. The method according to claim 5, 6, or 24 further comprising a delivery vehicle selected from the group consisting of a microsphere, a microparticle, a nanosphere, a nanoparticle, a liposome, a micelle and a gel, wherein the delivery vehicle comprises a macrophage and a macrophage-activating agent, or an activated macrophage, or a
macrophage, a macrophage-activating agent and a proteolytic enzyme, or an activated macrophage and a proteolytic enzyme.
26. The method according to claim 5 or 6 wherein the subject-in-need exhibits an inflammatory disorder.
27. The method according to claim 5 or 6 wherein the subject-in-need exhibits a disorder or condition selected from the group consisting of atherosclerosis, infectious complications of cystic fibrosis, endocarditis, chronic sinusitis, nasal polyposis, chronic otitis media, leptospirosis, kidney stones, osteomyelitis, necrosis of the jaw, a urinary tract infection, a bladder infection, a chronic wound, an artificial component of a joint, a temporary catheter and a permanent catheter.
28. The method according to claim 5 or 6 further comprising administering a prophylactically or therapeutically effective amount of an antibiotic.
29. The method according to claim 28 wherein the antibiotic is a member of a class of antibiotics selected from the group consisting of aminoglycosides, ansamycins, carbacephems, carbapenems, cephalosporins, glycopeptides, lincosamides, lipopeptides, macrolides, erythromycins, monobactams, nitrofurans, penicillins, penicillin combinations, antimicrobial polypeptides, quinolones, sulfonamides, tetracyclines and antimycobacterials.
30. The method according to claim 29 wherein the antibiotic is selected from the group consisting of amikacin, gentamicin, kanamycin, neomycin, roxithromycin, netilmicin, tobramycin, paromomycin, geldamycin, herimycin, loracarbef, ertapenem, doripenem, imipenem or cilastatin, meropenem, cefadroxil, cefazolin, cefalotin or cefalothin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefonicid, cefixime, cefdinir, cefditoren, cefotaxime, cefetamet, cefoperazone, cefpodoxime, ceftazidime, ceftibuten, ceftriaxone, cefepime, ceftaroline or fasamil, ceftobiprole, cephalothin, cefmetazole, cefotetan, ceftizoxime, cefsulodin, cephalexin, cinoxacin; teicoplanin, vancomycin, telavancin; clindamycin, lincomycin, daptomycin, azithromycin, clarithromycin, erythromycin, erythromycin estolate, erythromycin ethyl succinate, erythromycin glucoheptonate, erythromycin lactobionate, erythromycin stearate, roximthromycin, trolandomycin, telithromycin, spectinomycin, spriamycin, Aztreonam, furazolidone, nitrofurantoi, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin G, penicillin V, piperacillin, ticarcillin, amoxicillin and clavulanate, ampicillin and sulbactam, piperaccillin and tazobactam, ticarcillin and clavulanate; bacitracin, colistin, polymyxin B, ciprofloxacin, enoxacin, gatifloxacin, levofloxacin, lomefloxacin, moxifloxicin, ofloxacin, nalidixic acid, norfloxacin, ofloxicin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sufonamidochrysoidine, sulfacetamide, sufadiazine, silver sulfadiazine, sulfamethizole, sulfamethoxazole, sulfanilamide, sulfasalazine, sulfisoxazole, trimethoprim- sulfamethoxazole, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol, ethionamide, isoniazid, rifampicin or rifampin, rifabutin, rifapenine, streptomycin, arshenamine, chloramphenicol, fosfomycin, fusidic acid, linezolid, metronidazole, mupirocin, platensimycin, quinupristin or dalfopristin, rifaximin, thiamphenicol, tigercycline, tinidazole, trimethoprim, fleroxacin, nitrofurantoin, coamoxiclavuanate, and their salts, acids, and bases.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201161527773P | 2011-08-26 | 2011-08-26 | |
US61/527,773 | 2011-08-26 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2013032964A1 true WO2013032964A1 (en) | 2013-03-07 |
Family
ID=47756775
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2012/052421 WO2013032964A1 (en) | 2011-08-26 | 2012-08-26 | Compositions and methods for preventing and treating biofilms |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2013032964A1 (en) |
Cited By (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2015017402A1 (en) * | 2013-07-29 | 2015-02-05 | Board Of Regents Of The University Of Nebraska | Compositions and methods for the treatment of biofilm infections |
WO2015132392A1 (en) * | 2014-03-07 | 2015-09-11 | Reponex Pharmaceuticals Aps | Compositions for treating lung infections by airway administration |
WO2016145365A1 (en) * | 2015-03-11 | 2016-09-15 | Board Of Regents Of The University Of Nebraska | Conformationally stable analogs of the response selective c5a agonist ep67 |
US10105415B2 (en) | 2014-07-24 | 2018-10-23 | Reponex Pharmaceuticals A/S | Compositions comprising granulocyte-macrophage colony-stimulating factor for the treatment of inflammatory bowel disease |
WO2019099932A1 (en) * | 2017-11-19 | 2019-05-23 | Next Science IP Holdings Pty Ltd | Compositions and methods for treating intervertebral discs |
JP2019515947A (en) * | 2016-04-04 | 2019-06-13 | ケモセントリックス,インコーポレイティド | Soluble C5aR Antagonist |
WO2020023670A1 (en) * | 2018-07-24 | 2020-01-30 | University Of Virginia Patent Foundation | Compositions and methods for combating multidrug-resistant bacteria |
CN111479569A (en) * | 2017-10-18 | 2020-07-31 | 生物膜控制公司 | Use of cloxacillin for inhibiting/preventing biofilm formation |
CN112043720A (en) * | 2020-09-24 | 2020-12-08 | 厦门惠盈动物药业有限公司 | Spectinomycin lincomycin soluble powder capable of being completely dissolved in various solvents and preparation method thereof |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20110142812A1 (en) * | 2006-11-06 | 2011-06-16 | Stc.Unm | Suppressive macrophages, c-reactive protein and the treatment of systemic lupus erythematosus and immune thrombocytopenic purpura |
US20110195062A1 (en) * | 1998-09-18 | 2011-08-11 | Schering Corporation | Mammalian Cytokines; Related Reagents and Methods |
-
2012
- 2012-08-26 WO PCT/US2012/052421 patent/WO2013032964A1/en active Application Filing
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20110195062A1 (en) * | 1998-09-18 | 2011-08-11 | Schering Corporation | Mammalian Cytokines; Related Reagents and Methods |
US20110142812A1 (en) * | 2006-11-06 | 2011-06-16 | Stc.Unm | Suppressive macrophages, c-reactive protein and the treatment of systemic lupus erythematosus and immune thrombocytopenic purpura |
Non-Patent Citations (6)
Cited By (16)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2015017402A1 (en) * | 2013-07-29 | 2015-02-05 | Board Of Regents Of The University Of Nebraska | Compositions and methods for the treatment of biofilm infections |
US20160158353A1 (en) * | 2013-07-29 | 2016-06-09 | Board Of Regents Of The University Of Nebraska | Compositions and Methods for the Treatment of Biofilm Infections |
US10821178B2 (en) | 2013-07-29 | 2020-11-03 | Board Of Regents Of The University Of Nebraska | Methods of treating biofilm infections comprising administering inhibitors of myeloid-derived suppressor cells |
WO2015132392A1 (en) * | 2014-03-07 | 2015-09-11 | Reponex Pharmaceuticals Aps | Compositions for treating lung infections by airway administration |
EP3662924A1 (en) * | 2014-03-07 | 2020-06-10 | Reponex Pharmaceuticals A/S | Compositions for treating lung infections by airway administration |
US10105415B2 (en) | 2014-07-24 | 2018-10-23 | Reponex Pharmaceuticals A/S | Compositions comprising granulocyte-macrophage colony-stimulating factor for the treatment of inflammatory bowel disease |
WO2016145365A1 (en) * | 2015-03-11 | 2016-09-15 | Board Of Regents Of The University Of Nebraska | Conformationally stable analogs of the response selective c5a agonist ep67 |
CN107847452A (en) * | 2015-03-11 | 2018-03-27 | 内布拉斯加大学董事会 | Response selectivity C5a activators EP67 conformational stability analog |
JP2019515947A (en) * | 2016-04-04 | 2019-06-13 | ケモセントリックス,インコーポレイティド | Soluble C5aR Antagonist |
US11254695B2 (en) | 2016-04-04 | 2022-02-22 | Chemocentryx, Inc. | Soluble C5aR antagonists |
JP7121722B2 (en) | 2016-04-04 | 2022-08-18 | ケモセントリックス,インコーポレイティド | Soluble C5aR antagonist |
CN111479569A (en) * | 2017-10-18 | 2020-07-31 | 生物膜控制公司 | Use of cloxacillin for inhibiting/preventing biofilm formation |
WO2019099932A1 (en) * | 2017-11-19 | 2019-05-23 | Next Science IP Holdings Pty Ltd | Compositions and methods for treating intervertebral discs |
WO2020023670A1 (en) * | 2018-07-24 | 2020-01-30 | University Of Virginia Patent Foundation | Compositions and methods for combating multidrug-resistant bacteria |
CN112043720B (en) * | 2020-09-24 | 2021-07-02 | 厦门惠盈动物药业有限公司 | Spectinomycin lincomycin soluble powder capable of being completely dissolved in various solvents and preparation method thereof |
CN112043720A (en) * | 2020-09-24 | 2020-12-08 | 厦门惠盈动物药业有限公司 | Spectinomycin lincomycin soluble powder capable of being completely dissolved in various solvents and preparation method thereof |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2013032964A1 (en) | Compositions and methods for preventing and treating biofilms | |
Hanke et al. | Targeting macrophage activation for the prevention and treatment of Staphylococcus aureus biofilm infections | |
Reffuveille et al. | Staphylococcus aureus Biofilms and their Impact on the Medical Field | |
Zheng et al. | Colonization of medical devices by staphylococci | |
Blanchette et al. | Current therapies in treatment and prevention of fracture wound biofilms: why a multifaceted approach is essential for resolving persistent infections | |
John et al. | Reversible daptomycin tolerance of adherent staphylococci in an implant infection model | |
Wang et al. | Novel therapeutic interventions towards improved management of septic arthritis | |
AU2011343012A1 (en) | A composition comprising an antibiotic and a dispersant or an anti -adhesive agent | |
US10821178B2 (en) | Methods of treating biofilm infections comprising administering inhibitors of myeloid-derived suppressor cells | |
NO320884B1 (en) | Use of lysostaphin or variant thereof in the manufacture of a medicament for the treatment of infections. | |
Silveira et al. | Antibiofilm peptides: relevant preclinical animal infection models and translational potential | |
US9265820B2 (en) | Multivalent vaccine protection from Staphylococcus aureus infection | |
CN108883134A (en) | Activating stem cells and Systemic treatments for infected wound | |
KR102135648B1 (en) | Leoidin compound having anti-virulence activity and use thereof | |
CA2604494A1 (en) | Improved method and apparatus for treating bacterial infections in devices | |
JP2002523437A (en) | Methods for treating staphylococcal diseases | |
Chen et al. | The effect of Staphylococcus aureus on innate and adaptive immunity and potential immunotherapy for S. aureus-induced osteomyelitis | |
Kovacs et al. | Disruption of biofilm by bacteriophages in clinically relevant settings | |
Shunmugaperumal | Microbial colonization of medical devices and novel preventive strategies | |
JP7514493B2 (en) | Composition for improving the drug susceptibility of methicillin-resistant Staphylococcus aureus (MRSA) to antibacterial agents, composition for treating or preventing MRSA infection, and composition for reducing the pathogenicity of MRSA | |
Drago et al. | Biofilm formation and the biological response | |
WO2016146037A1 (en) | Inhibitor used to inhibit/disintegrate biofilm and application thereof | |
TW202122585A (en) | Methods for treating bacterial infections | |
Zakaria et al. | In vitro and in vivo studies of antibacterial effect of ceftriaxone moxifloxacin combination against methicillin resistant Staphylococcus aureus biofilms formed on biomedical implants | |
US20200215169A1 (en) | Antibacterial methods and related kits |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 12828261 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 12828261 Country of ref document: EP Kind code of ref document: A1 |