WO2023139248A1 - Inhibitors of acyl protein thioesterases against microbial infections - Google Patents
Inhibitors of acyl protein thioesterases against microbial infections Download PDFInfo
- Publication number
- WO2023139248A1 WO2023139248A1 PCT/EP2023/051499 EP2023051499W WO2023139248A1 WO 2023139248 A1 WO2023139248 A1 WO 2023139248A1 EP 2023051499 W EP2023051499 W EP 2023051499W WO 2023139248 A1 WO2023139248 A1 WO 2023139248A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- apt
- inhibitor
- cov
- formula
- Prior art date
Links
- 239000003112 inhibitor Substances 0.000 title claims abstract description 82
- 125000002252 acyl group Chemical group 0.000 title claims abstract description 39
- 102000005488 Thioesterase Human genes 0.000 title claims abstract description 37
- 108020002982 thioesterase Proteins 0.000 title claims abstract description 37
- 208000015181 infectious disease Diseases 0.000 title abstract description 92
- 230000000813 microbial effect Effects 0.000 title abstract description 8
- 238000011282 treatment Methods 0.000 claims abstract description 55
- 230000003612 virological effect Effects 0.000 claims abstract description 51
- 208000035143 Bacterial infection Diseases 0.000 claims abstract description 24
- 208000022362 bacterial infectious disease Diseases 0.000 claims abstract description 24
- 208000036142 Viral infection Diseases 0.000 claims abstract description 16
- 230000002265 prevention Effects 0.000 claims abstract description 16
- 241000700605 Viruses Species 0.000 claims abstract description 11
- 241000894006 Bacteria Species 0.000 claims abstract description 5
- 241001678559 COVID-19 virus Species 0.000 claims description 65
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 43
- 125000000592 heterocycloalkyl group Chemical group 0.000 claims description 43
- 239000003814 drug Substances 0.000 claims description 42
- 150000003839 salts Chemical class 0.000 claims description 33
- 125000003118 aryl group Chemical group 0.000 claims description 28
- 125000001072 heteroaryl group Chemical group 0.000 claims description 25
- 229910052739 hydrogen Inorganic materials 0.000 claims description 22
- 239000001257 hydrogen Substances 0.000 claims description 22
- 150000002431 hydrogen Chemical class 0.000 claims description 19
- YTPLMLYBLZKORZ-UHFFFAOYSA-N Thiophene Chemical group C=1C=CSC=1 YTPLMLYBLZKORZ-UHFFFAOYSA-N 0.000 claims description 16
- 201000003176 Severe Acute Respiratory Syndrome Diseases 0.000 claims description 15
- 229910052736 halogen Inorganic materials 0.000 claims description 14
- 150000002367 halogens Chemical class 0.000 claims description 14
- 125000000041 C6-C10 aryl group Chemical group 0.000 claims description 13
- 239000003937 drug carrier Substances 0.000 claims description 13
- 230000009385 viral infection Effects 0.000 claims description 13
- 239000008194 pharmaceutical composition Substances 0.000 claims description 12
- 125000001831 (C6-C10) heteroaryl group Chemical group 0.000 claims description 11
- 125000006374 C2-C10 alkenyl group Chemical group 0.000 claims description 11
- 241000711573 Coronaviridae Species 0.000 claims description 11
- 125000000217 alkyl group Chemical group 0.000 claims description 11
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 claims description 10
- 125000000008 (C1-C10) alkyl group Chemical group 0.000 claims description 9
- 125000003545 alkoxy group Chemical group 0.000 claims description 9
- 125000002541 furyl group Chemical group 0.000 claims description 8
- 208000030507 AIDS Diseases 0.000 claims description 6
- 208000031729 Bacteremia Diseases 0.000 claims description 6
- 206010040047 Sepsis Diseases 0.000 claims description 6
- 125000002915 carbonyl group Chemical group [*:2]C([*:1])=O 0.000 claims description 6
- 206010014599 encephalitis Diseases 0.000 claims description 6
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 6
- 229940124597 therapeutic agent Drugs 0.000 claims description 6
- 125000006823 (C1-C6) acyl group Chemical group 0.000 claims description 5
- 125000004191 (C1-C6) alkoxy group Chemical group 0.000 claims description 5
- 206010061192 Haemorrhagic fever Diseases 0.000 claims description 5
- 125000005257 alkyl acyl group Chemical group 0.000 claims description 5
- 125000004457 alkyl amino carbonyl group Chemical group 0.000 claims description 5
- 125000004448 alkyl carbonyl group Chemical group 0.000 claims description 5
- 239000003242 anti bacterial agent Substances 0.000 claims description 5
- 239000003443 antiviral agent Substances 0.000 claims description 5
- 230000001580 bacterial effect Effects 0.000 claims description 5
- 239000003085 diluting agent Substances 0.000 claims description 5
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 claims description 5
- 241000711466 Murine hepatitis virus Species 0.000 claims description 4
- 241000711484 Transmissible gastroenteritis virus Species 0.000 claims description 4
- 125000003282 alkyl amino group Chemical group 0.000 claims description 4
- 238000007918 intramuscular administration Methods 0.000 claims description 4
- 241001112741 Bacillaceae Species 0.000 claims description 3
- 241000606069 Chlamydiaceae Species 0.000 claims description 3
- 241000588921 Enterobacteriaceae Species 0.000 claims description 3
- 241000589246 Legionellaceae Species 0.000 claims description 3
- 241000593669 Pectobacteriaceae Species 0.000 claims description 3
- 241000947836 Pseudomonadaceae Species 0.000 claims description 3
- 208000025747 Rheumatic disease Diseases 0.000 claims description 3
- 241000315672 SARS coronavirus Species 0.000 claims description 3
- 206010039491 Sarcoma Diseases 0.000 claims description 3
- 206010022000 influenza Diseases 0.000 claims description 3
- 208000032839 leukemia Diseases 0.000 claims description 3
- 208000004429 Bacillary Dysentery Diseases 0.000 claims description 2
- 201000001178 Bacterial Pneumonia Diseases 0.000 claims description 2
- 241000711950 Filoviridae Species 0.000 claims description 2
- 241000710781 Flaviviridae Species 0.000 claims description 2
- 208000005577 Gastroenteritis Diseases 0.000 claims description 2
- 241001122120 Hepeviridae Species 0.000 claims description 2
- 241000700586 Herpesviridae Species 0.000 claims description 2
- 241000711467 Human coronavirus 229E Species 0.000 claims description 2
- 241000482741 Human coronavirus NL63 Species 0.000 claims description 2
- 206010022678 Intestinal infections Diseases 0.000 claims description 2
- 201000009906 Meningitis Diseases 0.000 claims description 2
- 241000127282 Middle East respiratory syndrome-related coronavirus Species 0.000 claims description 2
- 241000204034 Mycoplasmataceae Species 0.000 claims description 2
- 241000712464 Orthomyxoviridae Species 0.000 claims description 2
- 241000711504 Paramyxoviridae Species 0.000 claims description 2
- 241000606752 Pasteurellaceae Species 0.000 claims description 2
- 241000150350 Peribunyaviridae Species 0.000 claims description 2
- 241000711904 Pneumoviridae Species 0.000 claims description 2
- 241001248479 Pseudomonadales Species 0.000 claims description 2
- 241000712907 Retroviridae Species 0.000 claims description 2
- 241000711931 Rhabdoviridae Species 0.000 claims description 2
- 206010039438 Salmonella Infections Diseases 0.000 claims description 2
- 208000007893 Salpingitis Diseases 0.000 claims description 2
- 206010040550 Shigella infections Diseases 0.000 claims description 2
- 241000295644 Staphylococcaceae Species 0.000 claims description 2
- 241000194018 Streptococcaceae Species 0.000 claims description 2
- 241000710924 Togaviridae Species 0.000 claims description 2
- 208000037386 Typhoid Diseases 0.000 claims description 2
- 241000607493 Vibrionaceae Species 0.000 claims description 2
- 241000593672 Yersiniaceae Species 0.000 claims description 2
- 206010014665 endocarditis Diseases 0.000 claims description 2
- 206010014801 endophthalmitis Diseases 0.000 claims description 2
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 2
- 238000002663 nebulization Methods 0.000 claims description 2
- 206010039447 salmonellosis Diseases 0.000 claims description 2
- 208000013223 septicemia Diseases 0.000 claims description 2
- 201000005113 shigellosis Diseases 0.000 claims description 2
- 206010040872 skin infection Diseases 0.000 claims description 2
- 201000008297 typhoid fever Diseases 0.000 claims description 2
- 241001109669 Human coronavirus HKU1 Species 0.000 claims 1
- 241001428935 Human coronavirus OC43 Species 0.000 claims 1
- 238000000034 method Methods 0.000 abstract description 33
- 208000003322 Coinfection Diseases 0.000 abstract description 3
- 238000002360 preparation method Methods 0.000 abstract description 3
- 210000004027 cell Anatomy 0.000 description 160
- YVIJPELUPZUEJX-UHFFFAOYSA-N (5,5-dioxo-4h-thieno[3,2-c]thiochromen-2-yl)-[4-(4-methoxyphenyl)piperazin-1-yl]methanone Chemical compound C1=CC(OC)=CC=C1N1CCN(C(=O)C=2SC=3C4=CC=CC=C4S(=O)(=O)CC=3C=2)CC1 YVIJPELUPZUEJX-UHFFFAOYSA-N 0.000 description 87
- 150000001875 compounds Chemical class 0.000 description 66
- 102100040277 Acyl-protein thioesterase 2 Human genes 0.000 description 58
- 101710132083 Acyl-protein thioesterase 2 Proteins 0.000 description 56
- 230000010076 replication Effects 0.000 description 52
- 239000003053 toxin Substances 0.000 description 49
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 48
- 230000005764 inhibitory process Effects 0.000 description 48
- 231100000765 toxin Toxicity 0.000 description 47
- 108700012359 toxins Proteins 0.000 description 47
- 230000000694 effects Effects 0.000 description 43
- -1 tert Chemical group 0.000 description 40
- 241000193738 Bacillus anthracis Species 0.000 description 35
- 229940079593 drug Drugs 0.000 description 32
- 101000796085 Homo sapiens Anthrax toxin receptor 2 Proteins 0.000 description 28
- 238000003556 assay Methods 0.000 description 27
- 102100031325 Anthrax toxin receptor 2 Human genes 0.000 description 26
- 238000002474 experimental method Methods 0.000 description 25
- 102100040280 Acyl-protein thioesterase 1 Human genes 0.000 description 24
- 108020000999 Viral RNA Proteins 0.000 description 24
- 235000019439 ethyl acetate Nutrition 0.000 description 24
- 231100000673 dose–response relationship Toxicity 0.000 description 22
- 231100000566 intoxication Toxicity 0.000 description 22
- 230000035987 intoxication Effects 0.000 description 22
- 108050009514 Antigen peptide transporter 1 Proteins 0.000 description 21
- 230000003834 intracellular effect Effects 0.000 description 21
- 238000001262 western blot Methods 0.000 description 21
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 20
- 101710194807 Protective antigen Proteins 0.000 description 20
- 230000015572 biosynthetic process Effects 0.000 description 20
- 238000011081 inoculation Methods 0.000 description 20
- 238000011002 quantification Methods 0.000 description 20
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 18
- 230000002401 inhibitory effect Effects 0.000 description 18
- 230000001413 cellular effect Effects 0.000 description 17
- IAZDPXIOMUYVGZ-WFGJKAKNSA-N Dimethyl sulfoxide Chemical compound [2H]C([2H])([2H])S(=O)C([2H])([2H])[2H] IAZDPXIOMUYVGZ-WFGJKAKNSA-N 0.000 description 16
- 208000025721 COVID-19 Diseases 0.000 description 15
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 14
- 108020004459 Small interfering RNA Proteins 0.000 description 14
- 238000004458 analytical method Methods 0.000 description 14
- 238000003818 flash chromatography Methods 0.000 description 14
- 239000000463 material Substances 0.000 description 14
- 239000006228 supernatant Substances 0.000 description 14
- 101710118046 RNA-directed RNA polymerase Proteins 0.000 description 13
- 210000004962 mammalian cell Anatomy 0.000 description 13
- 239000000243 solution Substances 0.000 description 13
- 230000008685 targeting Effects 0.000 description 13
- 108091027544 Subgenomic mRNA Proteins 0.000 description 12
- 239000000203 mixture Substances 0.000 description 12
- 108090000623 proteins and genes Proteins 0.000 description 12
- 239000007787 solid Substances 0.000 description 12
- XDTMQSROBMDMFD-UHFFFAOYSA-N Cyclohexane Chemical compound C1CCCCC1 XDTMQSROBMDMFD-UHFFFAOYSA-N 0.000 description 11
- 230000037396 body weight Effects 0.000 description 11
- 230000006069 depalmitoylation Effects 0.000 description 11
- 239000003480 eluent Substances 0.000 description 11
- 239000011148 porous material Substances 0.000 description 11
- 238000000746 purification Methods 0.000 description 11
- 230000003833 cell viability Effects 0.000 description 10
- 230000014509 gene expression Effects 0.000 description 10
- 210000004072 lung Anatomy 0.000 description 10
- 239000002609 medium Substances 0.000 description 10
- 244000052769 pathogen Species 0.000 description 10
- NSGDYZCDUPSTQT-UHFFFAOYSA-N N-[5-bromo-1-[(4-fluorophenyl)methyl]-4-methyl-2-oxopyridin-3-yl]cycloheptanecarboxamide Chemical compound Cc1c(Br)cn(Cc2ccc(F)cc2)c(=O)c1NC(=O)C1CCCCCC1 NSGDYZCDUPSTQT-UHFFFAOYSA-N 0.000 description 9
- 208000037847 SARS-CoV-2-infection Diseases 0.000 description 9
- 238000001114 immunoprecipitation Methods 0.000 description 9
- 230000001717 pathogenic effect Effects 0.000 description 9
- 238000011156 evaluation Methods 0.000 description 8
- 210000001035 gastrointestinal tract Anatomy 0.000 description 8
- 241000701074 Human alphaherpesvirus 2 Species 0.000 description 7
- 241000699670 Mus sp. Species 0.000 description 7
- 238000011529 RT qPCR Methods 0.000 description 7
- 230000006191 S-acylation Effects 0.000 description 7
- 239000011324 bead Substances 0.000 description 7
- 230000000903 blocking effect Effects 0.000 description 7
- 125000004432 carbon atom Chemical group C* 0.000 description 7
- 230000001404 mediated effect Effects 0.000 description 7
- 108020004999 messenger RNA Proteins 0.000 description 7
- 102000004169 proteins and genes Human genes 0.000 description 7
- 239000000523 sample Substances 0.000 description 7
- 108091034117 Oligonucleotide Proteins 0.000 description 6
- 241000711975 Vesicular stomatitis virus Species 0.000 description 6
- 210000004556 brain Anatomy 0.000 description 6
- 231100000433 cytotoxic Toxicity 0.000 description 6
- 230000001472 cytotoxic effect Effects 0.000 description 6
- 230000001419 dependent effect Effects 0.000 description 6
- 201000010099 disease Diseases 0.000 description 6
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 6
- 239000002245 particle Substances 0.000 description 6
- 108020003175 receptors Proteins 0.000 description 6
- 102000005962 receptors Human genes 0.000 description 6
- 230000001360 synchronised effect Effects 0.000 description 6
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 6
- 238000011740 C57BL/6 mouse Methods 0.000 description 5
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 5
- 241000699666 Mus <mouse, genus> Species 0.000 description 5
- 239000000872 buffer Substances 0.000 description 5
- 239000013592 cell lysate Substances 0.000 description 5
- 230000012202 endocytosis Effects 0.000 description 5
- 238000000684 flow cytometry Methods 0.000 description 5
- XXSMGPRMXLTPCZ-UHFFFAOYSA-N hydroxychloroquine Chemical compound ClC1=CC=C2C(NC(C)CCCN(CCO)CC)=CC=NC2=C1 XXSMGPRMXLTPCZ-UHFFFAOYSA-N 0.000 description 5
- 229960004171 hydroxychloroquine Drugs 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 230000004048 modification Effects 0.000 description 5
- 238000012986 modification Methods 0.000 description 5
- 230000003389 potentiating effect Effects 0.000 description 5
- 239000000843 powder Substances 0.000 description 5
- 235000018102 proteins Nutrition 0.000 description 5
- 239000011541 reaction mixture Substances 0.000 description 5
- 210000002345 respiratory system Anatomy 0.000 description 5
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 4
- 102000003855 L-lactate dehydrogenase Human genes 0.000 description 4
- 108700023483 L-lactate dehydrogenases Proteins 0.000 description 4
- 229940124647 MEK inhibitor Drugs 0.000 description 4
- 239000007832 Na2SO4 Substances 0.000 description 4
- 108090001074 Nucleocapsid Proteins Proteins 0.000 description 4
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 4
- 125000003342 alkenyl group Chemical group 0.000 description 4
- 238000000540 analysis of variance Methods 0.000 description 4
- 230000036772 blood pressure Effects 0.000 description 4
- 239000012267 brine Substances 0.000 description 4
- 239000002775 capsule Substances 0.000 description 4
- 230000003013 cytotoxicity Effects 0.000 description 4
- 231100000135 cytotoxicity Toxicity 0.000 description 4
- 238000009472 formulation Methods 0.000 description 4
- 239000001963 growth medium Substances 0.000 description 4
- 239000010410 layer Substances 0.000 description 4
- 239000006166 lysate Substances 0.000 description 4
- 239000012139 lysis buffer Substances 0.000 description 4
- 238000004519 manufacturing process Methods 0.000 description 4
- 238000012544 monitoring process Methods 0.000 description 4
- 238000001543 one-way ANOVA Methods 0.000 description 4
- 210000000056 organ Anatomy 0.000 description 4
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 4
- 230000001681 protective effect Effects 0.000 description 4
- 229910052938 sodium sulfate Inorganic materials 0.000 description 4
- 235000011152 sodium sulphate Nutrition 0.000 description 4
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical compound O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 4
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 4
- 125000003107 substituted aryl group Chemical group 0.000 description 4
- 229910052717 sulfur Inorganic materials 0.000 description 4
- 230000001988 toxicity Effects 0.000 description 4
- 231100000419 toxicity Toxicity 0.000 description 4
- 239000003981 vehicle Substances 0.000 description 4
- VZSRBBMJRBPUNF-UHFFFAOYSA-N 2-(2,3-dihydro-1H-inden-2-ylamino)-N-[3-oxo-3-(2,4,6,7-tetrahydrotriazolo[4,5-c]pyridin-5-yl)propyl]pyrimidine-5-carboxamide Chemical compound C1C(CC2=CC=CC=C12)NC1=NC=C(C=N1)C(=O)NCCC(N1CC2=C(CC1)NN=N2)=O VZSRBBMJRBPUNF-UHFFFAOYSA-N 0.000 description 3
- DADSQEKJVIEJHZ-UHFFFAOYSA-N 4-chloro-2h-thiochromene-3-carbaldehyde Chemical compound C1=CC=C2C(Cl)=C(C=O)CSC2=C1 DADSQEKJVIEJHZ-UHFFFAOYSA-N 0.000 description 3
- FPDPHPRACHJYMB-UHFFFAOYSA-N 5,5-dioxo-4h-thieno[3,2-c]thiochromene-2-carboxylic acid Chemical compound C1S(=O)(=O)C2=CC=CC=C2C2=C1C=C(C(=O)O)S2 FPDPHPRACHJYMB-UHFFFAOYSA-N 0.000 description 3
- 101500000960 Bacillus anthracis Protective antigen PA-63 Proteins 0.000 description 3
- UHOVQNZJYSORNB-UHFFFAOYSA-N Benzene Chemical compound C1=CC=CC=C1 UHOVQNZJYSORNB-UHFFFAOYSA-N 0.000 description 3
- 241000494545 Cordyline virus 2 Species 0.000 description 3
- 102100023266 Dual specificity mitogen-activated protein kinase kinase 2 Human genes 0.000 description 3
- 101710146529 Dual specificity mitogen-activated protein kinase kinase 2 Proteins 0.000 description 3
- 101001038521 Homo sapiens Acyl-protein thioesterase 2 Proteins 0.000 description 3
- 108010052285 Membrane Proteins Proteins 0.000 description 3
- 102000018697 Membrane Proteins Human genes 0.000 description 3
- 241000283973 Oryctolagus cuniculus Species 0.000 description 3
- 229930182555 Penicillin Natural products 0.000 description 3
- RWRDLPDLKQPQOW-UHFFFAOYSA-N Pyrrolidine Chemical compound C1CCNC1 RWRDLPDLKQPQOW-UHFFFAOYSA-N 0.000 description 3
- 241000700584 Simplexvirus Species 0.000 description 3
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 3
- 230000019552 anatomical structure morphogenesis Effects 0.000 description 3
- 230000006287 biotinylation Effects 0.000 description 3
- 238000007413 biotinylation Methods 0.000 description 3
- 239000000969 carrier Substances 0.000 description 3
- 230000015556 catabolic process Effects 0.000 description 3
- 230000007541 cellular toxicity Effects 0.000 description 3
- 238000005119 centrifugation Methods 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 229910052801 chlorine Inorganic materials 0.000 description 3
- 239000012228 culture supernatant Substances 0.000 description 3
- 238000006731 degradation reaction Methods 0.000 description 3
- 229910052731 fluorine Inorganic materials 0.000 description 3
- 125000005842 heteroatom Chemical group 0.000 description 3
- 229910052740 iodine Inorganic materials 0.000 description 3
- 231100000252 nontoxic Toxicity 0.000 description 3
- 230000003000 nontoxic effect Effects 0.000 description 3
- 239000012044 organic layer Substances 0.000 description 3
- 229910052760 oxygen Inorganic materials 0.000 description 3
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 3
- 230000002829 reductive effect Effects 0.000 description 3
- 239000012679 serum free medium Substances 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 210000000952 spleen Anatomy 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 230000009885 systemic effect Effects 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 238000001890 transfection Methods 0.000 description 3
- 230000002485 urinary effect Effects 0.000 description 3
- 210000002845 virion Anatomy 0.000 description 3
- YMXHPSHLTSZXKH-RVBZMBCESA-N (2,5-dioxopyrrolidin-1-yl) 5-[(3as,4s,6ar)-2-oxo-1,3,3a,4,6,6a-hexahydrothieno[3,4-d]imidazol-4-yl]pentanoate Chemical compound C([C@H]1[C@H]2NC(=O)N[C@H]2CS1)CCCC(=O)ON1C(=O)CCC1=O YMXHPSHLTSZXKH-RVBZMBCESA-N 0.000 description 2
- AMFDITJFBUXZQN-KUBHLMPHSA-N (2s,3s,4r,5r)-2-(4-amino-5h-pyrrolo[3,2-d]pyrimidin-7-yl)-5-(hydroxymethyl)pyrrolidine-3,4-diol Chemical compound C=1NC=2C(N)=NC=NC=2C=1[C@@H]1N[C@H](CO)[C@@H](O)[C@H]1O AMFDITJFBUXZQN-KUBHLMPHSA-N 0.000 description 2
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 2
- DPRAYRYQQAXQPE-UHFFFAOYSA-N 2-bromohexadecanoic acid Chemical compound CCCCCCCCCCCCCCC(Br)C(O)=O DPRAYRYQQAXQPE-UHFFFAOYSA-N 0.000 description 2
- 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
- 102100025601 60S ribosomal protein L27 Human genes 0.000 description 2
- 102000007469 Actins Human genes 0.000 description 2
- 108010085238 Actins Proteins 0.000 description 2
- PAYRUJLWNCNPSJ-UHFFFAOYSA-N Aniline Chemical compound NC1=CC=CC=C1 PAYRUJLWNCNPSJ-UHFFFAOYSA-N 0.000 description 2
- 101100517196 Arabidopsis thaliana NRPE1 gene Proteins 0.000 description 2
- 241000711404 Avian avulavirus 1 Species 0.000 description 2
- 231100000699 Bacterial toxin Toxicity 0.000 description 2
- 101100190825 Bos taurus PMEL gene Proteins 0.000 description 2
- 241000282693 Cercopithecidae Species 0.000 description 2
- 208000001528 Coronaviridae Infections Diseases 0.000 description 2
- 241000725619 Dengue virus Species 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 108090000288 Glycoproteins Proteins 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- 208000009889 Herpes Simplex Diseases 0.000 description 2
- 101000719728 Homo sapiens 60S ribosomal protein L27 Proteins 0.000 description 2
- 101001038518 Homo sapiens Acyl-protein thioesterase 1 Proteins 0.000 description 2
- 241000701024 Human betaherpesvirus 5 Species 0.000 description 2
- 241000725303 Human immunodeficiency virus Species 0.000 description 2
- 102100032755 Leupaxin Human genes 0.000 description 2
- 101710156018 Leupaxin Proteins 0.000 description 2
- 208000025370 Middle East respiratory syndrome Diseases 0.000 description 2
- 241000714177 Murine leukemia virus Species 0.000 description 2
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine (DIPEA) Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 2
- 101100073341 Oryza sativa subsp. japonica KAO gene Proteins 0.000 description 2
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- GLUUGHFHXGJENI-UHFFFAOYSA-N Piperazine Chemical compound C1CNCCN1 GLUUGHFHXGJENI-UHFFFAOYSA-N 0.000 description 2
- NQRYJNQNLNOLGT-UHFFFAOYSA-N Piperidine Chemical compound C1CCNCC1 NQRYJNQNLNOLGT-UHFFFAOYSA-N 0.000 description 2
- 229920002556 Polyethylene Glycol 300 Polymers 0.000 description 2
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 2
- 241000710961 Semliki Forest virus Species 0.000 description 2
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 2
- 108010090804 Streptavidin Proteins 0.000 description 2
- 238000000692 Student's t-test Methods 0.000 description 2
- 102000006463 Talin Human genes 0.000 description 2
- 108010083809 Talin Proteins 0.000 description 2
- HEDRZPFGACZZDS-MICDWDOJSA-N Trichloro(2H)methane Chemical compound [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 2
- 102000003970 Vinculin Human genes 0.000 description 2
- 108090000384 Vinculin Proteins 0.000 description 2
- 108010067390 Viral Proteins Proteins 0.000 description 2
- 241000907316 Zika virus Species 0.000 description 2
- 238000005917 acylation reaction Methods 0.000 description 2
- 125000002877 alkyl aryl group Chemical group 0.000 description 2
- 125000005213 alkyl heteroaryl group Chemical group 0.000 description 2
- 150000001408 amides Chemical class 0.000 description 2
- RDOXTESZEPMUJZ-UHFFFAOYSA-N anisole Chemical compound COC1=CC=CC=C1 RDOXTESZEPMUJZ-UHFFFAOYSA-N 0.000 description 2
- 230000000840 anti-viral effect Effects 0.000 description 2
- 239000000688 bacterial toxin Substances 0.000 description 2
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N biotin Natural products N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 2
- 229910052794 bromium Inorganic materials 0.000 description 2
- 125000000484 butyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- 125000003917 carbamoyl group Chemical group [H]N([H])C(*)=O 0.000 description 2
- 125000002837 carbocyclic group Chemical group 0.000 description 2
- 229910052799 carbon Inorganic materials 0.000 description 2
- 238000003570 cell viability assay Methods 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 239000013000 chemical inhibitor Substances 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 239000000460 chlorine Substances 0.000 description 2
- 125000001309 chloro group Chemical group Cl* 0.000 description 2
- 238000000749 co-immunoprecipitation Methods 0.000 description 2
- 210000004748 cultured cell Anatomy 0.000 description 2
- 125000000113 cyclohexyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 2
- 125000000151 cysteine group Chemical class N[C@@H](CS)C(=O)* 0.000 description 2
- 210000004292 cytoskeleton Anatomy 0.000 description 2
- 238000002784 cytotoxicity assay Methods 0.000 description 2
- 231100000263 cytotoxicity test Toxicity 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 238000010494 dissociation reaction Methods 0.000 description 2
- 230000005593 dissociations Effects 0.000 description 2
- 239000002552 dosage form Substances 0.000 description 2
- 238000012377 drug delivery Methods 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- 150000002240 furans Chemical group 0.000 description 2
- 230000004927 fusion Effects 0.000 description 2
- 230000030279 gene silencing Effects 0.000 description 2
- 230000036039 immunity Effects 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 239000012678 infectious agent Substances 0.000 description 2
- 230000002458 infectious effect Effects 0.000 description 2
- 239000002054 inoculum Substances 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 238000002372 labelling Methods 0.000 description 2
- 210000002429 large intestine Anatomy 0.000 description 2
- 229940121292 leronlimab Drugs 0.000 description 2
- 238000012417 linear regression Methods 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 238000004020 luminiscence type Methods 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- CKJNUZNMWOVDFN-UHFFFAOYSA-N methanone Chemical compound O=[CH-] CKJNUZNMWOVDFN-UHFFFAOYSA-N 0.000 description 2
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 2
- 238000000386 microscopy Methods 0.000 description 2
- 125000001624 naphthyl group Chemical group 0.000 description 2
- 210000001331 nose Anatomy 0.000 description 2
- 125000002347 octyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- 229940049954 penicillin Drugs 0.000 description 2
- XHXFXVLFKHQFAL-UHFFFAOYSA-N phosphoryl trichloride Chemical compound ClP(Cl)(Cl)=O XHXFXVLFKHQFAL-UHFFFAOYSA-N 0.000 description 2
- 125000004194 piperazin-1-yl group Chemical group [H]N1C([H])([H])C([H])([H])N(*)C([H])([H])C1([H])[H] 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 238000012809 post-inoculation Methods 0.000 description 2
- 210000005000 reproductive tract Anatomy 0.000 description 2
- 230000002441 reversible effect Effects 0.000 description 2
- 101150005492 rpe1 gene Proteins 0.000 description 2
- 230000006807 siRNA silencing Effects 0.000 description 2
- 210000000813 small intestine Anatomy 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 229960005322 streptomycin Drugs 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 238000013268 sustained release Methods 0.000 description 2
- 239000012730 sustained-release form Substances 0.000 description 2
- 238000004448 titration Methods 0.000 description 2
- 210000003437 trachea Anatomy 0.000 description 2
- 230000001960 triggered effect Effects 0.000 description 2
- 210000001635 urinary tract Anatomy 0.000 description 2
- 210000004291 uterus Anatomy 0.000 description 2
- 230000007502 viral entry Effects 0.000 description 2
- 230000029812 viral genome replication Effects 0.000 description 2
- LXJXRIRHZLFYRP-VKHMYHEASA-L (R)-2-Hydroxy-3-(phosphonooxy)-propanal Natural products O=C[C@H](O)COP([O-])([O-])=O LXJXRIRHZLFYRP-VKHMYHEASA-L 0.000 description 1
- JHICIIDUQBMMRM-UHFFFAOYSA-N 1-(2,5-dichlorophenyl)piperazine Chemical compound ClC1=CC=C(Cl)C(N2CCNCC2)=C1 JHICIIDUQBMMRM-UHFFFAOYSA-N 0.000 description 1
- JIJMTXRHRBBARH-UHFFFAOYSA-N 1-(2,6-dichlorophenyl)piperazine Chemical compound ClC1=CC=CC(Cl)=C1N1CCNCC1 JIJMTXRHRBBARH-UHFFFAOYSA-N 0.000 description 1
- JDVUSTNITSGJOH-UHFFFAOYSA-N 1-(2,6-dimethylphenyl)piperazine Chemical compound CC1=CC=CC(C)=C1N1CCNCC1 JDVUSTNITSGJOH-UHFFFAOYSA-N 0.000 description 1
- PWZDJIUQHUGFRJ-UHFFFAOYSA-N 1-(2-chlorophenyl)piperazine Chemical compound ClC1=CC=CC=C1N1CCNCC1 PWZDJIUQHUGFRJ-UHFFFAOYSA-N 0.000 description 1
- WICKLEOONJPMEQ-UHFFFAOYSA-N 1-(2-methylphenyl)piperazine Chemical compound CC1=CC=CC=C1N1CCNCC1 WICKLEOONJPMEQ-UHFFFAOYSA-N 0.000 description 1
- AVJKDKWRVSSJPK-UHFFFAOYSA-N 1-(4-fluorophenyl)piperazine Chemical compound C1=CC(F)=CC=C1N1CCNCC1 AVJKDKWRVSSJPK-UHFFFAOYSA-N 0.000 description 1
- PVOAHINGSUIXLS-UHFFFAOYSA-N 1-Methylpiperazine Chemical compound CN1CCNCC1 PVOAHINGSUIXLS-UHFFFAOYSA-N 0.000 description 1
- FJDNYOZEHRTOLO-UHFFFAOYSA-N 1-[2-fluoro-4-(trifluoromethyl)phenyl]piperazine Chemical compound FC1=CC(C(F)(F)F)=CC=C1N1CCNCC1 FJDNYOZEHRTOLO-UHFFFAOYSA-N 0.000 description 1
- UDQMXYJSNNCRAS-UHFFFAOYSA-N 2,3-dichlorophenylpiperazine Chemical compound ClC1=CC=CC(N2CCNCC2)=C1Cl UDQMXYJSNNCRAS-UHFFFAOYSA-N 0.000 description 1
- CVQSWZMJOGOPAV-UHFFFAOYSA-N 2,3-dihydrothiochromen-4-one Chemical compound C1=CC=C2C(=O)CCSC2=C1 CVQSWZMJOGOPAV-UHFFFAOYSA-N 0.000 description 1
- KTHZVNMUARCVQZ-UHFFFAOYSA-N 2-(5-methoxyindol-1-yl)ethanamine Chemical compound COC1=CC=C2N(CCN)C=CC2=C1 KTHZVNMUARCVQZ-UHFFFAOYSA-N 0.000 description 1
- IZXIZTKNFFYFOF-UHFFFAOYSA-N 2-Oxazolidone Chemical class O=C1NCCO1 IZXIZTKNFFYFOF-UHFFFAOYSA-N 0.000 description 1
- ZCYVEMRRCGMTRW-UHFFFAOYSA-N 7553-56-2 Chemical group [I] ZCYVEMRRCGMTRW-UHFFFAOYSA-N 0.000 description 1
- 241001316595 Acris Species 0.000 description 1
- 101710132086 Acyl-protein thioesterase 1 Proteins 0.000 description 1
- 229920000936 Agarose Polymers 0.000 description 1
- 102100031323 Anthrax toxin receptor 1 Human genes 0.000 description 1
- WKBOTKDWSSQWDR-UHFFFAOYSA-N Bromine atom Chemical group [Br] WKBOTKDWSSQWDR-UHFFFAOYSA-N 0.000 description 1
- 125000005865 C2-C10alkynyl group Chemical group 0.000 description 1
- 101100356682 Caenorhabditis elegans rho-1 gene Proteins 0.000 description 1
- 229930186147 Cephalosporin Natural products 0.000 description 1
- 241001502567 Chikungunya virus Species 0.000 description 1
- 241000606153 Chlamydia trachomatis Species 0.000 description 1
- LXJXRIRHZLFYRP-VKHMYHEASA-N D-glyceraldehyde 3-phosphate Chemical compound O=C[C@H](O)COP(O)(O)=O LXJXRIRHZLFYRP-VKHMYHEASA-N 0.000 description 1
- 206010012735 Diarrhoea Diseases 0.000 description 1
- 206010059866 Drug resistance Diseases 0.000 description 1
- 231100000491 EC50 Toxicity 0.000 description 1
- 241001115402 Ebolavirus Species 0.000 description 1
- 241000283074 Equus asinus Species 0.000 description 1
- 241000588698 Erwinia Species 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 102000013446 GTP Phosphohydrolases Human genes 0.000 description 1
- 108091006109 GTPases Proteins 0.000 description 1
- 108010015899 Glycopeptides Proteins 0.000 description 1
- 102000002068 Glycopeptides Human genes 0.000 description 1
- 241000724675 Hepatitis E virus Species 0.000 description 1
- 101000652582 Homo sapiens Antigen peptide transporter 2 Proteins 0.000 description 1
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 241000712902 Lassa mammarenavirus Species 0.000 description 1
- 241000589242 Legionella pneumophila Species 0.000 description 1
- 208000004023 Legionellosis Diseases 0.000 description 1
- 208000035353 Legionnaires disease Diseases 0.000 description 1
- 208000007764 Legionnaires' Disease Diseases 0.000 description 1
- 108010028921 Lipopeptides Proteins 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- 241000712899 Lymphocytic choriomeningitis mammarenavirus Species 0.000 description 1
- 241001115401 Marburgvirus Species 0.000 description 1
- 241000712079 Measles morbillivirus Species 0.000 description 1
- 102000004232 Mitogen-Activated Protein Kinase Kinases Human genes 0.000 description 1
- 108090000744 Mitogen-Activated Protein Kinase Kinases Proteins 0.000 description 1
- 108010085220 Multiprotein Complexes Proteins 0.000 description 1
- 102000007474 Multiprotein Complexes Human genes 0.000 description 1
- 241000526636 Nipah henipavirus Species 0.000 description 1
- 229930040373 Paraformaldehyde Natural products 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 206010035718 Pneumonia legionella Diseases 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 229940096437 Protein S Drugs 0.000 description 1
- 241000589516 Pseudomonas Species 0.000 description 1
- 241000150264 Puumala orthohantavirus Species 0.000 description 1
- 238000001190 Q-PCR Methods 0.000 description 1
- 101150111584 RHOA gene Proteins 0.000 description 1
- 238000002123 RNA extraction Methods 0.000 description 1
- 241000711798 Rabies lyssavirus Species 0.000 description 1
- 241000725643 Respiratory syncytial virus Species 0.000 description 1
- 102100037486 Reverse transcriptase/ribonuclease H Human genes 0.000 description 1
- 229930189077 Rifamycin Natural products 0.000 description 1
- 241000008907 Rousettus bat coronavirus HKU9 Species 0.000 description 1
- 241001138501 Salmonella enterica Species 0.000 description 1
- 101000629318 Severe acute respiratory syndrome coronavirus 2 Spike glycoprotein Proteins 0.000 description 1
- 241000607768 Shigella Species 0.000 description 1
- 241000713311 Simian immunodeficiency virus Species 0.000 description 1
- 241000710960 Sindbis virus Species 0.000 description 1
- VMHLLURERBWHNL-UHFFFAOYSA-M Sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 1
- 101710198474 Spike protein Proteins 0.000 description 1
- 108010034396 Streptogramins Proteins 0.000 description 1
- 241000282898 Sus scrofa Species 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- 101710120037 Toxin CcdB Proteins 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- 231100000230 acceptable toxicity Toxicity 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 210000001552 airway epithelial cell Anatomy 0.000 description 1
- 125000005119 alkyl cycloalkyl group Chemical group 0.000 description 1
- 108700038111 alunacedase alfa Proteins 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 235000001014 amino acid Nutrition 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 229940126575 aminoglycoside Drugs 0.000 description 1
- 108010039069 anthrax toxin receptors Proteins 0.000 description 1
- 230000000845 anti-microbial effect Effects 0.000 description 1
- 230000001147 anti-toxic effect Effects 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 239000004599 antimicrobial Substances 0.000 description 1
- 229940121357 antivirals Drugs 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 229940065181 bacillus anthracis Drugs 0.000 description 1
- PXXJHWLDUBFPOL-UHFFFAOYSA-N benzamidine Chemical compound NC(=N)C1=CC=CC=C1 PXXJHWLDUBFPOL-UHFFFAOYSA-N 0.000 description 1
- 125000002619 bicyclic group Chemical group 0.000 description 1
- 125000004369 butenyl group Chemical group C(=CCC)* 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- 238000010804 cDNA synthesis Methods 0.000 description 1
- 150000001732 carboxylic acid derivatives Chemical class 0.000 description 1
- 239000012876 carrier material Substances 0.000 description 1
- 238000000423 cell based assay Methods 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 239000012094 cell viability reagent Substances 0.000 description 1
- 230000005889 cellular cytotoxicity Effects 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- 229940124587 cephalosporin Drugs 0.000 description 1
- 150000001780 cephalosporins Chemical class 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 239000003067 chemokine receptor CCR5 antagonist Substances 0.000 description 1
- 229940038705 chlamydia trachomatis Drugs 0.000 description 1
- 229960005091 chloramphenicol Drugs 0.000 description 1
- 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 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 238000011490 co-immunoprecipitation assay Methods 0.000 description 1
- 238000004440 column chromatography Methods 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 229940125782 compound 2 Drugs 0.000 description 1
- 239000013068 control sample Substances 0.000 description 1
- 238000013270 controlled release Methods 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 239000012043 crude product Substances 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- 125000004210 cyclohexylmethyl group Chemical group [H]C([H])(*)C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- 230000000120 cytopathologic effect Effects 0.000 description 1
- 210000000172 cytosol Anatomy 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 230000002354 daily effect Effects 0.000 description 1
- 230000020176 deacylation Effects 0.000 description 1
- 238000005947 deacylation reaction Methods 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 125000002704 decyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 238000010217 densitometric analysis Methods 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 239000008298 dragée Substances 0.000 description 1
- 229940088679 drug related substance Drugs 0.000 description 1
- 230000029578 entry into host Effects 0.000 description 1
- 230000026502 entry into host cell Effects 0.000 description 1
- PVBRSNZAOAJRKO-UHFFFAOYSA-N ethyl 2-sulfanylacetate Chemical compound CCOC(=O)CS PVBRSNZAOAJRKO-UHFFFAOYSA-N 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- PQVSTLUFSYVLTO-UHFFFAOYSA-N ethyl n-ethoxycarbonylcarbamate Chemical compound CCOC(=O)NC(=O)OCC PQVSTLUFSYVLTO-UHFFFAOYSA-N 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- ZCGNOVWYSGBHAU-UHFFFAOYSA-N favipiravir Chemical compound NC(=O)C1=NC(F)=CNC1=O ZCGNOVWYSGBHAU-UHFFFAOYSA-N 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 1
- 125000001153 fluoro group Chemical group F* 0.000 description 1
- 229950002031 galidesivir Drugs 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 125000001188 haloalkyl group Chemical group 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 125000003187 heptyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 125000004404 heteroalkyl group Chemical group 0.000 description 1
- IPCSVZSSVZVIGE-UHFFFAOYSA-M hexadecanoate Chemical compound CCCCCCCCCCCCCCCC([O-])=O IPCSVZSSVZVIGE-UHFFFAOYSA-M 0.000 description 1
- 125000006038 hexenyl group Chemical group 0.000 description 1
- 125000004051 hexyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 102000057851 human LYPLA2 Human genes 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000037041 intracellular level Effects 0.000 description 1
- 238000007917 intracranial administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 description 1
- 125000001972 isopentyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 229940115932 legionella pneumophila Drugs 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- 229940041028 lincosamides Drugs 0.000 description 1
- 230000029226 lipidation Effects 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- GLXDVVHUTZTUQK-UHFFFAOYSA-M lithium hydroxide monohydrate Substances [Li+].O.[OH-] GLXDVVHUTZTUQK-UHFFFAOYSA-M 0.000 description 1
- 229940040692 lithium hydroxide monohydrate Drugs 0.000 description 1
- 210000005265 lung cell Anatomy 0.000 description 1
- 239000003120 macrolide antibiotic agent Substances 0.000 description 1
- 229940041033 macrolides Drugs 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 230000008172 membrane trafficking Effects 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 1
- UZKWTJUDCOPSNM-UHFFFAOYSA-N methoxybenzene Substances CCCCOC=C UZKWTJUDCOPSNM-UHFFFAOYSA-N 0.000 description 1
- HTNPEHXGEKVIHG-ZJTJHKMLSA-N molnupiravir Chemical compound CC(C)C(=O)OC[C@H]1O[C@H](C(O)C1O)N1C=C\C(NC1=O)=N\O HTNPEHXGEKVIHG-ZJTJHKMLSA-N 0.000 description 1
- 229940075124 molnupiravir Drugs 0.000 description 1
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000003136 n-heptyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000000740 n-pentyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 108700043045 nanoluc Proteins 0.000 description 1
- 239000006199 nebulizer Substances 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 125000001971 neopentyl group Chemical group [H]C([*])([H])C(C([H])([H])[H])(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 239000002547 new drug Substances 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 239000012299 nitrogen atmosphere Substances 0.000 description 1
- 125000001400 nonyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 125000002868 norbornyl group Chemical group C12(CCC(CC1)C2)* 0.000 description 1
- 238000001821 nucleic acid purification Methods 0.000 description 1
- 125000004365 octenyl group Chemical group C(=CCCCCCC)* 0.000 description 1
- 229920002113 octoxynol Polymers 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 230000003204 osmotic effect Effects 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 230000026792 palmitoylation Effects 0.000 description 1
- 229920002866 paraformaldehyde Polymers 0.000 description 1
- 244000045947 parasite Species 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000007110 pathogen host interaction Effects 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 150000002960 penicillins Chemical class 0.000 description 1
- 125000002255 pentenyl group Chemical group C(=CCCC)* 0.000 description 1
- VUNXBQRNMNVUMV-UHFFFAOYSA-N phenyl(piperazin-1-yl)methanone Chemical compound C=1C=CC=CC=1C(=O)N1CCNCC1 VUNXBQRNMNVUMV-UHFFFAOYSA-N 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 231100000683 possible toxicity Toxicity 0.000 description 1
- 238000010149 post-hoc-test Methods 0.000 description 1
- 230000001323 posttranslational effect Effects 0.000 description 1
- 238000011533 pre-incubation Methods 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- 125000004368 propenyl group Chemical group C(=CC)* 0.000 description 1
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 239000003531 protein hydrolysate Substances 0.000 description 1
- 230000005180 public health Effects 0.000 description 1
- 238000004445 quantitative analysis Methods 0.000 description 1
- 150000007660 quinolones Chemical class 0.000 description 1
- 230000007115 recruitment Effects 0.000 description 1
- 238000004064 recycling Methods 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- RWWYLEGWBNMMLJ-YSOARWBDSA-N remdesivir Chemical compound NC1=NC=NN2C1=CC=C2[C@]1([C@@H]([C@@H]([C@H](O1)CO[P@](=O)(OC1=CC=CC=C1)N[C@H](C(=O)OCC(CC)CC)C)O)O)C#N RWWYLEGWBNMMLJ-YSOARWBDSA-N 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- BTVYFIMKUHNOBZ-QXMMDKDBSA-N rifamycin s Chemical class O=C1C(C(O)=C2C)=C3C(=O)C=C1NC(=O)\C(C)=C/C=C\C(C)C(O)C(C)C(O)C(C)C(OC(C)=O)C(C)C(OC)\C=C/OC1(C)OC2=C3C1=O BTVYFIMKUHNOBZ-QXMMDKDBSA-N 0.000 description 1
- 229940081192 rifamycins Drugs 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 150000003335 secondary amines Chemical class 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 230000001568 sexual effect Effects 0.000 description 1
- 239000002002 slurry Substances 0.000 description 1
- 235000017281 sodium acetate Nutrition 0.000 description 1
- 239000001632 sodium acetate Substances 0.000 description 1
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 1
- 235000017557 sodium bicarbonate Nutrition 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- QDRKDTQENPPHOJ-UHFFFAOYSA-N sodium ethoxide Chemical compound [Na+].CC[O-] QDRKDTQENPPHOJ-UHFFFAOYSA-N 0.000 description 1
- VZWGHDYJGOMEKT-UHFFFAOYSA-J sodium pyrophosphate decahydrate Chemical compound O.O.O.O.O.O.O.O.O.O.[Na+].[Na+].[Na+].[Na+].[O-]P([O-])(=O)OP([O-])([O-])=O VZWGHDYJGOMEKT-UHFFFAOYSA-J 0.000 description 1
- AKHNMLFCWUSKQB-UHFFFAOYSA-L sodium thiosulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=S AKHNMLFCWUSKQB-UHFFFAOYSA-L 0.000 description 1
- 235000019345 sodium thiosulphate Nutrition 0.000 description 1
- IBKZNJXGCYVTBZ-IDBHZBAZSA-M sodium;1-[3-[2-[5-[(3as,4s,6ar)-2-oxo-1,3,3a,4,6,6a-hexahydrothieno[3,4-d]imidazol-4-yl]pentanoylamino]ethyldisulfanyl]propanoyloxy]-2,5-dioxopyrrolidine-3-sulfonate Chemical compound [Na+].O=C1C(S(=O)(=O)[O-])CC(=O)N1OC(=O)CCSSCCNC(=O)CCCC[C@H]1[C@H]2NC(=O)N[C@H]2CS1 IBKZNJXGCYVTBZ-IDBHZBAZSA-M 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 239000012192 staining solution Substances 0.000 description 1
- 108010051423 streptavidin-agarose Proteins 0.000 description 1
- 229940041030 streptogramins Drugs 0.000 description 1
- 125000001424 substituent group Chemical group 0.000 description 1
- 229940124530 sulfonamide Drugs 0.000 description 1
- 150000003456 sulfonamides Chemical class 0.000 description 1
- 125000004434 sulfur atom Chemical group 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 230000031068 symbiosis, encompassing mutualism through parasitism Effects 0.000 description 1
- 208000011580 syndromic disease Diseases 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 229940040944 tetracyclines Drugs 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 description 1
- 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 description 1
- 229960001082 trimethoprim Drugs 0.000 description 1
- GPRLSGONYQIRFK-MNYXATJNSA-N triton Chemical compound [3H+] GPRLSGONYQIRFK-MNYXATJNSA-N 0.000 description 1
- 230000010415 tropism Effects 0.000 description 1
- 238000007492 two-way ANOVA Methods 0.000 description 1
- KCFYEAOKVJSACF-UHFFFAOYSA-N umifenovir Chemical compound CN1C2=CC(Br)=C(O)C(CN(C)C)=C2C(C(=O)OCC)=C1CSC1=CC=CC=C1 KCFYEAOKVJSACF-UHFFFAOYSA-N 0.000 description 1
- 229960004626 umifenovir Drugs 0.000 description 1
- 241000712461 unidentified influenza virus Species 0.000 description 1
- 238000002255 vaccination Methods 0.000 description 1
- 229960005486 vaccine Drugs 0.000 description 1
- 210000003501 vero cell Anatomy 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 125000000391 vinyl group Chemical group [H]C([*])=C([H])[H] 0.000 description 1
- 230000001018 virulence Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 239000012224 working solution Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/496—Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D307/00—Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
- C07D307/02—Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
- C07D307/34—Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
- C07D307/56—Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
- C07D307/68—Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D333/00—Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
- C07D333/02—Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
- C07D333/04—Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
- C07D333/26—Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
- C07D333/38—Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D495/00—Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
- C07D495/02—Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
- C07D495/04—Ortho-condensed systems
Definitions
- the present invention relates to inhibitors of acyl protein thioesterases (APT) and method of preparation thereof. It also relates to the use of inhibitors of APT in the treatment and/or prevention of a microbial infection such as viral or bacterial infections or coinfection by virus and bacteria.
- APT acyl protein thioesterases
- S-acylation a reversible post- translational lipidation
- S-acylation consists of the addition of an acyl chain, often palmitate, to cytosolic cysteines in target proteins via a thioester bond. This modification is reversible and therefore acts as a functional switch on target proteins (Zaballa & van der Goot, 2018). S-acylation and deacylation are crucial for membrane trafficking events and impact the cellular distribution, abundance, and function of proteins. During infection, S-acylation modifies numerous viral proteins from clinically relevant viruses (Veit, 2012), as well as multiple host receptors fundamental for internalization, and therefore virulence, of numerous pathogens and pathogenic molecules.
- Pathogens are highly opportunistic organisms. Viruses often exploit S-acylation of viral proteins or host products to promote pathogenesis.
- CoVs 3-Coronaviruses (CoVs) have enormous zoonotic potential being responsible for various outbreaks of severe acute respiratory syndrome (SARS) in recent years including the pandemic Coronavirus disease-2019 (COVID-19). CoVs depend on S-acylation to produce fully infectious viruses (Mesquita et al, 2021 ). SARS-CoV-2, the etiological agent of COVID 19, contains several essential pathogenic molecules that are modified by S-acylation.
- Vaccines, antibodies and pathogen-directed antibiotics or antivirals are effective therapeutic strategies that work by targeting the specific infectious agent(s) they were designed for.
- Host-directed anti-microbial drugs in the other hand, have the potential to broadly target infectious agents with a lower risk of rapid resistance development (Fernandez-Oliva et al, 2019; Kaufmann et al, 2018).
- the present invention provides an inhibitor of acyl protein thioesterase (APT) of or a pharmaceutically acceptable salt thereof, wherein Ring A is a thiophene ring or a furane ring;
- APT acyl protein thioesterase
- Each RAm is independently selected from the group comprising hydrogen, halogen, Ci- C10 alkyl, C2-C10 alkenyl, C1-C6 alkoxy, C1-C6 acyl, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, C6-C10 aryl, and/or C6-C10 heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups; and m is an integer from 0 to 2;
- Each R1 , R2, R4, R5 is independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, optionally substituted Ci-Ce alkyl acyl, optionally substituted Ci-Ce alkyl aminocarbonyl, optionally substituted carbonyl, optionally substituted Ci-Ce alkyl carbonyl, optionally substituted Ce-C aryl, ether, C2-C10 alkenyl, C6-C10 heteroaryl, C3- C10 cycloalkyl, C3-C10 heterocycloalkyl, Ci-Ce alkoxy, optionally substituted amino, and/or Ci-Ce alkyl amino; and
- R3 is selected from the group comprising
- the invention also provides said inhibitors of APT of formula (II), or pharmaceutically acceptable salts thereof, for use as a medicament.
- the invention provides said inhibitors of APT of formula (II), or pharmaceutically acceptable salts thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
- the invention also relates to a pharmaceutical composition
- a pharmaceutical composition comprising at least one of said inhibitors of APT, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, or excipient.
- the invention also provides a kit comprising said inhibitors of APT, or pharmaceutically acceptable salts thereof, and information for use thereof.
- the invention further provides an inhibitor of APT of formula (ML349), or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
- the invention further provides an inhibitor of APT of formula (ML348), or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
- Anthrax toxin Protective Antigen (PA) PA
- (D): MEK2 degradation. Quantification for each time point for cells pre-treated for 2 h and expressed as % of t 0 for each sample. All results are mean ⁇ SEM.
- Figure 4 (A), (B) and (C): Microscopy-based quantification assay of Vero E6 cells infected with SARS-CoV-2-Spike-pseudotyped particles (VSV-S-CoV2) (A, C) or VSV-G- pseudotyped particles (VSVG) (B).
- C Dose response curve calculated for ML348, ML349, and hydroxychloroquine. Results are mean ⁇ SEM (A,B), or SD (C).
- FIG. 5 (A): Infection with SARS-CoV-2 determined using a plaque assay of Vero E6 cells pre-treated with ML348 or ML349 and infected in the presence of increasing concentrations of each drug. Dose-response curves were obtained using non-linear regression.
- Figure 7 Quantification of viral RNA replication (E, RdRp and E subgenomic) from Vero E6 cells (A) and correspondent supernatants (B) infected with SARS-CoV-2 (MOI- 0.1 for 24 h). Cells were inoculated for 1 h with SARS-CoV-2, washed and subsequently incubated in complete medium with 12.5 pM of ML348 or ML349.
- C Cells were previously transfected with control or siRNA oligos targeting the expression of APT1 (siAPT) for 72 h.
- D Quantification of APT1 mRNA levels from the samples used in C.
- Figure 9 (A): LYPLA1 (APT1 ) and LYPLA2 (APT2) mRNA levels in different cell lines. Values are TAG per Million (TPM) corresponding to 10*6 X (reads mapped to transcript/transcript length)/ SUM (reads mapped to transcript/transcript length).
- TPM TAG per Million
- B and C Quantification of viral RNA replication (E, RdRp and E subgenomic) from Calu-3 cells and correspondent supernatants infected with SARS-CoV-2 (MOI-0.1 for 24 h), treated as indicated.
- D Quantification of APT2 mRNA levels from the samples used in C.
- each data point represents an individual experiment and results are mean (A) or mean ⁇ SEM (B, C, D). All p values comparing RNA levels in relation to control conditions were obtained by (B, C) one-way ANOVA with Dunnet’s multiple comparison or (D) Paired two- tailed student’s t-test (*p ⁇ 0.05, **p ⁇ 0.01 , ****p ⁇ 0.0001 )
- Figure 12 (A): dose response inhibitions of SARS-CoV-2 replication (infection at MOI «0.05-01 ) in human lung-derived Calu-3 cells by ML349-derived compounds C5, C6, and C7, 24 h post-viral inoculation, quantified by QPCR of total viral RNA (E, RdRp and E sub-genomic). (B): levels of housekeeping (HK) transcripts during infection, indicative of cell viability. For each concentration, results are mean ⁇ SEM of n >3. Dose inhibition curves were fitted using nonlinear regression, and EC50 ⁇ SD concentrations are indicated.
- Figure 13 shows replication of SARS-CoV-2 inoculated in human lung-derived Calu-3 cells (MOI «0.5, 1 h), incubated with infection media supplemented with 5 pM of compound C2 and 0.5 pg/ml of P2G3 anti-Spike antibody, from 1 h post viral inoculation.
- Cells were harvested at the indicated time points and infection was determined by monitoring the levels of intracellular (A): genomic (E and RdRp) and (B): sub-genomic E (E-subgen) RNA transcripts by QPCR. Values were normalised to 1 for the timepoint 1 h post viral inoculation (before drug treatment) and results are mean ⁇ SEM of n>3. Data is presented as equivalent side-by-side plot graphs with log or linear scale representation of viral RNA levels - Fold-infection.
- Figure 15(A): dose-response inhibition of replication of SARS-CoV-2 variant Omicron BA.5 (infection at MOI 0.05-01 ) in human lung-derived Calu-3 cells by compound 2 (C2), 24 h post-viral inoculation, quantified by QPCR of total viral RNA (E, RdRp and E sub- genomic).
- B levels of housekeeping (HK) transcripts during infection, indicative of cell viability. For each concentration, results are mean ⁇ SEM of n >3. The dose inhibition curve was fitted using nonlinear regression, and EC50 ⁇ SD concentrations are indicated.
- Figure 16 toxicity evaluation of inhibiting APT2 with ML349 and C2 in vivo in C57BL/6 mice.
- A Body weight (g);
- B blood pressure (Hgmm);
- C pulse (beats per minute - bpm) were monitored for the indicated time points for every mouse mock- treated or treated with 1 mg of ML349 or C2 per 25 g of body weight for several days through 13 days.
- Mock-treated control mice were inoculated in parallel with equivalent volumes of vehicle solution used to resuspend ML349 and C2.
- FIG 17(A) Schematic representation of the APT2-dependent mechanism of synchronized anthrax toxin entry into host target cells.
- the toxin binds to the S-acylated anthrax receptor (CMG2) at the cell surface; this leads to 2, the recruitment and binding of APT2 to CMG2, which, in turn, promotes, 3, the complete de-acylation of the receptor; and 4, its dissociation from the host cytoskeleton components (actin, vinculin, and talin) and binding to the host GTPase RhoA.
- CMG2 S-acylated anthrax receptor
- IP co-immunoprecipitation
- B western blot analysis of CMG2- immunoprecipitation (IP) fractions to monitor the co-immunoprecipitation (co-IP) of APT2- CMG2 complexes from cell lysates of control (untreated) or anthrax toxin-treated cells.
- FIG. 18 Western blot analysis of APT2-CMG2 complexes present in CMG2- immunoprecipitation (IP) fractions from cell lysates of control (untreated) or anthrax toxin- treated cells.
- A RPE-1 cells ectopically expressing APT2-FLAG for 24 h, left untreated or treated with anthrax toxin (protective antigen-PA subunit) in the presence of APT2 inhibitors, ML349 or compound 02, C3, or control drug carrier DMSO. Drugs were added at a final concentration of 2 pM, 4 h before and during toxin treatment.
- B quantification of APT2-FLAG levels in equivalent CMG2 fractions displayed in A.
- FIG. 19 Quantification of APT2-FLAG levels in equivalent CMG2 fractions from western blot analysis of APT2-CMG2 complexes present in CMG2-immunoprecipitation (IP) fractions from cell lysates of control (untreated) or PA-toxin-treated cells: RPE-1 cells ectopically expressing APT2-FLAG for 24 h, left untreated or treated with anthrax toxin (protective antigen-PA subunit) in the presence of APT2 inhibitors, ML349 or compound of formula (II) (C9, C12, C13, C16, and C17), or control drug carrier DMSO. Drugs were added at a final concentration of 5 pM, 4 h before and during toxin treatment. Data are normalized to 100% for WT, and results are mean or mean ⁇ SEM, and each data point represents an individual experiment.
- IP CMG2-immunoprecipitation
- Figure 20 Quantification of western blot analysis of the dynamics of toxin pore formation (PA-Pore) in RPE-1 mammalian cells treated with anthrax toxin (protective antigen-PA subunit) in the presence of APT2 inhibitors (C2, C9, C12, C13, C16 at 5 pM) for the indicated times (0, 30 and 60 min).
- Figure 21 Inhibition of SARS-CoV-2 replication in human lung-derived Calu-3 cells (MOI «0.05-0.1 , 1 h), in the presence of the APT2 inhibitors, ML349, C2, C9, C12, C13, C15, C16 and C17 (used at 5 pM), from 1 h post-viral inoculation. Culture supernatants and infected cells were harvested 24 h post-viral inoculation. The levels of viral nucleocapsid protein N: cell lysates, were monitored by Western blot.
- the present invention relates to an inhibitor of acyl protein thioesterase (APT) of formula (II): or a pharmaceutically acceptable salt thereof, wherein
- Ring A is a thiophene ring or a furane ring
- Each RAm is independently selected from the group comprising hydrogen, halogen, Ci- C10 alkyl, C2-C10 alkenyl, C1-C6 alkoxy, C1-C6 acyl, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, C6-C10 aryl, and/or C6-C10 heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups; and m is an integer from 0 to 2;
- Each R1 , R2, R4, R5 is independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, optionally substituted Ci-Ce alkyl acyl, optionally substituted Ci-Ce alkyl aminocarbonyl, optionally substituted carbony
- R3 is selected from the group comprising
- the ring A is a thiophene ring represented by formula: wherein: each R6, R7 are independently selected from the group comprising hydrogen, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, C6-C10 aryl, and/or C6-C10 heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups.
- the ring A is a thiophene ring represented by formula: wherein: each R6, R7 are independently selected from the group comprising hydrogen, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, wherein said cycloalkyl, or optionally substituted heterocycloalkyl, may be fused with ring A and with 1 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl group. Even more preferably, the ring A is a thiophene ring represented by formula: Even more preferably, the ring A is a thiophene ring represented by formula:
- Halogen refers to a fluorine atom, a chlorine atom, a bromine atom, or an iodine atom.
- C1-C10 alkyl refers to monovalent straight-chained and branched alkyl groups having 1 to 10 carbon atoms. This term is exemplified by groups such as methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec.
- Ci-Cio-heteroalkyl includes both straight-chained and branched C1-C10 alkyl groups according to the definition above, having at least one or more heteroatoms selected from S, O and N.
- Ci-Ce-haloalkyl includes both straight-chained and branched Ci-Ce alkyl groups according to the definition above, having at least one or more halogen selected from F, Cl, Br or I.
- Carbonyl refers to the group -C(O)R where R includes hydrogen, “Ci-Ce- alkyl”, “aryl”, substituted aryl, “heteroaryl”, “Ci-Ce-alkyl aryl”, “Ci-Ce-alkyl heteroaryl”, or substituted amine (-NH-R’) wherein R’ is optionally substituted aryl.
- Ci-Ce alkyl acyl refers to Ci-Ce alkyl groups having a Ci-Ce acyl substituent as defined herein.
- Ci-Ce alkyl carbonyl refers to Ci-Ce alkyl groups having a “carbonyl” substituent as defined herein.
- aminocarbonyl refers to the chemical moiety -C(0)NRR’ in which each of R and R’ is independently hydrogen, Ci-Ce alkyl, aryl, substituted aryl, heteroaryl, Ci-Ce alkyl aryl, or Ci-Ce alkyl heteroaryl.
- Ci-Ce alkyl aminocarbonyl refers to Ci-Ce alkyl groups having an “aminocarbonyl” substituent as defined herein.
- C3-C10 cycloalkyl refers to a saturated carbocyclic group of from 3 to 10 carbon atoms having a single ring (e.g., cyclohexyl) or multiple condensed rings (e.g., norborn yl).
- Preferred cycloalkyl include cyclopentyl, cyclohexyl, norbornyl and the like.
- Ci-Ce alkyl cycloalkyl refers to Ci-Ce-alkyl groups having a cycloalkyl substituent, including cyclohexylmethyl, cyclopentylpropyl, and the like.
- C3-C10 heterocycloalkyl refers to Cs-Cio-cycloalkyl group according to the definition above, in which up to 3 carbon atoms are replaced by heteroatoms chosen from the group consisting of O, S, NR, R being defined as hydrogen or methyl.
- Preferred heterocycloalkyl include cyclohexane in which 1 carbon atom is replaced by S(O)2, pyrrolidine, piperidine, piperazine, 1 -methylpiperazine, and the like.
- optionally substituted C3-C10 heterocycloalkyl refers to a C3-C10 heterocycloalkyl as defined above that may be fused with 0 to 2 further groups selected from cycloalkyl, heterocycloalkyl, aryl or heteroaryl group. Preferred groups are benzene, toluene, phenol, aniline, anisole, and the like.
- Ci-Ce-alkyl heterocycloalkyl refers to Ci-Ce-alkyl groups having a heterocycloalkyl substituent as defined herein.
- C2-Cio-alkenyl refers to alkenyl groups preferably having from 2 to 10 carbon atoms and having at least 1 or 2 sites of alkenyl unsaturation.
- Preferable alkenyl groups include ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl and octenyl, and the like.
- alkenyl in the meaning of the present invention includes the cis and trans isomers.
- Ci-C 6 alkoxy refers to group -O-R’ where R’ includes both straight-chained and branched "Ci-Ce alkyl” or " Ci-Ce haloalkyl” or " Ci-Ce heteroalkyl” or "aryl” or “heteroaryl” or "Ci-Ce-alkyl aryl” or “Ci-Ce-alkyl heteroaryl”.
- Preferred alkoxy groups include by way of example, methoxy, ethoxy, propoxy, butoxy, phenoxy and the like.
- Amino refers to -NRR’ in which each of R and R’ is independently hydrogen, Ci-Ce alkyl, substituted aryl, heteroaryl, Ci-Ce alkyl aryl, Ci-Ce alkyl heteroaryl, cycloalkyl, or heterocycloalkyl.
- Ci-Ce-alkyl amino refers to Ci-Ce-alkyl groups having an amino substituent as defined herein.
- C6-C10 aryl refers to an unsaturated aromatic carbocyclic group from 6 to 10 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl).
- Preferred aryl groups include phenyl, naphthyl, phenantrenyl and the like.
- C6-C10 aryl refers to a “C6-C10 aryl” as defined above that may be substituted with 0 to 3 further groups independently selected from halogen (Cl, Br, F, I), Ci-Ce-haloalkyl, and alkoxy as defined herein. Preferred groups are -Cl, -CF3 and -OCH3.
- C6-C10 heteroaryl refers to a monocyclic heteroaromatic, or a bicyclic or a tricyclic fused- ring heteroaromatic group in which up to 3 carbon atoms are replaced by heteroatoms chosen from the group consisting of O, S, or N.
- ether refers to -R-O-R’ in which each of R and R’ is independently Ci-Ce alkyl, “Ce-Cio aryl”, and Ci-Ce cycloalkyl.
- the present invention also relates to an inhibitor of APT of formula (II): or a pharmaceutically acceptable salt thereof, wherein
- Ring A is a thiophene ring or a furane ring
- Each RAm is independently selected from the group comprising hydrogen, halogen, Ci- C10 alkyl, C2-C10 alkenyl, Ci-Ce alkoxy, Ci-Ce acyl, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, Ce-Cio aryl, and/or Ce-Cio heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups; and m is an integer from 0 to 2;
- Each R1 , R2, R4, R5 is independently selected from the group comprising hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted Ce-Cio aryl; Preferably each R1 , R2, R4, R5 is hydrogen; and
- R3 is selected from the group comprising
- the ring A is a thiophene ring represented by formula:
- the present invention also relates to an inhibitor of APT of formula (II) selected from the group comprising or a pharmaceutically acceptable salt thereof.
- the inhibitor of APT of formula (II) is selected from the group comprising More preferably, the inhibitor of APT of formula (II) is
- the present invention also relates to an inhibitor of APT of formula (II), as described herein, or a pharmaceutically acceptable salt thereof, for use as a medicament.
- the present invention relates to an inhibitor of APT of formula (II) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
- the present invention also relates to a method of treating and/or preventing a viral and/or a bacterial infection, the method comprising administering an effective amount in a subject in need thereof of at least one inhibitor of APT of formula (II), or any pharmaceutically acceptable salt thereof as described herein.
- the present invention also relates to an inhibitor of APT of formula , or a pharmaceutically acceptable salt thereof for use in the treatment and/or prevention of a viral and/or a bacterial infection.
- the present invention also relates to a method of treating and/or preventing a viral and/or a bacterial infection, the method comprising administering an effective amount in a subject in need thereof an inhibitor of APT of formula , or any pharmaceutically acceptable salt thereof.
- the present invention provides inhibitors of acyl protein thioesterase (APT) of formula (II) as described herein, and/or an inhibitor of APT of formula or any pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of viral infections mediated by or resulting from acyl protein thioesterases activity, and/or bacterial infections mediated by or resulting from acyl protein thioesterases activity.
- APT acyl protein thioesterase
- acyl protein thioesterases are enzymes that cleave off lipid modifications on proteins, located on the sulfur atom of cysteine residues linked via a thioester bond.
- the APT such as acyl-protein thioesterase 1 (APT1 ) and acyl-protein thioesterase 2 (APT2) are involved in the depalmitoylation of proteins.
- Viral infections mediated by or resulting from acyl protein thioesterases activity are infections of multiple human systems including infections of the upper and lower respiratory tract, infections of the skin, infections of the eye, infections of the gastrointestinal tract, infections of the mouth, infections of the urinary genital tract, infections of the intestinal tract, infections of the brain (e.g. meningitidis or encephalitis), and also systemic, multi organ infections such as haemorrhagic fevers.
- viral infections or viral infections associated diseases mediated by or resulting from acyl protein thioesterases activity are selected from the group comprising COVID, SARS, flu, rheumatic diseases, encephalitis, haemorrhagic fevers, acquired immunodeficiency syndrome (AIDS), sarcoma and/or leukemia.
- Bacterial infections or bacterial infections mediated by or resulting from acyl protein thioesterases activity are associated with infections of the upper and lower respiratory tract, infections of the skin, infections of the eye, infections of the gastro-intestinal tract, infections of the mouth, infections of the urinary tract, infections of the intestinal tract, infections of the uterus, and infections of the brain (e.g., meningitidis).
- Bacterial infections associated diseases mediated by or resulting from acyl protein thioesterases activity are selected from the group comprising Typhoid fever, Salmonellosis, bacteria Gastroenteritis intestinal infection, bacteremia, Shigellosis, Bacteria pneumonia (e.g., legionnaires disease), endophthalmitis, bacteremia, septicemia, sepsis, endocarditis, salpingitis, skin infections, and/or meningitis.
- the present invention provides inhibitors of acyl protein thioesterase (APT) as described herein, or pharmaceutically acceptable salts thereof, for use in the treatment and/or prevention of a viral and/or bacterial infection selected from the group comprising, SARS, COVID, flu, rheumatic diseases, encephalitis, haemorrhagic fevers, acquired immunodeficiency syndrome (AIDS), sarcoma, leukemia, meningitidis, encephalitis, bacteraemia and sepsis.
- APT acyl protein thioesterase
- the present invention also provides inhibitors of acyl protein thioesterase (APT) of formula (II) as described herein, and/or an inhibitor of APT of formula or pharmaceutically acceptable salts thereof, for use in the treatment and/or prevention of a microbial infection, wherein said microbial infection is a co-infection comprising a viral infection and a bacterial infection.
- APT acyl protein thioesterase
- viruses or viral families here represented are responsible of a variety of human diseases that comprise the infection of multiple human systems, including: the respiratory tract (upper and lower), the skin, the eye, the gastrointestinal tract, the mouth, the urinary genital and intestinal tract, the brain (e.g., meningitidis or encephalitis), and also systemic, multi organ infections such as haemorrhagic fevers.
- said viral infection can be caused by a virus selected from the group comprising Coronaviruses (CoV) (such as severe acute respiratory syndrome coronavirus (SARS), Middle East respiratory syndrome (MERS), Mouse Hepatitis Virus (MHV), transmissible gastroenteritis virus (TGEV), Swine acute diarrhoea syndrome coronavirus (SADS-cov), Rousettus bat coronavirus HKU9, and any other bat CoVs), Influenza virus (such as A, B,C), Respiratory syncytial virus (hRSV), Newcastle disease virus (NDV), Measles virus, Sindbis virus, Semliki Forest virus (SFV), Rabies virus, Vesicular Stomatitis Virus (VSV), human immunodeficiency virus (HIV), murine leukemia viruses (MulV), Ebola virus, Marburg virus, Herpes simplex virus (HSV), Human cytomegalovirus (HCMV), Dengue virus (DENV), Zika virus (ZI
- Coronaviruses
- said Coronavirus (CoV) can be selected from SARS-CoV-2, SARS-CoV-1 , MHV, TGEV, SADS, MERS-CoV, HCoV-HKLH , HCoV-229E, HCoV-NL63 and/or HCoV- OC43.
- said bacterial infection is caused by bacterial strains selected from the group comprising Bacillaceae, Vibrionaceae, Pectobacteriaceae Yersiniaceae, Staphylococcaceae, Streptococcaceae, Legionellaceae, Pseudomonadaceae, Chlamydiaceae, Mycoplasmataceae, Enterobacteriaceae, Pseudomonadales and/or Pasteurellaceae.
- bacterial families or strains are associated with infections of the respiratory tract (upper and lower), the skin, the eye, the gastro-intestinal tract, the mouth, the urinary and intestinal tract, uterus infections and other sexual transmitted diseases, brain infections (e.g., meningitidis), bacteraemia and sepsis.
- a pathogen from family Bacillaceae is for example Bacillus anthracis
- a pathogen from family Pectobacteriaceae is for example Erwinia spp (carotovora)
- a pathogen from family Pseudomonadaceae is for example Pseudomonas spp (syringae)
- a pathogen from family Enterobacteriaceae is for example Salmonella enterica or Shigella spp
- a pathogen from family Legionellaceae is for example Legionella pneumophila
- a pathogen from family Chlamydiaceae is for example Chlamydia trachomatis.
- novel inhibitors of formula (II) as described herein have been synthesized (Example 1 1 , compounds C1 , C2, C3, C9, C12, C13, C16 and C17).
- novel inhibitors of formula (II) as described herein demonstrate an EC50 that is decreased by about 3 to 8-fold compared to compound ML-349 (Example 12, figures 1 1 A and B).
- compounds such as C5, C6, C7 that are not covered by formula (II) as described herein are not potent inhibitors of SARS-CoV2 replication in Calu-3 cells (Example 12, Figure 12).
- inhibitors of formula (II) are effective without inducing cytotoxic effects in vitro (Example 14, figure 14) or in vivo (Example 16, Figure 16).
- inhibitors of formula (II) as described herein such as C2, C3 or a compound of formula ML349, can inhibit the APT2-dependent anthrax toxin intoxication of host cells (Example 17, figure 17 and Example 18, figure 18). Results have also been demonstrated for different compounds of formula (II) such as C2, C9, C12, C13, C16 and C17 (Example 19, figures 19 and 20).
- the present invention provides a pharmaceutical composition
- a pharmaceutical composition comprising at least one inhibitor of APT of formula (II) as described herein, or pharmaceutically acceptable salts thereof, and a pharmaceutically acceptable carrier, diluent, or excipient.
- pharmaceutically acceptable carrier means a carrier or excipient that is useful in preparing a pharmaceutical composition that is generally safe and possesses acceptable toxicities. Acceptable carriers include those that are acceptable for veterinary use as well as human pharmaceutical use.
- pharmaceutically acceptable carrier as used in the specification and claims includes both one and more than one such carrier.
- the present invention also relates to a pharmaceutical composition, comprising at least one inhibitor of APT of formula (II) as described herein, or pharmaceutically acceptable salts thereof, further comprising one or more additional therapeutic agents and a pharmaceutically acceptable carrier, diluent or excipient.
- said one or more additional therapeutic agents are selected from the group comprising antibacterial agents and/or antiviral agents.
- the at least one or more additional therapeutic agent is an antibacterial agent selected from the group comprising sulfonamides, penicillins, cephalosporins, aminoglycosides, chloramphenicol, tetracyclines, macrolides, lincosamides, streptogramins, glycopeptides, rifamycins, nitroimidiazoles, quinolones, trimethoprim, oxazolidinones, and/or lipopeptides and combinations thereof.
- an antibacterial agent selected from the group comprising sulfonamides, penicillins, cephalosporins, aminoglycosides, chloramphenicol, tetracyclines, macrolides, lincosamides, streptogramins, glycopeptides, rifamycins, nitroimidiazoles, quinolones, trimethoprim, oxazolidinones, and/or lipopeptides and combinations thereof.
- the at least one or more additional therapeutic agent is an antiviral agent selected from the group comprising Aribidol (umifenovir), Favilavir, APN01 , CCR5 antagonist leronlimab (PRO 140), Remdesivir (GS-5734), Galidesivir (BCX4430), Molnupiravir (MK-4482 I FJDD-2801 ), MK-7110 (CD24Fc).
- Aribidol umifenovir
- Favilavir Favilavir
- APN01 APN01
- CCR5 antagonist leronlimab PRO 140
- Remdesivir GS-5734
- Galidesivir BCX4430
- Molnupiravir MK-4482 I FJDD-2801
- MK-7110 CD24Fc
- the compounds of the invention namely inhibitor of acyl protein thioesterase (APT) as described herein, or a pharmaceutically acceptable salt thereof, that can be used in the treatment and/or prevention of a microbial infection can be incorporated into a variety of formulations and medicaments for therapeutic administration. More particularly, one or more compound(s) as provided herein can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers, and can be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, pills, powders, granules, dragees, gels, slurries, ointments, solutions, suppositories, injections, inhalants and aerosols.
- API acyl protein thioesterase
- administration of the compounds can be achieved in various ways, including oral, intranasal, intramuscular, buccal, rectal, parenteral, intraperitoneal, intradermal, transdermal, intracranial and/or intratracheal administration.
- the compound can be administered in a local rather than systemic manner, in a depot or sustained release formulation.
- the compounds can be formulated with common excipients, diluents or carriers, and compressed into tablets, or formulated as elixirs or solutions for convenient oral administration or administered by the intramuscular or intravenous routes.
- the compounds can be administered transdermally and can be formulated as sustained release dosage forms and the like.
- the compounds can be administered alone, in combination with each other, or they can be used in combination with other known compounds.
- Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences (Mack Publishing Company (1985) Philadelphia, PA, 17th ed.), which is incorporated herein by reference. Moreover, for a brief review of methods for drug delivery, see, Langer, Science (1990) 249:1527-1533, which is incorporated herein by reference.
- said pharmaceutical composition is administered by oral, inhalation, nebulization, intranasal, intrapulmonary, intradermal and/or intramuscular route of administration.
- drug substances can be delivered to the respiratory system using various devices such as nebulizer or spray.
- suitable unit doses for the compounds of the present invention can, for example, preferably contain between 0.1 mg to about 1000 mg, between 1 mg to about 500 mg, and between 1 mg to about 300 mg of the active compound. In another example, the unit dose is between 1 mg to about 100 mg.
- Such unit doses can be administered more than once a day, for example, 2, 3, 4, 5 or 6 times a day, but preferably 1 or 2 times per day, so that the total dosage for a 70 kg human adult is in the range of 0.001 to about 15 mg per kg weight of subject per administration.
- a preferred dosage is 0.01 to about 1.5 mg per kg weight of subject per administration, and such therapy can extend for a number of weeks or months, and in some cases, years. It will be understood, however, that the specific dose level for any particular patient will depend on a variety of factors including the activity of the specific compound employed; the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rate of excretion; other drugs that have previously been administered; and the severity of the particular disease undergoing therapy, as is well understood by those of skill in the area.
- a typical dosage can be one 1 mg to about 100 mg tablet or 1 mg to about 300 mg taken once a day, or, multiple times per day, or one time-release capsule or tablet taken once a day and containing a proportionally higher content of active ingredient.
- the timerelease effect can be obtained by capsule materials that dissolve at different pH values, by capsules that release slowly by osmotic pressure, or by any other known means of controlled release. It can be necessary to use dosages outside these ranges in some cases as will be apparent to those skilled in the art.
- the present invention provides a kit comprising an inhibitor of APT of formula (II), as disclosed herein, or a pharmaceutically acceptable salt thereof, and information for use thereof.
- the kit may further comprise one or more additional therapeutic agents, such as antibacterial agents and/or antiviral agents.
- the present invention provides an inhibitor of acyl protein thioesterase (APT) of general formula (I): or a pharmaceutically acceptable salt thereof, wherein
- R’ is independently selected from the group comprising optionally substituted thiophene ring, optionally substituted furane ring, optionally substituted C3-C10 cycloalkyl, or optionally substituted C6-C10 aryl;
- Each R1 , R2, R3, R4, R5 is independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, optionally substituted Ci-Ce alkyl acyl, optionally substituted Ci-Ce alkyl aminocarbonyl, optionally substituted carbonyl, optionally substituted Ci-Ce alkyl carbonyl, optionally substituted C6-C10 aryl, ether, C2-C10 alkenyl, C6-C10 heteroaryl, C3- C10 cycloalkyl, C3-C10 heterocycloalkyl, Ci-Ce alkoxy, optionally substituted amino, and/or Ci-Ce alkyl amino; for use in the treatment and/or prevention of a viral and/or a bacterial infection.
- R’ is independently selected from the group comprising optionally substituted thiophene ring, or optionally substituted furane ring.
- R’ is a thiophene ring represented by formula: wherein: each R6, R7 are independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, C2-C10 alkenyl, C1-C6 alkoxy, C1-C6 acyl, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, C6-C10 aryl, and/or C6-C10 heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups.
- R’ is a thiophene ring represented by formula: wherein: each R6, R7 are independently selected from the group comprising hydrogen, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, wherein said cycloalkyl, or optionally substituted heterocycloalkyl, may be fused with ring A and with 1 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl group.
- R’ is a thiophene ring represented by formula:
- R’ is a thiophene ring represented by formula
- the present invention relates to an inhibitor of acyl protein thioesterase (APT) of formula or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
- APT acyl protein thioesterase
- R’ is a furane ring represented by formula: wherein: each R8, R9, are independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, C2-C10 alkenyl, C1-C6 alkoxy, C1-C6 acyl, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, C6-C10 aryl, and/or C6-C10 heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups.
- R’ is a furane ring represented by formula: wherein: each R8, R9 are independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, Ci-Ce alkoxy, Ci-Ce acyl, C3-C10 cycloalkyl, and/or optionally substituted C3-C10 heterocycloalkyl, wherein said cycloalkyl, or optionally substituted heterocycloalkyl, may be fused with ring A and with 1 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl group.
- the ring A is a furane ring represented by formula: wherein: each R8, R9 are independently selected from the group comprising hydrogen, C3-C10 cycloalkyl, and/or optionally substituted C3-C10 heterocycloalkyl, wherein said cycloalkyl, or optionally substituted heterocycloalkyl, may be fused with ring A and with 1 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl group.
- the ring A is a furane ring represented by formula:
- the present invention relates to an inhibitor of acyl protein thioesterase (APT) of formula (ML348), or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
- APT acyl protein thioesterase
- inhibitors of acyl protein thioesterases prevent intoxication of mammalian cells with Anthrax toxin by inhibiting the internalization and recycling of the toxin receptor capillary morphogenesis gene 2 (CMG2/ANTXR2) (Example 1 , Figure 1 ) and by blocking endocytosis and pore formation of the anthrax toxin (Example 2, Figure 2).
- CMG2/ANTXR2 toxin receptor capillary morphogenesis gene 2
- inhibitors of acyl protein thioesterase block SARS-CoV-2 cellular infection ( Figures 5 and 9) and replication ( Figures 6-7). It has also been demonstrated that inhibitors of acyl protein thioesterase block cellular infection by other type of virus such as HSV-2 ( Figure 8).
- a dose response was determined for ML349 in Calu-3 cells and the EC50 is 8.6 pM ( Figure 10A).
- RPE1 ATCC CRL-4000
- VERO E6 ATCC CVCL-0574
- CALU-3 ATCC HTB55
- Verified siRNA for human APT2 were purchased from Qiagen.
- APT2 target sequence 5’- CAGCTGCTTCTCAGTCATGAA-3’.
- a pool of verified siRNA was used to target monkey APT1 and purchased from Qiagen.
- APT1 target sequence 5’- TACCGACAGGACCCTGTGGAA-3’, 5’-GGTCACAGATATACGGTATNN-3’, 5’- CCGGTGTATGTGCGGCAATNN-3’.
- a sequence targeting the viral glycoprotein VSV-G 5’-ATTGAACAAACGAAACAAGGA-3’ was used.
- Transfections of 50 nM of siRNA were carried out using TRANSIT-X2 (MIRUS), and the cells were analysed at least 72 h after transfection.
- MIRUS TRANSIT-X2
- the antibodies were commercially available: rabbit anti-human CMG2 (Proteintech, RRID: AB_2056741 ); mouse anti-GAPDH (Acris Antibodies, 4A1 -MA0100, RRID_AB 1874646) ; mouse anti-LPXN (Sigma, SAB1400343), anti-Anthrax Protective Antigen (List Biological Laboratories, ref: 771 B); N-terminal rabbit anti-MEK2 (Santa Cruz, sc-524), rabbit anti-SPIKE (LIFESPAN, Cat#LS-C19510), HRP-conjugated secondary antibodies (Pierce); and for immunoprecipitation protein G beads were purchased from GE Healthcare, and Streptavidin beads from Sigma.
- ML348, ML349 (Cayman Chemical company) and synthesised compounds (C1 , C2, C3, C9, C12, C13, C16, C17) were used at indicated times and concentration. All compounds were solubilised in dimethyl sulfoxide (DMSO).
- DMSO dimethyl sulfoxide
- Wild type PA (Anthrax toxin Protective Antigen) and LF (Anthrax toxin Lethal Factor) were produced in our laboratory by overexpression in E. coli as described (Feld et aL, 2012). Western Blotting
- CMG2-APT2-Flaq Co-immunoprecipitation assays
- RPE-1 cells cultured in 10 cm dishes (80% confluency) were transfected with plasmids encoding APT2-Flag for 24 h.
- Transfected cells were pretreated with APT2 inhibitors at the indicated concentrations 4 h before toxin treatment (PA63 500 ng/ml and LF 50 ng/ml). Toxin treatment was carried out for the indicated time points in the presence of the drugs.
- DMSO Dimethyl sulfoxide
- VSV-CoV-2 Vesicular stomatitis virus (VSV)-based SARS-CoV-2 pseudotypes (VSV-CoV-2) generated according to (Gasbarri et al, 2020) expressing a 19 amino acids C-terminal truncated spike protein (NCBI Reference sequence:NC_045512.2) were produced in HEK293F and titrated in Vero-E6.
- Vero-E6 cells (13,000 cells per well) were seeded in a 96-well plate.
- Compounds were serially diluted in DMEM and incubated with VSV-CoV-2 (MOI, 0.001 infectious units /cell) for 1 h at 37 °C. The mixture was added on cells for 1 h at 37 °C.
- the monolayers were washed and overlaid with medium containing 2% FBS for 18 h.
- the following day cells were fixed with paraformaldehyde 4%, stained with DAPI, and visualized using an ImageXpress Micro XL (Molecular Devices, San Jose, CA, USA) microplate reader and a 10x S Fluor objective.
- the percentage of infected cells was estimated by counting the number of cells expressing GFP and the total number of cells (DAPI-positive cells) from four different fields per sample using MetaXpress software (Molecular Devices, San Jose, CA, USA).
- Infected cells from 6 and 12 well plates and correspondent supernatants (when indicated - 150 pl from 6 well plates), were harvested and lysed in 220-330 pl of Maxwell® RSC Viral Total Nucleic Acid Purification Kit-lysis buffer from Promega, incubated at 80°C for 10 min, and used for Viral RNA extraction according to manufacturer’s instructions.
- RNA concentration was measured and 500 ng or 1000 ng of total RNA was used for cDNA synthesis using iScript.
- a 1 :5 dilution of cDNA was used to perform quantitative real-time PCR (Q-PCR) using Applied Biosystems SYBR Green Master Mix on 7900 HT Fast Q-PCR System (Applied Biosystems) with SDS 2.4 Software.
- Infected cells (24 well plates) (cultured at 1 .5 x 10 A 5 cells per well, 24 h before infection) were harvested and lysed in 150 pl of lysis buffer (0.5% NP-40, 500 mM Tris-HCI pH7.4, 20 mM EDTA, 10 mM NaF, 30mM sodium pyrophosphate decahydrate, 2 mM benzamidine and protease inhibitor cocktail; Roche) for at least 30 min. Lysates were cleared by centrifugation (5000g, 3 min), and the protein content of the supernatant was determined.
- lysis buffer 0.5% NP-40, 500 mM Tris-HCI pH7.4, 20 mM EDTA, 10 mM NaF, 30mM sodium pyrophosphate decahydrate, 2 mM benzamidine and protease inhibitor cocktail; Roche
- Samples were resuspended in 50 pl Laemmli buffer, boiled for 5 min, and approximately 20-30 pg of protein lysate were used for Western blot analysis (as described) using antibodies against viral nucleocapsid N and host GAPDH. The percentage of infection was calculated as the densitometric ratio between N and GAPDH levels for each sample.
- the number of viable cells in culture was measured by determining the reducing potential of cultured cells, thus metabolism, in Calu-3 cells.
- the assay measures the reduction of a cell-permeable substrate, which is diffused to the medium and processed by an exogenously added reporter enzyme (NanoLuc® luciferase).
- the reaction produces a luminescent signal that correlates with the number of viable cells.
- Vero E6 cells were infected in 6-well plates [as mentioned previously]. Cells washed and scrapped in 1 ml PBS and recovered by mild centrifugation 400g 3 min. Supernatants were discard and cells were fixed in 4% PFA for at least 30 min, washed twice in PBS, and permeabilized for 5 min with 0.1 % Triton in PBS. The following blocking, antibody, and washing steps were all done with FACS buffer (2% FBS in PBS/EDTA) primarily at RT. Blocking was done for 15 min, primary antibody incubation with mouse anti-Spike antibody (LC-C19510) was performed at 1 :200 for 30 min at RT or at 1 :500 ON at 4 °C, and cells were washed twice.
- FACS buffer 2% FBS in PBS/EDTA
- Alexa-647-conjugated donkey anti-mouse was used as a secondary antibody at 1 :600 for 30 min at RT, after which cells were washed three times and kept cold before flow cytometry acquisition.
- Analytical flow cytometry was performed using an LSRII or LSR Fortessa (BD; Becton Dickinson) instrument and results were analyzed using the FlowCore package in R.
- Lactate dehydrogenase (LDH) cytotoxicity assays were performed, using In Situ Death Detection Kit (Roche, Indianapolis, IN), in Vero E6 or CALU-3 cells cultured in 12 well plates with equivalent culture conditions used for infection assays.
- Wild-type HSV-2 viral suspensions were inoculated onto cells pre-treated with the indicated drugs for 1 h, at a multiplicity of infection (MOI) of 0.01 plaque-forming units (PFU) as described (Cagno et al, 2018). Viral inoculum was removed, and cells washed after 2 h, and infection was continued in the presence of the drugs for evaluation through a plaque-based assay.
- MOI multiplicity of infection
- PFU plaque-forming units
- mice 1 1 -week-old male C57BL/6 mice were inoculated intraperitoneally with 400 to 600 pl of a 2 mg/ml working solution of C2 or ML349, resuspended in PEG300 50% (v/v) solution in PBS, to a final dose of 1 mg of compound per 25 g of body weight.
- Control mice were inoculated in parallel with an equivalent volume (per body weight) of vehicle PEG300 50% (v/v) in PBS solution.
- Drug and vehicle treatments were injected on days 1 , 3, 6, 8, and 10 of the experiment, whereas clinical parameters (body weight, blood pressure, and pulse) were monitored on days 1 , 2, 3, 6, 7, 8, 9, 10, and 13. After 13 days of treatment, mice were euthanatized, and different organs were harvested for histological analysis and weight measurements.
- Example 1 Inhibition of APT2 activity increases surface expression of ANTXR2/CMG2
- Example 2 Inhibition of APT2 activity prevents intoxication of mammalian cells with Anthrax toxin.
- Mammalian cells were left untreated (control) or co-treated with ML349 (10 pM) or ML348 (10 pM) during 0, 20, 40 and 60 minutes. Cells were pre-treated with ML349 or ML348 during 2 h ( Figure 2A) or 4 h ( Figure 2B) prior to and during exposure to Anthrax toxin Protective Antigen (PA).
- ML349 10 pM
- ML348 10 pM
- Treatment with ML349 blocks endocytosis of CMG2/ANTXR2, thus preventing pore formation of the anthrax toxin PA subunit.
- ML349 thus prevents the protease subunit of anthrax toxin to reach the cytosol and in turn prevents the cleavage of a major host cell signaling regulator, the MAP kinase-kinase MEK.
- Example 3 siRNA silencing of APT2 expression prevents intoxication of mammalian cells with Anthrax toxin.
- Example 4 Inhibition of APT1 activity with ML348 diminishes SARS-CoV-2 Spike- pseudotyped particles (PPs) entry into host cell.
- a cell-based assay was performed to determine the effects of different chemical inhibitors on the internalization of Vesicular Stomatitis Virus (VSV)-based particles pseudotyped with the spike fusion glycoprotein from SARS-CoV-2.
- VSV Vesicular Stomatitis Virus
- the effects of ML348 and ML349, inhibitors of APT1 and APT2 respectively, as well as Palmostatine, Bromopalmitate, Chlroroquine and Hydroxychloroquine were tested and are represented on Figure 4.
- Vero E6 cells were infected with SARS-CoV-2-Spike- pseudotyped particles (VSV-S-CoV2) or VSV-G-pseudotyped particles(VSV-G) ( Figures 4A, B and C). Vero cells were either pre-treated (PRE-TREAT, Figure 4C) for 4 h washed, and infected with VSV-S- CoV2 or VSV-G, or infected in the presence of the same concentration of the chemical inhibitors (PRE+CO-TREAT). GFP-expressing cells were quantified by fluorescent microscopy.
- the percentage of infection was calculated comparing the percentage of positive cells to the percentage in infected untreated controls (DMSO). Dose-response curve was calculated for ML348, ML349, and hydroxychloroquine. ( Figure 4C).
- ML348 specifically inhibited the entry of PPs typed with SARS-CoV-2 spike protein to a similar extend as hydroxychloroquine (Figure 4A).
- This inhibitory effect was dosedependent ( Figure 4C) and not observed upon treatment of target cells with unspecific inhibitors of palmitoylation and depalmitoylation such as Bromopalmitate and Palmostatine respectively, neither by treatment with the APT2 inhibitor ML349 ( Figure 4A and 4B).
- Example 5 Inhibition of APT1 activity with ML348 blocks SARS-CoV-2 cellular infection.
- the depalmitoylation-targeting drugs ML348, ML349 were tested during infection of Vero E6 cells with a clinical isolate of SARS-Cov-2. Infection was monitored using a standard plaque-based infection assay as described in materials and methods (Viral stock production and titration with plaque-based assays; SARS CoV-2 infections).
- Vero E6 cells were pre-treated with ML348 or ML349 for 1 h washed, and infected with SARS-CoV-2, in the presence of drug. Infection was determined using a plaque assay and a dose-response curve was established for ML348 and ML349 ( Figure 5A). The EC50 for ML348 is 13.3 pM. Then, Vero E6 cells were infected with SARS-CoV-2 (MOI 0.1 ) for 24 h, in the presence of 12.5 pM ( Figure 5B). Drugs were added 1 h after viral inoculation, and replication was monitored by determining the % of high Spike-positive cells by flow cytometry. Results are mean ⁇ SEM and each dot represents one individual biological replicate.
- Example 6 Inhibition of APT1 activity with ML348 inhibits SARS-CoV-2 intracellular replication.
- ML348 occurs within a single cellular infection cycle (up to 8 to 12 h)
- viral genomic RNA replication was monitored throughout time by probing for two viral genomic regions corresponding to the E and RdRp genes of SARS- CoV-2.
- Vero E6 cells were treated with ML348 1 h after viral inoculation and throughout the time of infection.
- Example 7 Inhibition of APT1 activity and silencing of APT1 expression blocks SARS- CoV-2 infection in Vero E6 cells.
- Viral RNA replication in Vero E6 cells was also monitored after 24h of infection to follow multiple rounds of cellular infection.
- the viral intracellular replication was monitored by determining the levels of the sub-genomic E RNA, a bona-fide marker of intracellular viral RNA replication.
- E Quantification of viral RNA replication
- SARS-CoV-2 MOI-O.1 for 24 h
- Control treated without drug
- Cells were inoculated for 1 h with SARS-CoV-2, washed and subsequently incubated in complete medium with 12.5 pM of ML348 or ML349 ( Figure 7 A and 7B).
- Example 8 Inhibition of APT1 activity with ML348 blocks HSV-2 cellular infection.
- ML348 or ML349 could limit infection by other type of viruses.
- a plaque-based assays was performed using herpes simplex virus type 2 (HSV-2) according to materials and methods (HSV infection and inhibition assays). Vero E6 cells were pre-treated for 1 h in the presence of drug, washed, and infected with HSV-2 ( Figure 8 A). Infection was determined using a plaque assay. Dose-response was determined for ML348 and ML349. The EC50 for ML348 is 23.2 pM.
- Example 9 Inhibition of APT2 activity blocks SARS-CoV-2 infection in Calu-3 lung- derived cells.
- APT2 is often more abundant than APT1.
- APT2 is highly enriched when compared to Vero E6 cells ( Figure 9A).
- LYPLA1 AAT1
- LYPLA2 AAT2
- mRNA levels are measured in different cell lines ( Figure 9A). Values are TAG per Million (TPM) corresponding to 10*6 X ((reads mapped to transcript/transcript length)/ SUM (reads mapped to transcript/transcript length)). Results are the mean of independent experiments each from n > 2 biological replicates.
- SARS-CoV-2 The replication of SARS-CoV-2 was monitored in Calu-3 cells at 24h post-inoculation and upon treatment with 12.5 pM of ML348 or ML349. This concentration is below the EC50 of ML348 for inhibition of viral replication in Vero E6.
- Figure 9B shows the quantification of viral RNA replication (E, RdRp and E subgenomic) from Calu-3 cells and correspondent supernatants infected with SARS-CoV-2 (MOI-0.1 for 24 h), treated with ML348 or ML349.
- the cells were inoculated for 1 h with SARS-CoV- 2, washed and subsequently incubated in complete medium with 12.5 pM of ML348 or ML349.
- Example 10 Dose-response inhibition of SARS-CoV-2 replication in Calu-3 cells by ML349 and cytotoxicity evaluation of ML348 and ML349.
- Calu-3 cells were infected with SARS-CoV-2. Infection was determined using Q-PCR by quantifying the mean levels of E and RdRp viral RNA (Figure 10A). Normalized results are mean ⁇ SEM. The dose-response was determined for ML349 and the EC50 is 8.6 pM.
- a Lactate dehydrogenase activity assay was performed as described in the material and methods section. Vero E6 cells or Calu-3 cells were treated with increasing concentrations of ML348 for 48 h or ML349 for 24 h, respectively. The results of the cell viability are mean ⁇ SD of 2 biological replicates. Importantly, neither treatment of Vero E6 with ML348 for 48h nor treatment of Calu-3 with ML349 for 24h, triggered significant cellular toxicity at different ranges of concentrations ( Figure 10B and 10C).
- Example 1 1 Synthesis of novel inhibitors of acyl protein thioesterase (APT) of general formula (II).
- Compound SPC-EPFL-1 (C1 ) was prepared following General Procedure A using 1 -(4- fluorophenyl)piperazine (32 mg, 1 Eq, 0.18 mmol) to afford (5,5-dioxido-4H-thieno[3,2- c]thiochromen-2-yl)(4-(4-fluorophenyl)piperazin-1 -yl)methanone (42.6 mg, 54 %) as a white solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-30% 5CV, 30-100% for 15CV, 100% for 3CV].
- Compound SPC-EPFL-2 (C2) was prepared following General Procedure A using 1 -(2- chlorophenyl)piperazine (35 mg, 1 Eq, 0.18 mmol) to give (4-(2-chlorophenyl)piperazin- 1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (41.4 mg, 51 %) as a white solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-30% 5CV, 30-100% for 15CV, 100% for 3CV].
- Compound SPC-EPFL-3 (C3) was prepared following General Procedure A using 1 -(2,3- dichlorophenyl)piperazine (41 mg, 1 Eq, 0.18 mmol) to give (4-(2,3- dichlorophenyl)piperazin-1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (30.4 mg, 35 %) as a solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-30% 5CV, 30-100% for 15CV, 100% for 3CV].
- Compound SPC-EPFL-5 was prepared following General Procedure A using phenyl(piperazin-1 -yl)methanone (34 mg, 1 Eq, 0.18 mmol) to give (4-benzoylpiperazin- 1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (65.1 mg, 81 %) as a solid after purification by flash column chromatography [Silica-CS 12g, eluent: DCM/MeOH (0% MeOH for 3CV, 0-5% 15CV, 5% for 3CV, 5-10% for 3CV].
- Compound SPC-EPFL-6 was prepared following General Procedure A using 2-(5- methoxy-1 H-indol-1 -yl)ethan-1 -amine (27 mg, 1 Eq, 0.14 mmol) to give N-(2-(5-methoxy- 1 H-indol-1 -yl)ethyl)-4H-thieno[3,2-c]thiochromene-2-carboxamide 5,5-dioxide (36.8 mg, 57 %) as a solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-30% 5CV, 30-100% for 15CV, 100% for 3CV].
- Compound SPC-EPFL-9 (C9) was prepared following General Procedure A using 1 -o- tolyl-Piperazine (19 mg, 1 Eq, 0.1 mmol) to afford (5,5-dioxido-4H-thieno[3,2- c]thiochromen-2-yl)(4-(o-tolyl)piperazin-1 -yl)methanone (31 mg, 66 %) as a light yellow solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-30% 5CV, 30-50% for 15CV, 50% for 3CV].
- Compound SPC-EPFL-13 (C13) was prepared following General Procedure A using 1 - (2,6-dimethylphenyl)piperazine (48 mg, 1 Eq, 0.25 mmol) to afford (4-(2,6- dimethylphenyl)piperazin-1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (42 mg, 74 %) as a yellow solid after purification by flash column chromatography [Silica- CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-50% for 15CV, 50% for 3CV]. Yellow powder (42 mg, 0.09 mmol, 74%).
- Compound SPC-EPFL-16 (C16) was prepared following General Procedure A using 1 - (2,6-dichlorophenyl)piperazine (58 mg, 1 Eq, 0.25 mmol) to afford (4-(2,6- dichlorophenyl)piperazin-1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (56 mg, 91 %) as a Light yellow solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-60% for 15CV, 60% for 3CV].
- Compound SPC-EPFL-17 (C17) was prepared following General Procedure A using 1 - (2,5-dichlorophenyl)piperazine (58 mg, 1 Eq, 0.25 mmol) to afford (4-(2,5- dichlorophenyl)piperazin-1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (51 mg, 83 %) as a Light yellow solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-50% for 15CV, 50% for 3CV].
- Example 12 Dose response inhibition of SARS-CoV-2 replication in Calu-3 cells by ML349 and novel inhibitors of acyl protein thioesterase (APT) according to formula (II).
- APT acyl protein thioesterase
- the improved inhibitory effect of the novel inhibitors of acyl protein thioesterase (APT) according to formula (II), such as C1 , C2 or C3 during SARS-CoV-2 replication in Calu-3 cells ( Figure 1 1 A) further demonstrate that ML349 and novel inhibitors of acyl protein thioesterase (APT) according to formula (II) are potent inhibitors of SARS-CoV-2 infection and that host cell depalmitoylation activity constitutes a safe strategy to inhibit viral infections.
- inhibitors of acyl protein thioesterase (APT) of formula II wherein R3 is a substituted phenyl ring such as C1 , C2 or C3 have an improved inhibitory activity of SARS-CoV-2 replication compared to ML349.
- molecules that are not covered by formula II wherein R3 is a substituted phenyl ring such as C5, C6 or C7 do not inhibit SARS-CoV-2 replication in Calu-3 cells ( Figure 12).
- Example 13 Inhibition of APT2 activity with inhibitor C2 inhibits SARS-CoV-2 intracellular replication.
- Cells were harvested at the time points 1 , 4, 6, 8 and 12 hours and host and viral RNA levels quantified by QPCR as described in materials and methods.
- the data was normalized to the intracellular viral RNA levels harvested 1 h after inoculation before drug treatment and expressed as fold RNA replication of both viral genomic and subgenomic RNA. Normalized results are the mean ⁇ SEM of three independent biological replicates displayed within two equivalent graph plots with either linear or logarithmic scale for the fold RNA replication.
- Example 14 Dose-response cytotoxicity evaluation of APT2 inhibition in Calu-3 cells by ML349 and novel inhibitors of acyl protein thioesterase (APT) according to formula (II).
- a RealTime-GloTM MT Cell Viability Assay consisting of a nonlytic, homogeneous, bioluminescent method to measure cell viability in real time, was performed as described in material and method.
- Calu-3 cells were treated with increasing concentration of ML349 and C2 or C3 over the course of 6 days. Culture medium was replaced every 2 days with freshly diluted compounds in order to ensure constant activity. The results of the cell viability represent the evolution of the number of viable cells in culture at the indicated time-points and are mean SEM of 4 biological replicates ( Figure 14A-C). Fitted curves for Control (no drug), non-toxic and cytotoxic compound concentrations are indicated.
- APT2 activity with novel inhibitors of acyl protein thioesterase (APT) effectively and safely inhibits viral infections, such as SARS-CoV-2.
- APT2 activity with novel inhibitors of acyl protein thioesterase (APT) effectively and safely inhibits viral infections, such as SARS-CoV-2.
- Example 15 Inhibition of APT2 activity with compound C2 inhibits replication of SARS- CoV-2 variant Omicron BA.5.
- Example 16 Toxicity evaluation of APT2 inhibition in vivo, in C57BL/6 mice by ML349 and compound C2.
- Example 14 Following the experiment confirming that inhibition of APT2 with ML349, compound C2 and C3 did not induce significant cytotoxicity in vitro (Example 14), an experiment was carried out to evaluate the potential toxicity effect of inhibiting APT2 using ML349 or C2 in vivo in C57BL/6 mice. Mice were treated with 1 mg of ML349 or C2 per 25 g of body weight or an equivalent volume of drug delivery vehicle (control), inoculated intraperitoneally at alternate days through a period of 13 days. Toxicity was evaluated by monitoring the body weight, blood pressure, and pulse daily throughout treatment (Figure 16A-C).
- Example 17 Evaluating APT2 inhibition mechanism by ML349, compound C2, or C3 during anthrax toxin cellular intoxication.
- APT2 activity during anthrax intoxication can be monitored by probing for the formation of protein complexes between the anthrax receptor CMG2 and APT2 triggered by the binding of anthrax toxin protective antigen (PA) to CMG2 (Figure 17A). Formation of these complexes occurs upon de-acylation of CMG2 and is concomitant to the dissociation of CMG2 from the cytoskeleton components actin, vinculin, and talin, which ultimately promotes the internalization of the toxin and the receptor.
- the levels of CMG2-APT2 complexes can be probed by western blot analysis of lysates from RPE-1 cells treated with PA ( Figure 17B).
- ML349, C2, and C3 can reduce SARS-CoV-2 replication and inhibit the APT2- dependent anthrax toxin intoxication of host cells.
- Example 18 Comparison of APT2 inhibition efficacy by ML349, compound C2, C3 during anthrax toxin cellular intoxication.
- CMG2-APT2 complexes formed upon anthrax toxin protective antigen (PA) intoxication were also evaluated in cells pre and co-treated with only 2 pM of each compound to compare the potency of ML349 and compound C2, C3. At such concentrations C2 and C3, but not ML349, prevented the formation of APT2-CMG2 complexes, blocking cellular intoxication ( Figure 18). Therefore, as observed for the inhibition of SARS-CoV-2 replication, C2 and C3 inhibit intoxication of host cells by anthrax-toxin at lower concentrations than ML349.
- PA anthrax toxin protective antigen
- Example 19 Anthrax toxin cellular intoxication assays with compounds of formula (II). Novel compounds of formula (II) as described herein have been synthesized (Example 1 1 ). These compounds were tested for their capacity to inhibit the formation of CMG2- APT2 complexes in anthrax toxin cellular intoxication assays. Western blot analysis of a single parallel synchronized cellular intoxication experiment showed that compounds C9, C12, C13, C16, and C17 (used at 5 pM) inhibit the formation of CMG2-APT2 complexes beyond 50% (Figure 19).
- Example 20 Inhibition of APT2 activity with compounds of formula (II) blocks SARS-CoV- 2 infection in Calu-3 lung derived cells
- Figure 21 shows the levels of viral nucleocapsid protein N: cell lysates, were monitored by Western blot. The % of infection was quantified as the ratio between the levels of N and host GAPDH for three independent replicates for each compound with relation to Control cells (infected in the presence of drug carrier DMSO).
- Figure 21 shows that 5 pM of compounds C9, C12, C13, C16, or C17 significantly inhibited SARS-CoV-2 replication beyond 50% and more potently than ML349. At 5 pM, C2 and C3 displayed the most potent inhibitory effect.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Virology (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Communicable Diseases (AREA)
- Oncology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Molecular Biology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention relates inhibitors of acyl protein thioesterases and method of preparation thereof. It also relates to the use of inhibitors of acyl protein thioesterases in the treatment and/or prevention of a microbial infection such as viral or bacterial infections or coinfection by virus and bacteria.
Description
INHIBITORS OF ACYL PROTEIN THIOESTERASES AGAINST MICROBIAL INFECTIONS
TECHNICAL FIELD
The present invention relates to inhibitors of acyl protein thioesterases (APT) and method of preparation thereof. It also relates to the use of inhibitors of APT in the treatment and/or prevention of a microbial infection such as viral or bacterial infections or coinfection by virus and bacteria.
BACKGROUND OF THE INVENTION
For infection to progress, pathogens such as viruses, bacteria or parasites must rely on interactions with membranes from target cells. Host-pathogen interactions frequently involve modifications of host or microbial proteins by a reversible post- translational lipidation called S-acylation, commonly referred to as S-palmitoylation.
S-acylation consists of the addition of an acyl chain, often palmitate, to cytosolic cysteines in target proteins via a thioester bond. This modification is reversible and therefore acts as a functional switch on target proteins (Zaballa & van der Goot, 2018). S-acylation and deacylation are crucial for membrane trafficking events and impact the cellular distribution, abundance, and function of proteins. During infection, S-acylation modifies numerous viral proteins from clinically relevant viruses (Veit, 2012), as well as multiple host receptors fundamental for internalization, and therefore virulence, of numerous pathogens and pathogenic molecules.
Pathogens are highly opportunistic organisms. Viruses often exploit S-acylation of viral proteins or host products to promote pathogenesis. [3-Coronaviruses (CoVs) have enormous zoonotic potential being responsible for various outbreaks of severe acute respiratory syndrome (SARS) in recent years including the pandemic Coronavirus disease-2019 (COVID-19). CoVs depend on S-acylation to produce fully infectious viruses (Mesquita et al, 2021 ). SARS-CoV-2, the etiological agent of COVID 19, contains several essential pathogenic molecules that are modified by S-acylation.
Vaccines, antibodies and pathogen-directed antibiotics or antivirals are effective therapeutic strategies that work by targeting the specific infectious agent(s) they were designed for. Host-directed anti-microbial drugs, in the other hand, have the potential to
broadly target infectious agents with a lower risk of rapid resistance development (Fernandez-Oliva et al, 2019; Kaufmann et al, 2018).
Large spectrum therapeutic strategies are also invaluable for susceptible patients that cannot develop adequate immunity or are not eligible for vaccination such as immunocompromised individuals.
The emergence of drug-resistance and of new pathogens, such as SARS CoV-2, highlights the importance of alternative approaches to be used as first line wide-ranging interventions before specific therapies may be elaborated or immunity emerges.
Thus, there is a continuous and urgent necessity for the development of new drugs and molecular targets to strengthen the global public health preparedness against viral, bacterial, and other infection outbreaks.
SUMMARY OF THE INVENTION
The present invention provides an inhibitor of acyl protein thioesterase (APT) of
or a pharmaceutically acceptable salt thereof, wherein Ring A is a thiophene ring or a furane ring;
Each RAm is independently selected from the group comprising hydrogen, halogen, Ci- C10 alkyl, C2-C10 alkenyl, C1-C6 alkoxy, C1-C6 acyl, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, C6-C10 aryl, and/or C6-C10 heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups; and m is an integer from 0 to 2;
Each R1 , R2, R4, R5 is independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, optionally substituted Ci-Ce alkyl acyl, optionally substituted Ci-Ce alkyl aminocarbonyl, optionally substituted carbonyl, optionally substituted Ci-Ce alkyl
carbonyl, optionally substituted Ce-C aryl, ether, C2-C10 alkenyl, C6-C10 heteroaryl, C3- C10 cycloalkyl, C3-C10 heterocycloalkyl, Ci-Ce alkoxy, optionally substituted amino, and/or Ci-Ce alkyl amino; and
The invention also provides said inhibitors of APT of formula (II), or pharmaceutically acceptable salts thereof, for use as a medicament.
Further, the invention provides said inhibitors of APT of formula (II), or pharmaceutically acceptable salts thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
The invention also relates to a pharmaceutical composition comprising at least one of said inhibitors of APT, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, or excipient.
The invention also provides a kit comprising said inhibitors of APT, or pharmaceutically acceptable salts thereof, and information for use thereof.
The invention further provides an inhibitor of APT of formula
(ML349), or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
The invention further provides an inhibitor of APT of formula
(ML348), or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 : Labelling of surface proteins with NHS-biotin in mammalian cells untreated or treated with ML349 (10 pM) for the indicated times. Analysis of surface proteins ANTXR2/CMG2 by western blot. The quantification of surface ANTXR2/CMG2 is expressed as % of T=0. Results are mean ±SD.
Figure 2 (A) and (B): Synchronized anthrax intoxication assays in mammalian cells untreated (control) or co-treated with ML349 or ML348 (10 pM) for the indicated times. Anthrax toxin Protective Antigen (PA); (C): The dynamics of toxin pore formation (PA Pore); (D): MEK2 degradation. Quantification for each time point for cells pre-treated for 2 h and expressed as % of t=0 for each sample. All results are mean ±SEM.
Figure 3 (A): Synchronized anthrax intoxication assays in mammalian cells transfected with control siRNA or siRNA oligos targeting the expression of APT2 (siAPT2) for 72 h; (B) The dynamics of toxin pore formation (PA Pore) were quantified for each time point and expressed as % of t=0 for each sample. All results are mean ±SEM.
Figure 4 (A), (B) and (C): Microscopy-based quantification assay of Vero E6 cells infected with SARS-CoV-2-Spike-pseudotyped particles (VSV-S-CoV2) (A, C) or VSV-G- pseudotyped particles (VSVG) (B). (C): Dose response curve calculated for ML348, ML349, and hydroxychloroquine. Results are mean ±SEM (A,B), or SD (C).
Figure 5 (A): Infection with SARS-CoV-2 determined using a plaque assay of Vero E6 cells pre-treated with ML348 or ML349 and infected in the presence of increasing concentrations of each drug. Dose-response curves were obtained using non-linear regression. (B): Vero E6 cells infected with SARS-CoV-2 (MOI 0.1 ) in the presence of 12.5 pM of ML348 or ML349. Replication was monitored by determining the % of high Spike-positive cells by flow cytometry. All results are mean ±SEM, and each data point (in B) represents an independent experiment.
Figure 6 (A, B): Vero E6 cells inoculated for 1 h with SARS-CoV-2, washed, and subsequently incubated in complete medium with 12.5 pM of ML348. Cells were harvested at the indicated time points and infection was determined by monitoring the levels of intracellular E and RNA-dependent RNA-polymerase-RdRp RNA transcripts by Q-PCR. T=0 was harvested 1 h after inoculation before drug treatment. All results are mean ±SEM.
Figure 7 (A, B): Quantification of viral RNA replication (E, RdRp and E subgenomic) from Vero E6 cells (A) and correspondent supernatants (B) infected with SARS-CoV-2 (MOI- 0.1 for 24 h). Cells were inoculated for 1 h with SARS-CoV-2, washed and subsequently incubated in complete medium with 12.5 pM of ML348 or ML349. (C): Cells were previously transfected with control or siRNA oligos targeting the expression of APT1 (siAPT) for 72 h. (D): Quantification of APT1 mRNA levels from the samples used in C. All results (A, B, C) are mean ±SEM and p values comparing RNA levels in relation to control conditions were obtained by (A, B, C) one-way ANOVA with Dunnet’s multiple comparison or (D) Paired two-tailed student’s t-test (*p < 0.05, ***p < 0.001 , ****p < 0.0001 )
Figure 8 (A): Vero E6 cells were pre-treated with drug for 1 h washed, and infected with HSV-2, in the presence of drug. Infection was determined using a plaque assay. Results are mean ±SEM, and dose-response curves were obtained using non-linear regression.
Figure 9 (A): LYPLA1 (APT1 ) and LYPLA2 (APT2) mRNA levels in different cell lines. Values are TAG per Million (TPM) corresponding to 10*6 X (reads mapped to transcript/transcript length)/ SUM (reads mapped to transcript/transcript length). (B and C): Quantification of viral RNA replication (E, RdRp and E subgenomic) from Calu-3 cells and correspondent supernatants infected with SARS-CoV-2 (MOI-0.1 for 24 h), treated as indicated. (D): Quantification of APT2 mRNA levels from the samples used in C. For all, each data point represents an individual experiment and results are mean (A) or mean ±SEM (B, C, D). All p values comparing RNA levels in relation to control conditions were obtained by (B, C) one-way ANOVA with Dunnet’s multiple comparison or (D) Paired two- tailed student’s t-test (*p < 0.05, **p < 0.01 , ****p < 0.0001 )
Figure 10: dose response inhibition of SARS-CoV-2 replication (infection at MQI=0.05- 01 ) in Calu-3 cells by ML349 and cytotoxicity evaluation of ML348 and ML349 in Vero E6 and CALU-3 cells, respectively. All results are mean ±SEM and the dose inhibition curve (A) was fitted using a nonlinear regression.
Figure 1 1 (A): dose-response inhibition of SARS-CoV-2 replication (infection at MQI=0.05-01 ) in human lung-derived Calu-3 cells by ML349 and compounds C1 , C2 and C3, 24 h post viral inoculation, quantified by QPCR of total viral RNA (E, RdRp and E sub-genomic). Drugs were added 1 h a post-viral inoculation (B): levels of housekeeping (HK) transcripts during infection, indicative of cell viability. For each individual concentration results are mean ±SEM of n >3. Dose inhibition curves were fitted using a nonlinear regression and EC50 ±SD concentrations are indicated
Figure 12 (A): dose response inhibitions of SARS-CoV-2 replication (infection at MOI «0.05-01 ) in human lung-derived Calu-3 cells by ML349-derived compounds C5, C6, and C7, 24 h post-viral inoculation, quantified by QPCR of total viral RNA (E, RdRp and E sub-genomic). (B): levels of housekeeping (HK) transcripts during infection, indicative of cell viability. For each concentration, results are mean ±SEM of n >3. Dose inhibition
curves were fitted using nonlinear regression, and EC50 ±SD concentrations are indicated.
Figure 13 shows replication of SARS-CoV-2 inoculated in human lung-derived Calu-3 cells (MOI«0.5, 1 h), incubated with infection media supplemented with 5 pM of compound C2 and 0.5 pg/ml of P2G3 anti-Spike antibody, from 1 h post viral inoculation. Cells were harvested at the indicated time points and infection was determined by monitoring the levels of intracellular (A): genomic (E and RdRp) and (B): sub-genomic E (E-subgen) RNA transcripts by QPCR. Values were normalised to 1 for the timepoint 1 h post viral inoculation (before drug treatment) and results are mean ±SEM of n>3. Data is presented as equivalent side-by-side plot graphs with log or linear scale representation of viral RNA levels - Fold-infection.
Figure 14 (A, B and C): evaluation of levels of viable Calu-3 cells in the presence of the cell permeable RealTime-GloTM-Cell Viability reagent and incubated with the indicated concentrations of ML349, C2 and C3 for 6 days. Culture medium was replaced every 2 days using fresh ML349, C2 and C3 concentrations. Luminescence, corresponding to the levels of viable cells, was monitored at the indicated time points and the results are mean ±SEM of n=4. Fitted curves indicate Control (full-line), non-toxic (dotted-lines) and cytotoxic (>25 pM dashed-lines) concentrations. (D): dose-dependent cell viability at day 6 after drug treatment monitored as the % of viable cells normalised to Control (no drug). A significant decrease in cell viability was verified for all compounds when used at 25 to 100 pM (p<0.05 obtained by Two-way ANOVA).
Figure 15(A): dose-response inhibition of replication of SARS-CoV-2 variant Omicron BA.5 (infection at MOI =0.05-01 ) in human lung-derived Calu-3 cells by compound 2 (C2), 24 h post-viral inoculation, quantified by QPCR of total viral RNA (E, RdRp and E sub- genomic). (B): levels of housekeeping (HK) transcripts during infection, indicative of cell viability. For each concentration, results are mean ±SEM of n >3. The dose inhibition curve was fitted using nonlinear regression, and EC50 ±SD concentrations are indicated.
Figure 16: toxicity evaluation of inhibiting APT2 with ML349 and C2 in vivo in C57BL/6 mice. (A): Body weight (g); (B): blood pressure (Hgmm); and (C): pulse (beats per minute - bpm) were monitored for the indicated time points for every mouse mock-
treated or treated with 1 mg of ML349 or C2 per 25 g of body weight for several days through 13 days. Mock-treated control mice were inoculated in parallel with equivalent volumes of vehicle solution used to resuspend ML349 and C2. At 13 days of treatment, mice were sacrificed, and the relative spleen weight (as % of body weight) was determined, (D): Results are mean ±SEM of n=8 mice per condition for A, B, and C and n=5 for D. In D, each data point represents an individual mouse.
Figure 17(A): Schematic representation of the APT2-dependent mechanism of synchronized anthrax toxin entry into host target cells. In brief, in 1 , the toxin binds to the S-acylated anthrax receptor (CMG2) at the cell surface; this leads to 2, the recruitment and binding of APT2 to CMG2, which, in turn, promotes, 3, the complete de-acylation of the receptor; and 4, its dissociation from the host cytoskeleton components (actin, vinculin, and talin) and binding to the host GTPase RhoA. These events enable 5, the endocytosis of the toxin and access to its host target. (B): western blot analysis of CMG2- immunoprecipitation (IP) fractions to monitor the co-immunoprecipitation (co-IP) of APT2- CMG2 complexes from cell lysates of control (untreated) or anthrax toxin-treated cells. Mammalian RPE-1 cells ectopically expressing FLAG-APT2, for 24 h, were left untreated or treated with anthrax toxin (protective antigen-PA subunit) in the presence of APT2 inhibitors, ML349 or compounds C2, C3, or control drug carrier DMSO. Drugs were added at a final concentration of 5 pM, 4 h before and during toxin treatment. B and respective quantification in C show that inhibition of APT2 activity with ML349 and compound of formula C2 and C3 reduces APT2-CMG2 complexes formed upon PA toxin treatment. (C): quantification of APT2-FLAG levels in equivalent CMG2 IP fractions. Results are mean ±SEM of n=4, (****p<0.0001 obtained by one-way ANOVA).
Figure 18: Western blot analysis of APT2-CMG2 complexes present in CMG2- immunoprecipitation (IP) fractions from cell lysates of control (untreated) or anthrax toxin- treated cells. (A): RPE-1 cells ectopically expressing APT2-FLAG for 24 h, left untreated or treated with anthrax toxin (protective antigen-PA subunit) in the presence of APT2 inhibitors, ML349 or compound 02, C3, or control drug carrier DMSO. Drugs were added at a final concentration of 2 pM, 4 h before and during toxin treatment. (B): quantification of APT2-FLAG levels in equivalent CMG2 fractions displayed in A.
Figure 19: Quantification of APT2-FLAG levels in equivalent CMG2 fractions from western blot analysis of APT2-CMG2 complexes present in CMG2-immunoprecipitation (IP) fractions from cell lysates of control (untreated) or PA-toxin-treated cells: RPE-1 cells ectopically expressing APT2-FLAG for 24 h, left untreated or treated with anthrax toxin (protective antigen-PA subunit) in the presence of APT2 inhibitors, ML349 or compound of formula (II) (C9, C12, C13, C16, and C17), or control drug carrier DMSO. Drugs were added at a final concentration of 5 pM, 4 h before and during toxin treatment. Data are normalized to 100% for WT, and results are mean or mean ±SEM, and each data point represents an individual experiment.
Figure 20: Quantification of western blot analysis of the dynamics of toxin pore formation (PA-Pore) in RPE-1 mammalian cells treated with anthrax toxin (protective antigen-PA subunit) in the presence of APT2 inhibitors (C2, C9, C12, C13, C16 at 5 pM) for the indicated times (0, 30 and 60 min). Toxin pore formation (PA Pore) was quantified for each time-point and normalized to equivalent levels of PA63. Data were expressed as % of t0=1 for each sample.
Figure 21 : Inhibition of SARS-CoV-2 replication in human lung-derived Calu-3 cells (MOI«0.05-0.1 , 1 h), in the presence of the APT2 inhibitors, ML349, C2, C9, C12, C13, C15, C16 and C17 (used at 5 pM), from 1 h post-viral inoculation. Culture supernatants and infected cells were harvested 24 h post-viral inoculation. The levels of viral nucleocapsid protein N: cell lysates, were monitored by Western blot. The % of infection was quantified as the ratio between the levels of N and host GAPDH for three independent replicates for each compound with relation to Control cells (infected in the presence of drug carrier DMSO). Results are mean ±SEM of n=3, and p values were obtained by one-way ANOVA analysis.
DETAILED DESCRIPTION OF THE INVENTION
In one aspect, the present invention relates to an inhibitor of acyl protein thioesterase (APT) of formula (II):
or a pharmaceutically acceptable salt thereof, wherein
Ring A is a thiophene ring or a furane ring;
Each RAm is independently selected from the group comprising hydrogen, halogen, Ci- C10 alkyl, C2-C10 alkenyl, C1-C6 alkoxy, C1-C6 acyl, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, C6-C10 aryl, and/or C6-C10 heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups; and m is an integer from 0 to 2; Each R1 , R2, R4, R5 is independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, optionally substituted Ci-Ce alkyl acyl, optionally substituted Ci-Ce alkyl aminocarbonyl, optionally substituted carbonyl, optionally substituted Ci-Ce alkyl carbonyl, optionally substituted C6-C10 aryl, ether, C2-C10 alkenyl, C6-C10 heteroaryl, C3- C10 cycloalkyl, C3-C10 heterocycloalkyl, Ci-Ce alkoxy, optionally substituted amino, and/or Ci-Ce alkyl amino; and
R3 is selected from the group comprising
Preferably, the ring A is a thiophene ring represented by formula:
wherein: each R6, R7 are independently selected from the group comprising hydrogen, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, C6-C10 aryl, and/or C6-C10 heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups.
More preferably, the ring A is a thiophene ring represented by formula:
wherein: each R6, R7 are independently selected from the group comprising hydrogen, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, wherein said cycloalkyl, or optionally substituted heterocycloalkyl, may be fused with ring A and with 1 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl group. Even more preferably, the ring A is a thiophene ring represented by formula:
Even more preferably, the ring A is a thiophene ring represented by formula:
The following definitions are supplied to facilitate the understanding of the present invention.
As used herein, the term “comprise” is generally used in the sense of include, permitting the presence of one or more features or components.
As used herein, the singular form “a”, “an” and “the” include plural references unless the context clearly dictates otherwise.
The following paragraphs provide definitions of the various chemical moieties that make up the compounds according to the invention and are intended to apply uniformly throughout the specification and claims unless an otherwise expressly set out definition provides a broader definition.
“Halogen” refers to a fluorine atom, a chlorine atom, a bromine atom, or an iodine atom.
“C1-C10 alkyl” refers to monovalent straight-chained and branched alkyl groups having 1 to 10 carbon atoms. This term is exemplified by groups such as methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec. butyl, tert, butyl, n-pentyl, isopentyl, neopentyl, hexyl, heptyl, octyl, 2-ethylhexyl, 1 ,1 ,3,3-tetramethylbutyl, n-heptyl, 2,4,4 trimethylpentyl, 2- ethylhexyl, octyl, nonyl, decyl, and the like.
“Ci-Cio-heteroalkyl” includes both straight-chained and branched C1-C10 alkyl groups according to the definition above, having at least one or more heteroatoms selected from S, O and N.
“Ci-Ce-haloalkyl” includes both straight-chained and branched Ci-Ce alkyl groups according to the definition above, having at least one or more halogen selected from F, Cl, Br or I.
“Ci-Ce acyl” or “Carbonyl” refers to the group -C(O)R where R includes hydrogen, “Ci-Ce- alkyl”, “aryl”, substituted aryl, “heteroaryl”, “Ci-Ce-alkyl aryl”, “Ci-Ce-alkyl heteroaryl”, or substituted amine (-NH-R’) wherein R’ is optionally substituted aryl.
“Ci-Ce alkyl acyl” refers to Ci-Ce alkyl groups having a Ci-Ce acyl substituent as defined herein.
“Ci-Ce alkyl carbonyl” refers to Ci-Ce alkyl groups having a “carbonyl” substituent as defined herein.
“aminocarbonyl” refers to the chemical moiety -C(0)NRR’ in which each of R and R’ is independently hydrogen, Ci-Ce alkyl, aryl, substituted aryl, heteroaryl, Ci-Ce alkyl aryl, or Ci-Ce alkyl heteroaryl.
“Ci-Ce alkyl aminocarbonyl” refers to Ci-Ce alkyl groups having an “aminocarbonyl” substituent as defined herein.
“C3-C10 cycloalkyl” refers to a saturated carbocyclic group of from 3 to 10 carbon atoms having a single ring (e.g., cyclohexyl) or multiple condensed rings (e.g., norborn yl). Preferred cycloalkyl include cyclopentyl, cyclohexyl, norbornyl and the like.
“Ci-Ce alkyl cycloalkyl” refers to Ci-Ce-alkyl groups having a cycloalkyl substituent, including cyclohexylmethyl, cyclopentylpropyl, and the like.
“C3-C10 heterocycloalkyl” refers to Cs-Cio-cycloalkyl group according to the definition above, in which up to 3 carbon atoms are replaced by heteroatoms chosen from the group consisting of O, S, NR, R being defined as hydrogen or methyl. Preferred heterocycloalkyl include cyclohexane in which 1 carbon atom is replaced by S(O)2, pyrrolidine, piperidine, piperazine, 1 -methylpiperazine, and the like.
As used herein, “optionally substituted C3-C10 heterocycloalkyl” refers to a C3-C10 heterocycloalkyl as defined above that may be fused with 0 to 2 further groups selected
from cycloalkyl, heterocycloalkyl, aryl or heteroaryl group. Preferred groups are benzene, toluene, phenol, aniline, anisole, and the like.
"Ci-Ce-alkyl heterocycloalkyl" refers to Ci-Ce-alkyl groups having a heterocycloalkyl substituent as defined herein.
“C2-Cio-alkenyl” refers to alkenyl groups preferably having from 2 to 10 carbon atoms and having at least 1 or 2 sites of alkenyl unsaturation. Preferable alkenyl groups include ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl and octenyl, and the like. The term "alkenyl" in the meaning of the present invention includes the cis and trans isomers.
“Ci-C6 alkoxy” refers to group -O-R’ where R’ includes both straight-chained and branched "Ci-Ce alkyl" or " Ci-Ce haloalkyl" or " Ci-Ce heteroalkyl" or "aryl" or "heteroaryl" or "Ci-Ce-alkyl aryl" or "Ci-Ce-alkyl heteroaryl". Preferred alkoxy groups include by way of example, methoxy, ethoxy, propoxy, butoxy, phenoxy and the like.
“Amino” refers to -NRR’ in which each of R and R’ is independently hydrogen, Ci-Ce alkyl, substituted aryl, heteroaryl, Ci-Ce alkyl aryl, Ci-Ce alkyl heteroaryl, cycloalkyl, or heterocycloalkyl.
"Ci-Ce-alkyl amino” refers to Ci-Ce-alkyl groups having an amino substituent as defined herein.
“C6-C10 aryl” refers to an unsaturated aromatic carbocyclic group from 6 to 10 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl). Preferred aryl groups include phenyl, naphthyl, phenantrenyl and the like.
As used herein, optionally substituted "C6-C10 aryl" refers to a “C6-C10 aryl” as defined above that may be substituted with 0 to 3 further groups independently selected from halogen (Cl, Br, F, I), Ci-Ce-haloalkyl, and alkoxy as defined herein. Preferred groups are -Cl, -CF3 and -OCH3.
“C6-C10 heteroaryl” refers to a monocyclic heteroaromatic, or a bicyclic or a tricyclic fused- ring heteroaromatic group in which up to 3 carbon atoms are replaced by heteroatoms chosen from the group consisting of O, S, or N.
“ether” refers to -R-O-R’ in which each of R and R’ is independently Ci-Ce alkyl, “Ce-Cio aryl”, and Ci-Ce cycloalkyl.
The present invention also relates to an inhibitor of APT of formula (II):
or a pharmaceutically acceptable salt thereof, wherein
Ring A is a thiophene ring or a furane ring;
Each RAm is independently selected from the group comprising hydrogen, halogen, Ci- C10 alkyl, C2-C10 alkenyl, Ci-Ce alkoxy, Ci-Ce acyl, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, Ce-Cio aryl, and/or Ce-Cio heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups; and m is an integer from 0 to 2;
Each R1 , R2, R4, R5 is independently selected from the group comprising hydrogen, optionally substituted Ci-Ce alkyl, optionally substituted Ce-Cio aryl; Preferably each R1 , R2, R4, R5 is hydrogen; and
R3 is selected from the group comprising
The present invention also relates to an inhibitor of APT of formula (II) selected from the group comprising
or a pharmaceutically acceptable salt thereof.
Preferably, the inhibitor of APT of formula (II) is selected from the group comprising
More preferably, the inhibitor of APT of formula (II) is
In another aspect, the present invention also relates to an inhibitor of APT of formula (II), as described herein, or a pharmaceutically acceptable salt thereof, for use as a medicament.
In a further aspect, the present invention relates to an inhibitor of APT of formula (II) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
The present invention also relates to a method of treating and/or preventing a viral and/or a bacterial infection, the method comprising administering an effective amount in a subject in need thereof of at least one inhibitor of APT of formula (II), or any pharmaceutically acceptable salt thereof as described herein.
In another aspect, the present invention also relates to an inhibitor of APT of formula
, or a pharmaceutically acceptable salt thereof for use in the treatment and/or prevention of a viral and/or a bacterial infection.
The present invention also relates to a method of treating and/or preventing a viral and/or a bacterial infection, the method comprising administering an effective amount in a subject in need thereof an inhibitor of APT of formula
, or any pharmaceutically acceptable salt thereof.
Further, the present invention provides inhibitors of acyl protein thioesterase (APT) of formula (II) as described herein, and/or an inhibitor of APT of formula
or any pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of viral infections mediated by or resulting from acyl protein thioesterases activity, and/or bacterial infections mediated by or resulting from acyl protein thioesterases activity.
As used herein, acyl protein thioesterases (APT) are enzymes that cleave off lipid modifications on proteins, located on the sulfur atom of cysteine residues linked via a thioester bond. In particular, the APT, such as acyl-protein thioesterase 1 (APT1 ) and acyl-protein thioesterase 2 (APT2) are involved in the depalmitoylation of proteins.
Viral infections mediated by or resulting from acyl protein thioesterases activity are infections of multiple human systems including infections of the upper and lower respiratory tract, infections of the skin, infections of the eye, infections of the gastrointestinal tract, infections of the mouth, infections of the urinary genital tract, infections of
the intestinal tract, infections of the brain (e.g. meningitidis or encephalitis), and also systemic, multi organ infections such as haemorrhagic fevers.
For example, viral infections or viral infections associated diseases mediated by or resulting from acyl protein thioesterases activity are selected from the group comprising COVID, SARS, flu, rheumatic diseases, encephalitis, haemorrhagic fevers, acquired immunodeficiency syndrome (AIDS), sarcoma and/or leukemia.
Bacterial infections or bacterial infections mediated by or resulting from acyl protein thioesterases activity are associated with infections of the upper and lower respiratory tract, infections of the skin, infections of the eye, infections of the gastro-intestinal tract, infections of the mouth, infections of the urinary tract, infections of the intestinal tract, infections of the uterus, and infections of the brain (e.g., meningitidis).
Bacterial infections associated diseases mediated by or resulting from acyl protein thioesterases activity are selected from the group comprising Typhoid fever, Salmonellosis, bacteria Gastroenteritis intestinal infection, bacteremia, Shigellosis, Bacteria pneumonia (e.g., legionnaires disease), endophthalmitis, bacteremia, septicemia, sepsis, endocarditis, salpingitis, skin infections, and/or meningitis.
Thus, the present invention provides inhibitors of acyl protein thioesterase (APT) as described herein, or pharmaceutically acceptable salts thereof, for use in the treatment and/or prevention of a viral and/or bacterial infection selected from the group comprising, SARS, COVID, flu, rheumatic diseases, encephalitis, haemorrhagic fevers, acquired immunodeficiency syndrome (AIDS), sarcoma, leukemia, meningitidis, encephalitis, bacteraemia and sepsis.
The present invention also provides inhibitors of acyl protein thioesterase (APT) of formula (II) as described herein, and/or an inhibitor of APT of formula
or pharmaceutically acceptable salts thereof, for use in the treatment and/or prevention of a microbial infection, wherein said microbial infection is a co-infection comprising a viral infection and a bacterial infection.
Together, the viruses or viral families here represented (e.g. Orthomyxoviridae, Paramyxoviridae, Togaviridae, Rhabdoviridae, Filoviridae, Pneumoviridae Coronaviridae, Retroviridae, Herpesviridae, Flaviviridae, Hepeviridae, Bunyaviridae) are responsible of a variety of human diseases that comprise the infection of multiple human systems, including: the respiratory tract (upper and lower), the skin, the eye, the gastrointestinal tract, the mouth, the urinary genital and intestinal tract, the brain (e.g., meningitidis or encephalitis), and also systemic, multi organ infections such as haemorrhagic fevers.
In the present invention, preferably said viral infection can be caused by a virus selected from the group comprising Coronaviruses (CoV) (such as severe acute respiratory syndrome coronavirus (SARS), Middle East respiratory syndrome (MERS), Mouse Hepatitis Virus (MHV), transmissible gastroenteritis virus (TGEV), Swine acute diarrhoea syndrome coronavirus (SADS-cov), Rousettus bat coronavirus HKU9, and any other bat CoVs), Influenza virus (such as A, B,C), Respiratory syncytial virus (hRSV), Newcastle disease virus (NDV), Measles virus, Sindbis virus, Semliki Forest virus (SFV), Rabies virus, Vesicular Stomatitis Virus (VSV), human immunodeficiency virus (HIV), murine leukemia viruses (MulV), Ebola virus, Marburg virus, Herpes simplex virus (HSV), Human cytomegalovirus (HCMV), Dengue virus (DENV), Zika virus (ZIKV), Chikungunya virus, Lassa virus, Nipah virus, Hepatitis E virus, Simian immunodeficiency virus, Lymphocytic choriomeningitis virus, and/or Puumala virus.
For example, said Coronavirus (CoV) can be selected from SARS-CoV-2, SARS-CoV-1 , MHV, TGEV, SADS, MERS-CoV, HCoV-HKLH , HCoV-229E, HCoV-NL63 and/or HCoV- OC43.
In the present invention, preferably said bacterial infection is caused by bacterial strains selected from the group comprising Bacillaceae, Vibrionaceae, Pectobacteriaceae Yersiniaceae, Staphylococcaceae, Streptococcaceae, Legionellaceae, Pseudomonadaceae, Chlamydiaceae, Mycoplasmataceae, Enterobacteriaceae, Pseudomonadales and/or Pasteurellaceae.
These bacterial families or strains are associated with infections of the respiratory tract (upper and lower), the skin, the eye, the gastro-intestinal tract, the mouth, the urinary and intestinal tract, uterus infections and other sexual transmitted diseases, brain infections (e.g., meningitidis), bacteraemia and sepsis.
A pathogen from family Bacillaceae is for example Bacillus anthracis, a pathogen from family Pectobacteriaceae is for example Erwinia spp (carotovora), a pathogen from family Pseudomonadaceae is for example Pseudomonas spp (syringae), a pathogen from family Enterobacteriaceae is for example Salmonella enterica or Shigella spp, a pathogen from family Legionellaceae is for example Legionella pneumophila, a pathogen from family Chlamydiaceae is for example Chlamydia trachomatis.
In the present invention, novel inhibitors of formula (II) as described herein, have been synthesized (Example 1 1 , compounds C1 , C2, C3, C9, C12, C13, C16 and C17).
The inhibition of SARS-CoV-2 intracellular replication has been observed in Calu-3 cells at EC50 concentrations below 2.5 pM without inducing cellular cytotoxicity. Advantageously, novel inhibitors of formula (II) as described herein, demonstrate an EC50 that is decreased by about 3 to 8-fold compared to compound ML-349 (Example 12, figures 1 1 A and B). In contrast, compounds such as C5, C6, C7 that are not covered by formula (II) as described herein, are not potent inhibitors of SARS-CoV2 replication in Calu-3 cells (Example 12, Figure 12).
Furthermore, the inhibition of SARS-CoV-2 infection with an inhibitor of formula (II), such as C2 has been observed as early as 4 h after viral entry into cells (Figure 13A and 13B), thus confirming the inhibitory effect of the intracellular viral RNA replication (Example 13).
In addition, inhibitors of formula (II) are effective without inducing cytotoxic effects in vitro (Example 14, figure 14) or in vivo (Example 16, Figure 16).
Furthermore, it has been demonstrated that inhibition of APT2 activity with an inhibitor of general formula (II), such as C2 or a compound of formula ML349, inhibits replication of SARS-CoV-2 variant Omicron BA.5 (Example 15, Figure 15). Results have been demonstrated for different compounds of formula (II) such as C2, C3, C9, C12, C13, C16 and C17 (Example 20, figure 21 ).
Besides, it has been demonstrated that inhibitors of formula (II) as described herein such as C2, C3 or a compound of formula ML349, can inhibit the APT2-dependent anthrax toxin intoxication of host cells (Example 17, figure 17 and Example 18, figure 18). Results have also been demonstrated for different compounds of formula (II) such as C2, C9, C12, C13, C16 and C17 (Example 19, figures 19 and 20).
In another aspect, the present invention provides a pharmaceutical composition comprising at least one inhibitor of APT of formula (II) as described herein, or pharmaceutically acceptable salts thereof, and a pharmaceutically acceptable carrier, diluent, or excipient.
As to the appropriate carriers, reference may be made to the standard literature describing these, e.g. to chapter 25.2 of Vol. 5 of “Comprehensive Medicinal Chemistry”, Pergamon Press 1990, and to “Lexikon der Hilfsstoffe fur Pharmazie, Kosmetik und angrenzende Gebiete”, by H.P. Fiedler, Editio Cantor, 2002. The term “pharmaceutically acceptable carrier” means a carrier or excipient that is useful in preparing a pharmaceutical composition that is generally safe and possesses acceptable toxicities. Acceptable carriers include those that are acceptable for veterinary use as well as human pharmaceutical use. A “pharmaceutically acceptable carrier” as used in the specification and claims includes both one and more than one such carrier.
The present invention also relates to a pharmaceutical composition, comprising at least one inhibitor of APT of formula (II) as described herein, or pharmaceutically acceptable salts thereof, further comprising one or more additional therapeutic agents and a pharmaceutically acceptable carrier, diluent or excipient.
Preferably, said one or more additional therapeutic agents are selected from the group comprising antibacterial agents and/or antiviral agents.
For example, the at least one or more additional therapeutic agent is an antibacterial agent selected from the group comprising sulfonamides, penicillins, cephalosporins, aminoglycosides, chloramphenicol, tetracyclines, macrolides, lincosamides, streptogramins, glycopeptides, rifamycins, nitroimidiazoles, quinolones, trimethoprim, oxazolidinones, and/or lipopeptides and combinations thereof.
For example, the at least one or more additional therapeutic agent is an antiviral agent selected from the group comprising Aribidol (umifenovir), Favilavir, APN01 , CCR5 antagonist leronlimab (PRO 140), Remdesivir (GS-5734), Galidesivir (BCX4430), Molnupiravir (MK-4482 I FJDD-2801 ), MK-7110 (CD24Fc).
The compounds of the invention, namely inhibitor of acyl protein thioesterase (APT) as described herein, or a pharmaceutically acceptable salt thereof, that can be used in the treatment and/or prevention of a microbial infection can be incorporated into a variety of formulations and medicaments for therapeutic administration. More particularly, one or more compound(s) as provided herein can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers, and can be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, pills, powders, granules, dragees, gels, slurries, ointments, solutions, suppositories, injections, inhalants and aerosols. As such, administration of the compounds can be achieved in various ways, including oral, intranasal, intramuscular, buccal, rectal, parenteral, intraperitoneal, intradermal, transdermal, intracranial and/or intratracheal administration. Moreover, the compound can be administered in a local rather than systemic manner, in a depot or sustained release formulation. The compounds can be formulated with common excipients, diluents or carriers, and
compressed into tablets, or formulated as elixirs or solutions for convenient oral administration or administered by the intramuscular or intravenous routes. The compounds can be administered transdermally and can be formulated as sustained release dosage forms and the like. The compounds can be administered alone, in combination with each other, or they can be used in combination with other known compounds. Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences (Mack Publishing Company (1985) Philadelphia, PA, 17th ed.), which is incorporated herein by reference. Moreover, for a brief review of methods for drug delivery, see, Langer, Science (1990) 249:1527-1533, which is incorporated herein by reference.
Preferably, said pharmaceutical composition is administered by oral, inhalation, nebulization, intranasal, intrapulmonary, intradermal and/or intramuscular route of administration.
In particular, drug substances can be delivered to the respiratory system using various devices such as nebulizer or spray.
The amount of a compound as provided herein that can be combined with a carrier material to produce a single dosage form will vary depending upon the disease treated, the subject in need thereof, and the particular mode of administration. However, as a general guide, suitable unit doses for the compounds of the present invention can, for example, preferably contain between 0.1 mg to about 1000 mg, between 1 mg to about 500 mg, and between 1 mg to about 300 mg of the active compound. In another example, the unit dose is between 1 mg to about 100 mg. Such unit doses can be administered more than once a day, for example, 2, 3, 4, 5 or 6 times a day, but preferably 1 or 2 times per day, so that the total dosage for a 70 kg human adult is in the range of 0.001 to about 15 mg per kg weight of subject per administration. A preferred dosage is 0.01 to about 1.5 mg per kg weight of subject per administration, and such therapy can extend for a number of weeks or months, and in some cases, years. It will be understood, however, that the specific dose level for any particular patient will depend on a variety of factors including the activity of the specific compound employed; the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rate of excretion; other drugs that have previously been administered; and the severity
of the particular disease undergoing therapy, as is well understood by those of skill in the area. A typical dosage can be one 1 mg to about 100 mg tablet or 1 mg to about 300 mg taken once a day, or, multiple times per day, or one time-release capsule or tablet taken once a day and containing a proportionally higher content of active ingredient. The timerelease effect can be obtained by capsule materials that dissolve at different pH values, by capsules that release slowly by osmotic pressure, or by any other known means of controlled release. It can be necessary to use dosages outside these ranges in some cases as will be apparent to those skilled in the art.
In another aspect, the present invention provides a kit comprising an inhibitor of APT of formula (II), as disclosed herein, or a pharmaceutically acceptable salt thereof, and information for use thereof. The kit may further comprise one or more additional therapeutic agents, such as antibacterial agents and/or antiviral agents.
In another aspect, the present invention provides an inhibitor of acyl protein thioesterase (APT) of general formula (I):
or a pharmaceutically acceptable salt thereof, wherein
R’ is independently selected from the group comprising optionally substituted thiophene ring, optionally substituted furane ring, optionally substituted C3-C10 cycloalkyl, or optionally substituted C6-C10 aryl; and
Each R1 , R2, R3, R4, R5 is independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, optionally substituted Ci-Ce alkyl acyl, optionally substituted Ci-Ce alkyl aminocarbonyl, optionally substituted carbonyl, optionally substituted Ci-Ce alkyl carbonyl, optionally substituted C6-C10 aryl, ether, C2-C10 alkenyl, C6-C10 heteroaryl, C3- C10 cycloalkyl, C3-C10 heterocycloalkyl, Ci-Ce alkoxy, optionally substituted amino, and/or Ci-Ce alkyl amino;
for use in the treatment and/or prevention of a viral and/or a bacterial infection.
Preferably, R’ is independently selected from the group comprising optionally substituted thiophene ring, or optionally substituted furane ring.
In one embodiment, R’ is a thiophene ring represented by formula:
wherein: each R6, R7 are independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, C2-C10 alkenyl, C1-C6 alkoxy, C1-C6 acyl, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, C6-C10 aryl, and/or C6-C10 heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups.
Preferably, R’ is a thiophene ring represented by formula:
wherein: each R6, R7 are independently selected from the group comprising hydrogen, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, wherein said cycloalkyl, or optionally substituted heterocycloalkyl, may be fused with ring A and with 1 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl group.
More preferably, R’ is a thiophene ring represented by formula:
Even more preferably, R’ is a thiophene ring represented by formula
Thus, the present invention relates to an inhibitor of acyl protein thioesterase (APT) of formula
or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
In another embodiment, R’ is a furane ring represented by formula:
wherein: each R8, R9, are independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, C2-C10 alkenyl, C1-C6 alkoxy, C1-C6 acyl, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, C6-C10 aryl, and/or C6-C10 heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups.
Preferably, R’ is a furane ring represented by formula:
wherein: each R8, R9 are independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, Ci-Ce alkoxy, Ci-Ce acyl, C3-C10 cycloalkyl, and/or optionally substituted C3-C10 heterocycloalkyl, wherein said cycloalkyl, or optionally substituted heterocycloalkyl, may be fused with ring A and with 1 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl group.
More preferably, the ring A is a furane ring represented by formula:
wherein: each R8, R9 are independently selected from the group comprising hydrogen, C3-C10 cycloalkyl, and/or optionally substituted C3-C10 heterocycloalkyl, wherein said cycloalkyl, or optionally substituted heterocycloalkyl, may be fused with ring A and with 1 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl group.
Thus, the present invention relates to an inhibitor of acyl protein thioesterase (APT) of formula
(ML348), or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
As shown in the examples, inhibitors of acyl protein thioesterases prevent intoxication of mammalian cells with Anthrax toxin by inhibiting the internalization and recycling of the toxin receptor capillary morphogenesis gene 2 (CMG2/ANTXR2) (Example 1 , Figure 1 ) and by blocking endocytosis and pore formation of the anthrax toxin (Example 2, Figure 2).
Furthermore, inhibitors of acyl protein thioesterase (APT1 and 2) block SARS-CoV-2 cellular infection (Figures 5 and 9) and replication (Figures 6-7). It has also been demonstrated that inhibitors of acyl protein thioesterase block cellular infection by other type of virus such as HSV-2 (Figure 8). A dose response was determined for ML349 in Calu-3 cells and the EC50 is 8.6 pM (Figure 10A).
Besides, any viral infections, bacterial infections, pharmaceutical composition, formulations, and combinations that have been described herein are incorporated in this aspect of the invention.
Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described. It is to be understood that the invention includes all such variations and modifications without departing from the spirit or essential characteristics thereof. The invention also includes all of the steps, features, compositions and compounds referred to or indicated in this specification, individually or collectively, and any and all combinations or any two or more of said steps or features. The present disclosure is therefore to be considered as in all aspects illustrated and not restrictive, the scope of the invention being indicated by the appended Claims, and all changes which come within the meaning and range of equivalency are intended to be embraced therein.
EXAMPLES
Materials and methods
Cell lines
RPE1 (ATCC CRL-4000), VERO E6 (ATCC CVCL-0574) and CALU-3 (ATCC HTB55) cells were grown in Dulbecco’s Modified Eagle Medium supplemented with 10% FBS, penicillin, and streptomycin. siRNA
Verified siRNA for human APT2 were purchased from Qiagen. APT2 target sequence: 5’- CAGCTGCTTCTCAGTCATGAA-3’. A pool of verified siRNA was used to target monkey APT1 and purchased from Qiagen. APT1 target sequence: 5’- TACCGACAGGACCCTGTGGAA-3’, 5’-GGTCACAGATATACGGTATNN-3’, 5’- CCGGTGTATGTGCGGCAATNN-3’. As a control siRNA, a sequence targeting the viral glycoprotein VSV-G (5’-ATTGAACAAACGAAACAAGGA-3’) was used. Transfections of 50 nM of siRNA were carried out using TRANSIT-X2 (MIRUS), and the cells were analysed at least 72 h after transfection.
Antibodies and Reagents
The antibodies were commercially available: rabbit anti-human CMG2 (Proteintech, RRID: AB_2056741 ); mouse anti-GAPDH (Acris Antibodies, 4A1 -MA0100, RRID_AB 1874646) ; mouse anti-LPXN (Sigma, SAB1400343), anti-Anthrax Protective
Antigen (List Biological Laboratories, ref: 771 B); N-terminal rabbit anti-MEK2 (Santa Cruz, sc-524), rabbit anti-SPIKE (LIFESPAN, Cat#LS-C19510), HRP-conjugated secondary antibodies (Pierce); and for immunoprecipitation protein G beads were purchased from GE Healthcare, and Streptavidin beads from Sigma.
ML348, ML349 (Cayman Chemical company) and synthesised compounds (C1 , C2, C3, C9, C12, C13, C16, C17) were used at indicated times and concentration. All compounds were solubilised in dimethyl sulfoxide (DMSO).
Wild type PA (Anthrax toxin Protective Antigen) and LF (Anthrax toxin Lethal Factor) were produced in our laboratory by overexpression in E. coli as described (Feld et aL, 2012). Western Blotting
Cells were washed three times in 1 x PBS at 4°C and lysed in Buffer (1 x PBS, 1 % Triton X-100, and protease inhibitor cocktail; Roche) for 30 min on ice. Lysates were then spun down at 5,000 rpm on a table-top centrifuge, and the protein content of supernatant were determined, the samples were boiled in Laemmli buffer for 5 min before separation via SDS-PAGE and western blotting against the different anti-bodies used in this study. Western blots were developed using the ECL protocol and imaged on a Fusion Solo from Vilber Lourmat. Densitometric analysis was performed using the software Bio-1 D from the manufacturer.
Surface biotinylation
Surface biotinylation was performed on RPE1 cells with endogenous CMG2. After toxin treatment (PA63 500ng/ml and LF 50ng/ml), cells were allowed to cool down shaking at 4°C for 15 min to arrest endocytosis. Cells were then washed three times with cold PBS and treated with EZ-Link Sulfo-NHS-SS-Biotin No weight (Thermo Scientific) for 30min shaking at 4°C. Cells were then washed 3 times for 5 min with 100mM NH4CI and lysed in lysis Buffer for 1 h at 4°C. Lysate were then centrifuged for 5 minutes at 5000rpm and the supernatant incubated with streptavidin agarose beads (Sigma) overnight on a wheel at 4°C. Western blots were performed.
Oligonucleotides, Plasmids and Sequences
CMG2-APT2-Flaq Co-immunoprecipitation assays For immunoprecipitations, RPE-1 cells cultured in 10 cm dishes (80% confluency) were transfected with plasmids encoding APT2-Flag for 24 h. Transfected cells were pretreated with APT2 inhibitors at the indicated concentrations 4 h before toxin treatment (PA63 500 ng/ml and LF 50 ng/ml). Toxin treatment was carried out for the indicated time points in the presence of the drugs. Cells were washed and lysed for 30 min at 4°C in PBS-1 % triton-X, with protease inhibitors cocktail (Roche), centrifuged for 3 min at 2000 g, and supernatants further precleared with protein G-agarose conjugated beads for 30 min. Cleared supernatants were incubated for 16 h at 4°C with CMG2 antibodies and beads. Beads were washed three times in lysis buffer, resuspended in 60 pl Laemmli
buffer, and boiled, and the CMG2 immunoprecipitated fractions were processed for Western blot analysis as described.
Viral Stock production and titration with plaque-based assays
All viral stocks were produced and isolated from supernatants of Vero E6 cells, cultured in T75 culture flasks to a confluency of 80-90%, and infected with an original passage 2 (P2) SARS-CoV-2 virus (lineage B.1 ), for 48 or 72 h, at MOI«0.05, in 10 ml DMEM supplemented with 2.5% FCS. Stocks were obtained from the following strain: hCoV- 19/Switzerland/GE-SNRCI-29943121/2020, (GISAID ID: EPI _ISL_ 414019). For Omicron stocks, cells were infected with BA.5 isolates (ID: EPI_ISL_12268493.2 to produce P2 working stocks. Passage 3 SARS-CoV-2 B.1 or passage 2 Omicron BA.5 supernatants were harvested, clear of cell debris by centrifugation (500 g 10 min) and filtration (0.45 pm), aliquoted and stored at -80 C. Viral titters were quantified by determining the number of individual plaque forming units after 48 h of infection in confluent Vero E6 cells. In brief, viral stocks were serially diluted (10-fold) in serum-free medium and (400 pl) inoculated in triplicate 48 wells, confluent Vero E6 cells (2.5 x 105) cells per well. After 1 h, inoculums were discarded, and cells overlaid with a mixture of 0.4% of Avicel-3515 (Dupont) (from 2% stock) in DMEM supplemented with 5% FCS and pen/strep for additional 48 h. Overlays were discarded, and cells fixed in 4% PFA for 30 min at RT. Fixed cells were washed in PBS and stained with 0.1 % crystal violet solution (in 20% ethanol/water) for 15 min. Staining solution was discard and wells washed twice in water. Plates were allowed to dry and analyzed for quantification of the cytopathic effect as number of individual Plaque forming units (PFU) per ml (Avg PFU*1/Volume*1 /dilution factor). Such un-concentrated viral stocks yielded between 0.5 to 3x106 PFU/ml.
SARS CoV-2 infections
All infections for experimental analysis were done using passage 3 SARS-CoV-2 (B.1 ) stocks. BA.5 passage 2 was used for experiments using the Omicron variant. Vero E6 cells or CALU-3 cells seeded to a confluency of 90 to 100%, were, washed twice in warm serum-free medium and inoculated with the indicated MOI of SARS-CoV-2, diluted in serum free medium (1 ml for 6-well plates and 500 pl for 12-well plates). 1 hour after inoculation cells were washed with complete medium and infection allowed to proceed for the indicated time points in DMEM supplemented with 2.5% FCS, penicillin and
streptomycin. For plaque-based assays culture medium was supplemented with Avicel overlay as described.
Drug treatments
All drug treatments were done as indicated in complete culture and/or infection medium. Equivalent volume of Dimethyl sulfoxide (DMSO), used as a solvent, was added for control samples.
VSV-CoV-2 production and Inhibition Assays
Vesicular stomatitis virus (VSV)-based SARS-CoV-2 pseudotypes (VSV-CoV-2) generated according to (Gasbarri et al, 2020) expressing a 19 amino acids C-terminal truncated spike protein (NCBI Reference sequence:NC_045512.2) were produced in HEK293F and titrated in Vero-E6. For infection, Vero-E6 cells (13,000 cells per well) were seeded in a 96-well plate. Compounds were serially diluted in DMEM and incubated with VSV-CoV-2 (MOI, 0.001 infectious units /cell) for 1 h at 37 °C. The mixture was added on cells for 1 h at 37 °C. The monolayers were washed and overlaid with medium containing 2% FBS for 18 h. The following day cells were fixed with paraformaldehyde 4%, stained with DAPI, and visualized using an ImageXpress Micro XL (Molecular Devices, San Jose, CA, USA) microplate reader and a 10x S Fluor objective. The percentage of infected cells was estimated by counting the number of cells expressing GFP and the total number of cells (DAPI-positive cells) from four different fields per sample using MetaXpress software (Molecular Devices, San Jose, CA, USA).
Analysis of intracellular viral RNA-Q-PCR
Infected cells (from 6 and 12 well plates) and correspondent supernatants (when indicated - 150 pl from 6 well plates), were harvested and lysed in 220-330 pl of Maxwell® RSC Viral Total Nucleic Acid Purification Kit-lysis buffer from Promega, incubated at 80°C for 10 min, and used for Viral RNA extraction according to manufacturer’s instructions. RNA concentration was measured and 500 ng or 1000 ng of total RNA was used for cDNA synthesis using iScript. A 1 :5 dilution of cDNA was used to perform quantitative real-time PCR (Q-PCR) using Applied Biosystems SYBR Green Master Mix on 7900 HT Fast Q-PCR System (Applied Biosystems) with SDS 2.4 Software. Primers used are described in Supplementary table 1 . All data (always in triplicate) were normalized to Ct values from three housekeeping (HK) genes ALAS-1 , Guss and TBP, except for
supernatants from replication experiments at 24 h, which Ct data were normalized using Ct values from correspondent cellular HK genes. Results were expressed as 2A(- AACt)*100%.
Analysis of intracellular viral Nucleocapsid-N levels by Western blot
Infected cells (24 well plates) (cultured at 1 .5 x 10A5 cells per well, 24 h before infection) were harvested and lysed in 150 pl of lysis buffer (0.5% NP-40, 500 mM Tris-HCI pH7.4, 20 mM EDTA, 10 mM NaF, 30mM sodium pyrophosphate decahydrate, 2 mM benzamidine and protease inhibitor cocktail; Roche) for at least 30 min. Lysates were cleared by centrifugation (5000g, 3 min), and the protein content of the supernatant was determined. Samples were resuspended in 50 pl Laemmli buffer, boiled for 5 min, and approximately 20-30 pg of protein lysate were used for Western blot analysis (as described) using antibodies against viral nucleocapsid N and host GAPDH. The percentage of infection was calculated as the densitometric ratio between N and GAPDH levels for each sample.
Realtime-Glo™ mt cell viability assay
The number of viable cells in culture was measured by determining the reducing potential of cultured cells, thus metabolism, in Calu-3 cells. The assay measures the reduction of a cell-permeable substrate, which is diffused to the medium and processed by an exogenously added reporter enzyme (NanoLuc® luciferase). The reaction produces a luminescent signal that correlates with the number of viable cells. The luminescence signal was monitored every day for 6 days in Calu-3 cells cultured in 96-well plates (50000 cells/well), 24 h prior addition of the substrate and increasing concentrations of ML349, C2, and C3. Two wells per compound per concentration were average for 4 independent biological replicates. Results are mean ±SEM of n=4. Dose-dependent viability curves, and cytotoxic concentration, were determined on day 6 of treatment using nonlinear regression and ANOVA analysis (Prism).
Analytical flow cytometry of SARS-CoV-2 infected cells
Vero E6 cells were infected in 6-well plates [as mentioned previously]. Cells washed and scrapped in 1 ml PBS and recovered by mild centrifugation 400g 3 min. Supernatants were discard and cells were fixed in 4% PFA for at least 30 min, washed twice in PBS, and permeabilized for 5 min with 0.1 % Triton in PBS. The following blocking, antibody,
and washing steps were all done with FACS buffer (2% FBS in PBS/EDTA) primarily at RT. Blocking was done for 15 min, primary antibody incubation with mouse anti-Spike antibody (LC-C19510) was performed at 1 :200 for 30 min at RT or at 1 :500 ON at 4 °C, and cells were washed twice. Alexa-647-conjugated donkey anti-mouse was used as a secondary antibody at 1 :600 for 30 min at RT, after which cells were washed three times and kept cold before flow cytometry acquisition. Analytical flow cytometry was performed using an LSRII or LSR Fortessa (BD; Becton Dickinson) instrument and results were analyzed using the FlowCore package in R.
LDH cytotoxicity assay
Lactate dehydrogenase (LDH) cytotoxicity assays were performed, using In Situ Death Detection Kit (Roche, Indianapolis, IN), in Vero E6 or CALU-3 cells cultured in 12 well plates with equivalent culture conditions used for infection assays.
HSV infection and inhibition assays
Wild-type HSV-2 viral suspensions were inoculated onto cells pre-treated with the indicated drugs for 1 h, at a multiplicity of infection (MOI) of 0.01 plaque-forming units (PFU) as described (Cagno et al, 2018). Viral inoculum was removed, and cells washed after 2 h, and infection was continued in the presence of the drugs for evaluation through a plaque-based assay.
Evaluation of Toxicity in vivo in C57BL/6 mice
1 1 -week-old male C57BL/6 mice were inoculated intraperitoneally with 400 to 600 pl of a 2 mg/ml working solution of C2 or ML349, resuspended in PEG300 50% (v/v) solution in PBS, to a final dose of 1 mg of compound per 25 g of body weight. Control mice were inoculated in parallel with an equivalent volume (per body weight) of vehicle PEG300 50% (v/v) in PBS solution. Drug and vehicle treatments were injected on days 1 , 3, 6, 8, and 10 of the experiment, whereas clinical parameters (body weight, blood pressure, and pulse) were monitored on days 1 , 2, 3, 6, 7, 8, 9, 10, and 13. After 13 days of treatment, mice were euthanatized, and different organs were harvested for histological analysis and weight measurements.
Quantification and statistical analysis
Unless otherwise indicated, all data were repeated at least three times independently. Unless otherwise stated, each data point corresponds to one independent experiment with consistent results. Statistical analysis was carried out using Prism software. Data representations and statistical details can be found in the description of the figures. For ANOVA analysis, p values were obtained by post hoc tests to compare every mean and pair of means (Tukey’s & Sidak’s) or to compare every mean to a control sample (Dunnet’s). Half-maximal effective concentrations (EC50) were obtained using dose inhibition curves fitted using nonlinear regression with a variable slope (Prism).
Targeting depalmitoylation inhibits intoxication by bacterial toxins
Example 1 : Inhibition of APT2 activity increases surface expression of ANTXR2/CMG2 Experiments were performed by surface biotinylation as described in materials and methods. Labelling of surface proteins with NHS-biotin was carried out in mammalian cells left untreated or treated with ML349 (10 pM) for the indicated times. The surface proteins were pulled down with streptavidin beads. The corresponding cell extracts were analyzed by western blot using antibodies and reagents as described in materials and methods. Leupaxin (LPXN) and glyceraldehyde-3-phosphate deshydrogenase (GAPDH) are blotted as positive and negative controls respectively. The quantification of surface ANTXR2/CMG2 is expressed as % of t=0 and results are mean ±SD (n>3) (Figure 1 ).
In these assays, pre-incubation of cells with the ML349 gradually blocks the internalization of the toxin receptor capillary morphogenesis gene 2 (CMG2/ANTXR2), causing its accumulation in the plasma membrane (Figure 1 ).
In this example, the inhibition of the thioesterase APT2 with ML349 prevents the endocytosis of the anthrax toxin receptor capillary morphogenesis gene 2. These results suggest that the inhibition of depalmitoylation through specific drugs that affect acyl protein thioesterases (APTs) could block the entry of pathogenic toxins into the host cells.
Example 2: Inhibition of APT2 activity prevents intoxication of mammalian cells with Anthrax toxin.
Experiments were performed by synchronized anthrax intoxication assays in mammalian cells. The results are represented on Western blots (Figure 2A and B).
Mammalian cells were left untreated (control) or co-treated with ML349 (10 pM) or ML348 (10 pM) during 0, 20, 40 and 60 minutes. Cells were pre-treated with ML349 or ML348
during 2 h (Figure 2A) or 4 h (Figure 2B) prior to and during exposure to Anthrax toxin Protective Antigen (PA).
The dynamics of toxin pore formation (PA Pore) (Figure 2C) and MEK2 degradation (Figure 2D) were quantified for each time point for cells pre-treated for 2 h and expressed as % of t=0 for each sample. Results are mean ±SD (n>3).
Treatment with ML349, blocks endocytosis of CMG2/ANTXR2, thus preventing pore formation of the anthrax toxin PA subunit. ML349 thus prevents the protease subunit of anthrax toxin to reach the cytosol and in turn prevents the cleavage of a major host cell signaling regulator, the MAP kinase-kinase MEK.
Example 3: siRNA silencing of APT2 expression prevents intoxication of mammalian cells with Anthrax toxin.
Experiments were performed using siRNA silencing APT2 as described in materials and methods in paragraph siRNA.
Synchronized anthrax intoxication assays were performed in mammalian cells transfected with control or siRNA oligos targeting the expression of APT2 (siAPT2) as described in materials and methods (siRNA). Mammalian cells were left in culture for 72 h after transfection and then intoxicated with Anthrax toxin Protective Antigen (PA). Results are represented on Western blot (Figure 3A). The dynamics of toxin pore formation (PA Pore) measured by western blot in Figure 3A were quantified for each time point 0, 20, 40 and 60 minutes, and values expressed as % of t=0 for each sample (Figure 3B). Results are mean ±SD (n>3).
The results show that silencing APT2 expression (Figure 3 A) recapitulates the effects of inhibiting its enzymatic activity with ML349 depicted in Figure 2A and 2B. Thus, inhibition of depalmitoylation can function as an effective strategy to block host cellular intoxication with bacterial toxins.
Targeting depalmitoylation inhibits Viral infections
Example 4: Inhibition of APT1 activity with ML348 diminishes SARS-CoV-2 Spike- pseudotyped particles (PPs) entry into host cell.
A cell-based assay was performed to determine the effects of different chemical inhibitors on the internalization of Vesicular Stomatitis Virus (VSV)-based particles pseudotyped with the spike fusion glycoprotein from SARS-CoV-2. The effects of ML348 and ML349, inhibitors of APT1 and APT2 respectively, as well as Palmostatine, Bromopalmitate, Chlroroquine and Hydroxychloroquine were tested and are represented on Figure 4.
Experiments were realized as described in materials and methods (VSV-CoV-2 production and Inhibition Assays). Vero E6 cells were infected with SARS-CoV-2-Spike- pseudotyped particles (VSV-S-CoV2) or VSV-G-pseudotyped particles(VSV-G) (Figures 4A, B and C). Vero cells were either pre-treated (PRE-TREAT, Figure 4C) for 4 h washed, and infected with VSV-S- CoV2 or VSV-G, or infected in the presence of the same concentration of the chemical inhibitors (PRE+CO-TREAT). GFP-expressing cells were quantified by fluorescent microscopy. The percentage of infection was calculated comparing the percentage of positive cells to the percentage in infected untreated controls (DMSO). Dose-response curve was calculated for ML348, ML349, and hydroxychloroquine. (Figure 4C). The 50% effective concentrations (EC50) were determined using Prism software (GraphPad Software, San Diego, CA): EC50 (ML348=19pM, EC50 (hydroxychloroquine)=1 1 pM).
ML348 specifically inhibited the entry of PPs typed with SARS-CoV-2 spike protein to a similar extend as hydroxychloroquine (Figure 4A). This inhibitory effect was dosedependent (Figure 4C) and not observed upon treatment of target cells with unspecific inhibitors of palmitoylation and depalmitoylation such as Bromopalmitate and Palmostatine respectively, neither by treatment with the APT2 inhibitor ML349 (Figure 4A and 4B).
Example 5: Inhibition of APT1 activity with ML348 blocks SARS-CoV-2 cellular infection. The depalmitoylation-targeting drugs ML348, ML349 were tested during infection of Vero E6 cells with a clinical isolate of SARS-Cov-2. Infection was monitored using a standard plaque-based infection assay as described in materials and methods (Viral stock production and titration with plaque-based assays; SARS CoV-2 infections).
Vero E6 cells were pre-treated with ML348 or ML349 for 1 h washed, and infected with SARS-CoV-2, in the presence of drug. Infection was determined using a plaque assay and a dose-response curve was established for ML348 and ML349 (Figure 5A). The
EC50 for ML348 is 13.3 pM. Then, Vero E6 cells were infected with SARS-CoV-2 (MOI 0.1 ) for 24 h, in the presence of 12.5 pM (Figure 5B). Drugs were added 1 h after viral inoculation, and replication was monitored by determining the % of high Spike-positive cells by flow cytometry. Results are mean ±SEM and each dot represents one individual biological replicate.
It has been found that SARS-CoV-2 cellular infection was inhibited when carried out in the presence of increasing micromolar doses of ML348 (EC50~13.3 pM) reaching full inhibition (Figure 5A). Complementary infection assays using a ML348 concentration at 12.5 pM below the determined EC50, demonstrated that treatment of infected cells with ML348, after viral inoculation and throughout the remaining time of infection, greatly reduced the number of Spike-positive infected cells, confirming the inhibitory effect of ML348 (Figure 5B).
Example 6: Inhibition of APT1 activity with ML348 inhibits SARS-CoV-2 intracellular replication.
To understand whether the effect of ML348 occurs within a single cellular infection cycle (up to 8 to 12 h), viral genomic RNA replication was monitored throughout time by probing for two viral genomic regions corresponding to the E and RdRp genes of SARS- CoV-2. Vero E6 cells were treated with ML348 1 h after viral inoculation and throughout the time of infection.
Vero E6 cells were inoculated for 1 h with SARS-CoV-2, washed and subsequently incubated in complete medium with 12.5 pM of ML348. Cells were harvested at the time points 0, 4, 8 and 12 hours (Figure 6A and 6B). Infection was determined by monitoring the levels of intracellular E and RNA-dependent RNA polymerase-RdRp transcripts by Q- PCR as described in materials and methods. t=0 was harvested 1 h after inoculation before drug treatment. Normalized results, with relation to control (w.r.t.) are the mean ± SD of three independent biological replicates.
ML348 blocked SARS-CoV-2 intracellular viral RNA replication as early as 8h after viral inoculation (Figure 6A and 6B).
Example 7: Inhibition of APT1 activity and silencing of APT1 expression blocks SARS- CoV-2 infection in Vero E6 cells.
Viral RNA replication in Vero E6 cells was also monitored after 24h of infection to follow multiple rounds of cellular infection. In these experiments, the viral intracellular replication was monitored by determining the levels of the sub-genomic E RNA, a bona-fide marker of intracellular viral RNA replication.
Quantification of viral RNA replication (E, RdRp and E subgenomic) was analyzed from Vero E6 cells or correspondent supernatants of Vero E6 cells infected with SARS-CoV-2 (MOI-O.1 for 24 h), treated without drug (Control), with ML348 or ML349. Cells were inoculated for 1 h with SARS-CoV-2, washed and subsequently incubated in complete medium with 12.5 pM of ML348 or ML349 (Figure 7 A and 7B).
As already observed in cells treated with ML348 after viral inoculation (see Example 6), the viral RNA replication was significantly reduced by ML348 both in cells and in culture supernatants containing released virions (Figures 7A and 7B).
In a further experiment, cells were previously transfected with control or siRNA oligos targeting the expression of APT1 (siAPT) for 72h as described in materials and methods (siRNA; Figure 7C).
The quantification of APT1 mRNA levels from the samples used in Figure 7C is represented on Figure 7D. The depletion is approximately 60%. All results were normalized to the mean of control samples and are mean ± SEM of n >3. p values were obtained by ANOVA analysis and *p<0.05, ***p<0.001 , ****p<0.0001 .
The effects caused by inhibition of the enzymatic activity of APT1 by ML348 (Figures 7A and 7B) can be recapitulated by carrying SARS-CoV-2 infections in Vero E6 cells siRNA- treated for monkey APT1 (Figure 7C). These experiments confirmed the specificity of ML348 inhibition during SARS-CoV-2 infection.
Example 8: Inhibition of APT1 activity with ML348 blocks HSV-2 cellular infection.
In this experiment, it was evaluated whether ML348 or ML349 could limit infection by other type of viruses. A plaque-based assays was performed using herpes simplex virus type 2 (HSV-2) according to materials and methods (HSV infection and inhibition assays).
Vero E6 cells were pre-treated for 1 h in the presence of drug, washed, and infected with HSV-2 (Figure 8 A). Infection was determined using a plaque assay. Dose-response was determined for ML348 and ML349. The EC50 for ML348 is 23.2 pM.
ML348 treatment reduces infection of HSV-2 in a dose-dependent manner (Figure 8). These data highlight the larger anti-viral scope of targeting depalmitoylation, using a specific inhibitor of APT1 activity.
Example 9: Inhibition of APT2 activity blocks SARS-CoV-2 infection in Calu-3 lung- derived cells.
The mRNA expression of APTs varies between cell lines. APT2 is often more abundant than APT1. In human airway epithelial cells, such as the lung-derived cell line, CALU-3, APT2 is highly enriched when compared to Vero E6 cells (Figure 9A).
Given the tropism of SARS-CoV-2 to lung cells, the effect of inhibiting depalmitoylation on viral replication was evaluated in Calu-3 cells.
The LYPLA1 (APT1 ) and LYPLA2 (APT2) mRNA levels are measured in different cell lines (Figure 9A). Values are TAG per Million (TPM) corresponding to 10*6 X ((reads mapped to transcript/transcript length)/ SUM (reads mapped to transcript/transcript length)). Results are the mean of independent experiments each from n > 2 biological replicates.
The replication of SARS-CoV-2 was monitored in Calu-3 cells at 24h post-inoculation and upon treatment with 12.5 pM of ML348 or ML349. This concentration is below the EC50 of ML348 for inhibition of viral replication in Vero E6.
Figure 9B shows the quantification of viral RNA replication (E, RdRp and E subgenomic) from Calu-3 cells and correspondent supernatants infected with SARS-CoV-2 (MOI-0.1 for 24 h), treated with ML348 or ML349. The cells were inoculated for 1 h with SARS-CoV- 2, washed and subsequently incubated in complete medium with 12.5 pM of ML348 or ML349.
In another experiment, the cells were previously transfected with control or siRNA oligos targeting the expression of APT2 (siAPT2) for 72h (Figure 9C). Figure 9D shows the
quantification of APT2 mRNA levels from the samples used in Figure 9C. All results were normalized to the mean of control samples and are mean ± SEM of n >3. p values were obtained by ANOVA analysis and *p<0.05, ***p<0.001 , ****p<0.0001 .
The treatment of cells with ML349 significantly diminishes viral intracellular replication and secretion to the culture supernatant (Figure 9B). Correspondently, partial depletion of APT2 in Calu-3 also diminished intracellular replication of SARS-CoV-2 after 24h of infection in Calu-3 cells (Figure 9C and 9D). Thus, these experiments in Calu-3 cells confirm that targeting depalmitoylation activity represents a valid strategy to inhibit SARS- CoV-2 infection.
Example 10: Dose-response inhibition of SARS-CoV-2 replication in Calu-3 cells by ML349 and cytotoxicity evaluation of ML348 and ML349.
In this experiment, the EC50 of ML349 inhibiting SARS-CoV-2 intracellular replication was determined in Calu-3. In addition, an analysis of the potential cytotoxic effects caused by the drug-treatment protocols was performed.
Calu-3 cells were infected with SARS-CoV-2. Infection was determined using Q-PCR by quantifying the mean levels of E and RdRp viral RNA (Figure 10A). Normalized results are mean ± SEM. The dose-response was determined for ML349 and the EC50 is 8.6 pM.
A Lactate dehydrogenase activity assay was performed as described in the material and methods section. Vero E6 cells or Calu-3 cells were treated with increasing concentrations of ML348 for 48 h or ML349 for 24 h, respectively. The results of the cell viability are mean ± SD of 2 biological replicates. Importantly, neither treatment of Vero E6 with ML348 for 48h nor treatment of Calu-3 with ML349 for 24h, triggered significant cellular toxicity at different ranges of concentrations (Figure 10B and 10C).
The inhibitory effect of ML349 during SARS-CoV-2 replication in Calu-3 cells (EC50 ~ 8.6 pM) (Figure 10A) and the one observed for ML348 in infection of Vero E6 cells (EC50~13.3 pM) demonstrate that both drugs ML348 and ML349 are potent inhibitors for SARS-CoV-2 infection.
Altogether, targeting host cell depalmitoylation activity constitutes a safe strategy to inhibit viral infections such as SARS-CoV-2, or HSV, and bacterial-mediated intoxication.
Example 1 1 : Synthesis of novel inhibitors of acyl protein thioesterase (APT) of general formula (II).
Synthesis of carboxylic acid precursor
Phosphorus oxychloride (647 pL, 1.14 Eq, 6.94 mmol) was added dropwise to ice-cold anhydrous DMF solution (12 mL, 0.48 molar), followed by dropwise addition of thiochroman-4-one (1.00 g, 6.09 mmol). The reaction mixture was stirred for 30 minutes at 0°C, then heated at 80°C for 2 hours, and quenched with 4mL of ice-cold aqueous sodium acetate (25%). The mixture was extracted with CH2CI2 (4 x 30mL), washed with brine, dried with Na2SO4 and concentrated. The crude product, 4-chloro-2H- thiochromene-3-carbaldehyde (1 .12 g, 5.34 mmol), was obtained as a brown oil.
1 H NMR (400MHz, Chloroform-d) 5 10.32 (s, 1 H), 7.95-7.88 (m, 1 H), 7.34 - 7.40 (m, 3H), 3.71 (s, 2H).
69% yield
4-chloro-2H-thiochromene-3-carbaldehyde (1.28 g, , 6.08 mmol) is slowly added to a mixture of ice-cold ethyl 2-mercaptoacetate (1 .33 mL, 2 Eq, 12.2 mmol), sodium ethoxide (1.65 g, 4 Eq, 24.3 mmol) in ethanol (7.06 mL, 0.86 molar). The reaction mixture was stirred overnight at room temperature and then heated at 80°C for 2 hours. Solvents were removed and EtOAc was added to the residue. The organic layer was washed with brine,
dried and purified by flash column chromatography (20% EtOAc, 80% Cyclohexane) to yield ethyl 4H-thieno[3,2-c]thiochromene-2-carboxylate (1 .33 g, 4.83 mmol, 80 %).
1 H NMR (400MHz, Chloroform-d) 5 7.57 (s, 1 H), 7.54 - 7.45 (m, 1 H), 7.39 - 7.33 (m, 1 H), 7.20 - 7.15 (m, 2H), 4.36 (q, J = 7.14 Hz, 2H), 3.94 (s, 2H), 1 .39 (t, J = 7.12 Hz, 3H).
Ethyl 4H-thieno[3,2-c]thiochromene-2-carboxylate 5,5-dioxide
mCPBA (3.75 g, 3.00 Eq, 21 .70 mmol) was added portionwise to an ice-cold solution of ethyl 4H-thieno[3,2-c]thiochromene-2-carboxylate (2.00 g, 1.00 Eq, 7.24 mmol) in CH2CI2 (80 mL). The reaction was warmed to room temperature and stirred until complete conversion (3 h, TLC). The reaction mixture was quenched by adding a mixture of sat. aq. Na2S2O3 (10 mL) and sat. aq. NaHCO3 (50 mL) and stirred for 1 h. The layers were separated and the aqueous layer was extracted with CH2CI2, dried over Na2SO4, filtered, and concentrated under vacuum. The compound was purified by flash column chromatography (CH2CI2 100% to CH2CI2:MeOH 95:5) to yield ethyl 4H-thieno[3,2- c]thiochromene-2-carboxylate 5,5-dioxide (710 mg, 7.24 mmol, 32%) as a yellow powder. 1 H NMR (400 MHz, DMSO-d6) 5 7.99 (ddd, J = 7.8, 1.3, 0.5 Hz, 1 H), 7.89 (dd, J = 7.8, 1 .2 Hz, 1 H), 7.81 (td, J = 7.6, 1 .3 Hz, 1 H), 7.70 (td, J = 7.6, 1 .2 Hz, 1 H), 4.96 (s, 1 H), 4.35 (q, J = 7.1 Hz, 1 H), 1 .33 (t, J = 7.1 Hz, 2H).
To a solution of ethyl 4H-thieno[3,2-c]thiochromene-2-carboxylate 5,5-dioxide (598 mg, 1 .00 Eq, 1 .94 mmol) in a mixture of THF/MeOH/H2O (2:1 :1 ) (30 mL) was added lithium hydroxide monohydrate (407 mg, 5.00 Eq, 9.70 mmol). The reaction mixture was stirred at room temperature for 16 h then partially concentrated under vacuum. Water was added and the solution was washed with EtOAc (2x) and acidified with aq. 6M HCL The aqueous
layer was washed with EtOAc. Finally, the organic layers were combined, washed with brine, dried over Na2SO4 and concentrated under vacuum. 4H-thieno[3,2- c]thiochromene-2-carboxylic acid 5,5-dioxide (533 mg, 1 .94 mmol, 98%) was obtained as a white powder.
1 H NMR (400 MHz, DMSO-d6) 5 13.53 (s, 1 H), 7.98 (ddd, J = 7.8, 1 .3, 0.5 Hz, 1 H), 7.89 - 7.76 (m, 3H), 7.68 (td, J = 7.6, 1 .3 Hz, 1 H), 4.94 (s, 2H).
Synthesis of amide derivatives
To a solution of 4H-thieno[3,2-c]thiochromene-2-carboxylic acid 5,5-dioxide (1.00 Eq) in anhydrous DMF (0.1 M) under N2 atmosphere, DIPEA (2.00 Eq), HBTU (3.00 Eq) and the secondary amine (2.00 Eq) were added. The mixture was stirred under N2 at room temperature for 16 h. Water was added to the solution and the aqueous layer was washed with EtOAc. The organic layer was washed with sat. aq. NH4CI, brine and, dried over Na2SO4. The solvent was evaporated and the crude residue was purified by column chromatography (cHex:EtOAc) to afford the final compound.
(4-(4-fluorophenyl)piperazin- 1 -yl)(4H-thieno[3,2-c]thiochromen-2-yl)methanone (SPC-
Compound SPC-EPFL-1 (C1 ) was prepared following General Procedure A using 1 -(4- fluorophenyl)piperazine (32 mg, 1 Eq, 0.18 mmol) to afford (5,5-dioxido-4H-thieno[3,2- c]thiochromen-2-yl)(4-(4-fluorophenyl)piperazin-1 -yl)methanone (42.6 mg, 54 %) as a white solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-30% 5CV, 30-100% for 15CV, 100% for 3CV].
1 H NMR (400MHz, DMSO-d6) 5 8.01 - 7.92 (m, 1 H), 7.81 -7.75 (m, 2H), 7.68 - 7.60 (m, 1 H), 7.55 (s, 1 H), 7.15 - 7.05 (m, 2H), 7.02 - 6.95 (m, 2H), 4.90 (s, 2H), 3.81 - 3.75 (m, 4H), 3.18 (app t, J = 5.2 Hz, 4H).
Compound SPC-EPFL-2 (C2) was prepared following General Procedure A using 1 -(2- chlorophenyl)piperazine (35 mg, 1 Eq, 0.18 mmol) to give (4-(2-chlorophenyl)piperazin- 1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (41.4 mg, 51 %) as a white solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-30% 5CV, 30-100% for 15CV, 100% for 3CV]. 1 H NMR (400MHz, DMSO-d6) 5 8.02 - 7.93 (m, 1 H), 7.83 - 7.76 (m, 2H), 7.70 - 7.60 (m, 1 H), 7.56 (s, 1 H), 7.45 (dd, J = 7.9, 1 .5 Hz, 1 H), 7.36 - 7.28 (m, 1 H), 7.19 (dd, J = 8.1 , 1 .5 Hz, 1 H), 7.09 (td, J = 7.6, 1 .5 Hz, 1 H), 4.89 (s, 2H), 3.89 - -3.81 (m, 4H), 3.06 (app t, J = 5Hz, 4H).
Compound SPC-EPFL-3 (C3) was prepared following General Procedure A using 1 -(2,3- dichlorophenyl)piperazine (41 mg, 1 Eq, 0.18 mmol) to give (4-(2,3- dichlorophenyl)piperazin-1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (30.4 mg, 35 %) as a solid after purification by flash column chromatography [Silica-CS
12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-30% 5CV, 30-100% for 15CV, 100% for 3CV].
1 H NMR (400MHz, DMSO-d6) 5 8.01 - 7.96 (m, 1 H), 7.83 - 7.74 (m, 2H), 7.70 - 7.60 (m, 1 H), 7.56 (s, 1 H), 7.37 - 7.31 (m, 2H), 7.19 - 7.1 1 (m, 1 H), 4.89 (s, 2H), 3.95 - 3.98 (m, 4H), 3.08 (app t, J = 4.91 Hz, 4H).
Compound SPC-EPFL-5 was prepared following General Procedure A using phenyl(piperazin-1 -yl)methanone (34 mg, 1 Eq, 0.18 mmol) to give (4-benzoylpiperazin- 1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (65.1 mg, 81 %) as a solid after purification by flash column chromatography [Silica-CS 12g, eluent: DCM/MeOH (0% MeOH for 3CV, 0-5% 15CV, 5% for 3CV, 5-10% for 3CV].
1 H NMR (400MHz, DMSO-d6) 5 7.97 (dd, J = 7.6 Hz, 1 H), 7.75-7.82 (m, 2H), 7.61 -7.67 (m, 1 H), 7.54 (s, 1 H), 7.40-7.50 (m, 5H), 4.88 (s, 2H), 3.35-3.90 (br m, 8H),
Compound SPC-EPFL-6 was prepared following General Procedure A using 2-(5- methoxy-1 H-indol-1 -yl)ethan-1 -amine (27 mg, 1 Eq, 0.14 mmol) to give N-(2-(5-methoxy- 1 H-indol-1 -yl)ethyl)-4H-thieno[3,2-c]thiochromene-2-carboxamide 5,5-dioxide (36.8 mg,
57 %) as a solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-30% 5CV, 30-100% for 15CV, 100% for 3CV]. 1 H NMR (400MHz, DMSO-d6) 5 8.87 (t, J = 5.8 Hz, 1 H), 7.95 (d, J = 7.6 Hz, 1 H), 7.75 - 7.80 (m, 2H), 7.67 - 7.61 (m, 2H), 7.41 (d, J = 8.8 Hz, 1 H), 7.28 (d, J = 3.0 Hz, 1 H), 7.04 (d, J = 2.4 Hz, 1 H), 6.78 (dd, J = 8.8, 2.4 Hz, 1 H), 6.32 (d, J = 3.0 Hz, 1 H), 4.94 (s, 2H), 4.32 (t, J = 6.0 Hz, 2H), 3.59 (q, J = 6.0 Hz, 2H), 3.74 (s, 3H).
(4-(3-aminobicyclo[ 1.1.1 ]pentan- 1 -yl)piperazin- 1 -yl)(4H-thieno[3,2-c]thiochromen-2- yl)methanone (SPC-EPFL-7)
Tert-butyl(4-(4-(5,5-dioxido-4H-thieno[3,2-c]thiochromene-2-carbonyl)piperazin-1 - yl)bicyclo[1 .1 .1 ] pentan-2-yl)carbamate was prepared following General Procedure A using tert-butyl (4-(piperazin-1 -yl)bicyclo[1 .1 .1 ]pentan-2-yl)carbamate (48 mg, 1 Eq, 0.18 mmol). The compound was purified by flash column chromatography [Silica-CS 12g, eluent: DCM/MeOH (0% MeOH for 3CV, 0-5% 15CV, 5% for 3CV, 5-10% for 3CV], To a solution of tert-butyl(3-(4-(5,5-dioxido-4H-thieno[3,2-c]thiochromene-2- carbonyl)piperazin-1 -yl)bicyclo[1 .1 .1 ]pentan-1 -yl)carbamate (60 mg, 1 Eq, 0.1 1 mmol) in 1 ,4-dioxane (1 .1 mL, 0.1 molar) was added HCI in dioxane 4M (0.42 mL, 4 molar, 15 Eq, 1.7 mmol) dropwise at 0°C. The reaction mixture was stirred at room temperature overnight. SPC-EPFL-7 (9 mg, 20%) was obtained as a solid after purification by flash column chromatography.
1 H NMR (400MHz, DMSO-d6) 5 7.97 (dt, J = 7.7, 1 .0 Hz, 1 H), 7.76-7.82 (m, 2H), 7.61 - 7.72 (m, 1 H), 7.49 (s, 1 H), 4.88 (s, 2H), 3.62 - 3.68 (m, 4H), 2.41 (app t, J = 5.1 Hz, 4H), 1.65 (s, 6H).
(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl) (4-(o-tolyl)piperazin- 1 -yl)methanone
Compound SPC-EPFL-9 (C9) was prepared following General Procedure A using 1 -o- tolyl-Piperazine (19 mg, 1 Eq, 0.1 mmol) to afford (5,5-dioxido-4H-thieno[3,2- c]thiochromen-2-yl)(4-(o-tolyl)piperazin-1 -yl)methanone (31 mg, 66 %) as a light yellow solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-30% 5CV, 30-50% for 15CV, 50% for 3CV]. 1 H NMR (400 MHz, CDCI3) 5 8.05 (d, J = 7.8 Hz, 1 H), 7.72 - 7.59 (m, 2H), 7.54 (td, J = 7.4, 1 .5 Hz, 1 H), 7.28 - 7.13 (m, 3H), 7.08 - 6.98 (m, 2H), 4.44 (s, 2H), 3.93 (app ft, J = 4.9 Hz, 4H), 2.98 (app t, J = 4.9 Hz, 4H), 2.35 (s, 3H).
(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)(4-(2-fluoro-4-
Compound SPC-EPFL-12 (C12) was prepared following General Procedure A using 1 - (2-fluoro-4-(trifluoromethyl)phenyl)piperazine (71 mg, 1 Eq, 0.28 mmol) to afford (5,5- dioxido-4H-thieno[3,2-c]thiochromen-2-yl)(4-(2-fluoro-4-
(trifluoromethyl)phenyl)piperazin-1 -yl)methanone (43 mg, 59 %) as a Pink powder after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (10% EtOAc for 3CV, 10-50% for 15CV, 50% for 3CV].
1 H NMR (400 MHz, CDCI3) 5 8.05 (dd, J = 7.8, 1.3 Hz, 1 H), 7.72 - 7.59 (m, 2H), 7.55 (td, J = 7.9, 7.2, 1 .4 Hz, 1 H), 7.40 - 7.27 (m, 2H), 7.23 (s, 1 H), 7.04 - 6.95 (m, 1 H), 4.45 (s, 2H), 4.07 - 3.75 (m, 4H), 3.23 (app t, J = 5.0 Hz, 4H).
(4-(2,6-dimethylphenyl)piperazin-1-yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2- yl)methanone (SPC-EPFL-13)
Compound SPC-EPFL-13 (C13) was prepared following General Procedure A using 1 - (2,6-dimethylphenyl)piperazine (48 mg, 1 Eq, 0.25 mmol) to afford (4-(2,6- dimethylphenyl)piperazin-1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (42 mg, 74 %) as a yellow solid after purification by flash column chromatography [Silica- CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-50% for 15CV, 50% for 3CV]. Yellow powder (42 mg, 0.09 mmol, 74%). 1 H NMR (400 MHz, DMSO- d6) 5 7.97 (d, J = 7.8 Hz, 1 H), 7.85 - 7.76 (m, 2H), 7.66 (ddd, J = 8.3, 6.3, 2.4 Hz, 1 H), 7.55 (s, 1 H), 7.04 - 6.91 (m, 3H), 4.90 (s, 2H), 3.80 (app t, J = 4.9 Hz, 4H), 3.1 1 (app t, J = 4.9 Hz, 4H), 2.31 (s, 6H).
(4-(2,6-dichlorophenyl)piperazin-1-yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2- yl)methanone (SPC-EPFL-16)
Compound SPC-EPFL-16 (C16) was prepared following General Procedure A using 1 - (2,6-dichlorophenyl)piperazine (58 mg, 1 Eq, 0.25 mmol) to afford (4-(2,6- dichlorophenyl)piperazin-1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (56 mg, 91 %) as a Light yellow solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-60% for 15CV, 60% for 3CV].
1 H NMR (400 MHz, DMSO- d6) 5 8.00 - 7.93 (m, 1 H), 7.85 - 7.75 (m, 2H), 7.65 (ddd, J = 7.8, 6.5, 2.2 Hz, 1 H), 7.55 (s, 1 H), 7.46 (d, J = 8.1 Hz, 2H), 7.20 (dd, J = 8.4, 7.7 Hz, 1 H), 4.89 (s, 2H), 3.82 (app t, J = 4.9 Hz, 4H), 3.27 - 3.20 (m, 4H).
(4-(2,5-dichlorophenyl)piperazin-1-yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2- yl)methanone (SPC-EPFL-17)
Compound SPC-EPFL-17 (C17) was prepared following General Procedure A using 1 - (2,5-dichlorophenyl)piperazine (58 mg, 1 Eq, 0.25 mmol) to afford (4-(2,5- dichlorophenyl)piperazin-1 -yl)(5,5-dioxido-4H-thieno[3,2-c]thiochromen-2-yl)methanone (51 mg, 83 %) as a Light yellow solid after purification by flash column chromatography [Silica-CS 12g, eluent: Cyclohexane/EtOAc (0% EtOAc for 3CV, 0-50% for 15CV, 50% for 3CV].
1 H NMR (400 MHz, DMSO- d6) 5 7.97 (dt, J = 7.8, 1 .0 Hz, 1 H), 7.85 - 7.75 (m, 2H), 7.65 (ddd, J = 7.8, 6.2, 2.5 Hz, 1 H), 7.56 (s, 1 H), 7.48 (d, J = 8.5 Hz, 1 H), 7.21 (d, J = 2.4 Hz, 1 H), 7.15 (dd, J = 8.5, 2.4 Hz, 1 H), 4.89 (s, 2H), 3.86 (br s, 4H), 3.09 (app t, J = 5.0 Hz, 4H).
Example 12: Dose response inhibition of SARS-CoV-2 replication in Calu-3 cells by ML349 and novel inhibitors of acyl protein thioesterase (APT) according to formula (II).
In figure 1 1 , the dose dependent inhibition curves and correspondent EC50 of ML349 and novel inhibitors of acyl protein thioesterase (APT) according to formula (II) (SPC-EPFL1 - C1 , SPC-EPFL2-C2, and SPC-EPFL3-C3) in inhibiting SARS-CoV-2 intracellular replication was determined in Calu-3 by quantifying total viral RNA at 24 h post viral inoculation with SARS-CoV-2 at MOI=0.05 to 0.1. Analysis of the potential cytotoxic effects caused by the drug-treatment during infection was assessed by comparing the levels of housekeeping transcripts during infection.
Infection and cell viability were determined using qPCR to quantify the mean levels of E, RdRp and Esub genomic viral RNA (Figure 1 1 A) and host ALAS-1 and RPL27 transcripts (Figure 1 1 B), respectively. Normalized results are mean ± SEM and the dose-response curves are indicated for the 4 compounds. The observed EC50 concentrations correspond to ML349EC50=7,9 pM, C1 EC5O=6.7 pM, C2EC5O=2.3 pM and C3EC5O=O.96 pM.
Advantageously, novel inhibitors of general formula (II) such as C2 or C3 demonstrate an EC50 that is decreased by about 3 to 8-fold compared to compound ML-349 (Example 12, figures 1 1 A and B).
The levels of housekeeping transcripts during infection remained constant through all tested concentrations of each studied compound. Therefore, treatment of SARS-CoV-2- infected Calu-3 cells with ML349, C1 , C2 or C3 for 24h, does not significantly induce cellular toxicity at different ranges of concentrations (Figure 1 1 B).
The improved inhibitory effect of the novel inhibitors of acyl protein thioesterase (APT) according to formula (II), such as C1 , C2 or C3 during SARS-CoV-2 replication in Calu-3 cells (Figure 1 1 A) further demonstrate that ML349 and novel inhibitors of acyl protein thioesterase (APT) according to formula (II) are potent inhibitors of SARS-CoV-2 infection and that host cell depalmitoylation activity constitutes a safe strategy to inhibit viral infections.
It has been demonstrated that inhibitors of acyl protein thioesterase (APT) of formula II wherein R3 is a substituted phenyl ring such as C1 , C2 or C3 have an improved inhibitory activity of SARS-CoV-2 replication compared to ML349. In contrast, molecules that are not covered by formula II wherein R3 is a substituted phenyl ring such as C5, C6 or C7 do not inhibit SARS-CoV-2 replication in Calu-3 cells (Figure 12).
Example 13: Inhibition of APT2 activity with inhibitor C2 inhibits SARS-CoV-2 intracellular replication.
It has been previously demonstrated that ML348 blocked SARS-CoV-2 intracellular viral RNA replication in Vero E6 as early as 8 h after viral inoculation (Figure 6A and 6B). To understand if targeting APT2 in Calu-3 cells was blocking infection in an equivalent manner (up to 8 to 12 h), viral genomic and sub-genomic RNA replication was monitored throughout time by probing for two viral genomic regions corresponding to the E and RdRp genes, and the E-sub genomic transcript of SARS-CoV-2. Calu-3 cells were first inoculated with SARS-CoV-2 at MOI=0.5 for 1 h, washed thoroughly in culture medium to removed extracellular virions and subsequently incubated in complete medium with 5 pM of C2 and 0.5 pg/ml of anti-Spike antibody (Fenwick et aL, 2022) to neutralized of virions released within the first round of viral cellular life cycle (approximately from 6 h
onwards in Calu-3) (Cortese et aL, 2020), restricting the quantification of the viral RNA replication to the initial round of infection.
Cells were harvested at the time points 1 , 4, 6, 8 and 12 hours and host and viral RNA levels quantified by QPCR as described in materials and methods. The data was normalized to the intracellular viral RNA levels harvested 1 h after inoculation before drug treatment and expressed as fold RNA replication of both viral genomic and subgenomic RNA. Normalized results are the mean ± SEM of three independent biological replicates displayed within two equivalent graph plots with either linear or logarithmic scale for the fold RNA replication.
The results show that blocking APT2 activity with C2 strongly inhibits SARS-CoV-2 infection as early as 4 h after viral entry into cells (Figure 13A and 13B), thus confirming an inhibitory effect of the intracellular viral RNA replication.
Example 14: Dose-response cytotoxicity evaluation of APT2 inhibition in Calu-3 cells by ML349 and novel inhibitors of acyl protein thioesterase (APT) according to formula (II).
A RealTime-Glo™ MT Cell Viability Assay, consisting of a nonlytic, homogeneous, bioluminescent method to measure cell viability in real time, was performed as described in material and method.
Calu-3 cells were treated with increasing concentration of ML349 and C2 or C3 over the course of 6 days. Culture medium was replaced every 2 days with freshly diluted compounds in order to ensure constant activity. The results of the cell viability represent the evolution of the number of viable cells in culture at the indicated time-points and are mean SEM of 4 biological replicates (Figure 14A-C). Fitted curves for Control (no drug), non-toxic and cytotoxic compound concentrations are indicated.
After 6 days of drug-treatment ML349, C2 and C3 were observed to induced cellular toxicity at very high concentrations (from 25 pM onwards, Figure 14D), at least 10-fold more concentrated than the determined EC50 (C1 EC5O=6.7 pM, C2EC5O=2.3 pM and C3EC50~0.96 pM).
Thus, blocking APT2 activity with novel inhibitors of acyl protein thioesterase (APT) according to the present invention effectively and safely inhibits viral infections, such as SARS-CoV-2.
Example 15: Inhibition of APT2 activity with compound C2 inhibits replication of SARS- CoV-2 variant Omicron BA.5.
An experiment was performed to evaluate if the compound C2 inhibits multiple SARS- CoV-2 strains. The dose-dependent effect of C2 was determined during the intracellular replication of SARS-CoV-2 variant of concern Omicron BA5 in Calu-3 cells by quantifying the total viral RNA at 24 h post-viral inoculation with SARS-CoV-2 BA5 at MOI =0.05 to 0.1. Infection and cell viability were measured using QPCR of E, RdRp, and E sub- genomic viral RNA (Figure 15A) and host ALAS-1 and RPL27 RNA (Figure 15B), respectively.
Normalized mean results show that C2 inhibits replication of SARS-CoV-2 BA5 strain with an EC50«2.031 pM (Figure 15A) without significantly affecting the levels of housekeeping transcripts during infection (Figure 15B). These data confirm that inhibition of APT2 activity with C2 potently blocks replication of different SARS-CoV-2 strains.
Example 16: Toxicity evaluation of APT2 inhibition in vivo, in C57BL/6 mice by ML349 and compound C2.
Following the experiment confirming that inhibition of APT2 with ML349, compound C2 and C3 did not induce significant cytotoxicity in vitro (Example 14), an experiment was carried out to evaluate the potential toxicity effect of inhibiting APT2 using ML349 or C2 in vivo in C57BL/6 mice. Mice were treated with 1 mg of ML349 or C2 per 25 g of body weight or an equivalent volume of drug delivery vehicle (control), inoculated intraperitoneally at alternate days through a period of 13 days. Toxicity was evaluated by monitoring the body weight, blood pressure, and pulse daily throughout treatment (Figure 16A-C). In addition, the relative spleen weight harvested after 13 days of treatment was also monitored (Figure 16D), and a histological analysis of multiple organs (nose, trachea, lung, brain, small and large intestine, liver, kidney) was also performed (data not shown). Inhibiting APT2 using ML349 or C2 did not significantly change body weight, blood pressure, and pulse throughout treatment, neither did it cause any detectable pathological signs in the spleen (relative weight) after 13 days of treatment (Figure 16). The histological analysis of nose, trachea, lung, brain, small and large intestine, liver and kidney also did not show any ML349- or C2- related change in tissue morphology (data not shown).
In conclusion, treatment of mice with APT2 inhibitors, ML349 and C2, at 1 mg of compound per 25 g of body weight, on alternate days does not induce any apparent toxic effect.
Example 17: Evaluating APT2 inhibition mechanism by ML349, compound C2, or C3 during anthrax toxin cellular intoxication.
Internalization of the anthrax toxin and degradation of its host target MEK2 requires APT2 activity (Figure 1 -3).
APT2 activity during anthrax intoxication can be monitored by probing for the formation of protein complexes between the anthrax receptor CMG2 and APT2 triggered by the binding of anthrax toxin protective antigen (PA) to CMG2 (Figure 17A). Formation of these complexes occurs upon de-acylation of CMG2 and is concomitant to the dissociation of CMG2 from the cytoskeleton components actin, vinculin, and talin, which ultimately promotes the internalization of the toxin and the receptor. The levels of CMG2-APT2 complexes can be probed by western blot analysis of lysates from RPE-1 cells treated with PA (Figure 17B). Inhibition of APT2 activity with 5 pM ML349, 4 h before and during toxin treatment, prevents the formation of CMG2-APT2 complexes (Figure 17B). Equivalent treatment of RPE-1 cells with compound C2 and C3, also blocks the formation of APT2-CMG2 complexes upon PA intoxication.
Thus ML349, C2, and C3 can reduce SARS-CoV-2 replication and inhibit the APT2- dependent anthrax toxin intoxication of host cells.
Example 18: Comparison of APT2 inhibition efficacy by ML349, compound C2, C3 during anthrax toxin cellular intoxication.
The levels of CMG2-APT2 complexes formed upon anthrax toxin protective antigen (PA) intoxication were also evaluated in cells pre and co-treated with only 2 pM of each compound to compare the potency of ML349 and compound C2, C3. At such concentrations C2 and C3, but not ML349, prevented the formation of APT2-CMG2 complexes, blocking cellular intoxication (Figure 18). Therefore, as observed for the inhibition of SARS-CoV-2 replication, C2 and C3 inhibit intoxication of host cells by anthrax-toxin at lower concentrations than ML349.
Example 19: Anthrax toxin cellular intoxication assays with compounds of formula (II).
Novel compounds of formula (II) as described herein have been synthesized (Example 1 1 ). These compounds were tested for their capacity to inhibit the formation of CMG2- APT2 complexes in anthrax toxin cellular intoxication assays. Western blot analysis of a single parallel synchronized cellular intoxication experiment showed that compounds C9, C12, C13, C16, and C17 (used at 5 pM) inhibit the formation of CMG2-APT2 complexes beyond 50% (Figure 19).
Experiments were also performed to evaluate the dynamics of the formation of the SDS- resistant toxin pore in the presence of the same compounds (Figure 20). In agreement with the observations from Figure 19, compounds C2, C9, C13, C16 and C17 reduced the formation of PA pores when mammalian cells were pre- and co-treated with 5 pM of each compound during PA treatment. Furthermore, the observed inhibition was equivalent to the inhibitory effect of C2 in the same assay.
Example 20: Inhibition of APT2 activity with compounds of formula (II) blocks SARS-CoV- 2 infection in Calu-3 lung derived cells
Experiments were conducted to compare the effect of inhibiting APT2 activity during SARS-CoV-2 replication in human lung-derived cells with ML349 and different compounds of formula (II). Calu-3 cells were inoculated with SARS-CoV-2 at MOI =0.05 to 0.1 for 1 h, washed, and co-treated with 5 pM of each APT2 inhibitor or an equivalent volume of drug vehicle. At 24 h post-inoculation, infected cells were harvested for quantitative analysis of the levels of the Nucleocapsid protein N. The intracellular levels of N were monitored by western blot as a readout for viral intracellular replication.
Figure 21 shows the levels of viral nucleocapsid protein N: cell lysates, were monitored by Western blot. The % of infection was quantified as the ratio between the levels of N and host GAPDH for three independent replicates for each compound with relation to Control cells (infected in the presence of drug carrier DMSO).
Figure 21 shows that 5 pM of compounds C9, C12, C13, C16, or C17 significantly inhibited SARS-CoV-2 replication beyond 50% and more potently than ML349. At 5 pM, C2 and C3 displayed the most potent inhibitory effect.
Altogether these data demonstrated that compounds of formula (II) as described herein have potent and non-toxic anti-viral and anti-toxin effects.
Claims
1 . An inhibitor of acyl protein thioesterase (APT) of formula (II):
or a pharmaceutically acceptable salt thereof, wherein
Ring A is a thiophene ring or a furane ring;
Each RAm is independently selected from the group comprising hydrogen, halogen, Ci-
C-io alkyl, C2-C10 alkenyl, C1-C6 alkoxy, C1-C6 acyl, C3-C10 cycloalkyl, optionally substituted C3-C10 heterocycloalkyl, C6-C10 aryl, and/or C6-C10 heteroaryl, wherein said cycloalkyl, optionally substituted heterocycloalkyl, aryl or heteroaryl groups may be fused with ring A and with 0 to 2 further cycloalkyl, heterocycloalkyl, aryl or heteroaryl groups; and m is an integer from 0 to 2;
Each R1 , R2, R4, R5 is independently selected from the group comprising hydrogen, halogen, C1-C10 alkyl, optionally substituted Ci-Ce alkyl acyl, optionally substituted Ci-Ce alkyl aminocarbonyl, optionally substituted carbonyl, optionally substituted Ci-Ce alkyl carbonyl, optionally substituted C6-C10 aryl, ether, C2-C10 alkenyl, C6-C10 heteroaryl, C3- C10 cycloalkyl, C3-C10 heterocycloalkyl, Ci-Ce alkoxy, optionally substituted amino, and/or Ci-Ce alkyl amino; and
R3 is selected from the group comprising
3. The inhibitor of acyl protein thioesterase (APT) of formula (II) according to any one of claims 1 -2, or a pharmaceutically acceptable salt thereof, wherein each R1 , R2, R4, R5 is hydrogen.
4. The inhibitor of APT of formula (II) according to any one of claims 1 -3, selected from the group comprising
10
5. An inhibitor of APT of formula (II) according to any one of claims 1 -4, or a pharmaceutically acceptable salt thereof, for use as a medicament.
6. An inhibitor of APT of formula (II) according to any one of claims 1 -4, or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of a viral and/or a bacterial infection.
8. The inhibitor of APT for use according to any one of claims 6-7, wherein said viral infection is caused by a virus selected from the group comprising Coronaviridae, Orthomyxoviridae, Paramyxoviridae, Togaviridae, Rhabdoviridae, Filoviridae, Pneumoviridae, Retroviridae, Herpesviridae, Flaviviridae, Hepeviridae, and/or Bunyaviridae.
9. The inhibitor of APT for use according to claim 8, wherein said Coronaviridae comprises SARS-CoV-2, SARS-CoV-1 , MHV, TGEV, SADS, MERS-CoV, HCoV-HKU1 , HCoV-229E, HCoV-NL63 and/or HCoV-OC43.
10. The inhibitor of APT for use according to any one of claims 6-7, wherein said bacterial infection is caused by bacterial strains selected from the group comprising Bacillaceae, Vibrionaceae, Pectobacteriaceae Yersiniaceae, Staphylococcaceae, Streptococcaceae, Legionellaceae, Pseudomonadaceae, Chlamydiaceae, Mycoplasmataceae, Enterobacteriaceae, Pseudomonadales and/or Pasteurellaceae.
11 . The inhibitor of APT for use according to any one of claims 6-10,
wherein said viral infection is selected from the group comprising COVID, SARS, flu, rheumatic diseases, encephalitis, haemorrhagic fevers, acquired immunodeficiency syndrome (AIDS), sarcoma and/or leukemia; and/or wherein said bacterial infection is selected from the group comprising Typhoid fever, Salmonellosis, bacteria Gastroenteritis intestinal infection, bacteremia, Shigellosis, Bacteria pneumonia, endophthalmitis, bacteremia, septicemia, sepsis, endocarditis, salpingitis, skin infections, and/or meningitis.
12. A pharmaceutical composition comprising at least one inhibitor of APT according to any one of claims 1 -4, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, or excipient.
13. The pharmaceutical composition according to claim 12, further comprising one or more additional therapeutic agents selected from the group comprising antibacterial agents and/or antiviral agents.
14. The pharmaceutical composition according to any one of claims 12-13, wherein said pharmaceutical composition is administered by oral, inhalation, nebulization, intranasal, intrapulmonary, intradermal and/or intramuscular route of administration.
15. A kit comprising an inhibitor of APT according to any one of claims 1 -4, and information for use thereof.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP22152748 | 2022-01-21 | ||
EP22152748.4 | 2022-01-21 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2023139248A1 true WO2023139248A1 (en) | 2023-07-27 |
Family
ID=79927550
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2023/051499 WO2023139248A1 (en) | 2022-01-21 | 2023-01-23 | Inhibitors of acyl protein thioesterases against microbial infections |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2023139248A1 (en) |
Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
FR2825706A1 (en) * | 2001-06-06 | 2002-12-13 | Pf Medicament | New benzothienyl and indole derivatives having prenyl transferase protein inhibiting activity, are useful for the prevention and treatment of cancers |
WO2005026175A1 (en) * | 2003-09-08 | 2005-03-24 | Aventis Pharmaceuticals Inc. | Thienopyrazoles |
US20070203154A1 (en) * | 2006-02-28 | 2007-08-30 | Xianbo Zhou | Therapeutic Compounds |
CN102558074A (en) * | 2009-03-31 | 2012-07-11 | 中国科学院广州生物医药与健康研究院 | Piperazine amide compound containing pyridazine substituent group |
CN108586485A (en) * | 2018-05-30 | 2018-09-28 | 沈阳药科大学 | 1- (4- hydroxyl imido grpup thieno [2,3-b] thiapyrans formoxyl) piperazine compounds and its application |
CN108822126A (en) * | 2018-05-30 | 2018-11-16 | 沈阳药科大学 | Thieno thiapyran formyl piperazine class compound and its medical usage |
WO2022246115A1 (en) * | 2021-05-19 | 2022-11-24 | Cornell University | Apt1 and apt2 inhibitors and uses thereof |
-
2023
- 2023-01-23 WO PCT/EP2023/051499 patent/WO2023139248A1/en unknown
Patent Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
FR2825706A1 (en) * | 2001-06-06 | 2002-12-13 | Pf Medicament | New benzothienyl and indole derivatives having prenyl transferase protein inhibiting activity, are useful for the prevention and treatment of cancers |
WO2005026175A1 (en) * | 2003-09-08 | 2005-03-24 | Aventis Pharmaceuticals Inc. | Thienopyrazoles |
US20070203154A1 (en) * | 2006-02-28 | 2007-08-30 | Xianbo Zhou | Therapeutic Compounds |
CN102558074A (en) * | 2009-03-31 | 2012-07-11 | 中国科学院广州生物医药与健康研究院 | Piperazine amide compound containing pyridazine substituent group |
CN108586485A (en) * | 2018-05-30 | 2018-09-28 | 沈阳药科大学 | 1- (4- hydroxyl imido grpup thieno [2,3-b] thiapyrans formoxyl) piperazine compounds and its application |
CN108822126A (en) * | 2018-05-30 | 2018-11-16 | 沈阳药科大学 | Thieno thiapyran formyl piperazine class compound and its medical usage |
WO2022246115A1 (en) * | 2021-05-19 | 2022-11-24 | Cornell University | Apt1 and apt2 inhibitors and uses thereof |
Non-Patent Citations (5)
Title |
---|
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY |
HERNANDEZ JEANNIE L ET AL: "APT2 Inhibition Restores Scribble Localization andS-Palmitoylation in Snail-Transformed Cells", CELL CHEMICAL BIOLOGY, ELSEVIER, AMSTERDAM, NL, vol. 24, no. 1, 5 January 2017 (2017-01-05), pages 87 - 97, XP029890377, ISSN: 2451-9456, DOI: 10.1016/J.CHEMBIOL.2016.12.007 * |
IGOR VUJIC ET AL: "Acyl protein thioesterase 1 and 2 (APT-1, APT-2) inhibitors palmostatin B, ML348 and ML349 have different effects on NRAS mutant melanoma cells", ONCOTARGET, vol. 7, no. 6, 9 February 2016 (2016-02-09), XP055638544, DOI: 10.18632/oncotarget.6907 * |
KAPLAN ALAN P. ET AL: "Identification of 5-(1-Methyl-5-(trifluoromethyl)-1 H -pyrazol-3-yl)thiophene-2-Carboxamides as Novel and Selective Monoamine Oxidase B Inhibitors Used to Improve Memory and Cognition", ACS CHEMICAL NEUROSCIENCE, vol. 8, no. 12, 20 September 2017 (2017-09-20), US, pages 2746 - 2758, XP093039221, ISSN: 1948-7193, DOI: 10.1021/acschemneuro.7b00282 * |
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2021207409A2 (en) | Small molecule inhibitors of sars-cov-2 viral replication and uses thereof | |
US20220257588A1 (en) | Methods of preventing or treating flavivirus virus infections and methods of inhibiting the entry of flvivirus, enterovirus or lentivirus into host cells | |
DK2731608T3 (en) | Inhibitors of the bacterial type III secretion system | |
WO2021225767A1 (en) | Methods and compositions for the treatment of sars-cov-2 | |
JP2020196704A (en) | Prophylactic and/or therapeutic agent for influenza virus infection or corona virus infection | |
JP5898061B2 (en) | Inhibitors of bacterial type III secretion system | |
Wang et al. | Synthesis and structure-activity optimization of 7-azaindoles containing aza-β-amino acids targeting the influenza PB2 subunit | |
CN114699419A (en) | Use of a PLpro protein inhibitor in a medicament for the treatment or prevention of a novel coronavirus infection | |
WO2023139248A1 (en) | Inhibitors of acyl protein thioesterases against microbial infections | |
US10226434B2 (en) | Design, synthesis and methods of use of acyclic fleximer nucleoside analogues having anti-coronavirus activity | |
US11376306B2 (en) | Peptides and uses therefor as antiviral agents | |
KR20230021009A (en) | Azelastine as an antiviral treatment | |
Cole | Baloxavir marboxil. Influenza virus cap-dependent endonuclease (CEN) inhibitor, Anti-influenza agent | |
Cannalire et al. | A Focus on Severe Acute Respiratory Syndrome (SARS) Coronavirus (SARS-CoVs) 1 and 2 | |
WO2021228037A1 (en) | Compositions and methods for broad spectrum anti-viral therapy | |
WO2020221894A1 (en) | Antiviral compounds | |
KR20180036523A (en) | Inhibitor of function of cyclophilin and use of the same | |
WO2024006949A2 (en) | Allosteric inhibitors of the main protease of sars-cov-2 | |
WO2022195296A1 (en) | Anti viral therapy | |
JP2023551018A (en) | Compounds for the treatment of enveloped virus infections | |
JP2023526200A (en) | Compatible Solutes for Prevention or Treatment of SARS-CoV-2 Infection | |
WO2023170187A1 (en) | INHIBITION OF INTRACELLULAR PATHOGEN UPTAKE BY INHIBITORS OF THE IKK-α/NIK COMPLEX | |
US10301258B2 (en) | Benzende sulfonamide derivatives as HIV integrase inhibitors | |
WO2022046642A1 (en) | Compounds and compositions for disrupting programmed ribosomal frameshifting | |
JP2017516858A (en) | Cyclic compounds having 1,3 diamino functionality for use in the treatment of HIV infection |
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: 23701496 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |