key: cord-0908465-swmkkdya authors: Santos, Lucianna H.; Kronenberger, Thales; Almeida, Renata G.; Silva, Elany B.; Rocha, Rafael E. O.; Oliveira, Joyce C.; Barreto, Luiza V.; Skinner, Danielle; Fajtová, Pavla; Giardini, Miriam A.; Woodworth, Brendon; Bardine, Conner; Lourenço, André Luiz; Craik, Charles S.; Poso, Antti; Podust, Larissa M.; McKerrow, James H.; Siqueira-Neto, Jair L.; O’Donoghue, Anthony J.; da Silva Júnior, Eufrânio N.; Ferreira, Rafaela S. title: Structure-based identification of naphthoquinones and derivatives as novel inhibitors of main protease Mpro and papain-like protease PLpro of SARS-CoV-2 date: 2022-01-05 journal: bioRxiv DOI: 10.1101/2022.01.05.475095 sha: 6c28762c7163840079da7cc8f73d2df9fa201679 doc_id: 908465 cord_uid: swmkkdya The worldwide COVID-19 pandemic caused by the coronavirus SARS-CoV-2 urgently demands novel direct antiviral treatments. The main protease (Mpro) and papain-like protease (PLpro) are attractive drug targets among coronaviruses due to their essential role in processing the polyproteins translated from the viral RNA. In the present work, we virtually screened 688 naphthoquinoidal compounds and derivatives against Mpro of SARS-CoV-2. Twenty-four derivatives were selected and evaluated in biochemical assays against Mpro using a novel fluorogenic substrate. In parallel, these compounds were also assayed with SARS-CoV-2 PLpro. Four compounds inhibited Mpro with half-maximal inhibitory concentration (IC50) values between 0.41 µM and 66 µM. In addition, eight compounds inhibited PLpro with IC50 ranging from 1.7 µM to 46 µM. Molecular dynamics simulations suggest stable binding modes for Mpro inhibitors with frequent interactions with residues in the S1 and S2 pockets of the active site. For two PLpro inhibitors, interactions occur in the S3 and S4 pockets. In summary, our structure-based computational and biochemical approach identified novel naphthoquinonal scaffolds that can be further explored as SARS-CoV-2 antivirals. transmission from human to human is undoubtedly the main source of contagion, which 61 occurs mainly through droplets, hand contact, or contact with contaminated surfaces [5] . 62 To control the spread of this pandemic virus, biosecurity and hygiene measures are now 63 worldwide applied [6] . Despite the rapid development and emergency authorization of 64 vaccines, viral escape mutants have emerged, and SARS-CoV-2 infections remain a 65 concern for the global community. Therefore, there is a continuing need to discover 66 structural frameworks for drugs that can be employed against . 67 Drug development efforts have targeted the SARS-CoV-2 main protease (Mpro) 68 also known as 3-chymotrypsin-like protease (3CLpro) or non-structural protein 5 (nsp5) 69 [8, 9] . Mpro is an essential cysteine protease that cleaves the precursor replicase 70 polyprotein in a coordinated manner [10] , to generate at least 11 non-structural proteins 71 [11] . As a target, Mpro is conserved among other coronaviruses, and has no closely 72 related human homolog [12] [13] [14] . Therefore, it has been intensively investigated as a drug 73 target for SARS and Middle East Respiratory Syndrome (MERS) [15] [16] [17] [18] were studied and evaluated according to their potential to act as anti-SARS-CoV-2. Imine 154 derivatives were also targeted in our studies and were placed in group 6 [67] . 155 Finally, groups 7 and 8 are formed by hydrazo, imidazole, and oxazole derivatives 156 [108] [109] [110] [111] . The compounds in these groups were prepared from the quinones described 157 above and represent our attempt to study quinone-derived heterocyclic compounds with 158 biological activity against various microorganisms and their effectiveness against the 159 virus that causes COVID-19. Prior to biochemically evaluating the compounds against Mpro, we designed a 287 fluorescent-quenched peptide substrate with the sequence ATLQAIAS that corresponds 288 to the P4 to P4ʹ amino acids of the nsp7-nsp8 cleavage site and the dash representing the 289 scissile bond. This substrate was chosen because the sequence most closely matches the 290 consensus sequence for all 11 viral polypeptide cleavage sites (Figure 5A and B) [126] . 291 ATLQAIAS was flanked by 7-methoxycoumarin-4-acetyl-L-lysine on the N-terminus, 292 dinitrophenyl-L-lysine on the C-terminus. The peptide contains several non-polar amino 293 acid residues and therefore two d-Arginine residues were added on the N-terminus to 294 increase solubility. Using a concentration range of 3 µM to 250 µM, the KM for this 295 substrate was calculated to be 52.1 µM ± 14.4 µM. 296 297 We evaluated the 24 hit compounds from our virtual screen in a biochemical assay 300 using recombinant SARS-CoV-2 Mpro. The enzyme was pre-incubated with each 301 compound at 10 µM and then assayed with the fluorogenic peptide substrate. To avoid 302 detecting aggregators as false positives [127, 128] , our assay was performed in the 303 presence of 0.01% Tween 20. Additionally, we evaluated the absorbance of MCA 304 fluorescence by the compounds, to make sure the observed enzyme inhibition was not an 305 artifact of fluorescence, another common cause of false positives in enzyme assays [129] . 306 From this screen, three 1,4-naphthoquinones derivatives, 379, 382, and 415, fully 307 inhibited Mpro, while two quinone-based 1,2,3 triazoles, 191 and 194, had 50% or more 308 inhibition. 668 was insoluble in assay buffer and was therefore eliminated from further 309 analysis, while the remaining compounds had inhibition profiles of less than 50% (Table 310 1). The most potent compounds were subsequently evaluated at a concentration range of 311 10 µM to 9.7 nM and the half-maximal inhibitory concentration (IC50) was calculated to 312 be 66 µM ± 22 for 191, an ortho-quinone-based 1,2,3 triazole, 5 µM ± 0.15 for 415, 0.63 313 µM ± 0.04 for 379, and 0.41 µM ± 0.015 for 382 (Table 1 and Figure 5) . The IC50 values observed in these two assay conditions were similar, with slightly lower 345 IC50 values upon preincubation (0.42 µM ± 0.02 upon incubation vs 0.80 µM ± 0.06 346 without incubation for 382 and 5.0 µM ± 0.2 upon incubation vs 16 µM ± 1 without 347 incubation for 415) (Figure 6A and B) , while for the positive control GC373 the IC50 was 348 ten-fold lower upon preincubation (0.003 µM ± 0.001). A dilution experiment was also 349 performed, to check whether the compounds were irreversible. We incubated the 350 inhibitors and Mpro at high concentrations and then diluted the incubation mixture, 351 resulting in inhibitor concentrations 10-fold lower than their apparent IC50. In this assay, 352 an irreversible inhibitor will maintain approximately 10% of enzymatic activity, while a 353 rapidly reversible inhibitor will dissociate from the enzyme to restore approximately 90% 354 of enzymatic activity following the dilution event [130, 131] . When this was performed 355 with Mpro and GC373, a covalent Mpro inhibitor, the enzyme remained inhibited upon 356 dilution. The same behavior was observed for compound 415 suggesting that this 357 inhibitor is an irreversible covalent inhibitor (see Figure 7 for the proposed binding 358 mechanism). However, when the same test was carried out with compound 382 enzyme 359 activity returned after dilution ( Figure 6C ). This suggested that the inhibition by 382 is 360 reversible. 361 experiments (data shown as spheres or squares for each experiment), in which the 366 compounds were pre-incubated with Mpro prior to substrate addition (black) and without 367 preincubation with the compounds (purple). Reversibility assay (C). After preincubation 368 of Mpro with compounds, at higher concentrations, the sample was diluted, and product 369 formation was monitored for 120 minutes. Compound 382 reduced the enzymatic reaction 370 rate by 26% compared to vehicle control (red), while the compound 415 reduced product 371 formation by 100%, and this activity was not restored over a 2h period post dilution, as 372 observed for the covalent inhibitor GC373 (black). 373 To gain insights into the proposed binding mode of our Mpro inhibitors and guide 374 future optimization efforts, we conducted docking and MD studies with compounds 382 375 and 415, representatives from two inhibitor scaffolds discovered. Our simulations 376 considered the 415 ligand covalently bound, given the proposed reaction mechanism 377 ( Figure 7A) , to both monomers in the Mpro dimer, and 382 freely. For the free 378 simulations, however, the loss of interactions with E166 resulted in ligands being expelled 379 from one of the binding sites within a few nanoseconds (~200 ns) of simulation 380 (Supporting Information Table S2 ). Our analysis is focused on the other binding site, that 381 retained the ligand with stable interactions along the analyzed trajectory. 382 For both ligands, the most representative binding modes observed in the MD 383 simulations (Figure 7A and 7B) retain key interactions proposed based on docking with 384 Glide (Supporting Information Figure S45 ). However, compound 415 showed a more 385 conserved binding mode throughout the trajectory, being well represented by a single 386 pose, in which the nitro group interacts with the S1 pocket and the 1,4 naphthoquinone 387 interacts with the catalytic H41 and S2 subsite residues ( Figure 7B ). On the other hand, 388 the higher variability in the orientation of compound 382 led to four clusters with 389 frequency between 17.5 and 31.7% (Supporting Information Figure S48 ). Overall, the 390 1,4-naphthoquinone ring of ligand 415 occupies the S1 pocket, but fluctuations in the ring 391 orientation reflect on varied positions for the phenyl substituents. In the most populated 392 cluster ( Figure 7C ), the methoxyphenyl substituents occupy the S1ʹ and S2 subsites. 393 Compounds 382, and 415 display stable polar contacts (hydrogen bond and water 394 bridges) with G143 and S144 in the S1 pocket and π-cation or π-π interactions with the 395 sidechain of H41. These interactions were more frequent in the covalent simulations. The 396 ligands also display stable polar interactions with the main-chain nitrogen from E166 and 397 electrostatic contacts with its side-chain ( Figure 7D and 7E) , a residue that adopts a stable 398 conformation due to an interaction between its sidechain and the S1 from the other 399 protomer (S1*). Hydrophobic interactions to M49 and M165 from the S2 pocket, are 400 seldomly observed for these inhibitors and frequent interactions with the side-chain of 401 C145 was seen for the covalent inhibitor. conditions the hit naphthoquinone compounds were tested in three concentrations (24 517 µM, 6 µM, and 1.4 µM) and showed no significant antiviral activity dissociated from host 518 cytotoxicity (Supporting Information Figure S51) . We decided to test some compounds 519 in serial dilution starting at 1 µM and in infected human-derived HeLa cells expressing 520 ACE2 in addition to infected Vero cells. For HeLa-ACE2 cells, remdesivir was more 521 potent with EC50 of 40 nM, however, cell cytotoxicity was also noted at concentrations 522 above 2.4 µM. At lower concentrations the naphthoquinone compounds had no 523 significant antiviral activity up to 1 µM (Supporting Information Figure S52 ). Therefore, 524 it is important to further study the mechanism of action to understand the cytotoxicity and 525 decouple from the direct antiviral activity. 526 In this study, we used computational and biochemical approaches to evaluate a 530 library of quinones to find inhibitors against SARS-CoV-2 proteases. The wealth of 531 structural information from Mpro and PLpro allowed us to generate patterns of common 532 protein-ligand interactions, which were helpful in two stages of our computational 533 analysis. First, the selection of computational hits was guided by protein-ligand 534 interactions frequently observed in Mpro crystallographic complexes. Thus, we 535 prioritized compounds that interacted with conserved water molecules, S1 and S2 536 residues, filling one or more of the subsites with minimum solvent exposure. This strategy 537 was successful as, among 24 compounds selected for inhibitory assays, three molecules 538 with two different scaffolds were confirmed as Mpro inhibitors with low micromolar or 539 The journey of remdesivir: From 1116 Ebola to COVID-19 Remdesivir 1125 for the Treatment of Covid-19 -Final Report Coronavirus disease 2019 treatment: A review of 1128 early and emerging options Antiviral activity of Embelia ribes Burm. f. against influenza virus in vitro Plant-derived antiviral drugs as novel hepatitis B virus inhibitors: Cell 1136 culture and molecular docking study Computational studies reveal 1139 mechanism by which quinone derivatives can inhibit SARS-CoV-2. Study of 1140 embelin and two therapeutic compounds of interest, methyl prednisolone and 1141 dexamethasone SARS-CoV 3CLpro inhibitory effects of quinone-methide 1145 triterpenes from Tripterygium regelii Synthesis and potent antitumor 1150 activity of new arylamino derivatives of nor-β-lapachone and nor-α-lapachone Trypanosoma cruzi activity of derivatives from nor-lapachones and lapachones The evaluation of quinonoid 1162 compounds against Trypanosoma cruzi: Synthesis of imidazolic anthraquinones, 1163 nor-β-lapachone derivatives and β-lapachone-based 1 3-Arylamino and 3-1168 alkoxy-nor-β-lapachone Derivatives: Synthesis and Cytotoxicity against Cancer 1169 Electrochemical study, on mercury, of a Meta-nitroarylamine derivative 1174 of nor-β-lapachone, an antitumor and trypanocidal compound Synthesis and 1179 antitumor activity of selenium-containing quinone-based triazoles possessing two 1180 redox centres, and their mechanistic insights Hybrid compounds with two 1184 redox centres: Modular synthesis of chalcogen-containing lapachones and studies 1185 on their antitumor activity Efficient Access to Functionalized Trypanocidal and Antitumor Compounds Synthesis of quinone imine and 1196 sulphur-containing compounds with antitumor and trypanocidal activities: Redox 1197 and biological implications Naphthoquinone-based chalcone hybrids and derivatives: Synthesis and 1202 potent activity against cancer cell lines Potent antileukemic 1207 action of naphthoquinoidal compounds: Evidence for an intrinsic death 1208 mechanism based on oxidative stress and inhibition of DNA repair Synthesis of quinoidal 1212 molecules: Strategies towards bioactive compounds with an emphasis on 1213 lapachones On the investigation of hybrid quinones: Synthesis, 1218 electrochemical studies and evaluation of trypanocidal activity Ruthenium-catalyzed C-H oxygenation of quinones by 1222 weak O-coordination for potent trypanocidal agents Quinonoid 1226 compounds via reactions of lawsone and 2-aminonaphthoquinone with α-1227 bromonitroalkenes and nitroallylic acetates: Structural diversity by C-ring 1228 modification and cytotoxic evaluation against cancer cells New 1232 photoadditions of 2-hydroxy-1,4-naphthoquinones with naphthols and their 1233 derivatiyes Strategies towards potent trypanocidal drugs: Application of ] cycloadditions, sulfonyl phthalide annulation and 1239 nitroalkene reactions for the synthesis of substituted quinones and their 1240 evaluation against Trypanosoma cruzi Catalyzed Double Annulation of Quinones: Step-Economical Access to Valuable 1245 Synthesis and biological evaluation of novel 1,4-naphthoquinone derivatives as 1249 antibacterial and antiviral agents Synthesis of 1253 hydroxy and methoxy perylene quinones, their spectroscopic and computational 1254 characterization, and their antiviral activity Computer-aided 1258 design of 1, 4-naphthoquinone-based inhibitors targeting cruzain and rhodesain 1259 cysteine proteases Naphthoquinoidal [1,2,3]-triazole, a new structural moiety active 1263 against Trypanosoma cruzi Cytotoxic, 1268 trypanocidal activities and physicochemical parameters of nor-β-lapachone-based 1269 1,2,3-triazoles On the search for potential anti-Trypanosoma cruzi drugs: Synthesis and 1275 biological evaluation of 2-hydroxy-3-methylamino and 1,2,3-triazolic 1276 naphthoquinoidal compounds obtained by click chemistry reactions Synthesis and evaluation of the cytotoxic activity 1281 of 1,2-furanonaphthoquinones tethered to 1,2,3-1H-triazoles in myeloid and 1282 lymphoid leukemia cell lines On the search for 1286 potential antimycobacterial drugs: Synthesis of naphthoquinoidal, phenazinic and 1287 1,2,3-triazolic compounds and evaluation against Mycobacterium tuberculosis Redox Center Modification of Lapachones towards 1293 the Synthesis of Nitrogen Heterocycles as Selective Fluorescent Mitochondrial Imaging Probes Molecular hybridization as a powerful tool towards 1299 multitarget quinoidal systems: Synthesis, trypanocidal and antitumor activities of 1300 naphthoquinone-based 5-iodo-1,4-disubstituted-, 1,4-and 1,5-disubstituted-1,2,3-1301 triazoles Synthesis of Selenium-Quinone Hybrid Compounds with Potential Antitumor 1306 Activity via Rh-Catalyzed C-H Bond Activation and Click Reactions Novel fluorescent lapachone-based BODIPY Synthesis, computational and electrochemical aspects, and subcellular 1312 localisation of a potent antitumour hybrid quinone On the synthesis of quinone-based BODIPY hybrids: New insights on antitumor 1317 activity and mechanism of action in cancer cells Synthesis 1327 and anti-Trypanosoma cruzi activity of naphthoquinone-containing triazoles: 1328 Electrochemical studies on the effects of the quinoidal moiety Synthesis of 2,3-1331 unsaturated alkynyl O-glucosides from tri-O-acetyl-d-glucal by using 1332 montmorillonite K-10/iron(III) chloride hexahydrate 3-dipolar cycloaddition Combining clays 1337 and ultrasound irradiation for an o-acetylation reaction of N-glucopyranosyl and 1338 other molecules Natural product-1342 inspired profluorophores for imaging NQO1 activity in tumour tissues A stepwise huisgen 1346 cycloaddition process: Copper(I)-catalyzed regioselective "ligation" of azides 1347 and terminal alkynes Quinone-derived π-extended phenazines as new fluorogenic 1352 probes for live-cell imaging of lipid droplets Chemoselective oxidation of benzophenazines by m-CPBA: N-1356 oxidation vs. oxidative cleavage iodination/organoyl-thiolation for the benzenoid A-ring modification of 1361 quinonoid deactivated systems: A new protocol for potent trypanocidal quinones Carbon nanotube-ruthenium hybrid towards mild oxidation of sulfides to 1366 sulfones: Efficient synthesis of diverse sulfonyl compounds Copper complexes and carbon nanotube-copper ferrite-catalyzed 1371 benzenoid A-ring selenation of quinones: An efficient method for the synthesis of 1372 trypanocidal agents Benzoquinones with Electrophiles: C-H Iodination, Bromination, and 1376 Phenylselenation Ruthenium(II)-Catalyzed C-H Alkenylation of Quinones: Diversity-Oriented 1381 Strategy for Trypanocidal Compounds Cytotoxicity of lapachol, β-lapachone and related 1386 synthetic 1,4-naphthoquinones against oesophageal cancer cells Phthalides with Nitroalkenes for the Synthesis of Fused and Spiro Heterocycles Fused Heterocycles via (4+4) Annulation of Sulfonylphthalide with o-1394 Hydroxystyrenyl Derivatives Cycloadditions with Benzoquinones: De Novo Access to Naphthoquinones for Overcoming naphthoquinone 1401 deactivation: Rhodium-catalyzed C-5 selective C-H iodination as a gateway to 1402 functionalized derivatives Design of 1406 hybrid molecules as antimycobacterial compounds: Synthesis of isoniazid-1407 naphthoquinone derivatives and their activity against susceptible and resistant 1408 strains of Mycobacterium tuberculosis Synthesis of 1413 aryl substituted imidazoles and oxazoles and their potent activity against Mycobacterium tuberculosis Fluorescent oxazoles from quinones for 1418 bioimaging applications Selective endocytic trafficking in live cells 1423 with fluorescent naphthoxazoles and their boron complexes Evaluating the 3C-like protease activity of SARS-Coronavirus: Recommendations for 1427 standardized assays for drug discovery Anti-SARS-CoV-2 activities in vitro of Shuanghuanglian preparations and bioactive ingredients A taxonomically-driven approach to development of potent, 1436 broad-spectrum inhibitors of coronavirus main protease including SARS-CoV-2 1437 (COVID-19) Structural and Evolutionary Analysis Indicate That the SARS-CoV-2 Mpro Is a 1440 Challenging Target for Small-Molecule Inhibitor Design Structural plasticity of SARS-CoV-2 3CL Mpro active site cavity revealed by room temperature X-ray crystallography Crystallographic and electrophilic fragment 1452 screening of the SARS-CoV-2 main protease Similarity Analysis of Protein Cavities to the Functional Classification of Protein 1456 Families Using Cavbase Identification of LASSBio-1945 as an 1459 inhibitor of SARS-CoV-2 main protease (MPRO) through in silico screening 1460 supported by molecular docking and a fragment-based pharmacophore model A Blueprint for High Affinity SARS-CoV-2 Mpro Inhibitors from Activity Based Compound Library Screening Guided by Analysis of Protein Dynamics Critical 1470 assessment of the important residues involved in the dimerization and catalysis of Molecular biology of severe acute respiratory syndrome coronavirus Identification of Conserved Water Sites 1477 in Protein Structures for Drug Design Glide: A New Approach for Rapid, Accurate Docking and Scoring Enrichment Factors in Database Screening AutoDock Vina: Improving the speed and accuracy of 1484 docking with a new scoring function, efficient optimization, and multithreading Identification 1487 of host cellular protein substrates of sars-cov-2 main protease A specific mechanism of 1490 nonspecific inhibition A detergent-based assay for the detection of 1493 promiscuous inhibitors Quantitative analyses of 1497 aggregation, autofluorescence, and reactivity artifacts in a screen for inhibitors of 1498 a thiol protease A common mechanism 1501 underlying promiscuous inhibitors from virtual and high-throughput screening Design of Gallinamide A Analogs 1505 as Potent Inhibitors of the Cysteine Proteases Human Cathepsin L and From SARS to MERS: crystallographic studies on coronaviral 1509 proteases enable antiviral drug design New substructure filters for removal of pan assay 1512 interference compounds (PAINS) from screening libraries and for their exclusion 1513 in bioassays Crystallographic 1517 structure of wild-type SARS-CoV-2 main protease acyl-enzyme intermediate 1518 with physiological C-terminal autoprocessing site Structure and dynamics of SARS coronavirus main proteinase (M 1522 pro Targeting the Dimerization of the Main Protease of terminus is indispensable for proteolytic activity but not for enzyme 1530 dimerization: Biochemical and thermodynamic investigation in conjunction with 1531 molecular dynamics simulations SARS-CoV-2 main protease: a molecular dynamics study A small 1539 molecule compound with an indole moiety inhibits the main protease of SARS CoV-2 and blocks virus replication Synthesis of mixed aryl 2,3-diarylsulphanyl-1,4-1543 naphthoquinones SARS-CoV-2 Mpro inhibitors with antiviral activity in a transgenic mouse 1551 model, Science It takes two 1556 to tango: synthesis of cytotoxic quinones containing two redox active centers 1557 with potential antitumor activity The Protein Data Bank A Language and Environment for Statistical Computing Bio3d: an R package for the comparative analysis of protein structures The PyMOL Molecular Graphics System Docking covalent inhibitors: A parameter free approach to pose prediction and 1571 scoring Novel procedure 1574 for modeling ligand/receptor induced fit effects Scalable Algorithms for Molecular Dynamics Simulations on Commodity 1579 SC '06 Proc. 2006 ACM/IEEE Conf. Supercomput OPLS3: A Force Field Providing Broad 1584 Coverage of Drug-like Small Molecules and Proteins SARS-COV-2 Mpro conformational changes induced by covalently bound 1588 ligands Comparison of simple potential functions for simulating liquid water Particle mesh Ewald: An N·log(N) method for Ewald sums in large systems Potent in vitro anti-SARS-CoV-2 activity by gallinamide A and analogues 1600 via inhibition of cathepsin L https://www.gilead.com/news-and-press/press-room/press-1106 releases/2020/5/gileads-investigational-antiviral-remdesivir-receives-us-food-1107 and-drug-administration-emergency-use-authorization-for-the-treatment-of-1108