ACS Publications. Most Trusted. Most Cited. Most Read
Discovery and Mechanism of SARS-CoV-2 Main Protease Inhibitors
My Activity
  • Open Access
Article

Discovery and Mechanism of SARS-CoV-2 Main Protease Inhibitors
Click to copy article linkArticle link copied!

  • Sarah Huff
    Sarah Huff
    Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    More by Sarah Huff
  • Indrasena Reddy Kummetha
    Indrasena Reddy Kummetha
    Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
  • Shashi Kant Tiwari
    Shashi Kant Tiwari
    Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
  • Matthew B. Huante
    Matthew B. Huante
    Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United States
  • Alex E. Clark
    Alex E. Clark
    Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
  • Shaobo Wang
    Shaobo Wang
    Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    More by Shaobo Wang
  • William Bray
    William Bray
    Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    More by William Bray
  • Davey Smith
    Davey Smith
    Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    More by Davey Smith
  • Aaron F. Carlin
    Aaron F. Carlin
    Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
  • Mark Endsley
    Mark Endsley
    Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United States
    More by Mark Endsley
  • Tariq M. Rana*
    Tariq M. Rana
    Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    *Email: [email protected]
Open PDFSupporting Information (2)

Journal of Medicinal Chemistry

Cite this: J. Med. Chem. 2022, 65, 4, 2866–2879
Click to copy citationCitation copied!
https://doi.org/10.1021/acs.jmedchem.1c00566
Published September 27, 2021

Copyright © 2021 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY-NC-ND 4.0 .

Abstract

Click to copy section linkSection link copied!

The emergence of a new coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presents an urgent public health crisis. Without available targeted therapies, treatment options remain limited for COVID-19 patients. Using medicinal chemistry and rational drug design strategies, we identify a 2-phenyl-1,2-benzoselenazol-3-one class of compounds targeting the SARS-CoV-2 main protease (Mpro). FRET-based screening against recombinant SARS-CoV-2 Mpro identified six compounds that inhibit proteolysis with nanomolar IC50 values. Preincubation dilution experiments and molecular docking determined that the inhibition of SARS-CoV-2 Mpro can occur by either covalent or noncovalent mechanisms, and lead E04 was determined to inhibit Mpro competitively. Lead E24 inhibited viral replication with a nanomolar EC50 value (844 nM) in SARS-CoV-2-infected Vero E6 cells and was further confirmed to impair SARS-CoV-2 replication in human lung epithelial cells and human-induced pluripotent stem cell-derived 3D lung organoids. Altogether, these studies provide a structural framework and mechanism of Mpro inhibition that should facilitate the design of future COVID-19 treatments.

This publication is licensed under

CC-BY-NC-ND 4.0 .
  • cc licence
  • by licence
  • nc licence
  • nd licence
Copyright © 2021 The Authors. Published by American Chemical Society

SPECIAL ISSUE

This article is part of the COVID-19 special issue.

Introduction

Click to copy section linkSection link copied!

Emergent β-coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes COVID-19, for which there is a great need for a new targeted treatment. SARS-CoV-2 produces a spike protein that binds to host cell receptor ACE2 for entry. (1,2) Upon entry, the positive genomic RNA of SARS-CoV-2 will attach directly to the host ribosome and translate two large polyproteins, which are then processed by proteolysis into components for packaging new virions. This proteolysis is controlled by two protease enzymes, the coronavirus main protease (Mpro) and the papain-like protease (PLpro). (2−6) An RNA-dependent RNA polymerase (RdRp) is also required to replicate the RNA genome. (2,7) As all four of these proteins are essential for viral replication, they are considered attractive drug targets for treating coronaviruses. Of these, Mpro has been widely called the virus’ “Achilles Heel” and is considered one of the most attractive targets for drug development against SARS-CoV-2. The main protease is an established drug target for similar (+)-ssRNA viruses such as HIV and hepatitis C, (8−14) although to date there are no approved therapies targeting SARS-CoV-2’s main protease.
Some early studies have identified crystal structures of potential small-molecule binders of SARS-CoV-2 Mpro, which are predominately peptide-based scaffolds. (15−20) Jin et al. determined the first crystal structure of Mpro in complex with the peptide inhibitor N3. (15) Additional crystal structures of two rationally designed covalent peptide inhibitors, 11a and 11b, were reported shortly thereafter by Dai et al. (16) Two additional reports demonstrated that the feline coronavirus drug GC376 also inhibits Mpro and prevents viral replication in infected Vero cells. (17,20) Like 11a and 11b, GC376 is a peptide inhibitor that covalently modifies active site Cys 145. Several nonpeptide small molecules have also been shown to inhibit Mpro proteolysis in vitro, including the organoselenium compound ebselen; however, to date there is only limited knowledge of their mechanism of action and the cell-based evaluation of small molecules. (15,19) At this time, most studies targeting Mpro are focused on screening existing compounds, and there has been little interest in further developing early hits using medicinal chemistry and rational drug design approaches. In this study, we sought to determine if such approaches could be applied to address the following questions: (1) can rational drug design and medicinal chemistry be used to improve the potencies of identified Mpro inhibitors? (2) Can an enzymatic mechanism of inhibition be established for rationally designed Mpro inhibitors? (3) Can rationally designed compounds inhibit viral replication in vitro? Finally, (4) how do rationally designed Mpro inhibitors compare to existing treatments such as remdesivir?
To address these questions, we used the structure of the organoselenium compound ebselen as the basis for the rational design of improved Mpro inhibitors. In silico docking at the SARS-CoV-2 Mpro active site identified a class of 2-phenyl-1,2-benzoselenazol-3-one inhibitors with nanomolar proteolytic activities against recombinant SARS-CoV-2 Mpro and improved physicochemical properties relative to those of their parent compound ebselen. Steady-state enzyme kinetics was used to determine that ebselen and its analogue E04 are competitive inhibitors of Mpro, and 2-phenyl-1,2-benzoselenazol-3-one inhibitors may follow either a noncovalent or reversible covalent mechanism. These compounds were also determined to show potent antiviral effects in multiple SARS-CoV-2-infected cell lines, with leads E24 and E25 reporting inhibitory activities comparable or better to that of the clinical antiviral treatment remdesivir (EC50(E24) = 0.8 ± 0.3 μM, EC50(E25) = 2.0 ± 1.1 μM, and EC50(remdesivir) = 1.8 ± 1.3 μM). Furthermore, E24 was found to reduce viral replication in both Calu-3 human lung epithelial cells (EC50(E24) = 1.3 ± 0.8 μM) and human iPSC-derived lung organoids infected with SARS-CoV-2.

Results

Click to copy section linkSection link copied!

Identification of the 2-Phenyl-1,2-benzoselenazol-3-one Class of SARS-CoV-2 Mpro Inhibitors

Recently, a study showed that the organoselenium compound ebselen was able to bind SARS-CoV-2 Mpro and inhibit proteolytic activity. (15) While it was determined that ebselen could partially covalently modify Mpro, the binding site was not identified. Using this information as a starting point, we sought to model ebselen in the active site of Mpro to determine if organoselenium compounds could be rationally designed to target the catalytic site. Docking was performed in the Schrödinger software suite. (21−23) A crystal structure of the SARS-CoV-2 Mpro bound to N3 (PDB ID 6UL7) was used to generate the docking grid as a 15 Å cube centered on the ligand N3. As observed in Figure 1A, the active site of Mpro is expected to accommodate ebselen. Ebselen is predicted to bind in close proximity to catalytic His 41 and Cys 145, with the benzoisoselenazole ring forming π–π stacking interactions with catalytic His 41. Binding is further stabilized by a hydrogen bond between the carbonyl oxygen atom of ebselen and Glu 166. These results are consistent with a recently reported crystal structure of SARS-CoV-2 Mpro in complex with ebselen, which indicates that it primarily binds to the catalytic site of the enzyme and forms a selenyl sulfide bond with Cys 145; this was further confirmed by LC/MRM-MS. (24)

Figure 1

Figure 1. Docking poses of ebselen and 2-phenyl-1,2-benzoselenazol-3-one analogues. (A) Docking pose of ebselen bound to the catalytic site of SARS-CoV-2 Mpro. A hydrogen bond is present between the carbonyl oxygen atom of E01 and Glu 166. The benzoisoselenazole ring forms π–π stacking interactions with catalytic His 41. (B) Docking pose of E19 bound to the catalytic site of SARS-CoV-2 Mpro. A hydrogen bond is present between the carbonyl oxygen atom of E19 and Glu 166. The benzoisoselenazole ring forms π–π stacking interactions with catalytic His 41. The chlorine atom of E19 forms a halogen bond interaction with Gly 143. (C) Docking pose of E20 bound to the catalytic site of SARS-CoV-2 Mpro. π–π stacking interactions are present between the benzoisoselenazole ring of E20 and catalytic His 41. A hydrogen bond is present between the carbonyl oxygen atom of E20 and Glu 166. (D) Docking pose of E25 bound to the catalytic site of SARS-CoV-2 Mpro. A hydrogen bond is present between the carbonyl oxygen atom of E25 and Glu 166. π–π stacking interactions are present between the benzoisoselenazole ring of E25 and catalytic His 41. The nitrile group was observed to accept a hydrogen bond from Cys 44.

A series of 2-phenyl-1,2-benzoselenazol-3-one analogues was designed with substitutions primarily focused on the N-phenyl moiety. Substitutions included halogens (F, Cl, and Br) and hydrophobic groups (CH3, CF3, SCH3, OCH3, and CH2CH3) as well as several lipophilic replacements of the phenyl moiety. Potential inhibitors were docked and ranked by their docking score. The docking poses of prospective analogues were evaluated for interactions with catalytic residues His 41 and Cys 145; compounds that were expected to form strong hydrogen bonds or π-interactions with these residues were prioritized for synthesis. Docking poses for ebselen and three of its analogues can be found in Figure 1. As observed in Figure 1 B, E19 is able to maintain the interactions with Glu 166 and catalytic His 41 that were observed for ebselen, while the p-chloro substituent group forms an additional halogen bond with the amide backbone of Gly 143. Similarly, the m-cyano group of E25 is able to accept a hydrogen bond from Cys 44 while maintaining the interactions of the parent compound with His 41 and Glu 166 (Figure 1 A and D). In Figure 1 C, E20 is expected to maintain the core interactions of the parent structure with His 41 and Glu 166, while m,m-dichloro substitutions occupy a larger region of the binding pocket to increase the entropy of the binding interaction. Docking poses for select additional compounds may be found in the Supporting Information (Figure S1).
A total of 30 2-phenyl-1,2-benzoselenazol-3-one compounds were selected for synthesis according to Scheme 1 (Figure 1 E). Analogues were synthesized from commercially available acid chlorides and anilines in two steps. Nucleophilic acyl substitution in the presence of triethylamine yielded intermediate 3. Ring closure to form the benzoisoselenazole ring proceeded in the presence of NaSeOtBu with selenium powder and sodium tert-butoxide in DMF to produce the final product. In total, 30 compounds were prepared according to this scheme, purified by column chromatography (hexane/ethyl acetate 70:30), and characterized by NMR and HRMS prior to the biological evaluation.

Scheme 1

Scheme 1. Synthesis of Ebselen Analogues

The 2-Phenyl-1,2-benzoselenazol-3-one Class of Compounds Are Potent Inhibitors of Recombinant Mpro

To determine if the 2-phenyl-1,2-benzoselenazol-3-one class of rationally designed compounds was also able to inhibit protease activity of SARS-CoV-2 Mpro, a FRET assay adapted from Muramatsu et al. (25) was used to assess the inhibitory potency of the compounds against SARS-CoV-2 Mpro at 10 μM. Ebselen was included as a positive control. (15) Of the 30 analogues tested, 22 inhibited Mpro proteolytic activity by >50%, while eight compounds showed no inhibition at this concentration. Full dose–response curves were generated for compounds showing >50% inhibition at 10 μM (Table 1). The IC50 of ebselen was observed to be 0.33 μM, which was similar to the previously reported value of 0.67 μM. (15) Most 2-phenyl-1,2-benzoselenazol-3-one analogues reported IC50 values between 1 and 7.4 μM. Compounds E04, E07, E19, E20, and E25 reported submicromolar IC50 valuess ranging from 0.38 to 0.90 μM (Table 1). Additional reports have identified two peptide inhibitors, 11a and 11b, that were designed to inhibit the main protease with more potent IC50 values (0.053 and 0.040 μM, respectively); (16) however, the observed IC50 valuess for compounds E04, E07, E19, E20, and E25 are similar in range to that reported for the feline coronavirus drug GC376 (0.19 ± 0.04 μM). (17) In general, meta-substitution of the N-phenyl moiety was favorable. Two of the most potent compounds, E19 and E20, featured a meta-substituted chlorobenzene, and the most potent analogue, E25, featured a meta-cyano group. As observed in Figure 1 B, the p-chloro substitution of E19 is positioned such that it can form a halogen bond with the amide backbone of Gly 143, while the 1,2-benzisoselenazole ring forms π–π interactions with catalytic His 41 and a hydrogen bond to Glu 166. In Figure 1 D, the m-cyano group of E25 is able to accept a hydrogen bond from Cys 44 while maintaining the core interactions with the 1,2-benzisoselenazole ring, π–π interactions with catalytic His 41, and a hydrogen bond to Glu 166. In both these examples, meta-substitution could enable additional favorable interactions with the binding site that are not supported by ortho- or para-substitution. In general, para-substitution of the N-phenyl ring was tolerated, with most IC50 values in the micromolar range, while ortho-substitution was generally unfavorable. Compound E15 featured the only variation to the 1,2-benzisoselenazole ring, a methoxy group at the C6 position. As observed in Figure 1 A, the 1,2-benzisoselenazole ring of ebselen is expected to form π–π interactions with catalytic His 41 and a hydrogen bond to Glu 166, with the C6 position oriented toward His 41. The addition of the methoxy group at this position is likely to impair the π–π interactions expected with catalytic His 41 and could reposition the compound within the binding site such that the hydrogen bond to Glu 166 is weaker. This substitution was observed to reduce the potency against SARS-CoV-2 Mpro by ∼20× relative to that of ebselen.
Table 1. Structures, Docking Scores, and In Vitro Data Summary of 2-Phenyl-1,2-benzoselenazol-3-one Analogues

2-Phenyl-1,2-benzoselenazol-3-one Inhibitors Can Inhibit Mpro Covalently or Noncovalently

The selenium atom of ebselen is capable of forming a selenylsulfide bond (−Se–S−); (26−29) in Singh et al., the substitution of the Se atom with S or the mutation of the target active site Cys was observed to decrease inhibition. (29) In most cases, proteins covalently modified by ebselen may be rescued by the sulfhydryl reducing agents glutathione (GSH) and dithiothreitol (DTT). Previously, Jin et al. reported that ebselen was able to partially modify Mpro Cys residues and concluded that ebselen could inhibit Mpro by both covalent and noncovalent mechanisms. (15) To determine if 2-phenyl-1,2-benzoselenazol-3-one analogs were also able to covalently modify the active site of SARS-CoV-2 Mpro, we performed a preincubation dilution experiment where SARS-CoV-2 Mpro was incubated with 10 μM inhibitor for 30 min prior to 10× dilution with the reaction buffer. Upon dilution, rapidly reversible inhibitors dissociate from the enzyme active site, and the resulting activity is proportional to that observed for the final inhibitor concentration, in this case 1 μM. If the inhibitor dissociation is slow, then the enzyme–inhibitor complexes formed at high concentrations will persist after dilution and the resulting activity will instead be proportional to the initial concentration of 10 μM. The concentration range of 10 and 1 μM was selected as all compounds tested in this experiment showed full enzymatic inhibition at 10 μM and some activity at 1 μM, typically between 15 and 50% wt activity. As previous reports have indicated that the covalent modification of Cys residues by ebselen is sensitive to reversal by reducing agents such as DTT or glutathione, we removed DTT from the assay buffer for these experiments.
The effect of dilution on the reaction rate of SARS-CoV-2 Mpro preincubated with a 10 μM concentration of compounds E01 (ebselen), E04, E07, E19, E20, and E25 can be observed in Figure 2. The effect of the simultaneous addition of the substrate and either 1 or 10 μM inhibitor on the rate of SARS-CoV-2 Mpro was included for each inhibitor as a comparison. As seen in Figure 2 A, C, and E, dilution of the protease-inhibitor reaction mixture did not alter the reaction rate when Mpro was incubated with ebselen, E07, or E20, indicating that these compounds likely inhibit Mpro by covalent modification. However, the dilution of Mpro treated with compounds E04 and E19 did result in a reaction rate similar to that observed for a 1 μM concentration of the undiluted compound, indicating that these analogues are rapidly reversible inhibitors of SARS-CoV-2 Mpro (Figure 2 B and D). Interestingly, the dilution of the Mpro/E25 reaction mixture did show some recovery of the enzymatic activity but not a complete recovery comparable to the activity observed for Mpro treated with 1 μM E25 without dilution (Figure 2 F). These results could suggest that the covalent modification of Mpro was incomplete in the 30 min preincubation period. To determine if Mpro inhibition by 2-phenyl-1,2-benzoselenazol-3-one analogues is sensitive to reducing agents, the experiment was repeated with the presence of 1 mM DTT in the assay buffer. Under these conditions, the dilution of the reaction mixtures was sufficient to recover the enzymatic activity, suggesting that any covalent adduct formed with Mpro Cys residues was reduced such that the enzyme–inhibitor complex dissociated. These results indicate that while ebselen and some 2-phenyl-1,2-benzoselenazol-3-one analogues are able to covalently modify active Mpro Cys residues, this covalent modification is reversible in a reducing environment. It is also possible that 2-phenyl-1,2-benzoselenazol-3-one analogues can inhibit Mpro noncovalently, as observed for compounds E04 and E20.

Figure 2

Figure 2. Ebselen and its analogues can covalently modify SARS-CoV-2 Mpro in the absence of reducing agents. (A) Ebselen is a reversible covalent inhibitor of SARS-CoV-2 Mpro. The product formation over time was reduced by approximately 65% by 1 μM ebselen (red) and by 89% by 10 μM ebselen (purple) relative to that with the DMSO control (black) when added simultaneously with the fluorogenic substrate. Upon 10× dilution in an assay buffer without DTT, the product formation of SARS-CoV-2 Mpro preincubated with 10 μM ebselen (green) was reduced by 91%, which is comparable to an undiluted concentration of 10 μM. When diluted in the presence of DTT, the product formation of SARS-CoV-2 Mpro preincubated with 10 μM of ebselen (blue) was reduced by 53% relative to that with the DMSO control, similar to that observed for a final concentration of 1 μM. These data indicate that ebselen can covalently modify SARS-CoV-2 Mpro, although this modification is reversible in the presence of strong reducing agents. (B) Compound E04 is a rapidly reversible inhibitor of SARS-CoV-2 Mpro. Upon 10× dilution, the product formation of SARS-CoV-2 Mpro preincubated with 10 μM E04 in the absence of DTT (green) was reduced by 48% relative to that with the DMSO control, similar to that observed for a final concentration of 1 μM E04 (44% inhibition), while the simultaneous addition of the substrate and 10 μM E04 reduced the product formation by 86%. Similar results were observed for SARS-CoV-2 Mpro preincubated with 10 μM E04 in the presence of DTT (blue, 43% inhibition). (C) Compound E07 is a reversible covalent inhibitor of SARS-CoV-2 Mpro. (D) Compound E19 is a rapidly reversible inhibitor of SARS-CoV-2 Mpro. (E) Compound E20 is a reversible covalent inhibitor of SARS-CoV-2 Mpro. (F) Compound E25 is a reversible covalent inhibitor of SARS-CoV-2 Mpro. Upon 10× dilution, the product formation of SARS-CoV-2 Mpro preincubated with 10 μM E25 in the absence of DTT (green) was reduced by 64% relative to that with the DMSO control. The simultaneous addition of the substrate and 10 μM 25 reduced product formation by 94%, while the simultaneous addition of the substrate and 1 μM E25 reduced product formation by 29%. These data suggest that while E25 may covalently modify SARS-CoV-2 Mpro, the formation of the covalent adduct was incomplete after 30 min of incubation.

2-Phenyl-1,2-benzoselenazol-3-one Inhibitor E04 and Ebselen Are Competitive Inhibitors of Mpro

The mechanism of Mpro inhibition by ebselen and E04 was further characterized by steady-state enzyme kinetics (Figure 3). The reaction rates of untreated (wild-type) Mpro at varying substrate concentrations and Mpro in the presence of 0.5, 1, 5, and 10 μM E01 and E04 were determined, and the data were globally fit to a generalized mixed-model equation as described in the Inhibition Mechanism section. In this mechanism, the binding of the inhibitor to the ES complex is described by αKi, where Ki is the dissociation constant. An value of α ≫ 1 provided a better fit of the data for both inhibitors, suggesting a weak binding to the ES complex and a preference for the free enzyme. A plot of the reaction velocity versus the substrate concentration shows that when Mpro is treated with either ebselen or E04, Vmax recovers to that observed for wild-type Mpro at substrate concentrations of 30 μM (Figure 3 A and C). A double-reciprocal plot shows that for both compounds Km is dependent on [I] while Vmax remains consistent (Figure 3 B and D). These results indicate that ebselen and E04 are likely competitive inhibitors of Mpro.

Figure 3

Figure 3. Ebselen and E04 are competitive inhibitors of the SARS-CoV-2 main protease. (A) Mpro exposed to varying concentrations of ebselen reached a common Vmax at 30 μM substrate (B) All ebselen conditions converge on a common y-intercept, indicating that Vmax is independent of the inhibitor concentration, which is consistent with a competitive mechanism of inhibition. (C) Mpro exposed to varying concentrations of E04 reached a common Vmax at 30 μM substrate (D) All E04 conditions converge on a common y-intercept, indicating that Vmax is independent of the inhibitor concentration, which is consistent with a competitive mechanism of inhibition. (E) Antiviral activity of select compounds. Vero E6 cells were pretreated with varying doses of the compound, then infected with SARS-CoV-2 to a multiplicity of infection (MOI) of 0.01. After 72 h, cell proliferation was determined. Remdesivir is included as a positive control (EC50 = 1.8 ± 1.3 μM).

2-Phenyl-1,2-benzoselenazol-3-one Analogues Are Potent Antiviral Agents In Vitro

To determine if 2-phenyl-1,2-benzoselenazol-3-oneanalogues were able to impair SARS-CoV-2 replication, the compounds were screened against infected Vero E6 cells at doses ranging from 20 to 0.1 μM to determine the cellular EC50 value (Figures 3 E and S2 and Table 2). Briefly, cells were pretreated with compound for 1 h, then infected with SARS-CoV-2 (USA_WA01/2020) at a multiplicity of infection (MOI) of 0.01 for 1.5 h. Cells were washed, the compounds were reapplied, and cells were then incubated for 72 h prior to recording cthe ell proliferation. As a virus infection reduces cell proliferation and leads to cell death, only those compounds that show increased cell survival relative to that of the DMSO negative control are considered to have antiviral activity. In this way, compounds that are likely to be cytotoxic to Vero E6 cells at the concentrations tested are excluded as potential hits. Ebselen and remdesivir were included as positive controls. The antiviral activity of each compound is reported in Table 1. Most compounds reported EC50 valuess in the micromolar range, and compounds E18, E21, and E25 showed antiviral potencies similar to that of remdesivir (Tables 1 and 2 and Figure 3 E). The most potent compound, E24, showed a nanomolar antiviral potency (EC50 = 0.84 ± 0.3 μM). This is within range of the most potent Mpro inhibitors reported to date, including the rationally designed peptidyl inhibitors 11a and 11b (EC50 = 0.53 ± 0.01 and 0.72 ± 0.09 μM, respectively) (16) and the feline coronavirus drug GC376 (EC50 = 0.92 μM). (17) Interestingly, all three of these inhibitors are peptides that contain a similar warhead for the covalent modification of active site Cys 145. We demonstrated that the 2-phenyl-1,2-benzoselenazol-3-one class of inhibitors may inhibit Mpro either covalently or noncovalently (Figure 2).
Table 2. Antiviral Activity and logD Values of Select 2-Phenyl-1,2-benzoselenazol-3-one Analoguesa
compoundantiviral EC50 (μM)logD pH 7.4compoundantiviral EC50 (μM)logD pH 7.4
ebselen>200.92remdesivir1.8 ± 1.2ND
E0411.2 ± 1.33.19E2018.2 ± 3.61.67
E0726.5 ± 1.20.85E215.2 ± 1.81.46
E186.5 ± 2.02.87E240.8 ± 0.33.18
E1917.4 ± 3.51.53E252.0 ± 1.11.45
a

ND, not determined.

For some compounds, the potencies of the 2-phenyl-1,2-benzoselenazol-3-one analogues were reduced in the Vero cell assay relative to those in the FRET assay using recombinant Mpro. For example, while E04, E19, E20, and E25 reported nanomolar IC50 valuess against recombinant Mpro (IC50 = 0.38–0.90 μM), their EC50 valuess in the antiviral assay were in the micromolar range (2.0–18.2 μM). As these compounds are not expected to prevent viral entry into the cells, their antiviral potency will be a function of a variety of factors, including cell membrane permeability, efflux rate, and cellular metabolic pathways. The antiviral assays were also conducted over a 72 h incubation period, while the FRET assay was performed in a 1 h time frame, further complicating a direct comparison of potencies across these two assay systems. As observed in the preincubation dilution assays, many of the 2-phenyl-1,2-benzoselenazol-3-one analogues are sensitive to reducing agents, and their inhibitory potency could be affected by intracellular reducing mechanisms. While the exact potencies varied, in general compounds that were inactive against recombinant Mpro were also inactive in the Vero E6 cell antiviral assays.

2-Phenyl-1,2-benzoselenazol-3-one Compounds Show Improved LogD Values Relative to Ebselen

LogD values were also determined for ebselen and 2-phenyl-1,2-benzoselenazol-3-onecompounds E04, E07, E18, E19, E20, E21, E24, and E25 (Table 2). Meta-analyses of pharmaceutical drug development projects have identified the importance of logD in identifying compounds that are more likely to feature favorable clearance rates and membrane permeabilities; one such study found that compounds with a molecular weight of 350 g/mol and a logD of 1.5 had a 25% success rate of being advanced to clinical trials. (30−34) By analyzing the logD values for these analogues at this stage, we are able to predict which compounds are most likely to show favorable pharmacokinetic properties in future in vivo models. We may also determine if logD optimization can be useful in refining these compounds as antiviral agents prior to an in vivo study. As observed in Table 2, logD values are generally in the ideal range for most orally available drugs (Table 2, entries1–3). (30−33) Furthermore, logD values were broadly observed to correlate with the antiviral activity, where compounds with higher logD values (E18, and E24) showed greater antiviral potencies than those with low logD values (E07, E19, and E20). These data indicate that the logD value is a critical parameter for optimizing the antiviral potencies of these compounds.

2-Phenyl-1,2-benzoselenazol-3-one Analogues Show Antiviral Properties in Human Lung Cells

To further confirm the antiviral properties observed in the Vero E6 cell assay, four compounds were selected foran additional evaluation in human lung epithelial cells. Ebselen and remdesivir were both included as positive controls. As before, Calu-3 cells were pretreated with 0.1–20 μM concentrations of the compound for 1 h prior to infection with SARS-CoV-2 (USA_WA01/2020; MOI = 0.1) for 1.5 h. Cells were washed, the compounds were reapplied, and cells were then incubated for 24 h prior to the isolation and quantification of intracellular and supernatant viral RNA by RT-qPCR. Table 3 lists the antiviral EC50 values of ebselen, remdesivir, E04, E18, E21, and E24 in Calu-3 cells.
Table 3. Antiviral Activity of Select 2-Phenyl-1,2-benzoselenazol-3-one Analogues in Calu-3 Cells
compoundEC50 (μM)compoundEC50 (μM)
ebselen5.0 ± 4.0remdesivir0.8 ± 0.6
E042.8 ± 1.3E2115.7 ± 4.3
E188.8 ± 5.4E241.3 ± 0.8
In this model, remdesivir appears to be slightly more potent than in the Vero E6 cell proliferation assays (EC50 = 0.8 ± 0.6 and 1.8 ± 1.2 μM, respectively); interestingly, this slight improvement in potency in human cell models was also observed for remdesivir by Dittmer et al. (35) and Jang et al. (36) in Calu-3 cells and by Choi et al. (37) in hESC- and hiPSC-derived cardiomyocytes. The difference in potency could be due to the variations in experimental conditions, including the cell lines and infection rates. Additionally, a recent comparison of commonly used cell models for SARS-CoV-2 antiviral assays by Dittmer et al. indicated that entry mechanisms vary for Calu-3 and Vero cells, where entry in Calu-3 cells was pH-independent and relied on transmembrane protease serine 2 (TMPRSS2) and entry to Vero cells was reliant upon acid-dependent endosomal proteases and low pH levels. (35) These variations in entry mechanisms resulted in significant alterations in the drug efficacy, particularly for compounds that targeted host proteases. (35) However, compounds that target the virus directly, like remdesivir, should remain active across cell types. As our 2-phenyl-1,2-benzoselenazol-3-one analogues target the SARS-CoV-2 main protease, we expected to retain antiviral activity for these compounds as well.
In Calu-3, ebselen had an EC50 of 5.0 μM, which is in the range of reported EC50 values observed by Vero E6 cell proliferation assays. (24) Of the remaining 2-phenyl-1,2-benzoselenazol-3-one analogues, E18 and E24 had EC50 values consistent with those observed in Vero E6 cells (E18 EC50 = 8.8 ± 5.4 μM; E24 EC50 = 1.3 ± 0.8 μM). E04 showed a marked improvement in potency, with a fourfold reduction in EC50 in Calu-3 cells (2.8 ± 1.3 μM) versus that in Vero E6 (11.2 ± 1.3 μM). E21, however, showed a threefold reduction in potency Calu-3 cells (EC50 = 15.7 ± 4.3 μM) relative to that in Vero E6 (EC50 = 5.2 ± 1.8 μM). This difference in potency observed for E04 and E21 could be due to changes in either the compound permeability or stability in various cells and culture conditions. Importantly, all compounds remained active antivirals in human lung epithelial cells, indicating that the antiviral properties of the analogues are robust across assay systems and relevant to human models.

Compound E24 Reduces SARS-CoV-2 Viral Infection in Human Lung Organoids

The most potent compound from our antiviral screens, E24, was evaluated in a lung organoid model derived from human-induced pluripotent stem cells (iPSC). Human iPSC-derived lung 3D organoids were generated as described by Tiwari et al. (38) Organoids were characterized as containing epithelial cells, alveolar types 1 and 2, and a high expression of both ACE2 and TMPRSS2 and were verified to permit SARS-CoV-2 infection as described previously. (38−40) Organoids were pretreated with 5 μM E24 for 2 h and infected with SARS-CoV-2 at MOI = 2 for 2 h at 37 °C. Then, the cells were washed, and a fresh medium was added. At 72 h postinfection, both intracellular viral RNA and viral RNA from the supernatant were isolated and quantified by RT-qPCR. As shown in Figure 4, E24 treatment reduced viral RNA levels by ∼50% in both the supernatant and intracellular fractions with no observable toxicity toward the organoids. These data confirm that E24 can impair the viral replication of SARS-CoV-2 in human lung organoids.

Figure 4

Figure 4. E24 inhibits SARS-CoV-2 replication in infected human iPSC-derived lung organoids. Sixty-day-old lung organoids were infected with the SARS-CoV-2 USA-WA1/2020 virus at a MOI of 2, and viral RNAs from (A) supernatant and (B) cellular fractions were quantified after 72 h of infection. Mean ± SEM of n = 3 organoids cultured and infected in different wells, *p < 0.05 by Student’s t-test.

Discussion and Conclusions

Click to copy section linkSection link copied!

In this study, we sought to apply medicinal chemistry and rational drug design approaches to the SARS-CoV-2 main protease to address the following specific goals: (1) to improve potency of early Mpro screening hits, (2) to establish an enzymatic mechanism of inhibition, (3) to rationally design compounds with potent antiviral activity, and (4) to compare rationally designed compounds against existing early SARS-CoV-2 antivirals. With this approach, we have identified a class of 2-phenyl-1,2-benzoselenazol-3-one compounds showing nanomolar potencies against recombinant SARS-CoV-2 Mpro and improved lipophilicities relative to the early Mpro inhibitor ebselen. Using steady-state enzyme kinetics, we were able to establish that some 2-phenyl-1,2-benzoselenazol-3-one compounds inhibit Mpro noncovalently, suggesting they are less likely to suffer from target promiscuity than covalent modifiers such as ebselen. Furthermore, lead compound E04 was found to inhibit Mpro through a competitive mechanism of inhibition, indicating that nonpeptidyl small molecules may bind to the active site of Mpro to inhibit proteolysis. Additionally, 2-phenyl-1,2-benzoselenazol-3-one compounds were determined to inhibit SARS-CoV-2 viral replication in infected Vero E6 and Calu-3 cells with comparable potencies to that of the clinical treatment remdesivir. The further evaluation of lead E24 in iPSC-derived human lung organoids confirmed that E24 was able to significantly reduce viral RNA levels in both supernatant and intracellular fractions at a 5 μM dose.
Ebselen is known to be a promiscuous protease inhibitor capable of binding a variety of viral proteases, including SARS-CoV-2 Mpro and PLpro, EV-A71 2Apro and 3Cpro, and EV-D68 2Apro and 3Cpro. (41,42) At this stage, it is unknown if the 2-phenyl-1,2-benzoselenazol-3-one analogues presented in this study are also able to bind and inhibit additional proteases such as the SARS-CoV-2 papain-like protease (PLpro), which cleaves nonstructural protein 1 (nsp1), nonstructural protein 2 (nsp2), and nonstructural protein 3 (nsp3) of the viral polyprotein. PLpro has been identified as a promising drug target for SARS-CoV-2 both for its essential role in promoting viral replication and also for its ability to promote immune escape by cleaving the host ubiquitin-like interferon-stimulated gene 15 protein (ISG15), a key regulator of the host innate immune response. (43−46) Without further studies to characterize the selectivity index of the 2-phenyl-1,2-benzoselenazol-3-one inhibitors, we cannot say if the antiviral activity observed in both the Vero E6 and Calu-3 assays and the human iPSC-derived lung organoids is due to the inhibition of Mpro, PLpro, or a combination of both. Additionally, while the mechanism of action has been evaluated for some benzoisoselenazole compounds in vitro, it remains unclear if the compounds are covalent or noncovalent modifiers intracellularly. Tandem MS/MS experiments to verify covalent modification at the active site should be completed prior to advancing these compounds as leads for clinical development. However, we believe that while E24 shows promise as a Mpro inhibitor, and the potency can be further optimized prior to establishing the selectivity and intracellular mechanism of action of this class of inhibitors.
While future studies are needed to determine toxicity and efficacy of the 2-phenyl-1,2-benzoselenazol-3-one class of compounds in animal models, these findings could potentially be relevant to the development of Mpro-targeted clinical candidates for the treatment of COVID-19.

Experimental Section

Click to copy section linkSection link copied!

In Silico Modeling of Ebselen Analogues with Schrödinger

In silico modeling of ebselen analogs was performed using the Glide docking module of the Schrödinger 11.5 modeling software suite. A crystal structure of the SARS-CoV-2 pro bound to N3 (PDB ID 6UL7) was first refined using Prime. (15,47,48) The optimized potentials for liquid simulations (OPLS) all-atom force field and the surface generalized born (SGB) continuum solution model were used to optimize and minimize the crystal structure, respectively. The docking grid was generated as a 15 Å cube centered on N3 (x = −14, y = 17.7, and z = 67.1). Ligprep was used to generate a minimized 3D structure for all antiviral compounds using the OPLS 2001 force field. Docking was performed with Glide XP. (21−23) Compounds were ranked by their docking score, and the docking poses were evaluated for interactions with key residues such as the catalytic dyad His 41 and Cys 145.

SARS-CoV-2 Main Protease Protein Expression and Purification

The GST-tagged SARS-CoV-2 main protease was expressed in Escherichia coli BL21 competent cells (New England Biolabs), which were transformed with the pET41b plasmid by heat shock and spread on a lysogeny broth (LB) kanamycin agar plate and then incubated overnight at 37 °C. Next, 2–3 colonies were picked and transferred to 5 mL of LB medium treated with kanamycin (0.5 mg/mL final concentration), then grown overnight with shaking at 37 °C. The overnight culture was then transferred to 2 L of the LB kanamycin medium and incubated at 37 °C until OD = 0.8. The culture was cooled at 4 °C for 20 min, induced with 0.5 mM isopropyl β-d-1-thiogalactopyranoside (IPTG), then grown with shaking at 16 °C. Cell pellets were collected by centrifugation (5000 g for 10 min at 4 °C), and the supernatant was discarded. The pellets were resuspended in lysis buffer (50 mM Tris-HCl pH 7.5, 500 mM NaCl, 1.0% Triton x-100, 0.5 mM EDTA, 1× complete protease inhibitor cocktail, and 1 mM β-mercaptoethanol in RNase-free water; 6 mL per gram) with DNase 1 (5 U/mL, RNase-free) and incubated at 4 °C for 1 h, then sonicated. The suspension was centrifuged at 10 000 g for 20 min, and the supernatant was transferred to a glutathione sepharose affinity resin column that had been pre-equilibrized with lysis buffer. The supernatant was incubated with the affinity resin column at 4 °C for 2 h with end-over-end rotation. After incubation, the column was washed with 5 bed volumes of lysis buffer, 3 × 5 bed volumes of wash buffer (50 mM Tris-HCl pH 7.5, 500 mM NaCl, 0.1% Triton x-100, 0.5 mM EDTA, 1× complete protease inhibitor cocktail, and 1 mM β-mercaptoethanol in RNase-free water), and 5 bed volumes of pre-elution buffer (50 mM Tris-HCl pH 7.5 and 100 mM NaCl in RNase-free water). The protease was eluted with elution buffer (50 mM Tris-HCl pH 7.5, 100 mM NaCl, and 40 mM glutathione in RNase-free water) and collected in 1 mL fractions until no further protein was collected (3–5 bed volumes total). The purity of the fractions was determined by SDS-page, and fractions of the purified protein were collected and concentrated to 2 mg/mL. The GST-tagged purified protein was incubated with TEV protease at 37 °C for 4 h to obtain the untagged SARS-CoV-2 protease. Excess GST and GST-TEV protease were removed by glutathione sepharose affinity column purification. The untagged proteases were eluted in 50 mM Tris-HCl pH 7.5 and 100 mM NaCl in RNase-free water. The purity of the fractions was determined by SDS-page, and fractions of purified protein were collected and concentrated to 5 μM. The protein solutions were transferred to a Slyde-A-Lyzer dialysis cassette (20 000 MWCO, Thermo Scientific) and dialyzed overnight at 4 °C against dialysis buffer (50 mM Tris-HCl pH 7.4, 200 mM NaCl, 1 mM EDTA, and 1 mM DTT in RNase-free water).

FRET Protease Activity Assay

The activity of the SARS-CoV-2 main proteases was determined using a fluorescence resonance energy transfer (FRET) assay adapted from Muramatsu et al. (25) The fluorogenic peptide Dabcyl-Val-Asn-Ser-Thr-Leu-Gln-Ser-Gly-Leu-Arg-Lys-EDANS was used as a substrate (AnaSpec Inc.). All reactions were performed in a black 96-well plate with 200 μL of assay buffer (50 mM Tris-HCl pH 7.4, 200 mM NaCl, 1 mM EDTA, and 1 mM DTT in RNase-free water), 25 nM SARS-CoV-2 protease, and 3 μM substrate. Ebselen analogs were screened at a concentration of 10 μM. All inhibitors were dissolved in DMSO and added to a final concentration of 0.2% DMSO. Fluorescent readings were normalized to 200 μL of assay buffer with 3 μM substrate and 10 μM inhibitor with a final concentration of 0.2% DMSO. Cleavage between the Gln and Ser residues was monitored by a BioTek Synergy plate reader with excitation at 380 nm and emission at 485 nm every 10 min for 1 h. The relative rate for wild-type protease, and each inhibitor concentration was averaged from three assay results. The relative reaction rates were normalized to the wild-type protease, and the relative activity was reported as a percentage of the wild-type protease activity in Table 1.
Additionally, dose–response curves were generated for compounds that showed significant inhibition at 10 μM using the assay conditions described above. The dose–response curve for ebselen (E01) was also generated as a positive control. Inhibitors were screened at six concentrations ranging from 0.100 to 10 μM. As before, cleavage between the Gln and Ser residues was monitored by a BioTek Synergy plate reader with excitation at 380 nm and emission at 485 nm every 10 min for 1 h. The relative rates for the wild-type protease and each inhibitor concentration were averaged from three assay results. The relative reaction rates were normalized to the wild-type protease, and dose–response curves were plotted in GraphPad Prism 6.
To determine if the ebselen analogs were covalent or rapid reversible inhibitors of SARS-CoV-2 Mpro, preincubation dilution experiments were performed. In assay buffer, 250 nM protease was incubated on ice for 30 min with 10 μM ebselen (E01), E04, E07, E19, E20, or E25. The reaction mixture was then diluted 10-fold with assay buffer, and the fluorogenic peptide substrate was added to a final concentration of 3 μM. The experiment was also repeated with assay buffer free of DTT to determine the effects of the reducing agent on the mechanism of inhibition. For each compound, the rate of product formation after preincubation and dilution was compared to the rate of product formation for Mpro with 0.2% DMSO, 1 μM compound, and 10 μM compound without dilution.

Inhibition Mechanism

The reaction velocities observed over a range of substrate (1.5, 3, 4.5, 6, 9, 15, 22.5, and 30 μM) and inhibitor concentrations (0, 0.5, 1, 5, and 10 μM) for either ebselen or E04 were globally fit to a generalized mixed-model of inhibition represented by the following equilibrium reaction scheme. This mechanism allows for inhibitor binding to both the free enzyme and the enzyme–substrate complex with varying affinity to be described by the following rate equation:
The term Ki describes the affinity of the inhibitor for free enzyme. The mechanism can be evaluated by the term α. When α approaches 1, the inhibitor is considered noncompetitive, and when α approaches infinity, the inhibitor is considered competitive.
(2)

logD Measurements

logD values were measured at pH 7.4 by the shake-flask method according to the literature. (49,50)

Antiviral Assays

Approximately 2 × 104 Vero E6 cells (ATCC) were plated in 100 μL of DMEM (Gibco) supplemented with 10% serum in each well of a 96-well flat bottom plate and allowed to adhere overnight. Cells were pretreated with 0.10–20 μM test compound in 100 μL of the same medium for 1 h, washed, and infected with SARS-CoV-2 (USA_WA01/2020 (World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch)) in 50 μL of medium at 0.01 MOI for 1.5 h. Cells were then washed, test compounds were reapplied at 0.10–20 μM in 100 μL of medium, and incubated at 37 °C in 5% CO2 for 72 h. Following incubation, 10 μL of room temperature WST8 reagent was added to each well, mixed, and incubated for an additional 3 h at 37 °C and 5% CO2. The OD at 460 nm was read on a BioTek plate reader.
For antiviral assays in Calu-3 human lung cells, approximately 1.5 × 105 Calu-3 cells (ATCC) were plated in 500 μL of EMEM (Gibco) supplemented with 10% FBS, 2 mM l-glutamine, and 1% penicillin/streptomycin in each well of 24-well flat bottom plates and allowed to adhere overnight. Cells were pretreated with 0.10–20 μM test compound in DMSO (0.2% final v/v) of the same medium for 1 h, washed, and infected with SARS-CoV-2 (USA_WA01/2020) in a reduced serum medium (2–3%) at 0.1 MOI for 1.5 h. Cells were then washed with PBS, and test compounds reapplied at 0.10–20 μM in 500 μL of the medium and incubated at 37 °C in 5% CO2 for 24 h. At 24 h postinfection, the supernatant and infected cells were collected and lysed using TRIzol, and RNA was extracted using a Direct-zol RNA Kit (Zymo) and quantified by RT-qPCR using SARS-CoV-2 N primers as previously described. (38−40) The primers are as follows: forward, CACATTGGCACCCGCAATC; reverse, GAGGAACGAGAAGAGGCTTG.

Lung Organoid Infection and Treatment with E24

Human iPSC-derived lung organoids were generated using previously published methods and characterized by the expression of ACE2, TMPRSS2, and alveolar cell epithelial markers (SFTPC, SFTPB, and HOPX). The 60D differentiated lung organoids were pretreated with 5 μM E24 for 2 h and infected with SARS-CoV-2 as described previously. (38) The SARS-CoV-2 isolate USA-WA1/2020 was obtained from BEI Resources. SARS-CoV-2 was propagated, and infectious units quantified by plaque assay using Vero E6 cells. Human iPSC-derived lung organoids, were infected with SARS-CoV-2 at MOI = 2 for 2 h at 37 °C. Then, cells were washed, and fresh medium was added. At 72 h post infection, the supernatant and infected organoids were collected and lysed using TRIzol, and RNA was extracted using a Direct-zol RNA kit (Zymo) and quantified by RT-qPCR using SARS-CoV-2 N primers.

General Procedure for the Synthesis of Compounds E01–30

A one-necked round-bottomed flask was fitted with a reflux condenser and a magnetic stirrer. The flask was charged with the corresponding SM 3 (1 mmol), followed by the addition of Se powder (5 mmol) and tBuONa (2 mmol). The reaction mixture was left to stir at 130 °C for 12 h. After the completion of reaction, the reaction mixture was diluted with excess water and extracted with ethyl acetate. The organic layer was separate and concentrated under reduced pressure. The crude material was purified by column chromatography (hexane/ethyl acetate 70:30) to yield compounds E01–30 as solids.

Compound Characterization

The Supporting Information provides a detailed description of the solvent purification and general methods for synthesis and purification of each analogue. All compounds were purified by column chromatography and characterized by 1H and 13C NMR and HRMS. Purity was determined by HPLC at >90% for all compounds, and spectra can be found in the Supporting Information.

2-Phenylbenzo[d][1,2]selenazol-3(2H)-one (E01)

1H NMR (600 MHz, CDCl3) δ 8.17 (d, J = 7.8 Hz, 1H), 7.72 (t, J = 7.8 Hz, 1H), 7.69 (dd, J = 8.4, 2.8 Hz, 3H), 7.52 (td, J = 6.2, 1.3 Hz, 1H), 7.48 (t, J = 8.4 Hz, 2H), 7.33 (dd, J = 14.1, 6.6 Hz, 1H). 13C NMR (151 MHz, CDCl3) δ 165.8, 139.2, 137.7, 132.6, 132.6, 129.5, 129.4, 127.6, 126.8, 126.6, 125.5, 125.5, 123.8. HRMS (ESI): m/z calculated for C13H9NOSe 274.9849, found 275.9927 [M + H]+. Purity >99%.

2-(4-Fluorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E02)

1H NMR (600 MHz, CDCl3) δ 8.11 (d, J = 7.8 Hz, 1H), 7.68–7.64 (m, 2H), 7.57 (dd, J = 8.6, 4.8 Hz, 2H), 7.48 (t, J = 6.6 Hz, 1H), 7.12 (t, J = 8.5 Hz, 2H). 13C NMR (151 MHz, CDCl3) δ 165.9, 161.9, 160.3, 137.7, 134.9, 132.7, 129.5, 127.7, 127.1, 126.7, 123.9, 116.3, 116.2. HRMS (ESI): m/z calculated for C13H8FNOSe 292.9755, found 293.9831[M + H]+. Purity >99%.

2-(2-(Methylthio)phenyl)benzo[d][1,2]selenazol-3(2H)-one (E03)

1H NMR (600 MHz, CDCl3) δ 8.19 (d, J = 7.8 Hz, 1H), 7.75–7.68 (m, 2H), 7.52 (t, J = 7.3 Hz, 1H), 7.47–7.41 (m, 2H), 7.37 (d, J = 8.0 Hz, 1H), 7.28 (t, J = 7.6 Hz, 1H), 2.48 (s, 3H). 13C NMR (151 MHz, CDCl3) δ 165.9, 138.8, 138.5, 134.8, 132.0, 129.4, 129.0, 129.0, 125.8, 125.7, 125.5, 125.0, 123.5, 14.6. HRMS (ESI): m/z calculated for C14H11NOSSe 320.9727, found 321.9805 [M + H]+. Purity >98%.

2-(4-(Trifluoromethyl)phenyl)benzo[d][1,2]selenazol-3(2H)-one (E04)

1H NMR (600 MHz, CDCl3) δ 8.13 (d, J = 7.8 Hz, 1H), 7.81 (d, J = 8.4 Hz, 2H), 7.70–7.67 (m, 4H), 7.49 (ddd, J = 8.0, 5.6, 2.6 Hz, 1H). 13C NMR (151 MHz, CDCl3) δ 167.0, 137.3, 133.2, 133.2, 129.7, 129.7, 127.4, 127.0, 127.0, 126.7, 126.6, 124.9, 123.9, 123.9. HRMS (ESI): m/z calculated for C14H8F3NOSe 342.9723, found 343.9798 [M + H]+. Purity >98%.

2-(3,4-Dimethylphenyl)benzo[d][1,2]selenazol-3(2H)-one (E05)

1H NMR (600 MHz, CDCl3) δ 8.16 (d, J = 7.6 Hz, 1H), 7.69 (dt, J = 7.9, 7.3 Hz, 2H), 7.51 (t, J = 7.3 Hz, 1H), 7.45 (s, 1H), 7.36 (d, J = 8.0 Hz, 1H), 7.23 (d, J = 8.0 Hz, 1H), 2.35 (s, 3H), 2.32 (s, 3H). 13C NMR (151 MHz, CDCl3) δ 165.8, 137.8, 137.8, 136.6, 135.8, 132.4, 130.4, 129.4, 127.6, 126.8, 126.5, 123.8, 123.1, 19.9, 19.5. HRMS (ESI): m/z calculated for C15H13NOSe 303.0162, found 304.0240 [M + H]+. Purity >98%.

2-(4-Fluorobenzyl)benzo[d][1,2]selenazol-3(2H)-one (E06)

1H NMR (600 MHz, CDCl3) δ 8.13 (d, J = 7.8 Hz, 1H), 7.63 (t, J = 6.4 Hz, 2H), 7.48 (td, J = 8.0, 4.2 Hz, 1H), 7.39 (dd, J = 8.0, 5.5 Hz, 2H), 7.10 (t, J = 8.5 Hz, 2H), 5.03 (s, 2H). 13C NMR (151 MHz, CDCl3) δ 167.3, 163.6, 161.0, 138.1, 133.2, 132.3, 130.5, 130.4, 129.1, 126.5, 124.2, 116.09, 115.9, 48.0. HRMS (ESI): m/z calculated for C14H10FNOSe 306.9912, found 307.9988 [M + H]+. Purity >99%.

2-(2,4-Dimethoxybenzyl)benzo[d][1,2]selenazol-3(2H)-one (E07)

1H NMR (600 MHz, CDCl3) δ 8.04 (d, J = 7.9 Hz, 1H), 7.54–7.51 (m, 2H), 7.39 (qd, J = 5.8, 3.0 Hz, 1H), 7.32 (d, J = 8.2 Hz, 1H), 6.46 (dt, J = 8.2, 2.2 Hz, 2H), 4.96 (s, 2H), 3.87 (d, J = 6.7 Hz, 3H), 3.81 (s, 3H). 13C NMR (151 MHz, CDCl3) δ 167.0, 161.3, 158.7, 138.5, 131.9, 131.7, 128.7, 127.9, 126.0, 123.8, 118.2, 104.4, 98.6, 55.5, 55.5, 43.2. HRMS (ESI): m/z calculated for C16H15NO3Se 349.0217, found 350.0296 [M + H]+. Purity >98%.

2-(2-Fluorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E08)

1H NMR (600 MHz, CDCl3) δ 8.19 (d, J = 7.8 Hz, 1H), 7.72 (d, J = 8.2 Hz, 2H), 7.55 (ddd, J = 8.0, 5.6, 2.6 Hz, 2H), 7.45–7.39 (m, 1H), 7.30–7.23 (m, 2H). 13C NMR (151 MHz, CDCl3) δ 166.6, 159.2, 157.5, −139.1, 132.8, 130.3, 130.1, 130.0, 129.6, 126.6, 124.8, 124.1, 117.1. HRMS (ESI): m/z calculated for C13H8FNOSe 292.9755, found 293.9834 [M + H]+. Purity >99%.

2-(4-(Trifluoromethyl)benzyl)benzo[d][1,2]selenazol-3(2H)-one (E09)

1H NMR (600 MHz, CDCl3) δ 8.15 (d, J = 7.8 Hz, 1H), 7.69–7.65 (m, 4H), 7.51 (t, J = 8.1 Hz, 3H), 5.13 (s, 2H). 13C NMR (151 MHz, CDCl3) δ 167.5, 137.8, 132.5, 129.3, 129.3, 128.7, 128.7, 126.9, 126.6, 126.6, 126.0, 126.0, 124.2, 124.2, 48.1. HRMS (ESI): m/z calculated for C15H10F3NOSe 356.9880, found 357.9957 [M + H]+. Purity >99%.

2-(4-Fluoro-2-(trifluoromethyl)phenyl)benzo[d][1,2]selenazol-3(2H)-one (E10)

1H NMR (600 MHz, CDCl3) δ 8.28 (d, J = 3.5 Hz, 1H), 8.05 (s, 1H), 7.79 (d, J = 7.8 Hz, 1H), 7.64 (d, J = 7.5 Hz, 1H), 7.51–7.47 (m, 1H), 7.44–7.40 (m, 2H). 13C NMR (151 MHz, CDCl3) δ 166.4, 160.1, 135.2, 133.7, 132.3, 132.2, 130.9, 127.7, 127.3, 127.1, 126.6, 120.1, 119.9, 113.8. HRMS (ESI): m/z calculated for C14H7F4NOSe 360.9629, found 361.9704 [M + H]+. Purity >90%.

2-(2,6-Difluorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E11)

1H NMR (600 MHz, CDCl3) δ 8.15 (d, J = 7.7 Hz, 1H), 7.82 (d, J = 7.4 Hz, 1H), 7.74 (s, 1H), 7.472 (d, J = 7.0 Hz, 1H), 7.38 (m, 1H), 7.06 (m, 2H). 13C NMR (151 MHz, CDCl3) δ 169.7, 166.7, 139.6, 133.5, 133.0, 132.2, 130.4, 129.7, 129.1, 126.6, 124.2, 112.5, 112.3. HRMS (ESI): m/z calculated for C13H7F2NOSe 310.9661, found 311.9737 [M + H]+. Purity >90%.

2-(4-Methoxybenzyl)benzo[d][1,2]selenazol-3(2H)-one (E12)

1H NMR (600 MHz, CDCl3) δ 8.12 (d, J = 7.8 Hz, 1H), 7.61 (d, J = 3.6 Hz, 2H), 7.50–7.45 (m, 1H), 7.35 (dt, J = 8.6, 2.1 Hz, 2H), 6.94 (dt, J = 8.6, 1.9 Hz, 2H), 5.00 (s, 2H), 3.86 (d, J = 6.4 Hz, 3H). 13C NMR (151 MHz, CDCl3) δ 167.1, 159.7, 138.2, 131.9, 130.2, 130.2, 129.4, 128.9, 127.8, 126.2, 124.0, 114.2, 114.2, 55.4, 48.3. HRMS (ESI): m/z calculated for C15H13NO2Se 319.0112, found 320.0190 [M + H]+. Purity >99%.

2-(4-Chloro-2-(trifluoromethoxy)phenyl)benzo[d][1,2]selenazol-3(2H)-one (E13)

1H NMR (600 MHz, CDCl3) δ 8.54 (d, J = 8.8 Hz, 1H), 8.18 (s, 1H), 7.78 (d, J = 7.8 Hz, 1H), 7.64 (dd, J = 7.6, 1.1 Hz, 1H), 7.48 (td, J = 7.7, 1.4 Hz, 1H), 7.42 (dd, J = 7.5, 0.6 Hz, 1H), 7.37–7.35 (m, 1H). 13C NMR (151 MHz, CDCl3) δ 166.0, 138.6, 135.4, 133.3, 132.4, 132.3, 129.6, 129.0, 127.8, 127.4, 127.3, 122.9, 120.8, 46.0. HRMS (ESI): m/z calculated for C14H7ClF3NO2Se 392.9283, found 393.9352 [M + H]+. Purity >95%.

2-(5-Chloro-2-methoxyphenyl)benzo[d][1,2]selenazol-3(2H)-one (E14)

1H NMR (600 MHz, CDCl3) δ 8.17 (d, J = 7.4 Hz, 1H), 7.70 (t, J = 5.9 Hz, 2H), 7.56 (d, J = 2.4 Hz, 1H), 7.53–7.49 (m, 1H), 7.39–7.35 (m, 1H), 6.99 (d, J = 8.9 Hz, 1H), 3.89 (s, 3H). 13C NMR (151 MHz, CDCl3) δ 166.8, 154.1, 139.2, 132.6, 129.9, 129.5, 129.5, 127.9, 126.4, 126.1, 125.5, 123.9, 113.4, 56.3. HRMS (ESI): m/z calculated for C14H10ClNO2Se 338.9565, found 339.9641 [M + H]+. Purity >93%.

2-(4-Chlorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E15)

1H NMR (600 MHz, (CD3)2SO) δ 8.08 (d, J = 8.0 Hz, 1H), 7.91 (dd, J = 7.8, 1.4 Hz, 2H), 7.68 (t, J = 7.7 Hz, 1H), 7.48 (t, J = 7.4 Hz, 1H), 7.36 (t, J = 8.2 Hz, 1H), 7.31 (t, J = 2.3 Hz, 1H), 7.16 (dd, J = 7.8, 1.2 Hz, 1H), 6.86 (dd, J = 8.3, 2.5 Hz, 1H), 3.79 (s, 3H). 13C NMR (151 MHz, (CD3)2SO) δ 165.0, 159.7, 140.9, 138.9, 132.3, 130.0, 128.6, 128.0, 126.3, 125.8, 116.7, 111.4, 110.4, 55.3. HRMS (ESI): m/z calculated for C14H11NO2Se 304.9955, found 309.3315 [M + H]+. Purity >90%.

2-(5-Fluoro-2-methoxyphenyl)benzo[d][1,2]selenazol-3(2H)-one (E16)

1H NMR (600 MHz, CDCl3) δ 8.12 (d, J = 7.7 Hz, 1H), 7.66–7.63 (m, 2H), 7.45 (ddd, J = 8.1, 5.7, 2.4 Hz, 1H), 7.33 (dd, J = 8.7, 2.8 Hz, 1H), 7.05 (td, J = 8.5, 3.1 Hz, 1H), 6.94 (dd, J = 9.0, 4.8 Hz, 1H), 3.84 (s, 3H). 13C NMR (151 MHz, CDCl3) δ 166.3, 156.6, 155.0, 151.9, 140.8, 132.7, 132.5, 128.2, 127.4 126.2, 116.7, 115.3, 114.0, 56.6. HRMS (ESI): m/z calculated for C14H10FNO2Se 322.9861, found 323.9937 [M + H]+. Purity >99%.

2-(4-Fluoro-2-methoxyphenyl)benzo[d][1,2]selenazol-3(2H)-one (E17)

1H NMR (600 MHz, CD3OD) δ 8.07 (d, J = 8.0 Hz, 1H), 7.86 (d, J = 7.3 Hz, 1H), 7.67 (ddd, J = 8.3, 7.2, 1.5 Hz, 1H), 7.46 (td, J = 7.5, 1.0 Hz, 1H), 7.37 (dd, J = 8.6, 6.4 Hz, 1H), 7.11 (dd, J = 11.0, 2.6 Hz, 1H), 6.85 (td, J = 8.4, 2.7 Hz, 1H), 3.78 (s, 3H). 13C NMR (151 MHz, CD3OD) δ 166.3, 140.9, 132.5, 131.5, 128.3, 127.6, 126.4, 126.4, 107.3, 107.1, 101.4, 101.3, 56.7, 36.3. HRMS (ESI): m/z calculated for C14H10FNO2Se 322.9861, found 323.9938 [M + H]+. Purity >95%.

2-(4-Ethylphenyl)benzo[d][1,2]selenazol-3(2H)-one (E18)

1H NMR (600 MHz, CDCl3) δ 8.12 (d, J = 7.7 Hz, 1H), 7.67–7.63 (m, 2H), 7.52 (dt, J = 8.2, 2.1 Hz, 2H), 7.47 (ddd, J = 7.9, 6.4, 1.8 Hz, 1H), 7.26 (d, J = 8.2 Hz, 2H), 2.67 (q, J = 7.6 Hz, 2H), 1.26 (t, J = 7.6 Hz, 3H). 13C NMR (151 MHz, CDCl3) δ 165.8, 143.2, 137.8, 136.6, 132.5, 129.4, 128.8, 128.8, 127.6, 126.5, 125.6, 125.6, 123.8, 28.5, 15.6. HRMS (ESI): m/z calculated for C15H13NOSe 303.0162, found 304.0240 [M + H]+. Purity >99%.

2-(3-Chlorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E19)

1H NMR (600 MHz, CDCl3 + CD3OD) δ 7.92 (d, J = 7.6 Hz, 1H), 7.69 (d, J = 8.0 Hz, 1H), 7.53 (dt, J = 8.2, 1.6 Hz, 2H), 7.35 (dd, J = 8.0, 1.1 Hz, 1H), 7.34 (t, J = 7.0 Hz, 1H), 7.24 (d, J = 8.0 Hz, 1H), 7.14 (ddd, J = 7.2, 4.0, 3.2 Hz, 1H). 13C NMR (151 MHz, CDCl3) δ 166.4, 140.1, 138.6, 134.7, 132.7, 130.2, 128.8, 127.3, 126.8, 126.5, 125.5, 124.3, 123.5. HRMS (ESI): m/z calculated for C13H8ClNOSe 308.9460, found 309.9536 [M + H]+. Purity >98%.

2-(3,5-Dichlorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E20)

1H NMR (600 MHz, (CD3)2SO) δ 8.11–8.06 (m, 2H), 7.91 (d, J = 2.2 Hz, 1H), 7.69 (t, J = 7.6 Hz, 2H), 7.58 (dd, J = 8.3, 2.7 Hz, 1H), 7.48 (t, J = 7.5 Hz, 1H). 13C NMR (151 MHz, (CD3)2SO) δ 166.4, 140.1, 138.6, 134.7, 132.7, 130.2, 128.8, 127.3, 126.8, 126.5, 125.5, 124.3, 123.54. HRMS (ESI): m/z calculated for C13H7Cl2NOSe 342.9070, found 344.2213 [M + H]+. Purity >95%.

2-(4-Bromophenyl)benzo[d][1,2]selenazol-3(2H)-one (E21)

1H NMR (600 MHz, CD3OD + CDCl3) δ 8.12 (d, J = 7.9 Hz, 1H), 7.90 (dd, J = 7.8, 1.4 Hz, 1H), 7.69 (ddd, J = 8.3, 7.2, 1.4, Hz, 1H), 7.64 (s, 4H), 7.48 (td, J = 7.5, 1.1 Hz, 1H). 13C NMR (151 MHz, CDCl3) δ 165.1, 139.3, 138.6, 132.4, 132.0, 132.0, 128.5, 128.0, 126.4, 126.4, 126.4, 126.1, 117.8. HRMS (ESI): m/z calculated for C13H8BrNOSe 352.8954, found 353.9028 [M + H]+. Purity >95%.

2-(p-Tolyl)benzo[d][1,2]selenazol-3(2H)-one (E22)

1H NMR (600 MHz,(CD3)2SO) δ 8.11 (d, J = 8.1 Hz, 1H), 7.789 (dd, J = 7.8, 1.4 Hz, 1H), 7.67 (t, J = 7.4 Hz, 1H), 7.51 (d, J = 8.1 Hz, 1H), 7.47 (t, J = 7.2 Hz, 2H), 7.25 (d, J = 8.1 Hz, 2H), 2.32 (s, 3H). 13C NMR (151 MHz, (CD3)2SO) δ 164.9, 138.8, 137.1, 135.3, 132.1, 129.6, 129.6, 128.6, 127.9, 126.2, 125.9, 124.7, 124.7, 20.6. HRMS (ESI): m/z calculated for C14H11NOSe 289.0006, found 290.0087 [M + H]+. Purity >96%.

2-(o-Tolyl)benzo[d][1,2]selenazol-3(2H)-one (E23)

1H NMR (600 MHz, CDCl3) δ 8.18 (d, J = 7.8 Hz, 1H), 7.74 (t, J = 7.2 Hz, 1H), 7.70 (t, J = 7.5 Hz, 1H), 7.53 (t, J = 7.5 Hz, 1H), 7.38–7.34 (m, 3H), 7.32 (d, J = 5.5 Hz, 1H), 2.29 (s, 3H). 13C NMR (151 MHz, CDCl3) δ 166.2, 139.1, 139.1, 137.7, 136.8, 132.4, 131.3, 129.4, 129.2, 129.1, 126.9, 126.5, 124.2, 18.3. HRMS (ESI): m/z calculated for C14H11NOSe 289.0006, found 290.0086 [M + H]+. Purity >98%.

2-(3-(Trifluoromethyl)phenyl)benzo[d][1,2]selenazol-3(2H)-one (E24)

1H NMR (600 MHz, CDCl3) δ 8.13 (d, J = 7.9 Hz, 1H), 7.95 (s, 1H), 7.84 (d, J = 7.8 Hz, 1H), 7.68 (d, J = 4.2 Hz, 2H), 7.56 (t, J = 7.8 Hz, 1H), 7.53 (d, J = 7.9 Hz, 1H), 7.49 (ddd, J = 8.0, 4.8, 3.4 Hz, 1H). 13C NMR (151 MHz, CDCl3) δ 166.0, 139.9, 137.4, 133.1, 130.1, 129.7, 128.6, 127.0, 127.0, 124.0, 123.4, 123.3, 122.2, 122.2. HRMS (ESI): m/z calculated for C14H8F3NOSe 342.9723, found 343.9800 [M + H]+. Purity >99%.

3-(3-Oxobenzo[d][1,2]selenazol-2(3H)-yl)benzonitrile (E25)

1H NMR (600 MHz, CDCl3) δ 8.15 (dt, J = 7.8, 1.1 Hz,1H), 7.78 (dd, J = 7.8, 1.4 Hz, 1H), 7.73–7.65 (m, 3H), 7.55 (dd, J = 8.1, 1.1 Hz, 1H), 7.50 (tdd, J = 6.1, 4.6, 2.9 Hz, 2H). 13C NMR (151 MHz, CDCl3) δ 166.5, 140.9, 138.6, 134.0, 133.8, 133.2, 129.9, 129.9, 128.7, 126.9, 125.4, 124.1, 116.1, 113.4. HRMS (ESI): m/z calculated for C14H8N2OSe 299.9802, found 300.9880 [M + H]+. Purity >98%.

2-(3-Methoxyphenyl)benzo[d][1,2]selenazol-3(2H)-one (E26)

1H NMR (600 MHz, CDCl3) δ 8.11 (d, J = 7.8 Hz, 1H), 7.65 (ddd, J = 10.0, 9.1, 4.4 Hz, 2H), 7.46 (ddd, J = 8.0, 6.8, 1.4 Hz, 1H), 7.360–7.28 (m, 2H), 7.16 (ddd, J = 7.9, 2.1, 0.9 Hz,1H), 6.83 (dd, J = 8.1, 2.3 Hz, 1H), 3.84 (s, 3H). 13C NMR (151 MHz, CDCl3) δ 165.8, 160.3, 140.3, 137.82, 132.7, 130.1, 129.5, 127.8, 126.7, 123.8, 117.6, 113.0, 111.0, 55.6. HRMS (ESI): m/z calculated for C14H11NO2Se 304.9955, found 306.0034 [M + H]+. Purity >99%.

2-(4-Methoxyphenyl)benzo[d][1,2]selenazol-3(2H)-one (E27)

1H NMR (600 MHz, CDCl3) δ 8.10 (d, J = 7.7 Hz, 1H), 7.64 (ddd, J = 12.3, 9.0, 4.5 Hz, 2H), 7.49 (d, J = 2. Hz, 1 H), 7.47 (td, J = 6.87, 1.1 Hz, 2H), 6.95 (dt, J = 8.9, 2.2 Hz, 2H), 3.83 (s, 3H). 13C NMR (151 MHz, CDCl3) δ 166.1, 158.5, 138.0, 132.4, 131.7, 129.3, 127.5, 127.3, 126.5, 123.9, 123.9, 114.5 114.5, 55.4. HRMS (ESI): m/z calculated for C14H11NO2Se 304.9955, found 306.0035 [M + H]+. Purity >99%.

2-Heptylbenzo[d][1,2]selenazol-3(2H)-one (E28)

1H NMR (600 MHz, CDCl3) δ 8.03 (d, J = 7.8 Hz, 1H), 7.63 (d, J = 7.9 Hz, 1H), 7.57 (td, J = 7.6, 1.4 Hz, 1H), 7.41 (t, J = 7.4 Hz, 1H), 3.84 (t, J = 7.3 Hz, 2H), 1.75–68 (m, 2H), 1.42–1.21 (m, 8H), 0.87 (t, J = 7.0 Hz, 3H). 13C NMR (151 MHz, CDCl3) δ 167.1, 137.6, 131.9, 128.9, 127.6, 126.3, 124.0, 44.8, 31.9, 30.4, 28.9, 26.3, 22.8, 14.1. HRMS (ESI): m/z calculated for C14H19NOSe 297.0632, found 298.0711 [M + H]+. Purity >98%.

2-Octylbenzo[d][1,2]selenazol-3(2H)-one (E29)

1H NMR (600 MHz, CDCl3) δ 8.04 (d, J = 7.8 Hz, 1H), 7.63 (dt, J = 8.0, 0.9 Hz, 1H), 7.57 (td, J = 7.1, 1.3 Hz, 1H), 7.42 (t, J = 7.2 Hz, 1H), 3.851 (t, J = 7.15 Hz, 2H), 1.72 (m, J = 2H), 1.43–1.27 (m, 4H), 1.29–1.25 (m, 2H), 1.27–1.20 (m, 4 H), 0.87 (t, J = 7.0 Hz, 3H). 13C NMR (151 MHz, CDCl3) δ 167.1, 137.7, 131.8, 128.9, 127.7, 126.2, 123.8, 44.9, 31.8, 30.7, 29.3, 29.1, 26.6, 22.6, 14.1. HRMS (ESI): m/z calculated for C15H21NOSe 311.0788, found 312.0868 [M + H]+. Purity >98%.

2-Pentylbenzo[d][1,2]selenazol-3(2H)-one (E30)

1H NMR (600 MHz, CDCl3) δ 8.04 (d, J = 7.8 Hz, 1H), 7.63 (d, J = 7.9 Hz, 1H), 7.57 (td, J = 7.7, 1.1 Hz, 1H), 7.42 (t, J = 7.5 Hz, 1H), 3.84 (t, J = 7.3 Hz, 2H), 1.72 (m, 2H), 1.37 (dd, J = 8.7, 5.5 Hz, 4H), 0.90 (t, J = 6.7 Hz, 3H). 13C NMR (151 MHz, CDCl3) δ 167.3, 137.7, 131.9, 128.9, 127.7, 126.2, 124.0, 44.9, 30.3, 28.8, 22.4, 14.0. HRMS (ESI): m/z calculated for C12H15NOSe 269.0319, found 270.0398 [M + H]+. Purity >99%.

Supporting Information

Click to copy section linkSection link copied!

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.1c00566.

  • Molecular formula strings(CSV)

  • Expanded synthetic methods and spectra for E01–30, structures and reported activities for known SARS-CoV-2 main protease inhibitors, docking poses, and individual antiviral dose–response curves for select compounds

    (PDF)

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

Click to copy section linkSection link copied!

  • Corresponding Author
    • Tariq M. Rana - Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesDivision of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesOrcidhttps://orcid.org/0000-0001-9558-5766 Email: [email protected]
  • Authors
    • Sarah Huff - Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    • Indrasena Reddy Kummetha - Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    • Shashi Kant Tiwari - Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    • Matthew B. Huante - Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United States
    • Alex E. Clark - Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    • Shaobo Wang - Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    • William Bray - Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    • Davey Smith - Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United States
    • Aaron F. Carlin - Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesOrcidhttps://orcid.org/0000-0002-1669-8066
    • Mark Endsley - Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United States
  • Author Contributions

    These authors contributed equally to this work. S.H. and I.R.K. designed and performed the experiments, analyzed the data, and wrote the manuscript; S.K.T., M.B.H., A.E.C., S.W., W.B., and M.E. performed experiments and analysis; D.S. provided resources; A.C. gave advice in experimental plans and performed experiments; T.M.R. conceived of and planned the project and participated in experimental design, data analysis, data interpretation, and manuscript writing.

  • Notes
    The authors declare the following competing financial interest(s): T.M.R. is a founder of ViRx Pharmaceuticals and has an equity interest in the company. The terms of this arrangement have been reviewed and approved by the University of California San Diego in accordance with its conflict of interest policies.

Acknowledgments

Click to copy section linkSection link copied!

We thank the Rao Zihe and Yang Haitao laboratories of ShanghaiTech University for providing the coordinates of the SARS-CoV-2 Mpro crystal structure (PDB ID 6UL7). We thank members of the Rana lab for helpful discussions and advice. This work was supported by a T32 fellowship from the National Cancer Institute of the National Institutes of Health (award no. T32CA121938 to S.H.), a Career Award for Medical Scientists from the Burroughs Wellcome Fund (USA), a grant from the National Institutes of Health USA (K08 AI130381) to A.F.C., the John and Mary Tu Foundation (USA), a CIRM award to T.M.R., and in part by grants from the National Institutes of Health (USA) (CA177322, DA039562, DA049524, and AI125103).

ABBREVIATONS

Click to copy section linkSection link copied!

ACE2

angiotensin-converting enzyme 2

COVID-19

coronavirus disease 2019

iPSC

induced pluripotent stem cells

ISG15

interferon-stimulated gene 15

MOI

multiplicity of infection

Mpro

main protease

nsp1

nonstructural protein 1

nsp2

nonstructural protein 2

nsp3

nonstructural protein 3

OPLS

optimized potentials for liquid simulations

PLpro

papain-like protease

RT-qPCR

real-time quantitative reverse transcription PCR

RdRp

RNA-dependent RNA polymerase

SARS-CoV-2

severe acute respiratory syndrome coronavirus 2

SGB

surface generalized born

(+)ssRNA

positive-sense single-stranded RNA

TMPRSS2

transmembrane protease serine 2

References

Click to copy section linkSection link copied!

This article references 50 other publications.

  1. 1
    Li, F.; Li, W.; Farzan, M.; Harrison, S. C. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science 2005, 309 (5742), 18641868,  DOI: 10.1126/science.1116480
  2. 2
    Phan, T. Novel coronavirus: From discovery to clinical diagnostics. Infect., Genet. Evol. 2020, 79, 104211,  DOI: 10.1016/j.meegid.2020.104211
  3. 3
    Pillaiyar, T.; Manickam, M.; Namasivayam, V.; Hayashi, Y.; Jung, S. H. An Overview of Severe Acute Respiratory Syndrome-Coronavirus (SARS-CoV) 3CL Protease Inhibitors: Peptidomimetics and Small Molecule Chemotherapy. J. Med. Chem. 2016, 59 (14), 6595628,  DOI: 10.1021/acs.jmedchem.5b01461
  4. 4
    Ziebuhr, J.; Snijder, E. J.; Gorbalenya, A. E. Virus-encoded proteinases and proteolytic processing in the Nidovirales. J. Gen. Virol. 2000, 81 (4), 853879,  DOI: 10.1099/0022-1317-81-4-853
  5. 5
    Báez-Santos, Y. M.; St. John, S. E.; Mesecar, A. D. The SARS-coronavirus papain-like protease: Structure, function and inhibition by designed antiviral compounds. Antiviral Res. 2015, 115, 2138,  DOI: 10.1016/j.antiviral.2014.12.015
  6. 6
    Lee, T. W.; Cherney, M. M.; Huitema, C.; Liu, J.; James, K. E.; Powers, J. C.; Eltis, L. D.; James, M. N. Crystal structures of the main peptidase from the SARS coronavirus inhibited by a substrate-like aza-peptide epoxide. J. Mol. Biol. 2005, 353 (5), 11371151,  DOI: 10.1016/j.jmb.2005.09.004
  7. 7
    Xu, X.; Liu, Y.; Weiss, S.; Arnold, E.; Sarafianos, S. G.; Ding, J. Molecular model of SARS coronavirus polymerase: implications for biochemical functions and drug design. Nucleic Acids Res. 2003, 31 (24), 71177130,  DOI: 10.1093/nar/gkg916
  8. 8
    De Clercq, E.; Li, G. Approved Antiviral Drugs over the Past 50 Years. Clin. Microbiol. Rev. 2016, 29 (3), 695747,  DOI: 10.1128/CMR.00102-15
  9. 9
    Kanters, S.; Socias, M. E.; Paton, N. I.; Vitoria, M.; Doherty, M.; Ayers, D.; Popoff, E.; Chan, K.; Cooper, D. A.; Wiens, M. O.; Calmy, A.; Ford, N.; Nsanzimana, S.; Mills, E. J. Comparative efficacy and safety of second-line antiretroviral therapy for treatment of HIV/AIDS: A systematic review and network meta-analysis. Lancet HIV 2017, 4 (10), e433e441,  DOI: 10.1016/S2352-3018(17)30109-1
  10. 10
    Deeks, E. D. Darunavir: a review of its use in the management of HIV-1 infection. Drugs 2014, 74 (1), 99125,  DOI: 10.1007/s40265-013-0159-3
  11. 11
    la Porte, C. J. Saquinavir, the pioneer antiretroviral protease inhibitor. Expert Opin. Drug Metab. Toxicol. 2009, 5 (10), 13131322,  DOI: 10.1517/17425250903273160
  12. 12
    Heo, Y. A.; Deeks, E. D. Sofosbuvir/Velpatasvir/Voxilaprevir: A Review in Chronic Hepatitis C. Drugs 2018, 78 (5), 577587,  DOI: 10.1007/s40265-018-0895-5
  13. 13
    Sanford, M. Simeprevir: a review of its use in patients with chronic hepatitis C virus infection. Drugs 2015, 75 (2), 183196,  DOI: 10.1007/s40265-014-0341-2
  14. 14
    Chopp, S.; Vanderwall, R.; Hult, A.; Klepser, M. Simeprevir and sofosbuvir for treatment of hepatitis C infection. Am. J. Health-Syst. Pharm. 2015, 72 (17), 14451455,  DOI: 10.2146/ajhp140290
  15. 15
    Jin, Z.; Du, X.; Xu, Y.; Deng, Y.; Liu, M.; Zhao, Y.; Zhang, B.; Li, X.; Zhang, L.; Peng, C.; Duan, Y.; Yu, J.; Wang, L.; Yang, K.; Liu, F.; Jiang, R.; Yang, X.; You, T.; Liu, X.; Yang, X.; Bai, F.; Liu, H.; Liu, X.; Guddat, L. W.; Xu, W.; Xiao, G.; Qin, C.; Shi, Z.; Jiang, H.; Rao, Z.; Yang, H. Structure of M(pro) from COVID-19 virus and discovery of its inhibitors. Nature 2020, 582, 289293,  DOI: 10.1038/s41586-020-2223-y
  16. 16
    Dai, W.; Zhang, B.; Jiang, X. M.; Su, H.; Li, J.; Zhao, Y.; Xie, X.; Jin, Z.; Peng, J.; Liu, F.; Li, C.; Li, Y.; Bai, F.; Wang, H.; Cheng, X.; Cen, X.; Hu, S.; Yang, X.; Wang, J.; Liu, X.; Xiao, G.; Jiang, H.; Rao, Z.; Zhang, L. K.; Xu, Y.; Yang, H.; Liu, H. Structure-based design of antiviral drug candidates targeting the SARS-CoV-2 main protease. Science 2020, 368 (6497), 13311335,  DOI: 10.1126/science.abb4489
  17. 17
    Vuong, W.; Khan, M. B.; Fischer, C.; Arutyunova, E.; Lamer, T.; Shields, J.; Saffran, H. A.; McKay, R. T.; van Belkum, M. J.; Joyce, M. A.; Young, H. S.; Tyrrell, D. L.; Vederas, J. C.; Lemieux, M. J. Feline coronavirus drug inhibits the main protease of SARS-CoV-2 and blocks virus replication. Nat. Commun. 2020, 11 (1), 4282,  DOI: 10.1038/s41467-020-18096-2
  18. 18
    Zhang, L.; Lin, D.; Sun, X.; Curth, U.; Drosten, C.; Sauerhering, L.; Becker, S.; Rox, K.; Hilgenfeld, R. Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved alpha-ketoamide inhibitors. Science 2020, 368 (6489), 409412,  DOI: 10.1126/science.abb3405
  19. 19
    Jin, Z.; Zhao, Y.; Sun, Y.; Zhang, B.; Wang, H.; Wu, Y.; Zhu, Y.; Zhu, C.; Hu, T.; Du, X.; Duan, Y.; Yu, J.; Yang, X.; Yang, X.; Yang, K.; Liu, X.; Guddat, L. W.; Xiao, G.; Zhang, L.; Yang, H.; Rao, Z. Structural basis for the inhibition of SARS-CoV-2 main protease by antineoplastic drug carmofur. Nat. Struct. Mol. Biol. 2020, 27 (6), 529532,  DOI: 10.1038/s41594-020-0440-6
  20. 20
    Fu, L.; Ye, F.; Feng, Y.; Yu, F.; Wang, Q.; Wu, Y.; Zhao, C.; Sun, H.; Huang, B.; Niu, P.; Song, H.; Shi, Y.; Li, X.; Tan, W.; Qi, J.; Gao, G. F. Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main protease. Nat. Commun. 2020, 11 (1), 4417,  DOI: 10.1038/s41467-020-18233-x
  21. 21
    Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S. Glide: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J. Med. Chem. 2004, 47 (7), 17391749,  DOI: 10.1021/jm0306430
  22. 22
    Halgren, T. A.; Murphy, R. B.; Friesner, R. A.; Beard, H. S.; Frye, L. L.; Pollard, W. T.; Banks, J. L. Glide: a new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening. J. Med. Chem. 2004, 47 (7), 17501759,  DOI: 10.1021/jm030644s
  23. 23
    Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren, T. A.; Sanschagrin, P. C.; Mainz, D. T. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes. J. Med. Chem. 2006, 49 (21), 61776196,  DOI: 10.1021/jm051256o
  24. 24
    Amporndanai, K.; Meng, X.; Shang, W.; Jin, Z.; Rogers, M.; Zhao, Y.; Rao, Z.; Liu, Z. J.; Yang, H.; Zhang, L.; O’Neill, P. M.; Samar Hasnain, S. Inhibition mechanism of SARS-CoV-2 main protease by ebselen and its derivatives. Nat. Commun. 2021, 12 (1), 3061,  DOI: 10.1038/s41467-021-23313-7
  25. 25
    Muramatsu, T.; Takemoto, C.; Kim, Y. T.; Wang, H.; Nishii, W.; Terada, T.; Shirouzu, M.; Yokoyama, S. SARS-CoV 3CL protease cleaves its C-terminal autoprocessing site by novel subsite cooperativity. Proc. Natl. Acad. Sci. U. S. A. 2016, 113 (46), 1299713002,  DOI: 10.1073/pnas.1601327113
  26. 26
    Sarma, B. K.; Mugesh, G. Antioxidant activity of the anti-inflammatory compound ebselen: a reversible cyclization pathway via selenenic and seleninic acid intermediates. Chem. - Eur. J. 2008, 14 (34), 1060310614,  DOI: 10.1002/chem.200801258
  27. 27
    Haenen, G. R.; De Rooij, B. M.; Vermeulen, N. P.; Bast, A. Mechanism of the reaction of ebselen with endogenous thiols: Dihydrolipoate is a better cofactor than glutathione in the peroxidase activity of ebselen. Mol. Pharmacol. 1990, 37 (3), 412422
  28. 28
    Wagner, G.; Schuch, G.; Akerboom, T. P.; Sies, H. Transport of ebselen in plasma and its transfer to binding sites in the hepatocyte. Biochem. Pharmacol. 1994, 48 (6), 11371144,  DOI: 10.1016/0006-2952(94)90150-3
  29. 29
    Singh, N.; Halliday, A. C.; Thomas, J. M.; Kuznetsova, O. V.; Baldwin, R.; Woon, E. C.; Aley, P. K.; Antoniadou, I.; Sharp, T.; Vasudevan, S. R.; Churchill, G. C. A safe lithium mimetic for bipolar disorder. Nat. Commun. 2013, 4, 1332,  DOI: 10.1038/ncomms2320
  30. 30
    Johnson, T. W.; Dress, K. R.; Edwards, M. Using the Golden Triangle to optimize clearance and oral absorption. Bioorg. Med. Chem. Lett. 2009, 19 (19), 55605564,  DOI: 10.1016/j.bmcl.2009.08.045
  31. 31
    Johnson, T. W.; Gallego, R. A.; Edwards, M. P. Lipophilic Efficiency as an Important Metric in Drug Design. J. Med. Chem. 2018, 61 (15), 64016420,  DOI: 10.1021/acs.jmedchem.8b00077
  32. 32
    Hopkins, A. L.; Keseru, G. M.; Leeson, P. D.; Rees, D. C.; Reynolds, C. H. The role of ligand efficiency metrics in drug discovery. Nat. Rev. Drug Discovery 2014, 13 (2), 105121,  DOI: 10.1038/nrd4163
  33. 33
    Raymer, B.; Bhattacharya, S. K. Lead-like Drugs: A Perspective. J. Med. Chem. 2018, 61 (23), 1037510384,  DOI: 10.1021/acs.jmedchem.8b00407
  34. 34
    Weiss, M. M.; Dineen, T. A.; Marx, I. E.; Altmann, S.; Boezio, A.; Bregman, H.; Chu-Moyer, M.; DiMauro, E. F.; Feric Bojic, E.; Foti, R. S.; Gao, H.; Graceffa, R.; Gunaydin, H.; Guzman-Perez, A.; Huang, H.; Huang, L.; Jarosh, M.; Kornecook, T.; Kreiman, C. R.; Ligutti, J.; La, D. S.; Lin, M. J.; Liu, D.; Moyer, B. D.; Nguyen, H. N.; Peterson, E. A.; Rose, P. E.; Taborn, K.; Youngblood, B. D.; Yu, V.; Fremeau, R. T., Jr. Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics While Mitigating Metabolic Liabilities. J. Med. Chem. 2017, 60 (14), 59695989,  DOI: 10.1021/acs.jmedchem.6b01851
  35. 35
    Dittmar, M.; Lee, J. S.; Whig, K.; Segrist, E.; Li, M.; Kamalia, B.; Castellana, L.; Ayyanathan, K.; Cardenas-Diaz, F. L.; Morrisey, E. E.; Truitt, R.; Yang, W.; Jurado, K.; Samby, K.; Ramage, H.; Schultz, D. C.; Cherry, S. Drug repurposing screens reveal cell-type-specific entry pathways and FDA-approved drugs active against SARS-Cov-2. Cell Rep. 2021, 35 (1), 108959,  DOI: 10.1016/j.celrep.2021.108959
  36. 36
    Jang, W. D.; Jeon, S.; Kim, S.; Lee, S. Y. Drugs repurposed for COVID-19 by virtual screening of 6,218 drugs and cell-based assay. Proc. Natl. Acad. Sci. U. S. A. 2021, 118 (30), e2024302118  DOI: 10.1073/pnas.2024302118
  37. 37
    Choi, S. W.; Shin, J. S.; Park, S. J.; Jung, E.; Park, Y. G.; Lee, J.; Kim, S. J.; Park, H. J.; Lee, J. H.; Park, S. M.; Moon, S. H.; Ban, K.; Go, Y. Y. Antiviral activity and safety of remdesivir against SARS-CoV-2 infection in human pluripotent stem cell-derived cardiomyocytes. Antiviral Res. 2020, 184, 104955,  DOI: 10.1016/j.antiviral.2020.104955
  38. 38
    Tiwari, S. K.; Wang, S.; Smith, D.; Carlin, A. F.; Rana, T. M. Revealing Tissue-Specific SARS-CoV-2 Infection and Host Responses using Human Stem Cell-Derived Lung and Cerebral Organoids. Stem Cell Rep. 2021, 16 (3), 437445,  DOI: 10.1016/j.stemcr.2021.02.005
  39. 39
    Li, N.; Hui, H.; Bray, B.; Gonzalez, G. M.; Zeller, M.; Anderson, K. G.; Knight, R.; Smith, D.; Wang, Y.; Carlin, A. F.; Rana, T. M. METTL3 regulates viral m6A RNA modification and host cell innate immune responses during SARS-CoV-2 infection. Cell Rep. 2021, 35 (6), 109091,  DOI: 10.1016/j.celrep.2021.109091
  40. 40
    Wang, S.; Li, W.; Hui, H.; Tiwari, S. K.; Zhang, Q.; Croker, B. A.; Rawlings, S.; Smith, D.; Carlin, A. F.; Rana, T. M. Cholesterol 25-Hydroxylase inhibits SARS-CoV-2 and other coronaviruses by depleting membrane cholesterol. EMBO J. 2020, 39 (21), e106057  DOI: 10.15252/embj.2020106057
  41. 41
    Ma, C.; Hu, Y.; Townsend, J. A.; Lagarias, P. I.; Marty, M. T.; Kolocouris, A.; Wang, J. Ebselen, Disulfiram, Carmofur, PX-12, Tideglusib, and Shikonin Are Nonspecific Promiscuous SARS-CoV-2 Main Protease Inhibitors. ACS Pharmacol Transl Sci. 2020, 3 (6), 12651277,  DOI: 10.1021/acsptsci.0c00130
  42. 42
    Weglarz-Tomczak, E.; Tomczak, J. M.; Talma, M.; Burda-Grabowska, M.; Giurg, M.; Brul, S. Identification of ebselen and its analogues as potent covalent inhibitors of papain-like protease from SARS-CoV-2. Sci. Rep. 2021, 11 (1), 3640,  DOI: 10.1038/s41598-021-83229-6
  43. 43
    Freitas, B. T.; Durie, I. A.; Murray, J.; Longo, J. E.; Miller, H. C.; Crich, D.; Hogan, R. J.; Tripp, R. A.; Pegan, S. D. Characterization and Noncovalent Inhibition of the Deubiquitinase and deISGylase Activity of SARS-CoV-2 Papain-Like Protease. ACS Infect. Dis. 2020, 6 (8), 20992109,  DOI: 10.1021/acsinfecdis.0c00168
  44. 44
    Shin, D.; Mukherjee, R.; Grewe, D.; Bojkova, D.; Baek, K.; Bhattacharya, A.; Schulz, L.; Widera, M.; Mehdipour, A. R.; Tascher, G.; Geurink, P. P.; Wilhelm, A.; van der Heden van Noort, G. J.; Ovaa, H.; Muller, S.; Knobeloch, K. P.; Rajalingam, K.; Schulman, B. A.; Cinatl, J.; Hummer, G.; Ciesek, S.; Dikic, I. Papain-like protease regulates SARS-CoV-2 viral spread and innate immunity. Nature 2020, 587 (7835), 657662,  DOI: 10.1038/s41586-020-2601-5
  45. 45
    Klemm, T.; Ebert, G.; Calleja, D. J; Allison, C. C; Richardson, L. W; Bernardini, J. P; Lu, B. G.; Kuchel, N. W; Grohmann, C.; Shibata, Y.; Gan, Z. Y.; Cooney, J. P; Doerflinger, M.; Au, A. E; Blackmore, T. R; Heden van Noort, G. J; Geurink, P. P; Ovaa, H.; Newman, J.; Riboldi-Tunnicliffe, A.; Czabotar, P. E; Mitchell, J. P; Feltham, R.; Lechtenberg, B. C; Lowes, K. N; Dewson, G.; Pellegrini, M.; Lessene, G.; Komander, D. Mechanism and inhibition of the papain-like protease, PLpro, of SARS-CoV-2. EMBO J. 2020, 39 (18), e106275  DOI: 10.15252/embj.2020106275
  46. 46
    Liu, G.; Lee, J.-H.; Parker, Z. M.; Acharya, D.; Chiang, J. J.; van Gent, M.; Riedl, W.; Davis-Gardner, M. E.; Wies, E.; Chiang, C.; Gack, M. U. ISG15-dependent activation of the sensor MDA5 is antagonized by the SARS-CoV-2 papain-like protease to evade host innate immunity. Nature Microbiology 2021, 6, 467478,  DOI: 10.1038/s41564-021-00884-1
  47. 47
    Jacobson, M. P.; Friesner, R. A.; Xiang, Z.; Honig, B. On the role of the crystal environment in determining protein side-chain conformations. J. Mol. Biol. 2002, 320 (3), 597608,  DOI: 10.1016/S0022-2836(02)00470-9
  48. 48
    Jacobson, M. P.; Pincus, D. L.; Rapp, C. S.; Day, T. J.; Honig, B.; Shaw, D. E.; Friesner, R. A. A hierarchical approach to all-atom protein loop prediction. Proteins: Struct., Funct., Genet. 2004, 55 (2), 351367,  DOI: 10.1002/prot.10613
  49. 49
    Donovan, S. F.; Pescatore, M. C. Method for measuring the logarithm of the octanol-water partition coefficient by using short octadecyl-poly(vinyl alcohol) high-performance liquid chromatography columns. J. Chromatogr A 2002, 952 (1–2), 4761,  DOI: 10.1016/S0021-9673(02)00064-X
  50. 50
    Box, K.; Bevan, C.; Comer, J.; Hill, A.; Allen, R.; Reynolds, D. High-throughput measurement of pKa values in a mixed-buffer linear pH gradient system. Anal. Chem. 2003, 75 (4), 883892,  DOI: 10.1021/ac020329y

Cited By

Click to copy section linkSection link copied!

This article is cited by 74 publications.

  1. Zhihao Gu, Yong Yan, Hanwen Liu, Di Wu, Hequan Yao, Kejiang Lin, Xuanyi Li. Discovery of Covalent Lead Compounds Targeting 3CL Protease with a Lateral Interactions Spiking Neural Network. Journal of Chemical Information and Modeling 2024, 64 (8) , 3047-3058. https://doi.org/10.1021/acs.jcim.3c01900
  2. Katarzyna Macegoniuk, Wojciech Tabor, Luca Mazzei, Michele Cianci, Mirosław Giurg, Kamila Olech, Małgorzata Burda-Grabowska, Rafał Kaleta, Agnieszka Grabowiecka, Artur Mucha, Stefano Ciurli, Łukasz Berlicki. Optimized Ebselen-Based Inhibitors of Bacterial Ureases with Nontypical Mode of Action. Journal of Medicinal Chemistry 2023, 66 (3) , 2054-2063. https://doi.org/10.1021/acs.jmedchem.2c01799
  3. Xi Feng, Lixia Pan, Zhiyu Qian, Dongyu Liu, Xin Guan, Li Feng, Bin Song, Xi Xu, Ninghua Tan, Yi Ma, Zhiyu Li, Zhe Wang, Jinlei Bian. Discovery of Selenium-Containing STING Agonists as Orally Available Antitumor Agents. Journal of Medicinal Chemistry 2022, 65 (22) , 15048-15065. https://doi.org/10.1021/acs.jmedchem.2c00634
  4. Gabriele La Monica, Alessia Bono, Antonino Lauria, Annamaria Martorana. Targeting SARS-CoV-2 Main Protease for Treatment of COVID-19: Covalent Inhibitors Structure–Activity Relationship Insights and Evolution Perspectives. Journal of Medicinal Chemistry 2022, 65 (19) , 12500-12534. https://doi.org/10.1021/acs.jmedchem.2c01005
  5. Xin-Xin Liang, Xing-Jie Zhang, Ying-Xin Zhao, Jian Feng, Jie-Chun Zeng, Qiang-Qiang Shi, Joseph Sakah Kaunda, Xiao-Li Li, Wei-Guang Wang, Wei-Lie Xiao. Aspulvins A–H, Aspulvinone Analogues with SARS-CoV-2 Mpro Inhibitory and Anti-inflammatory Activities from an Endophytic Cladosporium sp.. Journal of Natural Products 2022, 85 (4) , 878-887. https://doi.org/10.1021/acs.jnatprod.1c01003
  6. Wei Hou, Hongtao Xu. Incorporating Selenium into Heterocycles and Natural Products─From Chemical Properties to Pharmacological Activities. Journal of Medicinal Chemistry 2022, 65 (6) , 4436-4456. https://doi.org/10.1021/acs.jmedchem.1c01859
  7. Nouri Neamati. Advances Toward COVID-19 Therapies Special Issue. Journal of Medicinal Chemistry 2022, 65 (4) , 2713-2715. https://doi.org/10.1021/acs.jmedchem.2c00178
  8. Abdelali Chihab, Nabil El Brahmi, Abdelmoula El Abbouchi, Abdelaziz El Alaoui, Mostapha Bousmina, Elmostafa El Fahime, Saïd El Kazzouli. Synthesis, characterization, proteolytic activity inhibition, ADMET prediction, and molecular docking studies of novel indole derivatives as potential SARS-CoV-2 protease inhibitors. Journal of Molecular Structure 2025, 1323 , 140707. https://doi.org/10.1016/j.molstruc.2024.140707
  9. Aditya Trivedi, Tushar Kushwaha, Ishani, Sudhanshu Vrati, Dharmender Gupta, Sarala Rani Kayampeta, Mohammad Khalid Parvez, Krishna Kishore Inampudi, Mohan Babu Appaiahgari, Deepak Sehgal. Psoralidin acts as a dual protease inhibitor against PL pro and M pro of SARS ‐ CoV ‐2. The FEBS Journal 2025, 67 https://doi.org/10.1111/febs.17380
  10. Nabeel Ahmed, Aimen Sajid, Taseer Ahmad, Muhammad Ajmal Shah, Abdul Jabbar Shah. Coumarins as potential inhibitors targeting SARS-CoV-2. 2025, 151-182. https://doi.org/10.1016/B978-0-443-18757-5.00006-2
  11. Ilma Shakeel, Aaliya Taiyab, Aanchal Rathi, Taj Mohammad, Md. Imtaiyaz Hassan. Insights into SARS-CoV-2 structure: Approaches and action of SARS-CoV-2 main protease inhibitor. 2025, 89-117. https://doi.org/10.1016/B978-0-443-19170-1.00037-1
  12. S. k. Erfanul Haque, Sukanta Bhadra, Nishith Kumar Pal. Exploring potential therapeutic candidates against COVID-19: a molecular docking study. Discover Molecules 2024, 1 (1) https://doi.org/10.1007/s44345-024-00005-5
  13. Kristine Mae H. Bonalos, Khent Primo Alcantara, Pornchai Rojsitthisak, Ross D. Vasquez. Comprehensive In silico and In vitro evaluation of Plectranthus amboinicus essential oil: A promising inhibitor of SARS-CoV-2 B.1.1.529 Omicron variant, proinflammatory biomarkers and diabetes-related enzymes. Industrial Crops and Products 2024, 222 , 119905. https://doi.org/10.1016/j.indcrop.2024.119905
  14. Luca Sancineto, Francesca Mangiavacchi, Agnieszka Dabrowska, Agata J. Pacuła-Miszewska, Magdalena Obieziurska-Fabisiak, Cecilia Scimmi, Veronica Ceccucci, Juan Kong, Yao Zhao, Gianluca Ciancaleoni, Vanessa Nascimento, Bruno Rizzuti, Marco Bortoli, Laura Orian, Anna Kula-Pacurar, Haitao Yang, Jacek Ścianowski, Ying Lei, Krzysztof Pyrc, Claudio Santi. New insights in the mechanism of the SARS-CoV-2 Mpro inhibition by benzisoselenazolones and diselenides. Scientific Reports 2024, 14 (1) https://doi.org/10.1038/s41598-024-75519-6
  15. Chunru Liu, Xiaoqing Dong, Pinghuang Liu, Xi Lin. Advances in porcine respiratory and intestinal organoids: status and potential application for virus infections. One Health Advances 2024, 2 (1) https://doi.org/10.1186/s44280-024-00052-0
  16. Md. Kaderi Kibria, Md. Ahad Ali, Md. Nurul Haque Mollah, . Exploring bacterial key genes and therapeutic agents for breast cancer among the Ghanaian female population: Insights from In Silico analyses. PLOS ONE 2024, 19 (11) , e0312493. https://doi.org/10.1371/journal.pone.0312493
  17. Mianling Yang, Myoung Kyu Lee, Shenghua Gao, Letian Song, Hye‐Yeon Jang, Inseong Jo, Chun‐Chiao Yang, Katharina Sylvester, Chunkyu Ko, Shuo Wang, Bing Ye, Kai Tang, Junyi Li, Manyu Gu, Christa E. Müller, Norbert Sträter, Xinyong Liu, Meehyein Kim, Peng Zhan. Miniaturized Modular Click Chemistry‐enabled Rapid Discovery of Unique SARS‐CoV‐2 M pro Inhibitors With Robust Potency and Drug‐like Profile. Advanced Science 2024, 11 (43) https://doi.org/10.1002/advs.202404884
  18. Asma Fatima, Anupriya M. Geethakumari, Wesam S. Ahmed, Kabir H. Biswas. A potential allosteric inhibitor of SARS-CoV-2 main protease (Mpro) identified through metastable state analysis. Frontiers in Molecular Biosciences 2024, 11 https://doi.org/10.3389/fmolb.2024.1451280
  19. Borwornlak Toopradab, Wanting Xie, Lian Duan, Kowit Hengphasatporn, Ryuhei Harada, Silpsiri Sinsulpsiri, Yasuteru Shigeta, Liyi Shi, Phornphimon Maitarad, Thanyada Rungrotmongkol. Machine learning-based QSAR and LB-PaCS-MD guided design of SARS-CoV-2 main protease inhibitors. Bioorganic & Medicinal Chemistry Letters 2024, 110 , 129852. https://doi.org/10.1016/j.bmcl.2024.129852
  20. Ying Kuan, Hsu-Feng Chu, Pang-Hung Hsu, Kai-Cheng Hsu, Ta-Hsien Lin, Chun-Hsiang Huang, Wei-Yi Chen. Disulfiram inhibits coronaviral main protease by conjugating to its substrate entry site. International Journal of Biological Macromolecules 2024, 276 , 133955. https://doi.org/10.1016/j.ijbiomac.2024.133955
  21. Christos Papaneophytou. Breaking the Chain: Protease Inhibitors as Game Changers in Respiratory Viruses Management. International Journal of Molecular Sciences 2024, 25 (15) , 8105. https://doi.org/10.3390/ijms25158105
  22. Lei Han, Bing Wang, Kunhui Sun, Muqadas Sitara, Meifang Li, Ping Wang, Ning Chen, Xie-an Yu, Jiangwei Tian. A SARS-CoV-2 M pro fluorescent sensor for exploring pharmacodynamic substances from traditional Chinese medicine. The Analyst 2024, 149 (13) , 3585-3595. https://doi.org/10.1039/D4AN00372A
  23. Ya-Qi Xiao, Jiao Long, Shuang-Shuang Zhang, Yuan-Yuan Zhu, Shuang-Xi Gu. Non-peptidic inhibitors targeting SARS-CoV-2 main protease: A review. Bioorganic Chemistry 2024, 147 , 107380. https://doi.org/10.1016/j.bioorg.2024.107380
  24. Hezha O. Rasul, Noel Vinay Thomas, Dlzar D. Ghafour, Bakhtyar K. Aziz, Guillermo Salgado M., L. H. Mendoza-Huizar, Lorena Gerli Candia. Searching possible SARS-CoV-2 main protease inhibitors in constituents from herbal medicines using in silico studies. Journal of Biomolecular Structure and Dynamics 2024, 42 (8) , 4234-4248. https://doi.org/10.1080/07391102.2023.2220040
  25. Vimal K. Jain, K. Indira Priyadarsini. Selenium compounds as promising antiviral agents. New Journal of Chemistry 2024, 48 (15) , 6534-6552. https://doi.org/10.1039/D3NJ05180C
  26. Hongtao Li, Meng Sun, Fuzhi Lei, Jinfeng Liu, Xixiang Chen, Yaqi Li, Ying Wang, Jiani Lu, Danmei Yu, Yueqiu Gao, Jianrong Xu, Hongzhuan Chen, Man Li, Zhigang Yi, Xiao He, Lili Chen. Methyl rosmarinate is an allosteric inhibitor of SARS-CoV-2 3 CL protease as a potential candidate against SARS-cov-2 infection. Antiviral Research 2024, 224 , 105841. https://doi.org/10.1016/j.antiviral.2024.105841
  27. Md. Kaderi Kibria, Md. Ahad Ali, Muhammad Yaseen, Imran Ahmad Khan, Mashooq Ahmad Bhat, Md. Ariful Islam, Rashidul Alam Mahumud, Md. Nurul Haque Mollah. Discovery of Bacterial Key Genes from 16S rRNA-Seq Profiles That Are Associated with the Complications of SARS-CoV-2 Infections and Provide Therapeutic Indications. Pharmaceuticals 2024, 17 (4) , 432. https://doi.org/10.3390/ph17040432
  28. Prabuddha Bhattacharya, Anirban Mandal. Identification of amentoflavone as a potent SARS-CoV-2 M pro inhibitor: a combination of computational studies and in vitro biological evaluation. Journal of Biomolecular Structure and Dynamics 2024, 266 , 1-19. https://doi.org/10.1080/07391102.2024.2304676
  29. Samuel J.M. Santos, Antoninho Valentini. In silico investigation of Komaroviquinone as a potential inhibitor of SARS-CoV-2 main protease (Mpro): Molecular docking, molecular dynamics, and QM/MM approaches. Journal of Molecular Graphics and Modelling 2024, 126 , 108662. https://doi.org/10.1016/j.jmgm.2023.108662
  30. Frank Eric Tatsing Foka, Hazel Tumelo Mufhandu, . Predictive Assessment of the Antiviral Properties of Imperata cylindrica against SARS‐CoV‐2. Advances in Virology 2024, 2024 (1) https://doi.org/10.1155/2024/8598708
  31. Seshadri Reddy Varikasuvu, Subodh Kumar, Munikumar Manne, Mona Lisa, Saurabh Varshney, Pratima Gupta, Ashoo Grover, Vanita Lal, Harminder Singh, Shiv Kumar Mudgal, Konda Venkata Nagaraju. Covid-19 Clinical Outcomes and N-Acetylcysteine (Covinac Study): A Grade Compliant Meta-Analysis of Randomized Controlled Trials, Molecular Docking, and Dynamics Simulations with Mpro of Sars-Cov-2. 2024https://doi.org/10.2139/ssrn.4679821
  32. Xiaoli Shen, Hao Zhang, Pengyin Zhang, Xuerui Zhao, Chang Liu, Jianan Ju, Aijun Liu, Song Wang. The Inhibitory Mechanism of Sars-Cov-2 by Small Molecules of the Condensed Amino Thiourea Scaffold Based on Molecular Dynamics Simulation. 2024https://doi.org/10.2139/ssrn.4961580
  33. Kailasam Saravana Mani, Sundarraj Rajamanikandan, Balakrishnan Prabha. Discovery, synthesis and in silico approach of pyrrolo [3,4- c ]pyrroles as SARS-CoV-2 m pro inhibitors. Journal of Biomolecular Structure and Dynamics 2023, 41 (22) , 13454-13465. https://doi.org/10.1080/07391102.2023.2174599
  34. Ssemuyiga Charles, Mulumba Pius Edgar, Rajani Kanta Mahapatra. Artificial intelligence based virtual screening study for competitive and allosteric inhibitors of the SARS-CoV-2 main protease. Journal of Biomolecular Structure and Dynamics 2023, 41 (24) , 15286-15304. https://doi.org/10.1080/07391102.2023.2188419
  35. Shengchao Zhou, Kun Wang, Ziwei Hu, Tao Chen, Yao Dong, Rongmei Gao, Mengyuan Wu, Yuhuan Li, Xingyue Ji. Design, synthesis, and structure-activity relationships of a novel class of quinazoline derivatives as coronavirus inhibitors. European Journal of Medicinal Chemistry 2023, 261 , 115831. https://doi.org/10.1016/j.ejmech.2023.115831
  36. Shailima Rampogu, Tae Sung Jung, Min Woo Ha, Keun Woo Lee. Repurposing and computational design of PARP inhibitors as SARS-CoV-2 inhibitors. Scientific Reports 2023, 13 (1) https://doi.org/10.1038/s41598-023-36342-7
  37. Sunyu Song, Yeseul Kim, Kiwoong Kwak, Hyeonmin Lee, Hyunjae Park, Young Bong Kim, Hee-Jung Lee, Lin-Woo Kang. The N-terminal peptide of the main protease of SARS-CoV-2, targeting dimer interface, inhibits its proteolytic activity. BMB Reports 2023, 56 (11) , 606-611. https://doi.org/10.5483/BMBRep.2023-0153
  38. Jie Liu, Liang Xu, Wenjing Guo, Zoe Li, Md Kamrul Hasan Khan, Weigong Ge, Tucker A Patterson, Huixiao Hong. Developing a SARS-CoV-2 main protease binding prediction random forest model for drug repurposing for COVID-19 treatment. Experimental Biology and Medicine 2023, 82 https://doi.org/10.1177/15353702231209413
  39. Kun Zhou, Daquan Chen. Conventional Understanding of SARS‐CoV‐2 M pro and Common Strategies for Developing Its Inhibitors. ChemBioChem 2023, 24 (22) https://doi.org/10.1002/cbic.202300301
  40. Navid Kaboudi, Nadine Krüger, Maryam Hamzeh‐Mivehroud. Development of novel ligands against SARS‐CoV‐2 M pro enzyme: an in silico and in vitro Study. Molecular Informatics 2023, 42 (11) https://doi.org/10.1002/minf.202300120
  41. Wandong Liu, Juan Wang, Suyun Wang, Kairui Yue, Yu Hu, Xiaochun Liu, Lihao Wang, Shengbiao Wan, Ximing Xu. Discovery of new non-covalent and covalent inhibitors targeting SARS-CoV-2 papain-like protease and main protease. Bioorganic Chemistry 2023, 140 , 106830. https://doi.org/10.1016/j.bioorg.2023.106830
  42. Xin Li, Yongcheng Song. Structure and function of SARS-CoV and SARS-CoV-2 main proteases and their inhibition: A comprehensive review. European Journal of Medicinal Chemistry 2023, 260 , 115772. https://doi.org/10.1016/j.ejmech.2023.115772
  43. Jiaojiao Li, Lin Zhu, Zheng Qin, Zhengfu Li, Xun Gao, Jing Ji, Jinyang Shen. Discovery of Potential Compounds Against SARS-CoV-2 Based on 3CLpro/RdRp Dual-target: An In silico Approach. Letters in Drug Design & Discovery 2023, 20 (11) , 1846-1860. https://doi.org/10.2174/1570180819666220818145647
  44. Yinkai Duan, Haofeng Wang, Zhenghong Yuan, Haitao Yang. Structural biology of SARS-CoV-2 Mpro and drug discovery. Current Opinion in Structural Biology 2023, 82 , 102667. https://doi.org/10.1016/j.sbi.2023.102667
  45. Xiaojing Pang, Wei Xu, Yang Liu, Hua Li, Lixia Chen. The research progress of SARS-CoV-2 main protease inhibitors from 2020 to 2022. European Journal of Medicinal Chemistry 2023, 257 , 115491. https://doi.org/10.1016/j.ejmech.2023.115491
  46. Sabikeh Azimi, Muna S. Merza, Fatemeh Ghasemi, Hasan Ali Dhahi, Farid Baradarbarjastehbaf, Mehdi Moosavi, Pouya Ghamari Kargar, Christophe Len. Green and rapid and instrumental one-pot method for the synthesis of imidazolines having potential anti-SARS-CoV-2 main protease activity. Sustainable Chemistry and Pharmacy 2023, 34 , 101136. https://doi.org/10.1016/j.scp.2023.101136
  47. Andrea Citarella, Alessandro Dimasi, Davide Moi, Daniele Passarella, Angela Scala, Anna Piperno, Nicola Micale. Recent Advances in SARS-CoV-2 Main Protease Inhibitors: From Nirmatrelvir to Future Perspectives. Biomolecules 2023, 13 (9) , 1339. https://doi.org/10.3390/biom13091339
  48. Amanda Resende Tucci, Raquel Mello da Rosa, Alice Santos Rosa, Otávio Augusto Chaves, Vivian Neuza Santos Ferreira, Thamara Kelcya Fonseca Oliveira, Daniel Dias Coutinho Souza, Nathalia Roberto Resende Borba, Luciano Dornelles, Nayra Salazar Rocha, João Candido Pilar Mayer, João B. Teixeira da Rocha, Oscar Endrigo D. Rodrigues, Milene Dias Miranda. Antiviral Effect of 5′-Arylchalcogeno-3-aminothymidine Derivatives in SARS-CoV-2 Infection. Molecules 2023, 28 (18) , 6696. https://doi.org/10.3390/molecules28186696
  49. Tushar Saha, Shubhra Sinha, Rhodri Harfoot, Miguel E. Quiñones-Mateu, Shyamal C. Das. Spray-Dried Inhalable Microparticles Combining Remdesivir and Ebselen against SARS-CoV-2 Infection. Pharmaceutics 2023, 15 (9) , 2229. https://doi.org/10.3390/pharmaceutics15092229
  50. Swaraj Mohanty, Soumya Lipsa Rath, Poornima Sharma, Yasmin Ahmad. An Evidence of Drug Repurposing for COVID-19 Pandemic Based on In silico Investigation from Phenolic Derivatives of Silybum Marianum Against SARS-Cov-2 Proteins. OBM Genetics 2023, 07 (03) , 1-17. https://doi.org/10.21926/obm.genet.2303186
  51. Guilherme Wildner, Amanda Resende Tucci, Alessandro de Souza Prestes, Talise Muller, Alice dos Santos Rosa, Nathalia Roberto R. Borba, Vivian Neuza Ferreira, João Batista Teixeira Rocha, Milene Dias Miranda, Nilda Vargas Barbosa. Ebselen and Diphenyl Diselenide Inhibit SARS-CoV-2 Replication at Non-Toxic Concentrations to Human Cell Lines. Vaccines 2023, 11 (7) , 1222. https://doi.org/10.3390/vaccines11071222
  52. Hossein Mousavi, Behzad Zeynizadeh, Mehdi Rimaz. Green and efficient one-pot three-component synthesis of novel drug-like furo[2,3-d]pyrimidines as potential active site inhibitors and putative allosteric hotspots modulators of both SARS-CoV-2 MPro and PLPro. Bioorganic Chemistry 2023, 135 , 106390. https://doi.org/10.1016/j.bioorg.2023.106390
  53. Chamandi S. Dampalla, Matthew J. Miller, Yunjeong Kim, Alexandria Zabiegala, Harry Nhat Nguyen, Trent K. Madden, Hayden A. Thurman, Alexandra J. Machen, Anne Cooper, Lijun Liu, Kevin P. Battaile, Scott Lovell, Kyeong-Ok Chang, William C. Groutas. Structure-guided design of direct-acting antivirals that exploit the gem-dimethyl effect and potently inhibit 3CL proteases of severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2) and middle east respiratory syndrome coronavirus (MERS-CoV). European Journal of Medicinal Chemistry 2023, 254 , 115376. https://doi.org/10.1016/j.ejmech.2023.115376
  54. Samson Afewerki, Thiago Domingues Stocco, André Diniz Rosa da Silva, André Sales Aguiar Furtado, Gustavo Fernandes de Sousa, Guillermo U. Ruiz-Esparza, Thomas J. Webster, Fernanda R. Marciano, Maria Strømme, Yu Shrike Zhang, Anderson Oliveira Lobo. In vitro high-content tissue models to address precision medicine challenges. Molecular Aspects of Medicine 2023, 91 , 101108. https://doi.org/10.1016/j.mam.2022.101108
  55. J. Jeya Vandana, Cassandra Manrique, Lauretta A. Lacko, Shuibing Chen. Human pluripotent-stem-cell-derived organoids for drug discovery and evaluation. Cell Stem Cell 2023, 30 (5) , 571-591. https://doi.org/10.1016/j.stem.2023.04.011
  56. Xuming Tang, Dongxiang Xue, Tuo Zhang, Benjamin E. Nilsson-Payant, Lucia Carrau, Xiaohua Duan, Miriam Gordillo, Adrian Y. Tan, Yunping Qiu, Jenny Xiang, Robert E. Schwartz, Benjamin R. tenOever, Todd Evans, Shuibing Chen. A multi-organoid platform identifies CIART as a key factor for SARS-CoV-2 infection. Nature Cell Biology 2023, 25 (3) , 381-389. https://doi.org/10.1038/s41556-023-01095-y
  57. Matteo Pavan, Stefano Moro. Lessons Learnt from COVID-19: Computational Strategies for Facing Present and Future Pandemics. International Journal of Molecular Sciences 2023, 24 (5) , 4401. https://doi.org/10.3390/ijms24054401
  58. Luca Sancineto, Vanessa Nascimento. Chalcogen-containing Therapeutic Molecules. 2023, 592-602. https://doi.org/10.1039/BK9781839167386-00592
  59. Hongtao Xu, Wei Hou. Selenium-containing heterocycles. 2023, 915-930. https://doi.org/10.1016/B978-0-443-18611-0.00006-1
  60. Nasim Bakhtiyari, Sepideh Parvizpour, Yosef Masoudi-Sobhanzadeh, Ali Masoudi-Nejad. Computational-based strategies to deal with liver injuries induced by the repurposed drugs against SARS-CoV-2. Informatics in Medicine Unlocked 2023, 41 , 101336. https://doi.org/10.1016/j.imu.2023.101336
  61. Yong-Ming Yan, Xin Shen, Yan-Peng Li, Yong-Kai Cao, Jiao-Jiao Zhang, Yan Wang, Yong-Xian Cheng. Discovery of anti-SARS-CoV-2 agents from 38 Chinese patent drugs toward respiratory diseases <i>via</i> docking screening. Medicinal Plant Biology 2023, 2 (1) , 0-0. https://doi.org/10.48130/MPB-2023-0009
  62. Jiaojiao Zhang, Xin Shen, Yanpeng Li, Yongming Yan, Yan Wang, Yongxian Cheng. Discovery of anti-SARS-CoV-2 agents from commercially available flavor <i>via</i> docking screening. Medicinal Plant Biology 2023, 2 (1) , 0-0. https://doi.org/10.48130/MPB-2023-0010
  63. Kai Ni, Bo Che, Chongxin Yang, Youyuan Qin, Rong Gu, Chunhong Wang, Mingzhi Luo, Linhong Deng. Emerging toolset of three-dimensional pulmonary cell culture models for simulating lung pathophysiology towards mechanistic elucidation and therapeutic treatment of SARS-COV-2 infection. Frontiers in Pharmacology 2022, 13 https://doi.org/10.3389/fphar.2022.1033043
  64. Divya M. Teli, Bansari Patel, Mahesh T. Chhabria. Fragment-based design of SARS-CoV-2 Mpro inhibitors. Structural Chemistry 2022, 33 (6) , 2155-2168. https://doi.org/10.1007/s11224-022-02031-w
  65. Mahima Chauhan, Vijay Kumar Bhardwaj, Asheesh Kumar, Vinod Kumar, Pawan Kumar, M. Ghalib Enayathullah, Jessie Thomas, Joel George, Bokara Kiran Kumar, Rituraj Purohit, Arun Kumar, Sanjay Kumar. Theaflavin 3-gallate inhibits the main protease (Mpro) of SARS-CoV-2 and reduces its count in vitro. Scientific Reports 2022, 12 (1) https://doi.org/10.1038/s41598-022-17558-5
  66. Kunal Nepali, Ram Sharma, Sachin Sharma, Amandeep Thakur, Jing-Ping Liou. Beyond the vaccines: a glance at the small molecule and peptide-based anti-COVID19 arsenal. Journal of Biomedical Science 2022, 29 (1) https://doi.org/10.1186/s12929-022-00847-6
  67. Vijayan Ramachandran, Yanyun Liu, Qianying He, Andrew Tang, Patrick Ronaldson, Dominik Schenten, Rui Chang. Novel inhibitors against COVID-19 main protease suppressed viral infection. 2022https://doi.org/10.1101/2022.11.05.515305
  68. Kirsten McAulay, Alan Bilsland, Marta Bon. Reactivity of Covalent Fragments and Their Role in Fragment Based Drug Discovery. Pharmaceuticals 2022, 15 (11) , 1366. https://doi.org/10.3390/ph15111366
  69. Suhasini M. Iyengar, Kelly K. Barnsley, Hoang Yen Vu, Ian Jef A. Bongalonta, Alyssa S. Herrod, Jasmine A. Scott, Mary Jo Ondrechen. Identification and characterization of alternative sites and molecular probes for SARS-CoV-2 target proteins. Frontiers in Chemistry 2022, 10 https://doi.org/10.3389/fchem.2022.1017394
  70. Pushyaraga P. Venugopal, Omkar Singh, Debashree Chakraborty. Understanding the role of water on temperature-dependent structural modifications of SARS CoV-2 main protease binding sites. Journal of Molecular Liquids 2022, 363 , 119867. https://doi.org/10.1016/j.molliq.2022.119867
  71. Ly Hien Doan, Li-Wei Chu, Zi-Yi Huang, Anh Thuc Nguyen, Chia-Yin Lee, Chien-Ling Huang, Yu-Fen Chang, Wen-Yu Hsieh, Trang Thi Huyen Nguyen, Chao-Hsiung Lin, Chun-Li Su, Tsung-Hsien Chuang, Jin-Mei Lai, Feng-Sheng Wang, Chia-Jui Yang, Hui-Kang Liu, Yueh-Hsin Ping, Chi-Ying F. Huang. Virofree, an Herbal Medicine-Based Formula, Interrupts the Viral Infection of Delta and Omicron Variants of SARS-CoV-2. Frontiers in Pharmacology 2022, 13 https://doi.org/10.3389/fphar.2022.905197
  72. Nicola Gambacorta, Leonardo Caputo, Laura Quintieri, Linda Monaci, Fulvio Ciriaco, Orazio Nicolotti. Rational Discovery of Antiviral Whey Protein-Derived Small Peptides Targeting the SARS-CoV-2 Main Protease. Biomedicines 2022, 10 (5) , 1067. https://doi.org/10.3390/biomedicines10051067
  73. Supaphorn Seetaha, Phatcharin Khamplong, Panatda Wanaragthai, Thitinan Aiebchun, Siriluk Ratanabunyong, Sucheewin Krobthong, Yodying Yingchutrakul, Jatuporn Rattanasrisomporn, Kiattawee Choowongkomon. KERRA, Mixed Medicinal Plant Extracts, Inhibits SARS-CoV-2 Targets Enzymes and Feline Coronavirus. COVID 2022, 2 (5) , 621-632. https://doi.org/10.3390/covid2050046
  74. Zhonglei Wang, Liyan Yang. Broad‐spectrum prodrugs with anti‐SARS‐CoV‐2 activities: Strategies, benefits, and challenges. Journal of Medical Virology 2022, 94 (4) , 1373-1390. https://doi.org/10.1002/jmv.27517
  75. Yuling Han, Liuliu Yang, Lauretta A. Lacko, Shuibing Chen. Human organoid models to study SARS-CoV-2 infection. Nature Methods 2022, 19 (4) , 418-428. https://doi.org/10.1038/s41592-022-01453-y
  76. Guillem Macip, Pol Garcia-Segura, Júlia Mestres-Truyol, Bryan Saldivar-Espinoza, Gerard Pujadas, Santiago Garcia-Vallvé. A Review of the Current Landscape of SARS-CoV-2 Main Protease Inhibitors: Have We Hit the Bullseye Yet?. International Journal of Molecular Sciences 2022, 23 (1) , 259. https://doi.org/10.3390/ijms23010259
  77. Prasat Kittakoop, Dhanushka Darshana, Rapeepat Sangsuwan, Chulabhorn Mahidol. Alkaloids and Alkaloid-Like Compounds are Potential Scaffolds of Antiviral Agents against SARS-CoV-2 (COVID-19) Virus. HETEROCYCLES 2022, 105 (1) , 115. https://doi.org/10.3987/REV-22-SR(R)3

Journal of Medicinal Chemistry

Cite this: J. Med. Chem. 2022, 65, 4, 2866–2879
Click to copy citationCitation copied!
https://doi.org/10.1021/acs.jmedchem.1c00566
Published September 27, 2021

Copyright © 2021 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY-NC-ND 4.0 .

Article Views

17k

Altmetric

-

Citations

Learn about these metrics

Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.

  • Abstract

    Figure 1

    Figure 1. Docking poses of ebselen and 2-phenyl-1,2-benzoselenazol-3-one analogues. (A) Docking pose of ebselen bound to the catalytic site of SARS-CoV-2 Mpro. A hydrogen bond is present between the carbonyl oxygen atom of E01 and Glu 166. The benzoisoselenazole ring forms π–π stacking interactions with catalytic His 41. (B) Docking pose of E19 bound to the catalytic site of SARS-CoV-2 Mpro. A hydrogen bond is present between the carbonyl oxygen atom of E19 and Glu 166. The benzoisoselenazole ring forms π–π stacking interactions with catalytic His 41. The chlorine atom of E19 forms a halogen bond interaction with Gly 143. (C) Docking pose of E20 bound to the catalytic site of SARS-CoV-2 Mpro. π–π stacking interactions are present between the benzoisoselenazole ring of E20 and catalytic His 41. A hydrogen bond is present between the carbonyl oxygen atom of E20 and Glu 166. (D) Docking pose of E25 bound to the catalytic site of SARS-CoV-2 Mpro. A hydrogen bond is present between the carbonyl oxygen atom of E25 and Glu 166. π–π stacking interactions are present between the benzoisoselenazole ring of E25 and catalytic His 41. The nitrile group was observed to accept a hydrogen bond from Cys 44.

    Scheme 1

    Scheme 1. Synthesis of Ebselen Analogues

    Figure 2

    Figure 2. Ebselen and its analogues can covalently modify SARS-CoV-2 Mpro in the absence of reducing agents. (A) Ebselen is a reversible covalent inhibitor of SARS-CoV-2 Mpro. The product formation over time was reduced by approximately 65% by 1 μM ebselen (red) and by 89% by 10 μM ebselen (purple) relative to that with the DMSO control (black) when added simultaneously with the fluorogenic substrate. Upon 10× dilution in an assay buffer without DTT, the product formation of SARS-CoV-2 Mpro preincubated with 10 μM ebselen (green) was reduced by 91%, which is comparable to an undiluted concentration of 10 μM. When diluted in the presence of DTT, the product formation of SARS-CoV-2 Mpro preincubated with 10 μM of ebselen (blue) was reduced by 53% relative to that with the DMSO control, similar to that observed for a final concentration of 1 μM. These data indicate that ebselen can covalently modify SARS-CoV-2 Mpro, although this modification is reversible in the presence of strong reducing agents. (B) Compound E04 is a rapidly reversible inhibitor of SARS-CoV-2 Mpro. Upon 10× dilution, the product formation of SARS-CoV-2 Mpro preincubated with 10 μM E04 in the absence of DTT (green) was reduced by 48% relative to that with the DMSO control, similar to that observed for a final concentration of 1 μM E04 (44% inhibition), while the simultaneous addition of the substrate and 10 μM E04 reduced the product formation by 86%. Similar results were observed for SARS-CoV-2 Mpro preincubated with 10 μM E04 in the presence of DTT (blue, 43% inhibition). (C) Compound E07 is a reversible covalent inhibitor of SARS-CoV-2 Mpro. (D) Compound E19 is a rapidly reversible inhibitor of SARS-CoV-2 Mpro. (E) Compound E20 is a reversible covalent inhibitor of SARS-CoV-2 Mpro. (F) Compound E25 is a reversible covalent inhibitor of SARS-CoV-2 Mpro. Upon 10× dilution, the product formation of SARS-CoV-2 Mpro preincubated with 10 μM E25 in the absence of DTT (green) was reduced by 64% relative to that with the DMSO control. The simultaneous addition of the substrate and 10 μM 25 reduced product formation by 94%, while the simultaneous addition of the substrate and 1 μM E25 reduced product formation by 29%. These data suggest that while E25 may covalently modify SARS-CoV-2 Mpro, the formation of the covalent adduct was incomplete after 30 min of incubation.

    Figure 3

    Figure 3. Ebselen and E04 are competitive inhibitors of the SARS-CoV-2 main protease. (A) Mpro exposed to varying concentrations of ebselen reached a common Vmax at 30 μM substrate (B) All ebselen conditions converge on a common y-intercept, indicating that Vmax is independent of the inhibitor concentration, which is consistent with a competitive mechanism of inhibition. (C) Mpro exposed to varying concentrations of E04 reached a common Vmax at 30 μM substrate (D) All E04 conditions converge on a common y-intercept, indicating that Vmax is independent of the inhibitor concentration, which is consistent with a competitive mechanism of inhibition. (E) Antiviral activity of select compounds. Vero E6 cells were pretreated with varying doses of the compound, then infected with SARS-CoV-2 to a multiplicity of infection (MOI) of 0.01. After 72 h, cell proliferation was determined. Remdesivir is included as a positive control (EC50 = 1.8 ± 1.3 μM).

    Figure 4

    Figure 4. E24 inhibits SARS-CoV-2 replication in infected human iPSC-derived lung organoids. Sixty-day-old lung organoids were infected with the SARS-CoV-2 USA-WA1/2020 virus at a MOI of 2, and viral RNAs from (A) supernatant and (B) cellular fractions were quantified after 72 h of infection. Mean ± SEM of n = 3 organoids cultured and infected in different wells, *p < 0.05 by Student’s t-test.

  • References


    This article references 50 other publications.

    1. 1
      Li, F.; Li, W.; Farzan, M.; Harrison, S. C. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science 2005, 309 (5742), 18641868,  DOI: 10.1126/science.1116480
    2. 2
      Phan, T. Novel coronavirus: From discovery to clinical diagnostics. Infect., Genet. Evol. 2020, 79, 104211,  DOI: 10.1016/j.meegid.2020.104211
    3. 3
      Pillaiyar, T.; Manickam, M.; Namasivayam, V.; Hayashi, Y.; Jung, S. H. An Overview of Severe Acute Respiratory Syndrome-Coronavirus (SARS-CoV) 3CL Protease Inhibitors: Peptidomimetics and Small Molecule Chemotherapy. J. Med. Chem. 2016, 59 (14), 6595628,  DOI: 10.1021/acs.jmedchem.5b01461
    4. 4
      Ziebuhr, J.; Snijder, E. J.; Gorbalenya, A. E. Virus-encoded proteinases and proteolytic processing in the Nidovirales. J. Gen. Virol. 2000, 81 (4), 853879,  DOI: 10.1099/0022-1317-81-4-853
    5. 5
      Báez-Santos, Y. M.; St. John, S. E.; Mesecar, A. D. The SARS-coronavirus papain-like protease: Structure, function and inhibition by designed antiviral compounds. Antiviral Res. 2015, 115, 2138,  DOI: 10.1016/j.antiviral.2014.12.015
    6. 6
      Lee, T. W.; Cherney, M. M.; Huitema, C.; Liu, J.; James, K. E.; Powers, J. C.; Eltis, L. D.; James, M. N. Crystal structures of the main peptidase from the SARS coronavirus inhibited by a substrate-like aza-peptide epoxide. J. Mol. Biol. 2005, 353 (5), 11371151,  DOI: 10.1016/j.jmb.2005.09.004
    7. 7
      Xu, X.; Liu, Y.; Weiss, S.; Arnold, E.; Sarafianos, S. G.; Ding, J. Molecular model of SARS coronavirus polymerase: implications for biochemical functions and drug design. Nucleic Acids Res. 2003, 31 (24), 71177130,  DOI: 10.1093/nar/gkg916
    8. 8
      De Clercq, E.; Li, G. Approved Antiviral Drugs over the Past 50 Years. Clin. Microbiol. Rev. 2016, 29 (3), 695747,  DOI: 10.1128/CMR.00102-15
    9. 9
      Kanters, S.; Socias, M. E.; Paton, N. I.; Vitoria, M.; Doherty, M.; Ayers, D.; Popoff, E.; Chan, K.; Cooper, D. A.; Wiens, M. O.; Calmy, A.; Ford, N.; Nsanzimana, S.; Mills, E. J. Comparative efficacy and safety of second-line antiretroviral therapy for treatment of HIV/AIDS: A systematic review and network meta-analysis. Lancet HIV 2017, 4 (10), e433e441,  DOI: 10.1016/S2352-3018(17)30109-1
    10. 10
      Deeks, E. D. Darunavir: a review of its use in the management of HIV-1 infection. Drugs 2014, 74 (1), 99125,  DOI: 10.1007/s40265-013-0159-3
    11. 11
      la Porte, C. J. Saquinavir, the pioneer antiretroviral protease inhibitor. Expert Opin. Drug Metab. Toxicol. 2009, 5 (10), 13131322,  DOI: 10.1517/17425250903273160
    12. 12
      Heo, Y. A.; Deeks, E. D. Sofosbuvir/Velpatasvir/Voxilaprevir: A Review in Chronic Hepatitis C. Drugs 2018, 78 (5), 577587,  DOI: 10.1007/s40265-018-0895-5
    13. 13
      Sanford, M. Simeprevir: a review of its use in patients with chronic hepatitis C virus infection. Drugs 2015, 75 (2), 183196,  DOI: 10.1007/s40265-014-0341-2
    14. 14
      Chopp, S.; Vanderwall, R.; Hult, A.; Klepser, M. Simeprevir and sofosbuvir for treatment of hepatitis C infection. Am. J. Health-Syst. Pharm. 2015, 72 (17), 14451455,  DOI: 10.2146/ajhp140290
    15. 15
      Jin, Z.; Du, X.; Xu, Y.; Deng, Y.; Liu, M.; Zhao, Y.; Zhang, B.; Li, X.; Zhang, L.; Peng, C.; Duan, Y.; Yu, J.; Wang, L.; Yang, K.; Liu, F.; Jiang, R.; Yang, X.; You, T.; Liu, X.; Yang, X.; Bai, F.; Liu, H.; Liu, X.; Guddat, L. W.; Xu, W.; Xiao, G.; Qin, C.; Shi, Z.; Jiang, H.; Rao, Z.; Yang, H. Structure of M(pro) from COVID-19 virus and discovery of its inhibitors. Nature 2020, 582, 289293,  DOI: 10.1038/s41586-020-2223-y
    16. 16
      Dai, W.; Zhang, B.; Jiang, X. M.; Su, H.; Li, J.; Zhao, Y.; Xie, X.; Jin, Z.; Peng, J.; Liu, F.; Li, C.; Li, Y.; Bai, F.; Wang, H.; Cheng, X.; Cen, X.; Hu, S.; Yang, X.; Wang, J.; Liu, X.; Xiao, G.; Jiang, H.; Rao, Z.; Zhang, L. K.; Xu, Y.; Yang, H.; Liu, H. Structure-based design of antiviral drug candidates targeting the SARS-CoV-2 main protease. Science 2020, 368 (6497), 13311335,  DOI: 10.1126/science.abb4489
    17. 17
      Vuong, W.; Khan, M. B.; Fischer, C.; Arutyunova, E.; Lamer, T.; Shields, J.; Saffran, H. A.; McKay, R. T.; van Belkum, M. J.; Joyce, M. A.; Young, H. S.; Tyrrell, D. L.; Vederas, J. C.; Lemieux, M. J. Feline coronavirus drug inhibits the main protease of SARS-CoV-2 and blocks virus replication. Nat. Commun. 2020, 11 (1), 4282,  DOI: 10.1038/s41467-020-18096-2
    18. 18
      Zhang, L.; Lin, D.; Sun, X.; Curth, U.; Drosten, C.; Sauerhering, L.; Becker, S.; Rox, K.; Hilgenfeld, R. Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved alpha-ketoamide inhibitors. Science 2020, 368 (6489), 409412,  DOI: 10.1126/science.abb3405
    19. 19
      Jin, Z.; Zhao, Y.; Sun, Y.; Zhang, B.; Wang, H.; Wu, Y.; Zhu, Y.; Zhu, C.; Hu, T.; Du, X.; Duan, Y.; Yu, J.; Yang, X.; Yang, X.; Yang, K.; Liu, X.; Guddat, L. W.; Xiao, G.; Zhang, L.; Yang, H.; Rao, Z. Structural basis for the inhibition of SARS-CoV-2 main protease by antineoplastic drug carmofur. Nat. Struct. Mol. Biol. 2020, 27 (6), 529532,  DOI: 10.1038/s41594-020-0440-6
    20. 20
      Fu, L.; Ye, F.; Feng, Y.; Yu, F.; Wang, Q.; Wu, Y.; Zhao, C.; Sun, H.; Huang, B.; Niu, P.; Song, H.; Shi, Y.; Li, X.; Tan, W.; Qi, J.; Gao, G. F. Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main protease. Nat. Commun. 2020, 11 (1), 4417,  DOI: 10.1038/s41467-020-18233-x
    21. 21
      Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S. Glide: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J. Med. Chem. 2004, 47 (7), 17391749,  DOI: 10.1021/jm0306430
    22. 22
      Halgren, T. A.; Murphy, R. B.; Friesner, R. A.; Beard, H. S.; Frye, L. L.; Pollard, W. T.; Banks, J. L. Glide: a new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening. J. Med. Chem. 2004, 47 (7), 17501759,  DOI: 10.1021/jm030644s
    23. 23
      Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren, T. A.; Sanschagrin, P. C.; Mainz, D. T. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes. J. Med. Chem. 2006, 49 (21), 61776196,  DOI: 10.1021/jm051256o
    24. 24
      Amporndanai, K.; Meng, X.; Shang, W.; Jin, Z.; Rogers, M.; Zhao, Y.; Rao, Z.; Liu, Z. J.; Yang, H.; Zhang, L.; O’Neill, P. M.; Samar Hasnain, S. Inhibition mechanism of SARS-CoV-2 main protease by ebselen and its derivatives. Nat. Commun. 2021, 12 (1), 3061,  DOI: 10.1038/s41467-021-23313-7
    25. 25
      Muramatsu, T.; Takemoto, C.; Kim, Y. T.; Wang, H.; Nishii, W.; Terada, T.; Shirouzu, M.; Yokoyama, S. SARS-CoV 3CL protease cleaves its C-terminal autoprocessing site by novel subsite cooperativity. Proc. Natl. Acad. Sci. U. S. A. 2016, 113 (46), 1299713002,  DOI: 10.1073/pnas.1601327113
    26. 26
      Sarma, B. K.; Mugesh, G. Antioxidant activity of the anti-inflammatory compound ebselen: a reversible cyclization pathway via selenenic and seleninic acid intermediates. Chem. - Eur. J. 2008, 14 (34), 1060310614,  DOI: 10.1002/chem.200801258
    27. 27
      Haenen, G. R.; De Rooij, B. M.; Vermeulen, N. P.; Bast, A. Mechanism of the reaction of ebselen with endogenous thiols: Dihydrolipoate is a better cofactor than glutathione in the peroxidase activity of ebselen. Mol. Pharmacol. 1990, 37 (3), 412422
    28. 28
      Wagner, G.; Schuch, G.; Akerboom, T. P.; Sies, H. Transport of ebselen in plasma and its transfer to binding sites in the hepatocyte. Biochem. Pharmacol. 1994, 48 (6), 11371144,  DOI: 10.1016/0006-2952(94)90150-3
    29. 29
      Singh, N.; Halliday, A. C.; Thomas, J. M.; Kuznetsova, O. V.; Baldwin, R.; Woon, E. C.; Aley, P. K.; Antoniadou, I.; Sharp, T.; Vasudevan, S. R.; Churchill, G. C. A safe lithium mimetic for bipolar disorder. Nat. Commun. 2013, 4, 1332,  DOI: 10.1038/ncomms2320
    30. 30
      Johnson, T. W.; Dress, K. R.; Edwards, M. Using the Golden Triangle to optimize clearance and oral absorption. Bioorg. Med. Chem. Lett. 2009, 19 (19), 55605564,  DOI: 10.1016/j.bmcl.2009.08.045
    31. 31
      Johnson, T. W.; Gallego, R. A.; Edwards, M. P. Lipophilic Efficiency as an Important Metric in Drug Design. J. Med. Chem. 2018, 61 (15), 64016420,  DOI: 10.1021/acs.jmedchem.8b00077
    32. 32
      Hopkins, A. L.; Keseru, G. M.; Leeson, P. D.; Rees, D. C.; Reynolds, C. H. The role of ligand efficiency metrics in drug discovery. Nat. Rev. Drug Discovery 2014, 13 (2), 105121,  DOI: 10.1038/nrd4163
    33. 33
      Raymer, B.; Bhattacharya, S. K. Lead-like Drugs: A Perspective. J. Med. Chem. 2018, 61 (23), 1037510384,  DOI: 10.1021/acs.jmedchem.8b00407
    34. 34
      Weiss, M. M.; Dineen, T. A.; Marx, I. E.; Altmann, S.; Boezio, A.; Bregman, H.; Chu-Moyer, M.; DiMauro, E. F.; Feric Bojic, E.; Foti, R. S.; Gao, H.; Graceffa, R.; Gunaydin, H.; Guzman-Perez, A.; Huang, H.; Huang, L.; Jarosh, M.; Kornecook, T.; Kreiman, C. R.; Ligutti, J.; La, D. S.; Lin, M. J.; Liu, D.; Moyer, B. D.; Nguyen, H. N.; Peterson, E. A.; Rose, P. E.; Taborn, K.; Youngblood, B. D.; Yu, V.; Fremeau, R. T., Jr. Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics While Mitigating Metabolic Liabilities. J. Med. Chem. 2017, 60 (14), 59695989,  DOI: 10.1021/acs.jmedchem.6b01851
    35. 35
      Dittmar, M.; Lee, J. S.; Whig, K.; Segrist, E.; Li, M.; Kamalia, B.; Castellana, L.; Ayyanathan, K.; Cardenas-Diaz, F. L.; Morrisey, E. E.; Truitt, R.; Yang, W.; Jurado, K.; Samby, K.; Ramage, H.; Schultz, D. C.; Cherry, S. Drug repurposing screens reveal cell-type-specific entry pathways and FDA-approved drugs active against SARS-Cov-2. Cell Rep. 2021, 35 (1), 108959,  DOI: 10.1016/j.celrep.2021.108959
    36. 36
      Jang, W. D.; Jeon, S.; Kim, S.; Lee, S. Y. Drugs repurposed for COVID-19 by virtual screening of 6,218 drugs and cell-based assay. Proc. Natl. Acad. Sci. U. S. A. 2021, 118 (30), e2024302118  DOI: 10.1073/pnas.2024302118
    37. 37
      Choi, S. W.; Shin, J. S.; Park, S. J.; Jung, E.; Park, Y. G.; Lee, J.; Kim, S. J.; Park, H. J.; Lee, J. H.; Park, S. M.; Moon, S. H.; Ban, K.; Go, Y. Y. Antiviral activity and safety of remdesivir against SARS-CoV-2 infection in human pluripotent stem cell-derived cardiomyocytes. Antiviral Res. 2020, 184, 104955,  DOI: 10.1016/j.antiviral.2020.104955
    38. 38
      Tiwari, S. K.; Wang, S.; Smith, D.; Carlin, A. F.; Rana, T. M. Revealing Tissue-Specific SARS-CoV-2 Infection and Host Responses using Human Stem Cell-Derived Lung and Cerebral Organoids. Stem Cell Rep. 2021, 16 (3), 437445,  DOI: 10.1016/j.stemcr.2021.02.005
    39. 39
      Li, N.; Hui, H.; Bray, B.; Gonzalez, G. M.; Zeller, M.; Anderson, K. G.; Knight, R.; Smith, D.; Wang, Y.; Carlin, A. F.; Rana, T. M. METTL3 regulates viral m6A RNA modification and host cell innate immune responses during SARS-CoV-2 infection. Cell Rep. 2021, 35 (6), 109091,  DOI: 10.1016/j.celrep.2021.109091
    40. 40
      Wang, S.; Li, W.; Hui, H.; Tiwari, S. K.; Zhang, Q.; Croker, B. A.; Rawlings, S.; Smith, D.; Carlin, A. F.; Rana, T. M. Cholesterol 25-Hydroxylase inhibits SARS-CoV-2 and other coronaviruses by depleting membrane cholesterol. EMBO J. 2020, 39 (21), e106057  DOI: 10.15252/embj.2020106057
    41. 41
      Ma, C.; Hu, Y.; Townsend, J. A.; Lagarias, P. I.; Marty, M. T.; Kolocouris, A.; Wang, J. Ebselen, Disulfiram, Carmofur, PX-12, Tideglusib, and Shikonin Are Nonspecific Promiscuous SARS-CoV-2 Main Protease Inhibitors. ACS Pharmacol Transl Sci. 2020, 3 (6), 12651277,  DOI: 10.1021/acsptsci.0c00130
    42. 42
      Weglarz-Tomczak, E.; Tomczak, J. M.; Talma, M.; Burda-Grabowska, M.; Giurg, M.; Brul, S. Identification of ebselen and its analogues as potent covalent inhibitors of papain-like protease from SARS-CoV-2. Sci. Rep. 2021, 11 (1), 3640,  DOI: 10.1038/s41598-021-83229-6
    43. 43
      Freitas, B. T.; Durie, I. A.; Murray, J.; Longo, J. E.; Miller, H. C.; Crich, D.; Hogan, R. J.; Tripp, R. A.; Pegan, S. D. Characterization and Noncovalent Inhibition of the Deubiquitinase and deISGylase Activity of SARS-CoV-2 Papain-Like Protease. ACS Infect. Dis. 2020, 6 (8), 20992109,  DOI: 10.1021/acsinfecdis.0c00168
    44. 44
      Shin, D.; Mukherjee, R.; Grewe, D.; Bojkova, D.; Baek, K.; Bhattacharya, A.; Schulz, L.; Widera, M.; Mehdipour, A. R.; Tascher, G.; Geurink, P. P.; Wilhelm, A.; van der Heden van Noort, G. J.; Ovaa, H.; Muller, S.; Knobeloch, K. P.; Rajalingam, K.; Schulman, B. A.; Cinatl, J.; Hummer, G.; Ciesek, S.; Dikic, I. Papain-like protease regulates SARS-CoV-2 viral spread and innate immunity. Nature 2020, 587 (7835), 657662,  DOI: 10.1038/s41586-020-2601-5
    45. 45
      Klemm, T.; Ebert, G.; Calleja, D. J; Allison, C. C; Richardson, L. W; Bernardini, J. P; Lu, B. G.; Kuchel, N. W; Grohmann, C.; Shibata, Y.; Gan, Z. Y.; Cooney, J. P; Doerflinger, M.; Au, A. E; Blackmore, T. R; Heden van Noort, G. J; Geurink, P. P; Ovaa, H.; Newman, J.; Riboldi-Tunnicliffe, A.; Czabotar, P. E; Mitchell, J. P; Feltham, R.; Lechtenberg, B. C; Lowes, K. N; Dewson, G.; Pellegrini, M.; Lessene, G.; Komander, D. Mechanism and inhibition of the papain-like protease, PLpro, of SARS-CoV-2. EMBO J. 2020, 39 (18), e106275  DOI: 10.15252/embj.2020106275
    46. 46
      Liu, G.; Lee, J.-H.; Parker, Z. M.; Acharya, D.; Chiang, J. J.; van Gent, M.; Riedl, W.; Davis-Gardner, M. E.; Wies, E.; Chiang, C.; Gack, M. U. ISG15-dependent activation of the sensor MDA5 is antagonized by the SARS-CoV-2 papain-like protease to evade host innate immunity. Nature Microbiology 2021, 6, 467478,  DOI: 10.1038/s41564-021-00884-1
    47. 47
      Jacobson, M. P.; Friesner, R. A.; Xiang, Z.; Honig, B. On the role of the crystal environment in determining protein side-chain conformations. J. Mol. Biol. 2002, 320 (3), 597608,  DOI: 10.1016/S0022-2836(02)00470-9
    48. 48
      Jacobson, M. P.; Pincus, D. L.; Rapp, C. S.; Day, T. J.; Honig, B.; Shaw, D. E.; Friesner, R. A. A hierarchical approach to all-atom protein loop prediction. Proteins: Struct., Funct., Genet. 2004, 55 (2), 351367,  DOI: 10.1002/prot.10613
    49. 49
      Donovan, S. F.; Pescatore, M. C. Method for measuring the logarithm of the octanol-water partition coefficient by using short octadecyl-poly(vinyl alcohol) high-performance liquid chromatography columns. J. Chromatogr A 2002, 952 (1–2), 4761,  DOI: 10.1016/S0021-9673(02)00064-X
    50. 50
      Box, K.; Bevan, C.; Comer, J.; Hill, A.; Allen, R.; Reynolds, D. High-throughput measurement of pKa values in a mixed-buffer linear pH gradient system. Anal. Chem. 2003, 75 (4), 883892,  DOI: 10.1021/ac020329y
  • Supporting Information

    Supporting Information


    The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.1c00566.

    • Molecular formula strings(CSV)

    • Expanded synthetic methods and spectra for E01–30, structures and reported activities for known SARS-CoV-2 main protease inhibitors, docking poses, and individual antiviral dose–response curves for select compounds

      (PDF)


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.