CoViTris2020 and ChloViD2020 A Striking PDF
CoViTris2020 and ChloViD2020 A Striking PDF
CoViTris2020 and ChloViD2020 A Striking PDF
https://doi.org/10.1007/s11030-020-10169-0
ORIGINAL ARTICLE
Received: 7 August 2020 / Accepted: 4 December 2020 / Published online: 3 January 2021
© The Author(s), under exclusive licence to Springer Nature Switzerland AG part of Springer Nature 2021
Abstract
Designing anticoronavirus disease 2019 (anti-COVID-19) agents is the primary concern of medicinal chemists/drug designers
nowadays. Repurposing of known active compounds against the severe acute respiratory syndrome coronavirus 2 (SARS-
CoV-2) is a new effective and time-saving trend in anti-COVID-19 drug discovery. Thorough inhibition of the coronaviral-2
proteins (i.e., multitarget inhibition) is a possible powerful favorable strategy for developing effectively potent drugs for
COVID-19. In this new research study, I succeeded to repurpose the two antioxidant polyhydroxy-1,3,4-oxadiazole com-
pounds CoViTris2020 and ChloViD2020 as the first multitarget coronaviral protein blockers with extremely higher potencies
(reach about 65 and 304 times, for CoViTris2020, and 20 and 93 times, for ChloViD2020, more potent than remdesivir and
favipiravir, respectively). These two 2,5-disubstituted-1,3,4-oxadiazoles were computationally studied (through molecular
docking in almost all SARS-CoV-2 proteins) and biologically assessed (through a newly established robust in vitro anti-
COVID-19 assay) for their anticoronaviral-2 bioactivities. The data obtained from the docking investigation showed that both
ligands promisingly exhibited very strong inhibitory binding affinities with almost all docked enzymes (e.g., they displayed
extremely lower binding energies of − 12.00 and − 9.60 kcal/mol, respectively, with the SARS-CoV-2 RNA-dependent
RNA polymerase “RdRp”). The results of the biological assay revealed that CoViTris2020 and ChloViD2020 significantly
displayed very high anti-COVID-19 activities (anti-SARS-CoV-2 E C50 = 0.31 and 1.01 μM, respectively). Further in vivo/
clinical studies for the development of CoViTris2020 and ChloViD2020 as anti-COVID-19 medications are required. In
brief, the ascent of CoViTris2020 and ChloViD2020 as the two lead members of the novel family of anti-COVID-19 poly-
phenolic 2,5-disubstituted-1,3,4-oxadiazole derivatives represents a promising hope in COVID-19 therapy.
* Amgad M. Rabie
amgadpharmacist1@yahoo.com; dr.amgadrabie@gmail.com
1
Dr. Amgad Rabie’s Research Lab. for Drug Discovery
(DARLD), Mansoura, Egypt
2
Pharmaceutical Organic Chemistry Department, Faculty
of Pharmacy, Mansoura University, Mansoura 35516, Egypt
3
Dikernis, Egypt
13
Vol.:(0123456789)
1840 Molecular Diversity (2021) 25:1839–1854
Graphic abstract
CoViTris2020 and ChloViD2020 inhibit SARS-CoV-2 life cycle with surprising E C50 values of 0.31 and 1.01 μM, respec-
tively. CoViTris2020 strongly inhibits coronaviral-2 RdRp with exceptionally lower inhibitory binding energy of − 12.00
kcal/mol.
13
Molecular Diversity (2021) 25:1839–1854 1841
13
1842 Molecular Diversity (2021) 25:1839–1854
COVID-19 therapy nonstructural proteins (nsps) and struc- practical anti-COVID-19 biological evaluation (preliminary
tural proteins (sps) present in SARS-CoV-2 and human (up in vitro/in vivo assays and subsequent preclinical/clinical
to date) are presented in Figs. 2 and 3, respectively. In this trials), molecular docking of the molecules of both com-
research paper, I report the successful repurposing of the two pounds in the target SARS-CoV-2 nsps and sps (along with
previously synthesized antioxidant 1,3,4-oxadiazole com- one target human sp, ACE2) has been carried out using the
pounds, CoViTris2020 and ChloViD2020, as effective and docking engines of the most known and credible interna-
potent anti-COVID-19 agents (as the first potent antidotal tional molecular docking software programs (e.g., Discovery
multitarget anti-SARS-CoV-2/anti-COVID-19 drugs). Studio, LeDock, GemDock, and GOLD). Different docking
software programs were used to confidently and doubtlessly
ensure the results and to assess and guarantee their repro-
Methods ducibility. Integrating the expected pharmacophoric features
with the interaction energy analysis disclosed functionally
Computational molecular docking studies pivotal amino acid residues in the binding pockets/cavities of
of CoViTris2020 and ChloViD2020 (predictive the active and/or allosteric sites of the target SARS-CoV-2
anti‑COVID‑19 properties evaluation) enzymes (mainly) along with in silico predicted common
inhibitory binding modes with the highly potent refer-
To expectedly assess the anti-COVID-19 activities of the ence repurposed compounds (e.g., remdesivir, ivermectin,
two target antioxidant compounds of the current research, and favipiravir). The docking results were very brilliant to
CoViTris2020 and ChloViD2020, prior to their actual encourage me to utilize a new specific web-based server
Fig. 2 A detailed illustration of all the discovered target nsps present in SARS-CoV-2 and involved in COVID-19 therapy (up to date)
13
Molecular Diversity (2021) 25:1839–1854 1843
Fig. 3 A detailed illustration of all the discovered target sps present in SARS-CoV-2/human and involved in COVID-19 therapy (up to date)
designed in 2020, which is specialized in precise molecu- therapy (previously presented in Figs. 2 and 3) were accu-
lar docking of all COVID-19 target nsps/sps and prediction rately opted to be targeted and docked. This includes fifteen
of anti-COVID-19 activities and potencies of tested com- various nCoV protein targets and one human protein tar-
pounds. For this objective, the newly programmed COVID- get (the human enzyme ACE2). For docking of only one
19 Docking Server was used [25]. small molecule, the “Docking” mode box should be spe-
COVID-19 Docking Server web-based software (Auto- cifically selected as the computational module (type) for
Dock Vina is used as the main docking engine) is an inter- every specific target (this is the selection used in the current
active web server for docking small molecules, peptides, case). To get the best accurate results, an average exhaus-
or antibodies against potential protein targets of COVID-19 tiveness option of “12” was used. The detailed results of
in order to predict the binding modes between COVID-19 these estimations (binding energies in kcal/mol) are shown
targets and the ligands along with screening and evaluat- in Table 1. After docking, the structure of each enzyme/
ing the anti-COVID-19 activities of these ligands (i.e., the protein-compound complex was further examined and accu-
platform provides a free interactive modern tool using a rately analyzed for characterization by the help of the fully
very precise knowledge-based scoring function to evalu- automated comprehensive interactive tool of the famous Pro-
ate the candidate binding poses for the specific prediction tein–Ligand Interaction Profiler (PLIP) web server (https
of COVID-19 target-ligand interactions and the following ://projects.biotec.tu-dresden.de/plip-web) [26], and results
drug discovery for COVID-19). Generally, almost all SARS- were marginally tabulated for comparison, explication of
CoV-2 nsps (enzymes) and very few SARS-CoV-2/human the previous docking results, and then placing conclusions
sps as COVID-19 therapy targets are recommended for (see Table 2 as an example).
small molecule docking. The structures of all the functional
or structural protein targets involved in the SARS-CoV-2 Anti‑COVID‑19 biological activities (in vitro assay)
replication life cycle were collected by direct downloading of CoViTris2020 and ChloViD2020
from international web databases (e.g., from the Protein
Data Bank “PDB”) or constructed based on their known This novel and highly reliable anti-COVID-19 in vitro
homologs of coronaviruses (by using homology modeling assay is based upon the authentic procedures of Chu and
module of Maestro 10, website: www.schrodinger.com), coworkers with very slight modifications [5, 27]. The com-
and completely prepared ready for direct docking on this plete procedures were carried out in a specialized biosafety
web-based software. The utmost effective and influenced level 3 (BSL-3) laboratory (SARS-CoV-2 is classified as a
nsps and sps (almost are functional enzymes) among all the BSL-3 pathogen by the WHO and FDA) in Hong Kong SAR
detected and recognized proteins involved in COVID-19 (China). The assayed SARS-CoV-2 virus, BetaCoV/Hong
13
1844 Molecular Diversity (2021) 25:1839–1854
Table 1 Score values of the sixteen computationally predicted (remdesivir, ivermectin, and favipiravir), respectively, using COVID-
pharmacological anti-COVID-19-related activities (against SARS- 19 Docking Server methodology (the table demonstrates the top
CoV-2 nsps/sps and human sp ACE2) of the target 1,3,4-oxadiazoles docking binding model score value, i.e., the least predicted binding
(CoViTris2020 and ChloViD2020) and the three reference drugs energy value, in kcal/mol for each compound with each target protein)
Classification SARS-CoV-2/Human target protein Top pose score value for docking of nCoV protein targets
Inhibitory binding energies/affinities (kcal/mol)
CoViTris2020 ChloViD2020 Remdesivir Ivermectin Favipiravir
(B1a form)
Table 2 Summary of the main active amino acid residues of chains A respectively (pivotal catalytic residues of the expected active site are
and C “nsp12/7” (of the SARS-CoV-2 RdRp) interacted with CoViT- shown in italics)
ris2020, ChloViD2020, and remdesivir (active form) molecules,
Compound SARS-CoV-2 RdRp amino acid residues
Hydrogen bonds Hydrophobic interactions π-Cation/Halogen interactions
(of all types)
CoViTris2020 Chain A: ARG553, TYR619, LYS621 (2 H Chain A: PRO620, ASP623, ARG624, LYS798 Chain A: ARG553
bonds), CYS622, ASP623, SER682, THR687
(2 H bonds), ALA688, ASN691, SER759 (2 H
bonds), ASP760, SER795, LYS798
ChloViD2020 Chain A: ASP623, ASN691, ASP846, LYS849; Chain A: MET87, LYS411, ASN414, LYS417; Chain A: LYS417, ASP418
Chain C: VAL12, SER15, GLN18, GLN19, Chain C: MET90
MET90
Remdesivir Chain A: ARG555 (2 H bonds), CYS622, – Chain A: ARG555
ASP623, SER682, THR687, ALA688,
ASN691, ASP760
Kong/VM20001061/2020, was isolated from the fresh naso- gallic acid), purified (> 97% purity), fully characterized, and
pharynx aspirate and throat swab of a confirmed fifty-years- put in small dark brown glass containers to be ready for
aged COVID-19 male patient in Hong Kong using Vero E6 the assay [20]. The pure three reference compounds were
cells (ATCC CRL-1586). Stock virus ( 107.25 TCID50/mL) purchased from MedChemExpress (remdesivir), Sigma-
was prepared after three serial passages in Vero E6 cells in Aldrich (ivermectin, B 1a form), and Toyama Chemical “Fuji-
infection media (DMEM supplemented with 4.5 g/L D-glu- film group, Japan” (favipiravir). The stocks of the five tested
cose, 100 mg/L sodium pyruvate, 2% FBS, 100,000 U/L compounds were accurately prepared by dissolving each of
Penicillin–Streptomycin, and 25 mM HEPES). Following them in dimethylsulfoxide “DMSO” (to get a concentration
the original procedures in the literature, CoViTris2020 and of 100 mM of each of CoViTris2020, ChloViD2020, rem-
ChloViD2020 compounds were synthesized (starting from desivir, ivermectin, and favipiravir). To evaluate the in vitro
13
Molecular Diversity (2021) 25:1839–1854 1845
Table 3 Anti-COVID-19 (anticoronaviral-2) activities (along with human/mammalian cells toxicities) of CoViTris2020 and ChloViD2020
(using remdesivir, ivermectin, and favipiravir, respectively, as the reference drugs) against SARS-CoV-2 in Vero E6 cells
Classification Compound Name CC50a Inhibition of SARS-CoV-2 in vitro (μM)
(μM)
100% CPE Inhibitory con- 50% Reduction in infec- 50% Reduction
centration (CPEIC100)b tious virus (EC50)c in viral RNA
copy (EC50)d
13
1846 Molecular Diversity (2021) 25:1839–1854
CoViTris2020 molecule powerfully binds to the respec- include ACE2, nsp16 (all sites), N protein (NCB site),
tive proteins forming very stable inhibited (deactivated) nsp3 (both sites), helicase (both sites), Mpro, and RdRp
complexes with relatively amazing binding energies which without RNA (RNA site) (Fig. 4b), respectively.
are the lowest among all (i.e., significantly lower than the ChloViD2020 comes second, among all the five com-
binding energies of all the other four compounds in their pounds, in its inhibitory binding energies and affinities
complexes with the respective nsps and sps). Specifically, which are ranged from − 7.50 to − 9.90 kcal/mol. The bind-
CoViTris2020 molecule gives its best inhibitory bind- ing affinities of ChloViD2020 surpass those of all the three
ing affinities with the three nsps RdRp-RNA (RTP site) reference drugs (except for those of each of exoribonuclease
(− 12.00 kcal/mol), nsp14 (N7-MTase) (− 11.40 kcal/ and endoribonuclease with remdesivir, as there is binding
mol), and PLpro (dimer) (− 10.60 kcal/mol), respectively energies difference of − 0.20 kcal/mol in favor of remdesi-
(Fig. 4a–d), which are three of the most effective protein vir in both cases), as the potent antioxidant ChloViD2020
targets in hindering and stopping the life cycle of the molecule strongly binds to the respective proteins forming
SARS-CoV-2 through inhibition and deactivation of their very stable inhibited complexes with relatively lower bind-
active sites [24]. These exceptional results are very prom- ing energies (i.e., significantly lower than the binding ener-
ising as they indicate the high possibility of CoViTris2020 gies of all the three reference molecules in their complexes
to be a very potent inhibitor (blocker) of RdRp, nsp14, with the respective nsps and sps). ChloViD2020 molecule
and/or PLpro. Other proteins affording very encouraging specifically gives its best inhibitory binding affinities with
excellent binding affinities with CoViTris2020 molecule the four nsps nsp3 (207-379, MES site) (− 9.90 kcal/mol),
Fig. 4 Screenshots of COVID-19 Docking Server outputs of the ous colors; Trace Style). c SARS-CoV-2 nsp14 “N7-MTase” (PDB
top predicted binding model of docking of CoViTris2020 molecule code: 5C8S, 1J53 “for active site homology”; colored with other
(colored pink) in: a SARS-CoV-2 RdRp-RNA “RTP site” (PDB code: various colors; Cartoon Style). d SARS-CoV-2 PLpro “dimer” (PDB
7BV2; colored with other various colors; Cartoon Style). b SARS- code: 6WUU; colored with other various colors; Trace Style)
CoV-2 RdRp “RNA site” (PDB code: 7BV2; colored with other vari-
13
Molecular Diversity (2021) 25:1839–1854 1847
RdRp-RNA (RTP site) (− 9.60 kcal/mol), nsp14 (N7-MTase) and conformational flexibility when compared to those of
(− 9.30 kcal/mol), and PLpro (dimer) (− 9.30 kcal/mol), the other or reference anticoronaviral drugs (e.g., favipiravir,
respectively (Fig. 5a–d). Interestingly, ChloViD2020 remdesivir, hydroxychloroquine, ivermectin, and arbidol).
approaches and reaches the lower binding affinity of CoViT- These extraordinary unique flexibilities of CoViTris2020
ris2020 in only one enzyme which is N7-MTase (it is also and ChloViD2020 structures are apparently observed in
one of the most effective protein targets in stopping the the docking poses in SARS-CoV-2 target proteins (along
coronaviral-2 life cycle through blocking of its active site with human target ACE2) as shown in both Figs. 6 and
[24]) with the same binding energy value of -9.90 kcal/mol. 7, respectively. CoViTris2020 molecule (a trisubstituted
These promising results indicate the significant possibility of or three armed bulky derivative of citric acid) has higher
ChloViD2020 to be a very potent inhibitor ligand or antago- degree of flexibility than ChloViD2020 molecule (a typical
nist of nsp3, RdRp, nsp14, and/or PLpro. Other proteins giv- 2,5-disubstituted derivative of 1,3,4-oxadiazole ring) due
ing very encouraging binding affinities with ChloViD2020 to, mainly, its larger topological polar surface area (TPSA)
molecule include ACE2, N protein (NCB site), helicase and its higher number of atoms. This highly balanced flex-
(both sites), nsp16 (all sites), Mpro, Nsp15 (endoribonu- ibility is generally required for excellent and extreme posi-
clease), and RdRp without RNA (RNA site), respectively. tioning of the drug molecule to be more superimposable in
The chemical structures of CoViTris2020 and the active binding pockets and cavities of any enzyme or
ChloViD2020 molecules are exceptionally charac- protein (i.e., required for extreme lock-and-key positioning
terized by higher degrees of balanced orientational of the drug molecule after hitting and striking any protein
Fig. 5 Screenshots of COVID-19 Docking Server outputs of the with other various colors; Cartoon Style). c SARS-CoV-2 nsp14
top predicted binding model of docking of ChloViD2020 molecule “N7-MTase” (PDB code: 5C8S, 1J53 “for active site homology”;
(colored pink) in: a SARS-CoV-2 nsp3 “207-379, MES site” (PDB colored with other various colors; Cartoon Style). d SARS-CoV-2
code: 6W6Y; colored with other various colors; Cartoon Style). b PLpro “dimer” (PDB code: 6WUU; colored with other various
SARS-CoV-2 RdRp-RNA “RTP site” (PDB code: 7BV2; colored colors; Trace Style)
13
1848 Molecular Diversity (2021) 25:1839–1854
13
Molecular Diversity (2021) 25:1839–1854 1849
◂Fig. 6 Collective screenshots of COVID-19 Docking Server out- large number of hydrogen bonds (of all types) which exceeds
puts of the top predicted binding models resulted from the docking sixteen bonds. According to the previous literature, CoViT-
of CoViTris2020 molecule (colored pink) in different SARS-CoV-2
proteins (colored with other various colors; trace and cartoon styles),
ris2020 can be considered the only compound that strongly
showing the extremely balanced high degrees of orientational and binds with the SARS-CoV-2 nsp12 in this extensively effec-
conformational flexibility of the molecule during the hitting attempts tive inhibitory mode, making it an optimal potent SARS-
against all target coronaviral-2 proteins CoV-2 RdRp inhibitor candidate. Furthermore, this poten-
tial unique property gives CoViTris2020 an extra advantage
molecule), leading to more adequate and potent inhibition of having potent blocking nature in its action on the active
(i.e., antagonism or blocking) of the actions (either cata- site of the polymerase. On the other hand, ChloViD2020
lytic or whatever) done or mediated by the protein. Conse- is the only inhibitor among all the investigated compounds
quently, the highly balanced flexibilities of CoViTris2020 that interacts with both chains A (nsp12) and C (nsp7) of
and ChloViD2020 molecules greatly increase their abilities the RdRp complex structure, since it, additionally, creates
to be very potent anti-COVID-19 agents, respectively. interaction forces of the hydrogen-bond/hydrophobic types
Remdesivir, computationally, comes first among the three with the polymerase cofactor chain C (mainly with the resi-
reference drugs (and third among all tested compounds in dues VAL12, SER15, GLN18, GLN19, and MET90). To
general) in anti-COVID-19 activities with binding ener- the best of my knowledge, ChloViD2020 can be predictably
gies range of − 7.00 to − 9.10 kcal/mol. Remdesivir shows considered one of the rarest ligands that interacts with and
its best binding affinity with the target enzyme nsp14 inhibits more than one protein chain of the SARS-CoV-2
(N7-MTase), as it binds to this important SARS-CoV-2 RdRp complex (i.e., acts through a dual mode of action).
enzyme with a very good binding energy of − 9.10 kcal/ ChloViD2020 is also characterized by its unique interaction
mol. Ivermectin and favipiravir come last, with their best forces created between its chlorine atom and the residues
binding affinities observed with the two coronaviral-2 LYS417 and ASP418 of chain A. Table 2 summarizes all the
enzymes nsp14 (N7-MTase; − 7.20 kcal/mol) and RdRp main active amino acid residues involved in the inhibitory
(RTP site; − 6.90 kcal/mol), respectively. interactions of CoViTris2020, ChloViD2020, and remde-
The results of the inhibitory binding affinities of CoViT- sivir molecules, respectively.
ris2020 with the SARS-CoV-2 RdRp (RTP site) are rela- In short, the previous computational results of the pre-
tively the best among all the COVID-19 targets, thus the next dicted binding modes of the two inhibitors CoViTris2020
goal was to extensively investigate the specific interactions and ChloViD2020 with the SARS-CoV-2/human proteins
with the amino acids of the active site(s) of this crucial poly- extremely comply with and support my suggested hypotheti-
merase in deeper details (using the data files obtained from cal multitarget mechanism of anti-COVID-19 action of each
the same web server). These deep investigations revealed the of the two ligands.
high degree of similarity of the expected anti-RdRp mode of
action of the two ligands CoViTris2020 and ChloViD2020 Anti‑COVID‑19 biological activities (in vitro assay)
as compared to that of remdesivir and its active metabo- of CoViTris2020 and ChloViD2020
lite (some of the interacting residues of the active pockets
are the same and some others are very close) as shown in The results demonstrated in Table 3 obviously and
Fig. 8a–c. It was also found that CoViTris2020 molecule directly revealed the extremely higher and amazing anti-
has an apparent superiority over ChloViD2020 and rem- COVID-19 effectiveness and potency of CoViTris2020 and
desivir (in its active metabolic form) molecules (together ChloViD2020 (as two of the most potent anti-SARS-CoV-2
with the other two reference molecules) in the strength of compounds ever) relative to those of each of the reference
inhibitory binding forces with RdRp (e.g., CoViTris2020 drugs. All the five tested compounds were found to inhibit
has at least sixteen hydrogen bonds, more than four hydro- SARS-CoV-2 replication in Vero E6 cells with E C50 under
phobic interactions, and at least one π-cation interaction; 100 μM. Surprisingly, CoViTris2020 (EC50 = 0.31 μM)
ChloViD2020 has at least nine hydrogen bonds, five hydro- was found to be about 65, 171, and 303.5 times more
phobic interactions, and two halogen interactions; while on potent than remdesivir (EC 50 = 20.17 μM), ivermectin
the other hand, remdesivir active metabolite (the most active (EC50 = 53.00 μM), and favipiravir (EC50 = 94.09 μM),
drug among the three references) has only less than nine respectively, in the anti-SARS-CoV-2 activity (in
hydrogen bonds, no considerable hydrophobic interactions, vitro). With the same interesting results, ChloViD2020
and only one π-cation interaction), supporting its expected (EC50 = 1.01 μM) was found to be about 20, 52.5, and 93
promising comprehensive anti-SAR-CoV-2 activities. times more potent than remdesivir, ivermectin, and favi-
Exceptionally, CoViTris2020 is the only inhibitor among piravir, respectively, in the same assay. According to this
all the investigated compounds that strongly interacts with assay, CC50 of CoViTris2020 and ChloViD2020 is much
the RdRp complex structure (especially chain A) with that larger than 100 μM, thus both compounds are expected
13
1850 Molecular Diversity (2021) 25:1839–1854
Fig. 7 Collective screenshots of COVID-19 Docking Server out- showing the extremely balanced high degrees of orientational and
puts of the top predicted binding models resulted from the docking conformational flexibility of the molecule during the hitting attempts
of ChloViD2020 molecule (colored pink) in different SARS-CoV-2 against all target coronaviral-2 proteins
proteins (colored with other various colors; trace and cartoon styles),
13
Molecular Diversity (2021) 25:1839–1854 1851
Fig. 8 The inhibitory binding interactions, of a CoViTris2020; b ChloViD2020; c Remdesivir (active metabolite form), with the active amino
acids of the SARS-CoV-2 RdRp (2D and 3D representations, respectively)
to have very high safety margin and clinical selectiv- We should put into consideration the possibility that
ity index (SI; SI = CC50/EC50, SICoViTris2020 > 322.58 and CoViTris2020 and ChloViD2020 may undergo prior intra-
SIChloViD2020 > 99.01), while on the other hand, the refer- cellular metabolic transformation into more active forms
ences remdesivir, ivermectin, and favipiravir are expected (e.g., their triphosphate forms) by human cellular enzymes
to have narrow safety margin and clinical therapeutic index (e.g., kinases and transferases), which may differ among
(SIRemdesivir > 4.96, SIIvermectin > 1.89, and SIFavipiravir > 1.06). various cell types, thus evaluation of the pharmacological
CoViTris2020 and ChloViD2020 are also having interest- actions of these two target compounds in primary human
ingly very small values of the concentration that causes airway epithelial cells will clearly facilitate the interpreta-
100% inhibition of the SARS-CoV-2 cytopathic effects tion and clarification of the previous results. The metabolic
in vitro (CoViTris2020 has the best CPEIC100 value, among activation would almost add extra anti-COVID-19 activi-
all the tested compounds, of 0.99 μM, directly followed by ties to the two drugs through incorporation of human cell-
ChloViD2020 which has CPEIC100 value of 1.97 μM) and biocompatible and human cell-bioavailable moieties into
of the concentration that is required for 50% reduction in the chemical structures of both of them, and this would
the number of SARS-CoV-2 RNA copies in vitro (CoViT- consequently increase the net clinical effectiveness and
ris2020 has the best EC50 value, among all the tested com- efficacy of these two potent compounds. Importantly, the
pounds, of 0.33 μM, directly followed by ChloViD2020 three reference drugs (remdesivir, ivermectin, and favipira-
which has EC50 value of 1.23 μM). vir) are currently undergoing extensive and broad clinical
13
1852 Molecular Diversity (2021) 25:1839–1854
Fig. 8 (continued)
trials as anti-SARS-CoV-2/anti-COVID-19 agents world- (comprising the desirable advantages of both potent SARS-
wide. The very high values of CC50 of CoViTris2020 and CoV-2 inhibitors) in further assays (in vivo) and preclinical/
ChloViD2020 indicate that both compounds would be pre- clinical trials. Almost all the factual experimental results
dictably well tolerated in the human body. The significantly obtained and concluded, here, in the anti-COVID-19 anti-
desirable high values of SI (i.e., the extremely minute values viral biological evaluation are complying with the previous
of anti-SARS-CoV-2 E C50 along with the very high values of speculative theoretical results suggested and extracted from
mammalian cells C C50) of CoViTris2020 and ChloViD2020 the anti-SARS-CoV-2 computational molecular pharmaco-
reflect that both compounds evidently favor resistant RNA logical predictions for the two newly repurposed biologically
virus over DNA virus and mammalian cells, and this, in reevaluated compounds, CoViTris2020 and ChloViD2020,
turn, expectedly indicates and expresses the selective speci- and the three reference drugs.
ficity of these two compounds as anti-COVID-19 drugs
(specifically, CoViTris2020 can be considered as a unique
superpowerful SARS-CoV-2 antidote/killer). CoViT- Conclusions
ris2020 is apparently more potent and more promising than
ChloViD2020 as SARS-CoV-2 inhibitor. Using a combina- Thinking outside the box is very important to win our cur-
tion formula of CoViTris2020 and remdesivir is a possible rent challenge against the scary COVID-19 pandemic.
good experimental choice, as it may have exceptional and Potent multiblockade of, mainly, the novel SARS-CoV-2
striking combinational synergistic anti-COVID-19 action proteins including enzymes and receptors could be seen
13
Molecular Diversity (2021) 25:1839–1854 1853
as one of the most effective fertile approaches for compre- passes the in vivo bioassays and then the preclinical/clini-
hensive COVID-19 therapy through designing, discover- cal trials with highly effective and satisfactorily significant
ing, and searching for multitarget SARS-CoV-2 inhibitors, results as anti-COVID-19 agent, a possible combination
thus much efforts in drug discovery in 2020 were focused therapy (e.g., as an oral tablet/capsule, an intravenous/
on trying to successfully repurpose known drugs and com- intramuscular parenteral vial/ampoule, or a nasal/oral/ocu-
pounds in order to effectively inhibit this extremely resist- lar prophylactic gel/drops) with a second highly potent old
ant coronavirus. My specific efforts led to the discovery of anti-RNA virus drug, such as remdesivir, may be a recom-
two very promising potent multitarget SARS-CoV-2 inhibi- mended available choice for ultimate COVID-19 treatment
tors through the successful reevaluation and repurposing in the near future. In brief, in this new research paper, the
of the known antioxidant 1,3,4-oxadiazole compounds, antioxidant CoViTris2020 and ChloViD2020 molecules
previously synthesized by me, CoViTris2020 (1,2,3-tris[5- were successfully reevaluated, repurposed, and reported as
(3,4,5-trihydroxyphenyl)-1,3,4-oxadiazol-2-yl]propan-2-ol) very promising hit molecules (they could also be considered
and ChloViD2020 (5-[5-(7-chloro-4-hydroxyquinolin- as the first extremely potent anticoronaviral-2 polyphenolic
3-yl)-1,3,4-oxadiazol-2-yl]benzene-1,2,3-triol), which effec- 1,3,4-oxadiazole compounds “Coronavirus-2 Killers”) with
tively inhibited SARS-CoV-2 life cycle with IC50 values of general multitarget and very potent successful inhibition
0.31 (according to the used anti-COVID-19 assay, CoViT- against SARS-CoV-2 enzymes (mainly), and consequently,
ris2020 is the most potent SARS-CoV-2 inhibitor discov- both compounds are two of the first known promising under-
ered till now) and 1.01 μM, and amazingly presented about investigation candidate drugs for the effective and complete
65-/171-/303.5-fold and 20-/52.5-/93-fold anti-SARS-CoV-2 treatment of COVID-19.
activities and potencies more than remdesivir/ivermectin/
favipiravir, respectively. On the other hand, the discovery of Acknowledgements I gratefully thank and deeply acknowledge anyone
who gave a hand to make this new discovery and work coming out to
the very potent anti-COVID-19 activities of CoViTris2020 light.
and ChloViD2020 molecules through the successful bio-
logical reevaluation and repurposing opens the door for us Compliance with ethical standards
to establish the first class of anti-COVID-19 agents of the
type “polyphenolic 1,3,4-oxadiazoles” which will specifi- Conflict of interest I hereby declare that I totally have no known com-
cally comprise a series of 2,5-disubstituted-1,3,4-oxadiazole peting financial interests or personal relationships that could have ap-
derivatives (beginning with the first two effective members, peared to influence the work reported in this new research paper.
CoViTris2020 and ChloViD2020). Prior extensive com-
putational molecular studies showed that CoViTris2020
and ChloViD2020 have the ideal and balanced values of
the pharmacokinetic and druglikeness parameters required References
to be effectively potent anti-COVID-19 drugs inside the
1. Hui DS, Azhar EI, Madani TA, Ntoumi F, Kock R, Dar O,
human body. Computational molecular modeling analysis Ippolito G, Mchugh TD, Memish ZA, Drosten C, Zumla A,
of the best inhibitory docking binding modes of CoViT- Petersen E (2020) The continuing 2019-nCoV epidemic threat
ris2020 and ChloViD2020 molecules with the SARS- of novel coronaviruses to global health—the latest 2019 novel
CoV-2/human proteins showed that the 3,4,5-trihydroxy- coronavirus outbreak in Wuhan, China. Int J Infect Dis 91:264–
266. https://doi.org/10.1016/j.ijid.2020.01.009
phenyl moieties greatly increase the blocking affinities 2. Li JY, You Z, Wang Q, Zhou ZJ, Qiu Y, Luo R, Ge XY (2020)
and potencies at the active and/or allosteric sites of the The epidemic of 2019-novel-coronavirus (2019-nCoV) pneumo-
SARS-CoV-2/human enzymes or proteins (binding energies nia and insights for emerging infectious diseases in the future.
reach − 12.00 kcal/mol for CoViTris2020 and − 9.90 kcal/ Microbes Infect 22:80–85. https: //doi.org/10.1016/j.micin
f.2020.02.002
mol for ChloViD2020) when compared to those of the three 3. Jiang S, Du L, Shi Z (2020) An emerging coronavirus causing
references (which lack this effective antioxidant 3,4,5-tri- pneumonia outbreak in Wuhan, China: calling for developing
hydroxyphenyl moiety), remdesivir, ivermectin, and favi- therapeutic and prophylactic strategies. Emerg Microbes Infect
piravir (binding energies maximally reach − 9.10, − 7.20, 9:275–277. https://doi.org/10.1080/22221751.2020.1723441.
Erratum: Jiang S (2020) Correction. Emerg Microbes Infect
and − 6.90 kcal/mol, respectively). Surprisingly, CoViT- 9:539. https://doi.org/10.1080/22221751.2020.1737363
ris2020 surpassed ChloViD2020 and the three moderately 4. Dong L, Hu S, Gao J (2020) Discovering drugs to treat coronavi-
to highly potent reference drugs in the values of almost all rus disease 2019 (COVID-19). Drug Discov Ther 14:58–60. https
compared computational and experimental anti-COVID-19 ://doi.org/10.5582/ddt.2020.01012
5. Choy KT, Wong AY, Kaewpreedee P, Sia SF, Chen D, Hui KPY,
parameters, scores, and activities. Specifically, CoViT- Chu DKW, Chan MCW, Cheung PP, Huang X, Peiris M, Yen
ris2020 and ChloViD2020 give their best inhibitory binding HL (2020) Remdesivir, lopinavir, emetine, and homoharringto-
affinities results with the catalytic active site of the SARS- nine inhibit SARS-CoV-2 replication in vitro. Antiviral Res
CoV-2 RdRp. If the CoViTris2020 compound successfully 178:104786. https://doi.org/10.1016/j.antiviral.2020.104786
13
1854 Molecular Diversity (2021) 25:1839–1854
6. Elfiky AA (2020) Anti-HCV, nucleotide inhibitors, repur- 18. Abdul Amin S, Jha T (2020) Fight against novel coronavirus: a
posing against COVID-19. Life Sci 248:117477. https://doi. perspective of medicinal chemists. Eur J Med Chem 201:112559.
org/10.1016/j.lfs.2020.117477 https://doi.org/10.1016/j.ejmech.2020.112559
7. Elfiky AA (2020) Ribavirin, remdesivir, sofosbuvir, galidesivir, 19. Goyal B, Goyal D (2020) Targeting the dimerization of the main
and tenofovir against SARS-CoV-2 RNA dependent RNA poly- protease of coronaviruses: a potential broad-spectrum therapeu-
merase (RdRp): a molecular docking study. Life Sci 253:117592. tic strategy. ACS Comb Sci 22:297–305. https://doi.org/10.1021/
https : //doi.org/10.1016/j.lfs.2020.11759 2 . Erratum: Elfiky acscombsci.0c00058
AA (2020) Corrigendum. Life Sci 258:118350. https: //doi. 20. Rabie AM, Tantawy AS, Badr SMI (2016) Design, synthe-
org/10.1016/j.lfs.2020.118350 sis, and biological evaluation of novel 5-substituted-2-(3,4,5-
8. Shannon A, Le NT, Selisko B, Eydoux C, Alvarez K, Guillemot trihydroxyphenyl)-1,3,4-oxadiazoles as potent antioxidants. Am
JC, Decroly E, Peersen O, Ferron F, Canard B (2020) Remdesivir J Org Chem 6:54–80. https://doi.org/10.5923/j.ajoc.20160602.02
and SARS-CoV-2: structural requirements at both nsp12 RdRp 21. Ishida T (2019) Review on the role of Zn2+ ions in viral pathogen-
and nsp14 exonuclease active-sites. Antiviral Res 178:104793. esis and the effect of Zn2+ ions for host cell-virus growth inhibi-
https://doi.org/10.1016/j.antiviral.2020.104793 tion. Am J Biomed Sci Res 2:28–37. https://doi.org/10.34297/
9. Wang M, Cao R, Zhang L, Yang X, Liu J, Xu M, Shi Z, Hu Z, AJBSR.2019.02.000566
Zhong W, Xiao G (2020) Remdesivir and chloroquine effectively 22. te Velthuis AJ, van den Worm SH, Sims AC, Baric RS, Snijder
inhibit the recently emerged novel coronavirus (2019-nCoV) EJ, van Hemert MJ (2010) Zn2+ inhibits coronavirus and arterivi-
in vitro. Cell Res 30:269–271. https://doi.org/10.1038/s4142 rus RNA polymerase activity in vitro and zinc ionophores block
2-020-0282-0 the replication of these viruses in cell culture. PLoS Pathog
10. Yin W, Mao C, Luan X, Shen DD, Shen Q, Su H, Wang X, Zhou 6:e1001176. https://doi.org/10.1371/journal.ppat.1001176
F, Zhao W, Gao M, Chang S, Xie YC, Tian G, Jiang HW, Tao 23. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ (1997)
SC, Shen J, Jiang Y, Jiang H, Xu Y, Zhang S, Zhang Y, Xu HE Experimental and computational approaches to estimate solubil-
(2020) Structural basis for inhibition of the RNA-dependent RNA ity and permeability in drug discovery and development settings.
polymerase from SARS-CoV-2 by remdesivir. Science 368:1499– Adv Drug Deliv Rev 23:3–25. https://doi.org/10.1016/S0169
1504. https://doi.org/10.1126/science.abc1560 -409X(96)00423-1
11. Caly L, Druce JD, Catton MG, Jans DA, Wagstaff KM (2020) 24. Wu C, Liu Y, Yang Y, Zhang P, Zhong W, Wang Y, Wang Q, Xu
The FDA-approved drug ivermectin inhibits the replication of Y, Li M, Li X, Zheng M, Chen L, Li H (2020) Analysis of thera-
SARS-CoV-2 in vitro. Antiviral Res 178:104787. https://doi. peutic targets for SARS-CoV-2 and discovery of potential drugs
org/10.1016/j.antiviral.2020.104787 by computational methods. Acta Pharm Sin B 10:766–788. https
12. Cai Q, Yang M, Liu D, Chen J, Shu D, Xia J, Liao X, Gu Y, Cai ://doi.org/10.1016/j.apsb.2020.02.008
Q, Yang Y, Shen C, Li X, Peng L, Huang D, Zhang J, Zhang S, 25. COVID-19 Docking Server Web-based Software, COVID-19
Wang F, Liu J, Chen L, Chen S, Wang Z, Zhang Z, Cao R, Zhong Docking Server (Homepage on the Internet), Shan Change Lab.,
W, Liu Y, Liu L (2020) Experimental treatment with favipiravir Institute of Bioinformatics and Medical Engineering, School of
for COVID-19: an open-label control study. Engineering 6:1192– Electrical and Information Engineering, Jiangsu University of
1198. https://doi.org/10.1016/j.eng.2020.03.007 Technology, Changzhou 213001, China, 2020; Available from
13. Du YX, Chen XP (2020) Favipiravir: pharmacokinetics and con- COVID-19 Docking Server on the Web (homepage: http://ncov.
cerns about clinical trials for 2019-nCoV infection. Clin Pharma- schanglab.org.cn); Results obtained through using interactive
col Ther 108:242–247. https://doi.org/10.1002/cpt.1844 docking tool in COVID-19 Docking Server (Copyright© 2018–
14. Shiraki K, Daikoku T (2020) Favipiravir, an anti-influenza drug 2023, Shan Chang; Version 2020) on this website (accessed and
against life-threatening RNA virus infections. Pharmacol Ther cited in 2020, 11–22 June), and references cited therein.
209:107512. https://doi.org/10.1016/j.pharmthera.2020.107512 26. Salentin S, Schreiber S, Haupt VJ, Adasme MF, Schroeder M
15. Derwand R, Scholz M (2020) Does zinc supplementation enhance (2015) PLIP: fully automated protein-ligand interaction profiler.
the clinical efficacy of chloroquine/hydroxychloroquine to win Nucleic Acids Res 43:W443–W447. https://doi.org/10.1093/nar/
today’s battle against COVID-19? Med Hypotheses 142:109815. gkv315
https://doi.org/10.1016/j.mehy.2020.109815 27. Chu DKW, Pan Y, Cheng SMS, Hui KPY, Krishnan P, Liu Y, Ng
16. Liu J, Cao R, Xu M, Wang X, Zhang H, Hu H, Li Y, Hu Z, Zhong DYM, Wan CKC, Yang P, Wang Q, Peiris M, Poon LLM (2020)
W, Wang M (2020) Hydroxychloroquine, a less toxic derivative Molecular diagnosis of a novel coronavirus (2019-nCoV) causing
of chloroquine, is effective in inhibiting SARS-CoV-2 infec- an outbreak of pneumonia. Clin Chem 66:549–555. https://doi.
tion in vitro. Cell Discov 6:16. https://doi.org/10.1038/s4142 org/10.1093/clinchem/hvaa029
1-020-0156-0 28. Zhang L, Lin D, Kusov Y, Nian Y, Ma Q, Wang J, von Brunn
17. Wang X, Cao R, Zhang H, Liu J, Xu M, Hu H, Li Y, Zhao L, Li W, A, Leyssen P, Lanko K, Neyts J, de Wilde A, Snijder EJ, Liu H,
Sun X, Yang X, Shi Z, Deng F, Hu Z, Zhong W, Wang M (2020) Hilgenfeld R (2020) α-Ketoamides as broad-spectrum inhibitors
The anti-influenza virus drug, arbidol is an efficient inhibitor of of coronavirus and enterovirus replication: structure-based design,
SARS-CoV-2 in vitro. Cell Discov 6:28. https://doi.org/10.1038/ synthesis, and activity assessment. J Med Chem 63:4562–4578.
s41421-020-0169-8 https://doi.org/10.1021/acs.jmedchem.9b01828
13