You are on page 1of 20

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/349846647

Development of adamantane scaffold containing 1,3,4-thiadiazole


derivatives: Design, synthesis, anti-proliferative activity and molecular
docking study targeting EGFR

Article  in  Bioorganic Chemistry · March 2021


DOI: 10.1016/j.bioorg.2021.104794

CITATION READS

1 37

6 authors, including:

Yousry Ahmed Ammar Gameel A M Elhag Ali


Al-Azhar University Al-Azhar University
190 PUBLICATIONS   2,188 CITATIONS    40 PUBLICATIONS   298 CITATIONS   

SEE PROFILE SEE PROFILE

Amany Belal Ahmed B M Mehany


Taif University Al Azhar univerisity
39 PUBLICATIONS   361 CITATIONS    47 PUBLICATIONS   326 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

COVID-19 View project

New designed compounds with double target View project

All content following this page was uploaded by Gameel A M Elhag Ali on 22 April 2021.

The user has requested enhancement of the downloaded file.


Bioorganic Chemistry 110 (2021) 104794

Contents lists available at ScienceDirect

Bioorganic Chemistry
journal homepage: www.elsevier.com/locate/bioorg

Development of adamantane scaffold containing 1,3,4-thiadiazole


derivatives: Design, synthesis, anti-proliferative activity and molecular
docking study targeting EGFR
Mohammed M.S. Wassel a, Yousry A. Ammar b, *, Gameel A.M. Elhag Ali b, Amany Belal c, d,
Ahmed B.M. Mehany e, Ahmed Ragab b, *
a
Department of Foot and Mouth Disease, Veterinary Serum and Vaccine Research Institute (VSVRI), Abbasia, Cairo, Egypt
b
Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, 11884 Cairo, Egypt
c
Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
d
Medicinal Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
e
Zoology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt

A R T I C L E I N F O A B S T R A C T

Keywords: A new series of 1,3,4-thiadiazolo-adamantane derivatives were synthesized through molecular hybridization
Adamantane approach, then used as starting material to synthesize chloro and cyano acetamide-thiadiazole derivatives 2, 3.
1,3,4-Thiadiazole derivatives The newly designed compounds 1–3 were treated with different reagents to design 5-adamantyl thiadiazole
Anti-proliferative activity
derivatives 4–17 and evaluate their in vitro anti-proliferative activity against three cancer cell lines (MCF-7,
EGFR
Apoptosis and molecular docking
HepG-2 and A549). Doxorubicin was used as a positive control. The most promising compounds 5, 6, 10a, 10b,
14b, 14c, and 17 showed up-regulation for BAX and down-regulation of Bcl-2, these findings proved their role as
hopeful apoptotic inducers. In addition, the inhibitory activity against both wild EGFRWT and mutant EGFRL858R-
TK
for these derivatives revealed that compounds 5, 14c, and 17 have IC50 value ranging from 85 nM to 71.5 nM
against wild EGFRWT and 37.85–41.19 nM against the mutant type, Lapatinib was used as a reference standard
with IC50 values of 31.8 nM and 39.53 nM, respectively. The most potent derivatives were subjected to further
evaluation against double mutant EGFR L858R/T790M and showed good IC50 values between (0.27–0.78 nM)
compared to Lapatinib (0.18 nM) and Erlotinib (0.21 nM). Among them, thiazolo-thiadiazole adamantane de­
rivative 17 exhibited the strongest inhibitory activity to the EGFR. Molecular docking studies were performed
inside the active site of EGFR (1M17), and binding energy scores ranged between (− 19.19 to –22.07 Kcal/mol)
compared to Erlotinib (− 19.10 Kcal/mol). Furthermore, oral bioavailability beside some pharmacokinetics
properties of these derivatives were also investigated in this research work.

1. Introduction increased anticancer drug resistance and decreased apoptosis capacity


[8–13]. Hence, inhibition of apoptosis will play a major role in cancer
Cancer is emerging as one of the fastest-growing diseases in the progression during carcinogenesis. There are different types of molec­
world and a leading cause of death following cardiovascular diseases ular pathways used by the cancer cells to inhibit apoptosis [12]. BAX
[1–3]. Cancer is the world’s second-largest cause of death, accounting and Bcl-2 are respectively apoptotic and anti-apoptotic proteins, and the
for 9.6 million deaths in 2018, with approximately one in nine deaths ratio of those two proteins determines the frequency in apoptosis in cells
due to cancer [4,5]. Current cancer care approaches include surgery, [14–17]. An important determinant of cell survival is the relationship
radiotherapy, and chemotherapy either alone or in combination, how­ between the apoptotic and anti-apoptotic levels [18]. EGFR belongs to a
ever, metastasis (spread of disease to other areas in the body) is still family of four related receptor tyrosine kinases including EGFR (HER1/
representing a challenge [6]. DNA-damaging agents, such as radiation ErbB1), ErbB2 (HER2/neu), ErbB3 (HER3), and ErbB4 (HER4) that act
and chemotherapeutic drugs, can induce programmed cell death as a key mediator in cell signaling pathways involving cell proliferation,
(apoptosis) [7]. Many studies have shown a close association between apoptosis, angiogenesis, and metastatic spread [19,20]. Although each

* Corresponding authors.
E-mail addresses: yossry@azhar.edu.eg (Y.A. Ammar), ahmed_ragab@azhar.edu.eg (A. Ragab).

https://doi.org/10.1016/j.bioorg.2021.104794
Received 9 June 2020; Received in revised form 28 February 2021; Accepted 1 March 2021
Available online 5 March 2021
0045-2068/© 2021 Published by Elsevier Inc.
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

member of the EGFR family is the potent mediator of normal cell growth including human beings [50,51] (Fig. 1).
and development, overexpression of EGFR appears in various human Molecular hybridization has become an appealing technique for the
tumors [21,22]. The advantage of the adamantyl group confers addi­ rational design of anticancer agents with improved affinity and efficacy
tional potency in drugs in which it was present. The property underlined in comparison with parent drugs and increased activity [52]. The hybrid
was the lipophilicity of adamantane which increases the ease with, drug is a new term in development which involves combining two or
which the compound reaches the activity site as well as the bulkiness more pharmaceutical cores to produce a new hybrid drug which can
property that indicates a steric factor is also necessary at the activity site interact with multiple targets or minimize side effects using single-target
for the interactions[23]. Adamantane derivatives have several enticing agents [53,54]. Based on the previous findings and our ongoing efforts
pharmacological activities, such as antibacterial [24,25], antifungal to develop a new heterocyclic compound with biological activity
[26], antiviral [27–29], antidiabetic [30], carbonic anhydrase inhibitors [55–59]. It was conceived that binding more than one pharmacophores
[31], and anticancer effects [32–35]. Amantadine and many similar as adamantane and thiadiazole moieties to design one scaffold using
compounds with three-dimensional ‘box-like’ adamantane structure are drug design strategy would be of great interest developing potent anti­
employed in the treatment of certain neurological disorders [36]. It was cancer agents as described previously [60,61]. Nowadays, the creation
found that N-1-adamantylcitraconimide (ACI) (I), N-1-ada­ of hybrid molecules is a phenomenon that aims to combine multiple
mantylmaleimide (AMI) (II) and dimethyl adamantyl maleimide pharmacophore fragments in the same molecule with different biolog­
(DMAMI) (III) exhibited modest growth-inhibitory activity against four ical potentials [62–68,84,85]. Our study’s rationale came up from the
cancer cell lines (Colo 205, HepG-2, SK-BR-3 and Molt-4). Also, AMI was great importance and synthetic accessibility of adamantly-thiadiazole
also successful in inhibiting the growth of both in vitro and in vivo hybrids incorporating some amide, azomethane, amine, pyridine de­
human gastric cancer cells and induced in vitro apoptosis [37]. Dimethyl rivatives, and some new fused ring with thiadiazole as imidazo and
adamantyl maleimide (DMAMI), the AMI derivative, induces apoptosis pyrimidino derivatives. The synthesized derivatives were evaluated as
and inhibits the development of human colon cancer Colo 205 in SCID anti-proliferative activity against three cell lines, mitochondrial
mice [38]. In addition, 1,3,4-thiadiazoles are heterocyclic scaffolds apoptosis by regulating BAX and Bcl-2 proteins’ expression, and EGFR
found in several compounds that have various pharmacological activ­ inhibition. Besides, study cell cycle, apoptosis assay, and finally, some
ities as such as fungicidal [39], insecticidal [40], anticonvulsant [41], physicochemical and pharmacokinetic properties as well as molecular
anti-tuberculosis [42], anticancer [43,44], antiviral [45], antibacterial docking study for the most active derivatives were determined hoping to
[46]. The wide range of pharmaceutically suitable biological activities is generate potential anticancer molecules.
due to the existence of the NCS group, a hydrogen-binding domain with
two-electron donor systems, as well as sulfur atom that enhances the 2. Results and discussion
liposolubility of the molecule [47–49]. The broad and wide biological
activities of 1,3,4-thiadiazole derivatives are due to the aromaticity of 2.1. Chemistry
the ring, which also provides great in vivo stability to this five-
membered ring system and low toxicity for higher vertebrates, The synthetic procedures adopted to obtain the target compounds

Fig. 1. Rationale design for the target molecules.

2
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

are depicted in Schemes 1–3. The starting compound, 5-(adamantan-1- that exchangeable by D2O, in addition to three signals at δ 8.03, 7.59,
yl)-1,3,4-thiadiazol-2-amine (1) was prepared according to the previ­ and 7.49 ppm for aromatic protons. The 13C NMR spectrum exhibited
ously reported procedure [69]. Thus, 2-aminothiadiazole derivative 1 signals for carbonyl carbon at δ 174.22 and aromatic at δ 133.31,
was subjected to chloroacetylation through its reaction with chlor­ 132.27, 129.72, 129.08, 129.02, and 128.80 ppm as well as two singlet
oacetyl chloride in the presence of trimethylamine as a catalyst to afford signal at δ 167.78 and 159.17 ppm for two C– – N groups in thiadiazole
the corresponding 2-chloro acetamide derivative 2. Structure of the moiety. Condensation of compound 1 with isatin in boiling ethanol
latter product was confirmed based on analytical and spectral data. The furnished Schiff’s base 5 in a good yield. The chemical structure of
IR spectrum showed absorption bands at ν 3160, 2905, 2849, and 1714 compound 5 was elucidated based on elemental analysis and spectral
cm− 1 due to NH, aliphatic-CH and C– – O groups. Its 1H NMR spectrum data. The IR spectrum indicated the presence of a strong absorption
showed two singlet signals at δ 12.79 and 4.42 ppm corresponding to NH band at 1727 cm− 1 due to the carbonyl group. The 1H NMR spectrum
and CH2 groups. The adamantane protons were observed at δ 2.06, 2.01, showed the presence of indolinone part at 11.02, 7.48, 7.39, 6.96, and
and 1.76 ppm. 13C NMR for this compound exhibited signals at δ 174.6, 6.83 ppm for NH and four aromatic protons. Although, its 13C NMR
165.66, 157.88, and 43.12 ppm corresponding for C– – O, two C–– N, and exhibited signals at 168.89, 168.27, 138.84, 130.84, 125.13, 123.21,
CH2 groups respectively, while the adamantane-C were observed at δ 118.28, and 112.71 ppm due to the indolinone core.
42.76, 37.77, 36.28, and 28.30 ppm. Furthermore, we investigated the reactivity of 2-aminothiadiazole
In addition, N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-cyanoa­ derivative 1 towards some halogenated compounds to prepare imi­
cetamide (3) was obtained through cyano-acetylation of compound 1 dazo[2,1-b][1,3,4]thiadiazole derivatives. Thus, the reaction of thia­
with 3-(3,5-dimethyl-1H-pyrazol-1-yl)-3-oxopropane-nitrile in toluene. diazole derivative 1 with 3-chloroacetylacetone and ethyl 3-
Elemental analysis, IR, 1H NMR, and 13C NMR are in agreement with the chloroacetoacete in ethanol containing piperidine as a catalyst for
proposed structure. Its IR spectrum exhibited two new bands at 2263 obtaining the cyclic structure but a cyclic structures 6 and 7 were ob­
and 1720 cm− 1 due to CN and C– – O respectively. 1H NMR spectrum tained. Structure of the latter products was confirmed on the basis of
displayed a singlet signal at δ 4.07 ppm attributed to CH2. The 13C NMR correct elemental analyses and spectral data. The IR spectrum of com­
spectrum characterized signals at δ 174.48, 115.63, and 56.48 ppm pound 6 indicated the presence of two strong absorption bands at 1699
assigned to the C–– O, CN, and CH2 groups, respectively. and 1676 cm− 1 due to two carbonyl groups, while the carbonyl of ester
N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)benzamide (4) was ob­ group in compound 7 was observed at 1698 cm− 1. The 1H NMR spec­
tained by reaction of 2-aminothiadiazole derivative 1 with benzoyl trum of compound 6 showed the presence of NH proton at δ 9.48 and
chloride under reflux condition. The IR spectrum of compound 4 showed three singlet signals at δ 3.10, 2.15, and 2.04 ppm corresponding for two
a new absorption band at 1670 cm− 1 due to the carbonyl group. Its 1H methyl groups and methine proton in addition to the expected signals for
NMR spectrum revealed a singlet signal at δ 12.62 ppm due to NH proton thiadiazole adamantane moiety. In the similar manner compound 7

Scheme 1. Synthesis of thiadiazole amide, indolin-2-one and acyclic derivatives containing adamantane pharmacophore.

3
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

Scheme 2. Reaction of 2-chloroacetamide with a different reagent to produce imidazo[2,1-b]thiadiazol, substituted amide derivatives as well as thiazole containing
thiadiazolo-adamantane amine moiety.

Scheme 3. Condensation and cyclization reactions of cyno-acetamide derivative into pyrimidinone, aryl cyan-acrylamide, pyridine, chromenone and thiazolidinone
derivatives containing adamantyl thiadiazole moiety.

showed signals at δ 2.87 and 3.03 ppm for methine proton and methyl ethyl group carbons appear at δ 77.82, 60.56, and 14.66 ppm as well as
group in addition to ester group that observed as quartet and triplet at δ adamantane carbons.
4.23 and 1.45 ppm with coupling constant (J = 6.8 Hz). In addition, its N-(5-(Adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-chloroacetamide (2)
13
C NMR exhibited signals at δ 175.15, 168.29, 158.92, and 151.27 ppm is a versatile reagent and has been extensively used as a synthetic in­
for two carbonyls and C–– N respectively. Moreover, methine carbon and termediate for the synthesis of thiadiazole derivatives of potential

4
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

biological activity. It was thus of interest to study the reactivity of δ 3.88 ppm due to OCH3 protons beside signals in the region of δ 7.15 to
compound 2 towards a variety of chemical reagents. Thus, upon 8.39 ppm for Aromatic and NH2 protons as well as the expected ada­
refluxing of compound 2 in dioxane containing trimethylamine as a mantane protons that appear as three singlet signals at δ 1.74, 1.99 and
catalyst caused self-cyclization to furnish the corresponding 2-(ada­ 2.18 ppm.
mantan-1-yl)imidazo[2,1-b][1,3,4]thiadiazol-6-one (8). The chemical Reactivity of cyano-acetamide derivative with the 1,3-dicarbonyl
structure of compound 8 was established on the basis of its elemental compound was studied in the aim of formation of pyridine derivatives
analysis and spectral data. Its 1H NMR spectrum displayed two singlet with potential biological activities [70,71]. Thus, compound 3 reacted
signals at δ 12.82, and 4.05 ppm for hydroxy and methine proton and its with acetylacetone to give the pyridine derivative 15. Structure of the
13
C NMR spectrum was characterized by signal at δ 174.61 assigned to latter product was based on analytical and spectral data. The IR spec­
the carbon attached to the hydroxyl group. Amination of compound 2 trum of compound 15 showed the exchangeable NH band and exhibited
with piperidine as secondary amine and p-anisidine as well as p-tolui­ two absorption bands at 2223 and 1668 cm− 1 due to CN and C– –O
dine as primary amine afforded the corresponding amino derivatives groups. The 1H NMR spectrum revealed the appearance of three new
compounds 9 and 10. The structure of the amino products 9, and 10 singlets at δ 2.09, 2.42 and 6.56 ppm assigned to two methyl protons and
were verified by elemental analyses and spectroscopic methods. For the pyridinone H-5. Furthermore, cyclocondensation of compound 3
example, 1H NMR spectrum of compound 10a displayed aromatic pro­ with salicylaldehyde in boiling ethanol containing a catalytic amount of
ton signals which appeared as two doublets with coupling constant (J = piperidine afforded the coumarin derivative 16. The IR spectrum of
8.8 Hz) at 6.71, and 6.52 ppm. Addition to four singlet signals at δ 12.40, compound 16 showed the absence of cyano band that already present in
5.67, 3.98, and 3.62 ppm characteristic for the 2NH, CH2, and OCH3, the starting compound and the formed product exhibited the presence of
respectively, besides the expected protons of adamantane moiety and its two strong absorption bands at 1712 and 1695 cm− 1 which prove the
13
C NMR showed signals at δ 174.08, 170.65, 157.89, 55.74 and 47.41 formation chromone ring. Its 1H NMR spectrum revealed the absence of
ppm corresponding to C– – O, two C– – N, OCH3, and CH2 groups. Treat­ methylene signal and the presence of two singlet signals at δ 8.99 and
ment of 2-chloro acetamide derivative 2 with ammonium thiocyanate in 4.35 ppm due to chromone H-4, NH of amide derivatives. Besides, the
absolute ethanol afforded the thiazolidinone derivative 11 containing presence of aromatic and adamantane protons. Its 13C NMR spectrum is
thiadiazolo-adamantane Scheme 2. The IR spectrum of compound 11 an agreement with the expected structure.
revealed a strong absorption band at 1740 cm− 1 due to thiazolidine Finally, treatment of cyano-acetanilide thiadiazole derivative 3 with
carbonyl function. Its 1H NMR revealed one D2O exchangeable proton at phenyl isothiocyanate in DMF, in alkaline medium followed by treat­
δ 12.24 and a singlet signal at δ 4.07 ppm due to CH2 protons. Moreover, ment with methyl chloroacetate afforded the corresponding thiazolidi­
its 13C NMR exhibited signals at δ 177.10, 174.49, and 66.82 ppm for none derivative 17. The structure of thiazolidinone containing
two carbonyl group moreover endocyclic methylene group of thiazol-4- thiadiazolo-adamantane 17 was established by the appearance of a
one derivative. strong absorption band at 1732 cm− 1 for new carbonyl formed in the IR
Furthermore, we also investigated the reactivity of N-(5-(ada­ spectrum. This band displayed a strong confirmation for the thiazolidi­
mantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-cyanoacetamide (3) towards none nucleus formation. Another evidence for the cyclization occurred is
some selective reagents with the aim of preparing some bioactive de­ the presence of a singlet signal at δ 4.05 ppm in the 1H NMR spectrum
rivatives. Thus, compound 3 underwent cyclization when heated under equivalent to two protons for endocyclic methylene as well as signals at
reflux in dioxane to afford thiadiazolo[3,2-a]pyrimidin-7-one derivative δ 7.13 to 7.52 ppm related to phenyl group and in 13C NMR spectra
12. The structure of 12 was confirmed based on its elemental analysis aromatic carbons displayed between δ 124.05 to 136.44 ppm.
and spectral data. The IR spectrum of compound 12 showed absorption
bands at 3156 and 1718 cm− 1 indicating the presence of NH and C– –O 2.2. Biological evaluations
groups, respectively and absent of the cyan-group. Moreover, its 1H
NMR showed two singlets at δ 12.80 and 4.42 ppm due to the presence of 2.2.1. Effect of 1,3,4-thiadiazole derivatives on anti-proliferative activity
NH and CH2 protons. Furthermore, 13C NMR exhibited the presence of and SAR study
C–– O and CH2 carbons at δ 174.57 and 46.72 ppm, respectively in The in vitro anti-proliferative activity of twenty-one 1,3,4-thiadiazole
addition to the expected carbons of the thiadiazolo-adamantane moiety. derivatives, 2 to 17 containing adamantane moiety were evaluated
Condensation of cyano-acetanilide thiadiazole derivative 3 with against a panel of three cell lines including mammary gland breast
some aromatic aldehydes namely 4-methoxy or 4-chloro or 4-fluoro cancer cell line (MCF-7), human hepatocellular carcinoma cell line
benzaldehyde afforded the corresponding acrylamide derivatives 13a- (HepG-2) as well as human lung adenocarcinoma epithelial cells (A549)
c. The chemical structure of 13 was assigned on the basis of the spectral by using sulforhodamine B colorimetric (SRB) assay according to re­
and elemental analyses. The IR spectrum of compound 13a as an ported methods [17,72]. Doxorubicin (DOX) is one of the commonly
example showed absorption bands at 3175 and 1652 cm− 1 due to NH used small molecular anti-cancer drugs, was selected to be the reference
and amidic carbonyl functions, respectively. The 1H NMR spectrum of drug. The in vitro cytotoxicity of all the synthetized compounds are
compound 13a revealed the lack of CH2 signal and the presence of one summarized in Table 1 and IC50 values expressed by (μM) that means the
singlet signal exchangeable with D2O at δ 13.89 ppm due to NH proton, concentration of compound or drug required to inhibit the growth of
beside a singlet signal at δ 8.38 for –
– CH methine and two doublets at δ cancer cells by 50%.
8.05 and δ 7.16 ppm with coupling constant (J = 8.8 Hz) for aromatic The resulting data from Table 1 showed that adamantane-containing
protons and singlet signal at δ 3.88 corresponding for OCH3. In addition, 1,3,4-thiadiazole had moderate to potent growth inhibitory effect
13
C NMR of compound 13a exhibited signals at δ 177.11, 115.41, and against tested cell lines (MCF-7, HepG-2 and A549) with IC50 values
56.17 ppm corresponding for C– – O, CN and OCH3 carbons over the ranged between (5.71 ± 0.37 μM to 21.47 ± 1.5 μM), (4.28 ± 0.35 μM to
presence of aromatic and adamantane carbons. On the other hand, the 17.38 ± 1.35 μM) and (4.11 ± 0.28 μM to 16.31 ± 1.2 μM) μM in
interaction of cyano-acetamide derivative 3 with arylidenemalononi­ comparison to Doxorubicin (8.19 ± 0.72 μM, 7.46 ± 0.12 μM and 3.58
triles in the presence of piperidine affected cyclization to yield the ± 0.03 μM), respectively. Compounds 2, 3, 4, 9, 10a, and 10b that
corresponding pyridine carbonitrile derivatives 14a-c. The structure of containing thiadiazole having amide derivatives in position two showed
compound 14 was confirmed on the basis of its elemental analysis and that aromatic acetamide 10a, 10b revealed the highest activity de­
spectral data. The IR spectrum of 14a as the example showed absorption rivatives followed by benzamide, piperidinyl, chloro and cyanoaceta­
bands at 3314, 3172, 2223, and 1706 cm− 1 indicating the presence of mide derivatives (10a ˃ 10b ˃ 4 ˃ 9 ˃ 2 ˃ 3), and 4-methoxy phenyl
amino, two CN, and carbonyl groups, respectively. The 1H NMR spec­ acetamide derivative 10a was more active than 4-methyl derivative 10b
trum revealed the lack of CH2 signal and the presence of singlet signal at with IC50 values (6.44 ± 0.52 μM, 4.92 ± 0.35 μM, and 4.61 ± 0.31 μM)

5
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

Table 1 cells. The most potent compounds showed IC50 values from 89.75 to
Cytotoxic screening of the newly designed compounds 2–17 and Doxorubicin 225.7 μM. From Table 2, the selectivity index (SI) data indicated that
against three cancer cell lines (IC50 values are expressed in μM ± S.E). adamantane core that attached to 1,3,4-thiadiazole derivatives exhibits
Compound no. IC50 μM ± S.E* a high degree of cytotoxic selectivity. For instance, compound 6 with
MCF-7 HepG-2 A 549
pentane-2,3-dione alkyl group attached to the amino of 2-aminothiadia­
zole adamantane derivatives exhibited the safest compound with IC50
2 14.32 ± 1.13 11.46 ± 0.93 10.67 ± 0.84
(225.7 μM) and highest SI (26.84, 36.70 and 39.12) respectively.
3 17.44 ± 1.32 15.72 ± 1.1 16.31 ± 1.2
4 12.37 ± 0.97 9.45 ± 0.76 10.18 ± 0.82 Simultaneously, N-1,2,4-thiadiazolo-di-cyanopyridine derivative 14c
5 7.27 ± 0.68 5.83 ± 0.42 4.26 ± 0.35 with 4-chlorophenyl moiety in position four of pyridine showed IC50
6 8.41 ± 0.74 6.15 ± 0.5 5.77 ± 0.4 (89.75 μM) with SI (15.72, 20.97, and 21.84) to the tested cell line
7 18.22 ± 1.41 15.37 ± 1.05 13.81 ± 0.98 respectively.
8 11.28 ± 0.95 10.17 ± 0.92 9.74 ± 0.86
9 17.55 ± 1.23 13.64 ± 0.97 11.37 ± 0.95
10a 6.44 ± 0.52 4.92 ± 0.35 4.61 ± 0.31 2.2.1.2. Effect of compounds on mitochondrial apoptosis by regulating the
10b 9.31 ± 0.85 7.67 ± 0.64 5.74 ± 0.44 expression of BAX and Bcl-2 proteins. The effect of the new designed
11 14.61 ± 1.1 10.36 ± 0.91 9.38 ± 0.84 compounds on the induced apoptotic process via mitochondria medi­
12 15.84 ± 1.14 13.49 ± 0.97 10.75 ± 0.85
13a 12.45 ± 0.98 10.77 ± 0.95 8.49 ± 0.75
ated pathway up-regulation of BAX and down-regulation of Bcl-2 was
13b 17.62 ± 1.35 14.92 ± 1.14 15.02 ± 1.16 evaluated and the obtained data are represented in Table 3. Treatment
13c 10.45 ± 0.93 8.22 ± 0.7 6.75 ± 0.55 of human lung adenocarcinoma epithelial cells (A549) with the most
14a 12.41 ± 1.1 10.37 ± 0.92 9.12 ± 0.76 promising compounds 5, 6, 10a, 10b, 14b, 14c, and 17 expressed high
14b 8.16 ± 0.69 6.75 ± 0.57 5.79 ± 0.41
levels of BAX and lower levels of Bcl-2 compared to untreated cells.
14c 5.71 ± 0.37 4.28 ± 0.35 4.11 ± 0.28
15 17.39 ± 1.4 14.89 ± 1.1 12.55 ± 0.95 From Table 3, the condensation of 2-aminothiadiazole with isatin to
16 21.47 ± 1.5 17.38 ± 1.35 14.62 ± 1.2 produce thiadiazole containing 2-oxoindole 5 exhibited the highest
17 6.51 ± 0.48 4.47 ± 0.3 4.32 ± 0.25 apoptosis induced by (BAX = 415.84 pg/mL with 8.57 folds) than un­
Dox. 8.19 ± 0.72 7.46 ± 0.12 3.58 ± 0.03 treated cells, followed by enaminonitrile containing thiadiazole 14c
*
Three independent experiments were performed for each tested compound (BAX = 411.32 pg/mL, 8.47 folds), and thiadiazole hybridized with N-
for each concentration. phenyl thiazole 17 (BAX = 375.19 pg/mL, 7.73 folds) and the other
derivatives induced apoptosis from 6.13 to 7.02 folds higher than con­
(9.31 ± 0.85 μM, 7.67 ± 0.64 μM, and 5.74 ± 0.44 μM), respectively. trol. By the same way, the most promising derivative among the thia­
Thiazol-4-one derivatives 11 and 17 that were obtained from chloro and diazole derivatives that demonstrated the highest value in suppressing
cyanoacetamide of thiadiazole adamantane derivatives by reacting them apoptosis against BCL-2 is thiadiazole containing enaminonitrile deriv­
with different reagents, and it was found that N-phenyl thiazol-4-one ative 14c with Bcl-2 value 1.42 pg/mL (4.13 folds). Whereas compounds
derivative 17 exhibited cytotoxic activity higher than its analogue 11 5, 6, 10a, 10b, 14b, and 17 showed decrease in Bcl-2 levels from 3.06
with IC50 values ranged between (4.32 ± 0.25 μM to 6.51 ± 0.48 μM), pg/mL to 1.75 pg/mL that decreased the level by 1.92 to 3.35 folds.
and in comparison, to Doxorubicin (3.58 ± 0.03 μM to 8.19 ± 0.72 μM). Our study indicated that compounds 5, 6, 10a, 10b, 14b, 14c, and
Amino thiadiazole derivatives with diacetyl derivative 6 demonstrated a 17 exhibited chemo suppression/induction cancerous cells to apoptosis
significant activity than 2-aminothiazole 7 with ethyl acetoacetate core induction by changes in Bcl-2 and BAX gene levels, depending on the
where IC50 values (8.41 ± 0.74, μM 6.15 ± 0.5 μM, and 5.77 ± 0.4 μM) variety of substituents that were attached to 1,3,4-thiadiazolo
and (18.22 ± 1.41 μM, 15.37 ± 1.05 μM, and 13.81 ± 0.98 μM) against adamantane.
the tested cell line (MCF-7, HepG-2, and A549), respectively. That is due
to the presence of ethyl acetate group that decrease activity than acetyl 2.2.1.3. Effect of compounds on EGFR inhibition. Epidermal growth
group. factor receptor (EGFR) are considered as an attractive and clinically
2-Cyanoacrylamide 1,3,4-thiadiazolo-adamantane derivatives 13a validated drug targets in cancer therapy because it is important for
to 13c showed moderate activity and chloro derivatives showed to be various biological processes and cancers development and progression,
the most active member with IC50 values (10.45 ± 0.93 μM, 8.22 ± 0.7 such as cell proliferation, adhesion, migration, differentiation, and
μM, and 6.75 ± 0.55 μM) for MCF-7, HepG-2, and A549 cell lines, survival. EGFR can cause dysregulation by two different mechanisms
respectively. N-thiadiazole pyridine derivatives 14 and 15 exhibited one of them is the high expression of EGFR that lead to increase in re­
variable activity, and the presence of dicyanopyridine derivative 14 ceptor signaling output. In contrast, the second mechanism involves
exhibited more activity than mono-cyano derivative 15, as well as the overexpression of the ligand and therefore increase in EGFR signaling
presence of aryl derivatives in compounds 14a to 14c. Also, the presence activity despite normal or even low levels of receptor expression
of chloro-derivative caused higher activity than both fluro- and methoxy [73–75]. Depending on the results of antiproliferative activity, we
derivatives in position four in dicyanopyridine derivative 14. Simulta­ selected the most promising compounds 5, 6, 10a, 10b, 14b, 14c and 17
neously, the 3-amide-2-oxo-chromene derivative 16 decreased the po­ for EGFR enzymatic activity assays by using A549 cancer cell to
tency than other adamantane derivatives.
Finally, seven compounds 5, 6, 10a, 10b, 14b, 14c, and 17 with 5-
(adamantan-1-yl)-1,3,4-thiadiazole backbone core showed potent cyto­ Table 2
toxicity with low micromolar (<10 μM) and most of these derivatives IC50 values in μM ± S.E. for WI-38 cell line for the most promising compounds.
showed IC50 lower than Doxorubicin on both MCF-7 and HepG-2 and Cpd. No. IC50 μM WI-38 Cells SI*
nearly to the reference drug against A 549 cell line with growth inhib­ MCF-7 HepG-2 A 549
itory ranged from 4.11 μM to 5.79 μM, besides, compounds 8, 11, 13a,
5 97.55 ± 0.68 13.42 16.73 22.90
13c, and 14a with IC50 values from 6.75 μM to 9.74 μM. 6 225.7 ± 1.25 26.84 36.70 39.12
10a 128.39 ± 0.98 19.94 26.10 27.85
2.2.1.1. Effects of compounds on normal cells. The cytotoxic activities of 10b 178.41 ± 0.67 19.16 23.26 31.08
the new promising compounds 5, 6, 10a, 10b, 14b, 14c and 17 were 14b 185.9 ± 1.75 22.78 27.54 32.11
14c 89.75 ± 0.24 15.72 20.97 21.84
evaluated on normal non-cancer cells (WI-38 cells) to determine the 17 112.75 ± 0.55 17.32 25.22 26.10
selectivity of the 2-amino-1,3,4-thidiazole derivatives against cancer
*
SI: Selectivity index = (IC50 of WI-38)/(IC50 of the tested cancer cell line).

6
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

Table 3 except for 6, and 14b against EGFR L858R-TK.


Effect of compounds 5, 6, 10a, 10b, 14b, 14c, and 17 on expression of BAX and To study the most promising three derivatives 5, 14c, and 17 against
Bcl-2 that expressed by IC50 values in pg/mL ± S.E. for apoptosis markers. further kinase EGFR L858R/T790M double mutant with Lapatinib and
Cpd. No Bax (Pg/mL) Fold Bcl2 (Pg/mL) Fold Erlotinib as reference drugs (both drugs are known to target EGFR so
5 415.84 ± 3.53 8.57 1.75 ± 0.12 3.35
that they were selected to act as positive controls in this assay) and as
6 331.75 ± 2.19 6.83 2.54 ± 0.24 2.31 expected they inhibited the enzymatic activity against EGFR L858R/T790M
10a 340.55 ± 1.98 7.02 2.19 ± 0.17 2.64 with IC50 values ranged between (0.27 nM to 0.78 nM) compared to
10b 329.71 ± 2.98 6.79 2.71 ± 0.36 2.16 Lapatinib (0.18 nM) and Erlotinib (0.22 nM). Thiazolidinone derivative
14b 297.46 ± 3.10 6.13 3.06 ± 0.19 1.92
17 exhibited the highest value with 0.28 nM in comparison to enami­
14c 411.32 ± 1.25 8.47 1.42 ± 0.11 4.13
17 375.19 ± 2.65 7.73 1.88 ± 0.27 3.12 nonitrile pyridine derivative 14c (0.47 nM) and Schiff bases 5 (0.79
A549 cells 48.51 ± 1.04 1 5.87 ± 0.31 1 nM), and that may be due to presence of thiazolidinone moiety beside
thiadiazolo-adamantane backbone core.

elucidate the mechanism of these active compounds.


2.2.1.4. Cell cycle analysis. Depending on the antiproliferative activ­
From Table 4, the results of wide type EGFR on A549 cancer cells
ities, BAX, Bcl-2 gene expression as well as single and double mutant
expressed as the half-maximal inhibitory concentration IC50 (μM). All
EGFR results obtained previously. The most promising three derivatives
the promising compounds were investigated in EGFRWT inhibitory assay
5, 14c, and 17 were selected and investigated for cell cycle progression
and showed values <2 μM. The 3,5-dicyano-pyridine that containing 4-
and the apoptosis percentage induced by the help of adamantane de­
chlorophenyl derivatives as well as backbone core 1,3,4-thiadiazole
rivatives on human lung adenocarcinoma epithelial cells A549 cell when
adamantane 14c exhibited the most potent activity with IC50 value
treated with 5 μM. The obtained data represented in Table 5 and Fig. 2
85 nM while the fluro derivative 14b showed to be the least inhibitor
and Fig. 3.
towards EGFR in comparison to the tested compounds and Lapatinib
It is observed after 24 h; that the tested compounds exhibited PreG1
that used as a positive control due to its declared activity as a drug that
apoptosis and cell growth arrest in A549 at G2/M phase. Thiadiazole
can target EGFR, its IC50 (31.8 nM) against EGFRWT. Schiff base deriv­
containing both isatin or thiazole as hybrid molecule derivatives 5 and
ative 5 of 1,3,4-thiadiazole adamantane with isatin displayed 50% in­
17 beside adamantane core causes accumulation of A549 cell at G0-G1
hibition of EGFRWT expression at 35.2 nM. Moreover, compound 17
phases by 41.29% and 39.58%, respectively as well as, increasing G2/M
bearing 2-cyano-2(N-phenylthiazolidin-2-ylidene-4-one)acetamide de­
phase percentage by 26.64% and 26.29% in comparison to untreated
rivatives were found to be active inhibition (IC50 = 71.5 nM) against
cells 14.38%. While, enaminonitrile pyridine derivatives that are having
overexpression of EGFRWT. Compounds 6, 10a, and 10b inhibited the
both p-chlorophenyl moiety and adamantyl thiadiazole core revealed
enzymatic activity against EGFRWT with IC50 values (1.32, 0.954, and
cell cycle arrest by 42.09%, 29.67%, and 28.24% for G2/M, S, and G0-
1.27 μM).
G1 phases respectively. Also, enaminonitrile pyridine derivative 14c
Due to the importance of EGFR that can be defined as member of the
cause apoptosis and detain the cell cycle in both G2/M and preG1 by
membrane-bound receptor tyrosine kinase family and when activated,
74.50%. Although, both thiadiazole adamantane derivatives 5 and 17
transduce mitogenic signals [48]. The most promising compounds were
causes increase the content of DNA in G0-G1 and S phases by nearly
selected for further in vitro mutant EGFR L858R-TK that were expressed by
73.36% and 73.71%, respectively. Surprisingly, compounds 5, 14c, and
IC50 (nM) to evaluate the activity and selectivity of newly designed
17 induced apoptosis at pre-G1 phases by ranged from 16.59% to
compounds. Compound 14c showed the most potent inhibitory activity
32.41% in comparison to A549 cell by 1.86%.
against EGFR L858R-TK with IC50 (37.85 nM) in comparison to Lapatinib
Finally, the thiadiazolo-adamantane derivatives with different sub­
as a positive control (IC50 = 39.53 nM). Both compounds 5 and 17
stituent 5, 14c and 17 used as inhibitors for cancer cell and induced
showed equipotent to the standard drug with IC50 values (41.19 nM, and
apoptosis in different phases.
40.58 nM), respectively compared to Erlotinib (59.56 nM). Chlor­
oacetamide thiadiazole derivatives 2, when reacted with different aro­
2.2.1.5. Effect of compounds on apoptotic assay. To determine the
matic amines (4-methoxy and 4-methyl) to produce acetamide
mechanism of cell death and apoptosis-inducing effect double staining
derivative with aromatic core reveled activity with 52.11 nM and 55.28
Annexin V/propidium iodide was used through follow cytometry
nM with 30% less than Lapatinib and still having inhibitory activity
method [16,17], where apoptosis can be defined as an active and regular
better than Erlotinib 59.56 nM. Surprisingly, compound 14b demon­
pathway. At the same time, necrosis is a passive and accident cell death
strated the least inhibition towards EGFRWT, the observed IC50 = 70.49
[76]. Three thiadiazolo-adamantane derivatives 5, 14c, 17 were treated
nM against EGFR L858R-TK with nearly 56% less than the positive control.
with 5 μM and incubated for 24 h on A549 cell, and the obtained data
Most of the tested compounds reveled IC50 values higher than Erlotinib
showed in Table 6 and Fig. 4.
We detected an increase in the total percentage of apoptosis because
of the adamantane derivatives that exhibited total apoptosis ranged
Table 4
IC50 values in μM ± S.E. for EGFR kinases assay for the active compounds.
from 16.59% to 32.41% as compared to 1.86% for standard A549 cells.
By the same way, the enaminonitrile pyridine derivative 14c was the
Cpd. No. Enzyme inhibitory activity IC50 *
highest induction of total cell apoptosis (32.41%) by nearly 17.42 folds.
EGFRWT (μM) EGFR L858R-TK (nM) EGFR L/T**

5 0.0352 ± 0.008 41.19 ± 3.08 0.788


Table 5
6 1.323 ± 0.024 63.48 ± 2.89 –
Cell cycle analysis by percentage in different phases after treatment the lung
10a 0.954 ± 0.014 52.11 ± 3.54 –
10b 1.274 ± 0.026 55.28 ± 2.98 – cancer cell A549 with 10 μM of thiadiazolo-adamantane derivatives 5, 14c, and
14b 1.961 ± 0.023 70.49 ± 2.71 – 17.
14c 0.085 ± 0.010 37.85 ± 1.54 0.467 Compound No. Results
17 0.0715 ± 0.019 40.58 ± 1.25 0.278
Lapatinib 0.0318 ± 0.014 39.53 ± 1.62 0.182 %G0-G1 %S %G2/M %Pre-G1
Erlotinib 0.0656 ± 0.009 59.56 ± 2.31 0.218 5 41.29 32.07 26.64 19.11
*
Data are average of three independent results. 14c 28.24 29.67 42.09 32.41
** 17 39.58 34.13 26.29 16.59
EGFR L/T = double mutant EGFR L858R/T790M, (− ) mean not
cont. A549 46.93 38.69 14.38 1.86
determined.

7
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

Fig. 2. Diagram illustrated the percentage of cell cycle arrest for the most potency thiadiazole-adamantane derivatives 5, 14c, and 17 at different phases.

Moreover, the percentage in early-stage varies from 2.61%, 5.13% and the active site revealed binding energy between –22.07 to − 19.19 Kcal/
8.26% for 5, 17, and 14c, respectively. mol higher than Erlotinib (Figs. 6–9 and Table 8). Hydrazone containing
Finally, the effect of three derivatives 5, 17, and 14c treated with 2-oxo-indole as well as thiadiazolo-adamantane derivative demonstrate
A549 cell lead to an increase in the sensitivity to apoptosis by different one hydrogen bond donor from side chain Asp831 with NH of isatin
percentage in different stages and therefore increased total apoptosis moiety through bond length (2.54 ◦ A) and displayed binding energy S =
percentage and these data support the higher values on antiproliferative − 19.19 Kcal/mol (Figs. 7a and 7b). Enaminonitrile pyridine derivative
against A549 cell (<5 μM). Besides, the thiazolo-thiadiazole ada­ 14c showed the highest binding energy score S = –22.07 Kcal/mol with
mantane derivative 17 exhibited the most active derivative (Fig. 5). one hydrogen bond backbone acceptor between Met769 (the same
amino acid binding with Erlotinib) with cyano group in enaminonitrile
2.2.1.6. In silico studies derivative with bond length 3.18◦ A (22%) (Figs. 8a and 8b). Hybrid
2.2.1.6.1. Evaluation of physicochemical, and pharmacokinetic pre­ structure 17 that containing N-phenyl thiazole beside backbone moiety
diction on active compounds. Physicochemical parameters (oral (thiadiazolo-adamantane) displayed binding energy S = − 21.78 Kcal/
bioavailability) for both Lipinski’s and Veber rules were determined for mol through two amino acid residue Asp 831 and Lys721 with bond
the most promising compounds as well as Lapatinib and Erlotinib using length 2.81◦ A (45%), 2.60◦ A (51%) respectively (Figs. 9a and 9b).
Swiss ADME: a free web tool [77]. From data in Table 7, the most active Finally, the most promising derivatives investigated binding energy
compounds 5, 14c, and 17 obey RO5 criteria without no violations from higher than the co-crystalized inhibitor (Erlotinib) as well as ada­
Lipinski’s Rule, and both enaminonitrile 14c and thiazolo-thiadiazole mantane moiety displayed hydrophobic interaction inside the active site
adamantane derivative 17 failed to pass through Veber rule by Topo­ of pocket.
logical polar surface area (TPSA) higher than 140 Å2. For two standard
drugs, Erlotinib pass in both rule of five and Veber rule while Lapatinib 3. Conclusion
met the RO5 with one violation in MW > 500 and passed in Veber rule.
By the same way, some Pharmacokinetic properties were studies A novel series of twenty-one 1,3,4-thiadiazolo-adamantane de­
depending on Swiss ADME: a free web tool involving gastrointestinal rivatives were designed and synthetized starting from 5-(adamantan-1-
absorption and blood brain barrier. Compounds 14c and 17, as well as yl)-1,3,4-thiadiazol-2-amine (1). The newly synthetized derivatives 2 to
Lapatinib, showed low gastrointestinal absorption with no permeation 17 were evaluated in vitro for their anti-proliferative activity against the
to blood brain barrier while Schiff’s base 5 and Erlotinib exhibited high selected three cancer cell lines. The structure of the new designed
absorption to gastrointestinal track. Furthermore, Erlotinib pass through compounds was established and confirmed based on its elemental
the blood brain barrier but the most promising displayed no promotion. analysis and spectral data. Cytotoxic values showed a broad spectrum of
Finally, our new compounds exhibited not only promising activity, the designed compounds against the tested cell lines (MCF-7, HepG-2
but also some oral bioavailability beside pharmacokinetic properties as and A 549), and seven compounds 5, 6, 10a, 10b, 14b, 14c and 17
Lapatinib and Erlotinib. with 5-(adamantan-1-yl)-1,3,4-thiadiazole backbone core showed
2.2.1.6.2. Effect of compounds on molecular docking study. To potent cytotoxicity with low IC50 values (<10 μM). Moreover, the most
explore the possible binding conformation and got deeper insights of the potent compounds showed IC50 values from 89.75 to 225.7 μM against
structure activity relationship as well as understanding the potency of normal non-cancer cells WI-38. Besides, they have exhibited chemo
the newly designed thiadiazolo-adamantane derivatives 5, 14c and 17 suppression/induction to cancers cells e.g. apoptosis induction by
molecular docking simulation was performed with the ATP binding making changes in Bcl-2 and BAX gene levels. All promising compounds
pocket of EGFR PDB (1M17) [78]. From Table 8, it was found the co- investigated EGFRWT inhibitory assay<2 μM. Besides, compound 14c
crystalized ligand 4-anilinoquinazoline inhibitor (Erlotinib) exhibited showed the most potent inhibitory activity against EGFR L858R-TK with
self-docking process with small RMSD = 1.99 ◦ A and binding energy S = IC50 (37.85 nM) in comparison to Lapatinib as a positive control (IC50 =
− 19.10 Kcal/mol, through one hydrogen bond backbone acceptor with 39.53 nM) and Erlotinib (IC50 = 59.56 nM). Both compounds 5 and 17
Met769 with the nitrogen of quinazoline with bond length 3.13 ◦ A and showed equipotent activity to the standard drug with IC50 values (41.19
bound strength 16% (Figs. 6a and 6b). All the tested compound inside nM, and 40.58 nM), respectively. Furthermore, the most promising

8
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

Fig. 3. Cell cycle analysis (a) Control A549, (b) 5/A549, (c) 14c/A549 and (d) 17/A549.

derivatives 5, 14c, and 17 exhibited IC50 values ranged between (0.27 induced apoptosis and arrested the cell cycle by 74.50% in both G2/M
nM to 0.78 nM) compared to Lapatinib (0.18 nM) and Erlotinib (0.22 and preG1. While both thiadiazole adamantane derivatives 5 and 17 had
nM) against EGFR L858R/T790M double mutant kinase. Studying cell cycle caused increase in DNA content in G0-G1 and S phases by nearly
analysis revealed that the enaminonitrile pyridine derivative 14c 73.36%, and 73.71%, respectively. The most potent adamantane

9
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

Table 6 1,3,4-thiadiazol-2-yl)-2-cyano-2-(4-oxo-3-phenylthiazo-lidin-2-ylidene)
Percentage of cell death after treatment with compounds 5, 14c, and 17. acetamide (17) that has exhibited good EGFR inhibitors with lower IC50
Cpd. No. Apoptosis Necrosis values, that may be attributed to the presence of thiazolidinone moiety
beside thiadiazolo-adamantane backbone core. Finally, molecular
Total Early Late
docking study revealed that the promising compounds deeply bounded
5 19.11 2.61 5.41 11.09 inside the active site of the pocket as Erlotinib with binding energy
14c 32.41 8.26 14.79 9.36
17 16.59 5.13 7.35 4.11
between –22.07 to − 19.19 Kcal/mol which is higher than Erlotinib S =
A549 1.86 0.31 0.15 1.4 − 19.10 Kcal/mol, as well as the assessment of oral bioavailability
(Lipinski’s and Veber rules) beside other pharmacokinetics properties.

derivatives 5, 14c, and 17 showed increase in the total percentage of


apoptosis from 16.59% to 32.41% compared to 1.86% for standard A549
cells. Among the three promising derivatives, N-(5-(adamantan-1-yl)-

Fig. 4. Illustrated the different percentage of cell death by thiadiazolo-adamantane derivatives 5, 14c, and 17.

Fig. 5. Showed the results of the most active thiazolo-thiadiazole adamantane derivative 17.

Table 7
In silico the physicochemical and pharmacokinetic properties of the thiadiazolo-adamantane derivatives 5, 14c, and 17 as well as standard drugs (Lapatinib and
Erlotinib).
Cpd. No. Lipinski’s Rule Veber filter Pharmacokinetics

MW < 500 MLogP ≤ 4.15 nHBA ≤ 10 nHBD ≤ 5 nRB ≤ 10 TPSA ≤ 140 Å2 GI BBB

5 364.46 3.35 4 1 2 95.48 High No


14c 488.99 3.26 5 1 3 149.62 Low No
17 477.60 2.46 5 1 5 152.52 Low No
Lapatinib 581.06 3.44 8 2 11 114.73 Low No
Erlotinib 393.44 1.89 6 1 10 74.73 High yes

* MW; Molecular weight, MLogP; an octanol–water partition coefficient, nHBA; no. of hydrogen bond acceptor, nHBD; no of hydrogen bond donor, nRB; no. of
rotatable bond.

10
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

Table 8 4. Experimental section


Docking results of the adamantane derivatives with binding energy and inter­
acting group with specific amino acids in residue inside active side 1M17. 4.1. Chemistry
Cpd. No. (S) Amino Interacting groups Length Strength
(Kcal/ acids (◦ A) % Uncorrected melting points were recorded on digital Gallen Kamp
mol) MFB-595 instrument. The IR spectra (KBr) (cm− 1) were measured on a
Erlotinib − 19.10 Met769 N of quinazoline 3.13 16 Shimadzu 440 spectrophotometer.1H NMR spectra (δ, ppm) were ob­
5 − 19.19 Asp831 NH of isatin 2.54 49 tained in deuterated dimethyl sulfoxide (DMSO–d6) and 13C NMR at
14c Met769 CN of 3.18 22
–22.07
101 MHz, spectra were obtained on a Bruker spectrometer (400 MHz)
Enaminonitrile
17 − 21.78 Asp831 NH of 2- 2.81 45 spectrometer, using TMS as an internal standard; chemical shifts are
Lys721 aminothiazole 2.60 51 reported as δ ppm units. The data were presented as follows: chemical
CN of shift, multiplicity (s = singlet, d = doublet, t = triplet, q = quartet, m =
cyanoacetanilide multiplet, br = broad, app = apparent), coupling constant(s) in Hertz
(Hz), and integration. Mass spectra were recorded on Thermo Scientific

Fig. 6a. 2D interaction maps of Erlotinib inside 1M17 active site.

Fig. 6b. 3D interaction maps of Erlotinib inside 1M17 active site.

11
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

Fig. 7a. 2D interaction maps of compound 5 inside 1M17 active site.

Fig. 7b. 3D interaction maps of compound 5 inside 1M17 active site.

ISQLT mass spectrometer at the Regional Center for Mycology and Yield (80%) as white powder from ethanol; mp: 158–160 ◦ C; IR:
Biotechnology, Al-Azhar University. Elemental analyses were carried ν/cm− 1: 3160 (NH), 2905, 2849 (CH-aliph.), 1714 (C–
– O) 1612 (C–
– N);
1
out at Micro Analytical Unit, Cairo University, Cairo, Egypt. H NMR (400 MHz, DMSO) δ 12.79 (s, 1H, NH, exchangeable by D2O),
5-(Adamantan-1-yl)-1,3,4-thiadiazol-2-amine (1) was prepared 4.42 (s, 2H, CH2), 2.06 (s, 4H, 2CH2), 2.01 (s, 5H, 3CH + CH2), 1.76 (s,
according to the reported methods [23,69]. 6H, 3CH2); 13C NMR (101 MHz, DMSO) δ 174.60 (C– – O), 165.66,
Synthesis of N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2- 157.88(2C– – N), 43.12 (CH2), 42.76, 37.77, 36.28, 28.30 (Adamant. Cs);
chloroacetamide (2) [23] MS (EI, 70 eV): m/z (%) = 311.81 [M+] (28.75%), 111.18 (100%); Anal.
To a solution of the 5-(adamantan-1-yl)-1,3,4-thiadiazol-2-amine (1) Calcd for C14H18ClN3OS (311.83): C, 53.93; H, 5.82; N, 13.48; Found: C,
(5 mmol) in dioxane (20 mL) containing triethylamine (3 drops) as a 53.88; H, 5.84; N, 13.35%.
catalyst, (5 mmoL) of chloro-acetyl chloride in dioxane (5 mL) was Synthesis of N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-
added the reaction mixture was stirred for 2hs, then poured into crushed cyanoacetamide (3)
ice and the solid product was filtered off, washed with water to afford A mixture of 5-(adamantan-1-yl)-1,3,4-thiadiazol-2-amine (1) (5
the desired product. mmol) and 3-(3,5-dimethyl-1H-pyrazol-1-yl)-3-oxopropanenitrile (5

12
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

Fig. 8a. 2D interaction maps of compound 14c inside 1M17 active site.

Fig. 8b. 3D interaction maps of compound 14c inside 1M17 active site.

mmol) in toluene (20 mL) was heated under reflux for 30 min., after 162.73, 158.13 (2C–– N), 115.63(CN), 56.48(CH2), 43.10, 37.77, 36.27,
cooling the formed precipitate was filtered off, dried. 28.30, 26.50 (Adamant. Cs); MS (EI, 70 eV): m/z (%) = 302.48 [M+]
Yield (70%) as white powder from ethanol; mp: 180–182 ◦ C; IR: (10.13%), 73.99 (100%); Anal. Calcd for C15H18N4OS (302.40): C,
ν/cm− 1: 3348 (NH), 2965, 2850 (CH-aliph.), 2263 (CN), 1720 (C– – O) 59.58; H, 6.00; N, 18.53; Found: C, 59.78; H, 6.02; N, 18.65%.
1
1617 (C– N); H NMR (400 MHz, DMSO) δ 6.95 (s, 1H, NH, exchange­
– Synthesis of N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)ben­
able by D2O), 4.07 (s, 2H, CH2), 2.05 (s, 4H, 2CH2), 2.00 (s, 5H, 3CH + zamide (4)
CH2), 1.74 (s, 6H, 3CH2); 13C NMR (101 MHz, DMSO) δ 174.48 (C– – O), A mixture of 5-(adamantan-1-yl)-1,3,4-thiadiazol-2-amine (1) (5

13
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

Fig. 9a. 2D interaction maps of compound 17 inside 1M17 active site.

Fig. 9b. 3D interaction maps of compound 17 inside 1M17 active site.

mmol) and benzoyl chloride (5 mmol) in dioxane (20 mL) was heated (339.46): C, 67.23; H, 6.24; N, 12.38; Found: C, 67.11; H, 6.28; N,
under reflux for 10 hs, and the reaction mixture was left to cool, the 12.18%.
formed precipitate was filtered off then dried. Synthesis of 3-((5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)
Yield (65%) as white powder from ethanol; mp: 258–260 ◦ C; IR: imino)indolin-2-one (5)
ν/cm− 1: 3138 (NH), 2899, 2847 (CH-aliph.), 1670 (C– – O) 1602 (C– – N); A mixture of 5-(adamantan-1-yl)-1,3,4-thiadiazol-2-amine (1) (5
1
H NMR (400 MHz, DMSO) δ 12.62 (s, 1H, NH, exchangeable by D2O), mmol) and isatin (5 mmol), containing acetic acid (3 drops) as a catalyst
8.03 (d, J = 7.2 Hz, 2H, Ar- H), 7.59 (t, J = 7.4 Hz, 1H, Ar- H), 7.49 (t, J in absolute ethanol (20 mL) was exposed to reflux for 32hs, the reaction
= 7.6 Hz, 2H, Ar- H), 2.00 (s, 4H, 2CH2), 1.97 (s, 5H, 3CH + CH2), 1.69 mixture was left to cool, and the solid product so formed was collected
(s, 6H, 3CH2); 13C NMR (101 MHz, DMSO) δ 174.22(C– – O), 167.78, by filtration and crystallized from the proper solvent.
159.17(C– – N), 133.31, 132.27, 129.72, 129.08, 129.02, 128.80 (6Ar- Yield (70%) as Orange powder from ethanol; mp: 155–157 ◦ C; IR:
Cs), 43.15, 37.72, 36.32, 28.34 (Adamant. Cs); MS (EI, 70 eV): m/z (%) ν/cm− 1: 3246 (NH), 2890, 2845 (CH-aliph.), 1727 (C– – O) 1614 (C–– N);
1
= 339.3 [M+] (28.99%), 72.91 (100%); Anal. Calcd for C19H21N3OS H NMR (400 MHz, DMSO) δ 11.02 (s, 1H, NH, exchangeable by D2O),

14
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

7.48 (t, J = 7.6 Hz, 1H, Ar- H), 7.39 (d, J = 7.2 Hz, 1H, Ar- H), 6.96 (t, J (NH), 2900, 2847 (CH-aliph.), 1677 (C– – O) 1627 (C– – N); 1H NMR (400
= 7.4 Hz, 1H, Ar- H), 6.83 (d, J = 7.6 Hz, 1H, Ar- H), 1.93 (s, 4H, 2CH2), MHz, DMSO) δ 8.67 (s, 1H, NH, exchangeable by D2O), 4.23 (s, 2H,
1.81 (s, 5H, 3CH + CH2), 1.61 (s, 6H, 3CH2); 13C NMR (101 MHz, CH2), 2.79–2.76 (m, 2H, CH2-Pip.), 2.31–2.28 (m, 2H, CH2-Pip.), 1.85
DMSO) δ 168.89 (C– – O), 168.27, 159.78, 151.24(3C– – N), 138.84, (s, 4H, 2CH2), 1.79 (m, 5H, 3CH + CH2), 1.55 (s, 6H, 3CH2), 1.49–1.44
130.84, 125.13, 123.21, 118.28, 112.71(6Ar-Cs), 42.29, 38.04, 36.09, (m, 4H, 2CH2-Pip.), 1.38 (m, 2H, CH2-Pip.); 13C NMR (101 MHz, DMSO)
28.11 (Adamant. Cs); MS (EI, 70 eV): m/z (%) = 364.22 [M+] (33.21%), δ 174.25(C– – O), 157.71(C– – N), 60.14(CH2), 53.99 (2CH2-Pip.), 43.97,
119.26 (100%); Anal. Calcd for C20H20N4OS (364.47): C, 65.91; H, 5.53; 43.14, 41.91, 37.72, 36.30, 28.31, 28.06 (Adamant. Cs), 25.04, 22.56,
N, 15.37; Found: C, 65.94; H, 5.68; N, 15.51%. 22.16(3CH2-Pip.); MS (EI, 70 eV): m/z (%) = 360.22 [M+] (50.89%),
Synthesis of 3-((5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl) 205.71 (100%); Anal. Calcd for C19H28N4OS (360.52): C, 63.30; H, 7.83;
amino)pentane-2,4-dione (6) N, 15.54; Found: C, 63.14; H, 7.74; N, 15.59%.
To a solution of 5-(adamantan-1-yl)-1,3,4-thiadiazol-2-amine (1) (5 Synthesis of 2-amino-substituted-N-(5-(adamantan-1-yl)-1,3,4-
mmol) in absolute ethanol (20 mL) 3-chloroacetyl acetone (5 mmol) thiadiazol-2-yl)acetamide derivatives (10a, b)
containing piperidine (2 drops) as a catalyst was added then heated A mixture of N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-chlor­
under reflux for 8 hs, the reaction mixture was left to cool, and the oacetamide (2) (5 mmol), appropriate aromatic amine namely: p-methyl
obtained product was filtered and crystallized from proper solvent. aniline (5 mmol) and p-anisidine (5 mmol) containing triethylamine (3
Yield (72%) as brown powder from ethanol; mp: 120–122 ◦ C; IR: drops) as a catalyst in 20 mL of absolute ethanol was heated under reflux
ν/cm− 1: 3199 (NH), 2948, 2844, 2804 (CH-aliph.), 1699, 1676 (C– – O) for 12hs, the reaction mixture was left to cool and the formed precipitate
1617 (C– – N); 1H NMR (400 MHz, DMSO) δ 9.48 (s, 1H, NH, exchange­ was filtered off, dried, and recrystallized from proper solvent.
able by D2O), 3.10 (s, 1H, CH-methylinic), 2.15 (s, 3H, CH3), 2.04 (s, 3H, N-(5-(Adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-((p-methox­
CH3), 1.91 (s, 5H, 3CH + CH2), 1.79 (s, 4H, 2CH2), 1.72 (s, 6H, 3CH2); yphenyl)amino)acetamide (10a)
13
C NMR (101 MHz, DMSO) δ 168.94, 168.15(C– – O), 162.83, 154.89 Yield (72%) as dark brown powder from ethanol; mp: 245–247 ◦ C;
(C– N), 81.83 (CH ), 43.90, 42.14, 38.07, 36.04, 28.08, 27.97
– – IR: ν/cm− 1: 3186 (NH), 2902, 2847 (CH-aliph.), 1705 (C– – O) 1615
(Adamant. Cs), 22.49, 22.18 (2CH3); MS (EI, 70 eV): m/z (%) = 333.45 (C–– N); 1H NMR (400 MHz, DMSO) δ 12.40 (s, 1H, NH, exchangeable by
[M+] (9.02%), 123.69 (100%); Anal. Calcd for C17H23N3O2S (333.45): D2O), 6.71 (d, J = 8.8 Hz, 2H, Ar-H), 6.52 (d, J = 8.8 Hz, 2H, Ar-H), 5.67
C, 61.23; H, 6.95; N, 12.60; Found: C, 61.35; H, 7.05; N, 12.51%. (s, 1H, NH, exchangeable by D2O), 3.98 (s, 2H, CH2), 3.62 (s, 3H, OCH3),
Synthesis of ethyl 2-((5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl) 2.03 (s, 4H, 2CH2), 1.98 (s, 5H, 3CH + CH2), 1.73 (s, 6H, 3CH2); 13C
amino)-3-oxobutanoate (7) NMR (101 MHz, DMSO) δ 174.08 (C– – O), 170.65, 157.89 (C– – N),
A mixture of 5-(adamantan-1-yl)-1,3,4-thiadiazol-2-amine 1 (5 151.72, 142.50, 115.07, 113.74 (6Ar-Cs), 55.74 (OCH3), 47.41(CH2),
mmol), ethyl 3-chloroacetoacete (5 mmol) containing piperidine (2 43.14, 37.68, 36.29, 28.31(Adamant. Cs); MS (EI, 70 eV): m/z (%) =
drops) as a catalyst in 20 mL of absolute ethanol was heated under reflux 398.48 [M+] (10.70%), 102.30 (100%); Anal. Calcd for C21H26N4O2S
for 8 hs, the reaction mixture was left to cool, and the formed precipitate (398.53): C, 63.29; H, 6.58; N, 14.06; Found: C, 63.22; H, 6.48; N,
was filtered off, dried, and recrystallized from ethanol. 14.18%.
Yield (65%) as yellow powder; mp: 140–142 ◦ C; IR: ν/cm− 1: 3208 N-(5-(Adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-(p-tolylamino)
(NH), 2901, 2849 (CH-aliph.), 1698 (C– – O) 1620 (C– – N); 1H NMR (400 acetamide (10b)
MHz, DMSO) δ 8.69 (s, 1H, NH, exchangeable by D2O), 4.23 (q, 2H, J = Yield (82%) as yellow powder from ethanol; mp: 280–282 ◦ C; IR:
6.60, 2H, CH3–CH2–), 3.03 (s, 1H, CH-methylinic), 2.87 (s, 3H, CH3), ν/cm− 1: 3186 (NH), 2902, 2849 (CH-aliph.), 1708 (C– – O) 1619 (C–
– N);
1
1.93 (s, 4H, 2CH2), 1.81 (s, 5H, 3CH + CH2), 1.62 (s, 6H, 3CH2), 1.24 (t, H NMR (400 MHz, DMSO) δ 12.42 (s, 1H, NH, exchangeable by D2O),
J = 6.8 Hz, 3H, CH3–CH2–); 13C NMR (101 MHz, DMSO) δ 175.15, 6.89 (d, J = 8.4 Hz, 2H, Ar-H), 6.48 (d, J = 8.4 Hz, 2H, Ar-H), 5.83 (s,
168.29 (2C– – O), 158.92, 151.27(C– – N), 77.82 (CH–), 60.56 (CH3- 1H, NH, exchangeable by D2O), 4.00 (s, 2H, CH2), 2.13 (s, 3H, CH3),
CH2–), 43.90, 42.43, 42.25, 37.97, 36.14, 36.06, 28.15 (Adamant. Cs), 2.03 (s, 4H, 2CH2), 1.98 (s, 5H, 3CH + CH2), 1.72 (s, 6H, 3CH2); 13C
22.51(CH3), 14.66(CH3–CH2–); MS (EI, 70 eV): m/z (%) = 363.89 NMR (101 MHz, DMSO) δ 170.51(C– – O), 165.67, 157.89 (C– – N),
[M+] (14.09%), 56.18 (100%); Anal. Calcd for C18H25N3O3S (363.48): 146.11, 129.81, 125.48, 112.82 (6Ar-Cs), 46.92 (CH2), 43.14, 42.76,
C, 59.48; H, 6.93; N, 11.58; Found: C, 59.23; H, 7.18; N, 11.48%. 37.76, 37.68, 36.29, 28.31 (Adamant. Cs), 20.50 (CH3); MS (EI, 70 eV):
Synthesis of 2-(adamantan-1-yl)imidazo[2,1-b][1,3,4]thiadia­ m/z (%) = 381.99 [M+] (16.17%), 70.40 (100%); Anal. Calcd for
zol-6-ol (8) C21H26N4OS (382.53): C, 65.94; H, 6.85; N, 14.65; Found: C, 65.91; H,
A mixture of N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-chlor­ 6.73; N, 14.68%.
oacetamide (2) (5 mmol) in dioxane (15 mL) containing triethyl amine Synthesis of 2-((5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)
(3 drops) was refluxed for 8hs, and then poured into crushed ice to amino)thiazol-4(5H)-one (11)
afford precipitate which was filtered off, washed with water. A solution of N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-chlor­
Yield (75%) as white powder from ethanol; mp: 320–322 ◦ C; IR: oacetamide (2) (5 mmol) and ammonium thiocyanate (5 mmol) in
ν/cm− 1: 3532 (OH), 2901, 2849 (CH-aliph.), 1571 (C– – N); 1H NMR ethanol (20 mL) was heated under reflux for 6 hs. The precipitate formed
(400 MHz, DMSO) δ 12.82 (s, 1H, OH, exchangeable by D2O), 4.05 (s, upon cooling was isolated by filtration, then dried.
1H, CH–), 2.04 (s, 4H, 2CH2), 1.99 (s, 5H, 3CH + CH2), 1.74 (s, 6H, Yield (77%) as faint brown powder from ethanol; mp: 298–300 ◦ C;
3CH2); 13C NMR (101 MHz, DMSO) δ 174.61(C–OH), 162.67, 158.04 IR: ν/cm− 1: 3121 (NH), 2902, 2846 (CH-aliph.), 1740 (C– – O) 1603
(C–– N), 115.55 (CH–), 43.11, 37.98, 37.78, 36.27, 28.30, 27.92, 26.47 (C–– N); 1H NMR (400 MHz, DMSO) δ 12.24 (s, 1H, NH, exchangeable by
(Adamant. Cs); MS (EI, 70 eV): m/z (%) = 275.40 [M+] (17.17%), D2O), 4.07 (s, 2H, CH2), 2.05 (s, 4H, 2CH2), 1.99 (s, 5H, 3CH + CH2),
118.98 (100%); Anal. Calcd for C14H17N3OS (275.37): C, 61.06; H, 6.22; 1.74 (s, 6H, 3CH2); 13C NMR (101 MHz, DMSO) δ 177.10, 174.49
N, 15.26; Found: C, 61.01; H, 6.17; N, 15.16%. (C–– O), 169.73, 165.49(C– – N), 66.82 (CH2), 38.34, 36.25, 35.98, 28.29
Synthesis of N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2- (Adamant. Cs); MS (EI, 70 eV): m/z (%) = 334.47 [M+] (13.68%), 83.32
(piperidin-1-yl)acetamide (9) (100%); Anal. Calcd for C15H18N4OS2 (334.46): C, 53.87; H, 5.42; N,
To a solution of N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2- 16.75; Found: C, 53.82; H, 5.38; N, 16.79%.
chloroacetamide (2) (5 mmol) in dioxane (10 mL) and piperidine (5 Synthesis of 2-(adamantan-1-yl)-5-imino-5,6-dihydro-7H-
mmol) with three drops of triethyl amine was refluxed for 8 hs, the re­ [1,3,4]thiadiazolo[3,2-a]pyri-midin-7-one (12)
action mixture was left to cool, and the formed precipitate was filtered A mixture of N-(5-(adamantan-1-yl)-1, 3, 4-thiadiazol-2-yl)-2-cya­
off, dried, and recrystallized from ethanol. noacetamide (3) (5 mmol) in dioxane (15 mL) was refluxed for 8 hs,
Yield (66%) as creamy powder; mp: 200–202 ◦ C; IR: ν/cm− 1: 3136 and then poured into crushed ice to afford precipitate which was

15
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

filtered, washed with water. ethanol containing piperidine (2 drops) as a catalyst was heated under
Yield (72%) as faint yellow powder from ethanol; mp: 302–304 ◦ C; reflux for 10–16 hs. The resulting solid was filtered off, dried, and
IR: ν/cm− 1: 3156 (NH), 2903, 2849 (CH-aliph.), 1718(C– – O), 1625 recrystallized from methanol.
1
(C– N); H NMR (400 MHz, DMSO) δ 12.80 (s, 1H, NH, exchangeable by
– 1-(5-(Adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-6-amino-4-(4-
D2O), 4.42 (s, 2H, CH2), 2.06 (s, 4H, 2CH2), 2.01 (s, 5H, 3CH + CH2), methoxyphenyl)-2-oxo-1,2-dihydropyridine-3,5-dicarbonitrile
1.75 (s, 6H, 3CH2); 13C NMR (101 MHz, DMSO) δ 174.57 (C– – O), (14a)
165.64, 162.49, 157.90 (C– – N), 46.72 (CH2), 43.12, 37.77, 36.28, Yield (69%) as yellow powder from methanol; mp: 160–162 ◦ C; IR:
36.08, 28.31, 28.07 (Adamant. Cs); MS (EI, 70 eV): m/z (%) = 303.83 ν/cm− 1: 3314, 3172 (NH2), 3029 (CH-aro), 2904, 2848 (CH-aliph.),
[M+] (12.11%), 135.28 (100%); Anal. Calcd for C15H18N4OS (302.40): 2223(CN), 1706(C– – O), 1605 (C–– N); 1H NMR (400 MHz, DMSO) δ 8.39
C, 59.58; H, 6.00; N, 18.53; Found: C, 59.42; H, 6.08; N, 18.68%. (s, 1H, Ar-H), 8.04 (d, J = 8.0 Hz, 1H, Ar-H), 7.96 (s, 1H, Ar-H), 7.19 (s,
Synthesis of 3-substituted N-(5-(adamantan-1-yl)-1,3,4-thiadia­ 2H, NH2 exchangeable by D2O), 7.15 (d, J = 8.0 Hz, 1H, Ar-H), 3.88 (s,
zol-2-yl)-2-cyanoacrylamide derivatives (13a-c) 3H, –OCH3), 2.18 (s, 4H, 2CH2), 1.99 (s, 5H, 3CH + CH2), 1.74 (s, 6H,
A mixture of 2-cyanoacetamide derivative 3 (5 mmol) and the 3CH2); 13C NMR (101 MHz, DMSO) δ 164.82(C– – O), 163.49, 160.93
appropriate benzaldehyde derivatives (namely 4-methoxy or 4-chloro or (C–– N), 152.18, 150.57, 143.81, 133.84, 133.45, 132.93, 124.84,
4-fluorobenzaldehyde) (5 mmol) in 20 mL absolute ethanol containing 124.59, 115.68, 115.40 (2CN), 104.67, 77.32 (C-CN), 56.40(–OCH3),
piperidine (2 drops) as a catalyst was heated under reflux for 5 hs. The 43.05, 42.55, 37.95, 37.77, 36.26, 36.21, 28.29, 28.21(Adamant. Cs);
obtained product was filtered and recrystallized from the proper solvent. MS (EI, 70 eV): m/z (%) = 484.79 [M+] (76.06%), 409.21 (100%); Anal.
N-(5-(Adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-cyano-3-(4- Calcd for C26H24N6O2S (484.58): C, 64.44; H, 4.99; N, 17.34; Found: C,
methoxyphenyl)acrylamide (13a) 64.34; H, 5.07; N, 17.46%.
Yield (70%) as yellow crystal from ethanol; mp: 136–138 ◦ C; IR: 1-(5-(Adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-6-amino-4-(4-flu­
ν/cm− 1: 3175 (NH), 3007,3003 (CH-arom), 2903, 2846 (CH-aliph.), orophenyl)-2-oxo-1,2-dihydro-pyridine-3,5-dicarbonitrile (14b)
2245(CN), 1652(C– – O), 1586(C– – N); 1H NMR (400 MHz, DMSO) δ Yield (66%) as yellow powder from methanol; mp: 170–172 ◦ C; IR:
13.89 (s, 1H, NH, exchangeable by D2O), 8.38 (s, 1H, CH-methylinic), ν/cm− 1: 3280, 3142(NH2), 3077 (CH-aro), 2903, 2849 (CH-aliph.),
8.05 (d, J = 8.8 Hz, 2H, Ar-H), 7.16 (d, J = 8.8 Hz, 2H, Ar-H), 3.88 (s, 2227(CN), 1679 (C– – O), 1606 (C– – N); 1H NMR (400 MHz, DMSO) δ
3H, OCH3), 2.07 (s, 4H, 2CH2), 2.00 (s, 5H, 3CH + CH2), 1.76 (s, 6H, 8.06–7.97 (m, 1H, Ar-H), 7.73–7.68 (m, 1H, Ar-H), 7.50 (d, J = 8.8 Hz,
3CH2); 13C NMR (101 MHz, DMSO) δ 177.11 (C– – O), 163.49, 157.46 1H, Ar-H), 7.43 (s, 2H, NH2 exchangeable by D2O), 7.23 (d, J = 8.8 Hz,
(C–– N), 152.15, 147.91, 133.45, 131.95, 124.87, 117.52, 115.41(CN), 1H, Ar-H), 2.04 (s, 4H, 2CH2), 1.98 (s, 5H, 3CH + CH2), 1.73 (s, 6H,
105.69, 56.17 (OCH3), 42.55, 37.96, 36.21, 28.21 (Adamant. Cs); MS 3CH2); 13C NMR (101 MHz, DMSO) δ 176.33(C– – O), 173.87, 162.28
(EI, 70 eV): m/z (%) = 420.81 [M+] (34.50%), 294.16 (100%); Anal. (C–– N), 158.76, 154.28, 149.77, 134.13, 133.98, 133.53, 133.15,
Calcd for C23H24N4O2S (420.53): C, 65.69; H, 5.75; N, 13.32; Found: C, 131.91, 131.87, 130.70, 128.58, 117.51, 116.04, 115.83, 114.47 (2CN),
65.69; H, 5.77; N, 13.41%. 76.46 (C-CN), 43.16, 37.98, 36.33, 36.22, 28.33, 27.92 (Adamant. Cs);
N-(5-(Adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-cyano-3-(4-flu­ MS (EI, 70 eV): m/z (%) = 472.13 [M+] (12.29%), 91.64 (100%); Anal.
orophenyl) acrylamide (13b) Calcd for C25H21FN6OS (472.54): C, 63.54; H, 4.48; N, 17.79; Found: C,
Yield (78%) as orange crystal from ethanol; mp: 144–146 ◦ C; IR: 63.45; H, 4.52; N, 17.97%.
ν/cm− 1: 3150 (NH), 3058 (CH-aro), 2901, 2847 (CH-aliph.), 2249(CN), 1-(5-(Adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-6-amino-4-(4-
1678(C– – O), 1621 (C–– N); 1H NMR (400 MHz, DMSO) δ 11.93 (s, 1H, chlorophenyl)-2-oxo-1,2-dihydro pyridine-3,5-dicarbonitrile (14c)
NH, exchangeable by D2O), 8.76 (s, 1H, CH– methylinic), 8.39–8.25 Yield (68%) as yellow powder from methanol; mp: 150–152 ◦ C; IR:
(m, 1H, Ar-H), 8.04 (m, 1H, Ar-H), 7.80 (s, 1H, Ar-H), 7.38 (d, J = 8.8 ν/cm− 1: 3325, 3139 (NH2), 3034 (CH-aro), 2904, 2847 (CH-aliph.),
Hz, 1H, Ar-H), 1.90 (s, 4H, 2CH2), 1.82 (s, 5H, 3CH + CH2), 1.62 (s, 6H, 2226(CN), 1660 (C– – O), 1622 (C– – N); 1H NMR (400 MHz, DMSO) δ
3CH2); 13C NMR (101 MHz, DMSO) δ 176.33 (C– – O), 173.87, 168.42 7.94 (d, J = 8.4 Hz, 1H, Ar-H), 7.69 (d, J = 8.8 Hz, 1H, Ar-H), 7.63 (d, J
(C–– N), 158.68, 151.39, 133.99, 133.70, 117.15, 116.93, 115.57 (CN), = 8.4 Hz, 1H, Ar-H), 7.56 (d, J = 8.5 Hz, 1H, Ar-H), 7.43 (s, 2H, NH2
104.82, 43.99, 43.10, 42.81, 42.48, 37.97, 37.85, 37.59, 36.27, 28.24, exchangeable by D2O), 2.10 (s, 4H, 2CH2), 1.96 (s, 5H, 3CH + CH2),
27.91 (Adamant. Cs); MS (EI, 70 eV): m/z (%) = 408.81 [M+] (30.51%), 1.72 (s, 6H, 3CH2); 13C NMR (101 MHz, DMSO) δ 171.82(C– – O), 168.04,
105.02 (100%); Anal. Calcd for C22H21FN4OS (408.50): C, 64.69; H, 160.59(C– – N), 157.50, 150.80, 143.14, 139.51, 137.59, 132.60, 130.56,
5.18; N, 13.72; Found: C, 64.73; H, 5.11; N, 13.54%. 130.17, 129.11, 116.95, 114.33 (2CN), 103.63, 82.68 (C-CN), 44.01,
N-(5-(Adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-3-(4-chlor­ 43.16, 42.52, 37.99, 36.22, 35.95, 28.30, 28.22, 27.98 (Adamant. Cs);
ophenyl)-2-cyanoacryl amide (13c) MS (EI, 70 eV): m/z (%) = 488.12 [M+] (16.83%), 139.21 (100%); Anal.
Yield (73%) as yellow crystal from ethanol; mp: 112–114 ◦ C; IR: Calcd for C25H21ClN6OS (488.99): C, 61.41; H, 4.33; N, 17.19; Found: C,
ν/cm− 1: 3172 (NH), 3049 (CH-aro), 2902, 2847 (CH-aliph.), 2218(CN), 61.28; H, 4.39; N, 17.25%.
1681(C– – O), 1626 (C–– N); 1H NMR (400 MHz, DMSO) δ 12.52 (s, 1H, Synthesis of 1-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-4,6-
NH, exchangeable by D2O), 8.26 (s, 1H, CH– methylinic), 7.93 (d, J = dimethyl-2-oxo-1,2-dihydro-pyridine-3-carbonitrile (15)
8.4 Hz, 2H, Ar-H), 7.56 (d, J = 8.4 Hz, 2H, Ar-H), 1.96 (s, 4H, 2CH2), A mixture of N-(5-(adamantan-1-yl)-1, 3, 4-thiadiazol-2-yl)-2-cya­
1.90 (s, 5H, 3CH + CH2), 1.66 (s, 6H, 3CH2); 13C NMR (101 MHz, noacetamide (3) (5 mmol) and acetylacetone (5 mmol) in absolute
DMSO) δ 171.68 (C– – O), 168.74, 168.03 (C– – N), 165.92, 149.66, ethanol (20 mL) containing piperidine (2 drops) as catalyst was heated
137.17, 132.29, 131.62, 129.79, 117.41, 110.92 (CN), 103.63, 44.06, under reflux for 8 hs, the resulting solid was filtered off, dried, and
43.23, 42.72, 37.90, 36.32, 28.34, 28.28, 27.86 (Adamant. Cs); MS (EI, recrystallized from ethanol.
70 eV): m/z (%) = 424.20 [M+] (12.54%), 256.57 (100%); Anal. Calcd Yield (73%) as creamy powder from ethanol; mp: 102–104 ◦ C; IR:
for C22H21ClN4OS (424.95): C, 62.18; H, 4.98; N, 13.18; Found: C, ν/cm− 1: 2905, 2848 (CH-aliph.), 2223(CN), 1668 (C– – O), 1598 (C–– N);
1
62.22; H, 4.88; N, 13.28%. H NMR (400 MHz, DMSO) δ 6.56 (s, 1H, –CH pyridine), 2.42 (s, 3H,
Synthesis of 1-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-6- –CH3), 2.09 (s, 3H, –CH3), 2.08 (s, 4H, 2CH2), 1.89 (s, 5H, 3CH +
amino-4-(4-substituted)-2-oxo-1, 2-dihydropyridine-3, 5-dicarbo­ CH2), 1.77 (s, 6H, 3CH2); 13C NMR (101 MHz, DMSO) δ 162.47(C– – O),
nitrile derivatives (14a-c) 160.49, 158.30(C– – N), 151.70, 118.62, 115.41, 110.56, 98.96, 43.23,
Equimolar quantities of 2-cyanoacetamide derivative 3 and the 43.05, 36.41, 36.11, 28.33, 28.26, 21.48, 20.80 (Adamant. Cs); MS (EI,
appropriate 2-(arylidene)malono-nitrile (namely 2-(4-methox­ 70 eV): m/z (%) = 366.27 [M+] (32.49%), 80.31 (100%), Anal. Calcd
ybenzylidene)malononitrile, 2-(4-chlorobenzylidene)malono-nitrile, for C20H22N4OS (366.48): C, 65.55; H, 6.05; N, 15.29; Found: C, 65.62;
and 2-(4-fluorobenzylidene)malononitrile (5 mmol) in 20 mL absolute H, 6.21; N, 15.36%.

16
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-oxo-2H-chro­ CRediT authorship contribution statement


mene-3-carboxamide (16)
A mixture of N-(5-(adamantan-1-yl)-1, 3, 4-thiadiazol-2-yl)-2-cya­ Mohammed M.S. Wassel: Conceptualization, Methodology, Inves­
noacetamide 3 (5 mmol) and salicylaldehyde (5 mmol) in 20 mL abso­ tigation, Resources, Writing - original draft. Yousry A. Ammar:
lute ethanol containing piperidine (2 drops) as catalyst was heated Conceptualization, Methodology, Software, Formal analysis, Writing -
under reflux for 5 hs., the resulting solid was filtered off, dried, and original draft, Writing - review & editing, Supervision, Project admin­
recrystallized from dioxane. istration. Gameel A.M. Elhag Ali: Conceptualization, Methodology,
Yield (77%) as yellow crystal; mp: 128–130 ◦ C; IR: ν/cm− 1: 3349 Investigation, Resources, Writing - original draft, Supervision. Amany
(NH), 3042 (CH-aro), 2936, 2831 (CH-aliph.), 1712, 1695 (2C– – O), Belal: Methodology (Biology) and participation in the revise of manu­
1610 (C– – N); 1H NMR (400 MHz, DMSO) δ 8.99 (s, 1H, CH-chromene), script. Ahmed B.M. Mehany: Methodology (Biology). Ahmed Ragab:
8.04 (d, J = 7.5 Hz, 1H, Ar-H), 7.83 (t, J = 7.8 Hz, 1H, Ar-H), 7.58 (d, J Conceptualization, Software, Formal analysis, Validation, Methodology,
= 8.0 Hz, 1H, Ar-H), 7.51 (t, J = 7.3 Hz, 1H, Ar-H), 4.35 (s, 1H, NH Investigation, Data curation, Writing - original draft, Writing - review &
exchangeable by D2O), 2.08 (s, 4H, 2CH2), 2.05 (s, 5H, 3CH + CH2), editing, Visualization.
1.78 (s, 6H, 3CH2); 13C NMR (101 MHz, DMSO) δ 175.18, 160.52(C– – O),
157.29, 154.57 (C– – N), 149.16, 135.49, 131.11, 125.98, 118.71,
Declaration of Competing Interest
118.49, 116.91, 43.14, 37.86, 36.28, 28.32 (Adamant. Cs); MS (EI, 70
eV): m/z (%) = 407.33 [M+] (21.25%), 91.55 (100%); Anal. Calcd for The authors declare that they have no known competing financial
C22H21N3O3S (407.49): C, 64.85; H, 5.19; N, 10.31; Found: C, 64.68; H, interests or personal relationships that could have appeared to influence
5.09; N, 10.56%. the work reported in this paper.
Synthesis of N-(5-(adamantan-1-yl)-1,3,4-thiadiazol-2-yl)-2-
cyano-2-(4-oxo-3-phenylthiazo-lidin-2-ylidene)acetamide (17)
Acknowledgment
To a solution of finely powdered potassium hydroxide (0.01 mol) in
dry DMF (10 mL), 2-cyanoacetamide derivative 3 (5 mmol) was added
Dr. Mohammed M. S. Wassel wants to thank Dr. Ahmed Ragab,
and stirred for 0.5-1hr., then phenyl isothiocyanate was added slowly
lecturer of Organic Chemistry, Faculty of Science (Boys), Al-Azhar
after complete addition, the reaction mixture was stirring for an addi­
University, for his real sharing, valuable creative thoughts, honest
tional 6 hs. Then methyl chloroacetate (5 mmol) and the reaction
advice, helpful supervision, sincere encouragement, and continual help
mixture was stirred for further 4 h, then poured into crushed ice. The
throughout the development of this work. Besides, all authors thank Dr.
resulting precipitate was filtrated off, dried, and recrystallized from
Amany Belal and Dr. Ahmed B. M. Mehany for their efforts in the bio­
ethanol.
logical part (methodology) as well as their participation in revise of the
Yield (72%) as dark green powder ; mp: 250–252 ◦ C; IR: ν/cm− 1:
manuscript.
3136 (NH), 3055 (CH-Aro), 2901, 2847 (CH-aliph.), 2216(CN), 1732,
1677 (C– – O), 1629 (C– – N); 1H NMR (400 MHz, DMSO) δ 9.84 (s, 1H,
Appendix A. Supplementary material
NH, exchangeable by D2O), 7.52 (d, J = 7.6 Hz, 2H, Ar-H), 7.34 (d, J =
7.2 Hz, 2H, Ar-H), 7.13 (t, J = 7.3 Hz, 1H, Ar-H), 4.05 (s, 2H, CH2), 2.01
Supplementary data to this article can be found online at https://doi.
(s, 4H, 2CH2), 1.97 (s, 5H, 3CH + CH2), 1.75 (s, 6H, 3CH2); 13C NMR
org/10.1016/j.bioorg.2021.104794.
(101 MHz, DMSO) δ 180.06 (C– – C–S), 174.00, 169.75 (C– – O), 166.27,
163.09 (C– – N), 150.96, 139.94, 136.44, 135.78, 129.68, 128.88,
127.26, 124.05, 114.68 (CN), 73.58, 43.18, 43.11, 42.26, 41.49, 37.99, References
36.41, 36.27, 28.31, 28.18, 27.99 (Adamant. Cs + CH2); MS (EI, 70 eV):
[1] S.I. Farooqi, N. Arshad, P.A. Channar, F. Perveen, A. Saeed, F.A. Larik, A. Javeed,
m/z (%) = 477.80 [M+] (20.43%), 303.40 (100%); Anal. Calcd for Synthesis, theoretical, spectroscopic and electrochemical DNA binding
C24H23N5O2S2 (477.60): C, 60.36; H, 4.85; N, 14.66; Found: C, 60.48; H, investigations of 1, 3, 4-thiadiazole derivatives of ibuprofen and ciprofloxacin:
cancer cell line studies, J. Photochem. Photobiol. B Biol. 189 (2018) 104–118,
4.82; N, 14.45%.
https://doi.org/10.1016/j.jphotobiol.2018.10.006.
[2] M. Jankowski, Nutritional treatment improves the effectiveness of anti-cancer
therapy, Nowotwory. J. Oncol. 67 (2017) 313–315, https://doi.org/10.5603/
4.2. Biological evaluation NJO.2017.0052.
[3] C.P.W. B.W. Stewart, International Agency for Research on Cancer, France, WHO
press, Switzerland, 2014, World Cancer Rep., 2014. https://publications.iarc.fr/
The method of all biological evaluation were performed according to Non-Series-Publications/World-Cancer-Reports/World-Cancer-Report-2014.
our reported methods [79–83] and represented in supplementary [4] A. Molinari, A. Oliva, M. Arismendi-Macuer, L. Guzmán, W. Acevedo, D. Aguayo,
R. Vinet, A. San Feliciano, Antiproliferative benzoindazolequinones as potential
material. cyclooxygenase-2 inhibitors, Molecules 24 (2019) 2261, https://doi.org/10.3390/
molecules24122261.
[5] F. Bray, J. Ferlay, I. Soerjomataram, R.L. Siegel, L.A. Torre, A. Jemal, Global cancer
statistics, GLOBOCAN estimates of incidence and mortality worldwide for 36
4.3. Molecular docking simulation cancers in 185 countries, CA. Cancer J. Clin. 68 (2018) (2018) 394–424, https://
doi.org/10.3322/caac.21492.
Molecular docking targeting EGFR was performed using Molecular [6] R.K. Thalji, K. Raha, D. Andreotti, A. Checchia, H. Cui, G. Meneghelli, R. Profeta,
F. Tonelli, S. Tommasi, T. Bakshi, Structure-guided design of antibacterials that
Operating Environment software 10.2008 (MOE), Chemical Computing allosterically inhibit DNA gyrase, Bioorg. Med. Chem. Lett. 29 (2019) 1407–1412,
Group Inc., Montreal, Quebec, Canada. X-ray crystallography structure https://doi.org/10.1016/j.bmcl.2019.03.029.
of EGFRwt co-crystalized with original ligand 4-anilinoquinazoline in­ [7] S. Chakraborty, K.B. Mir, N.D. Seligson, D. Nayak, R. Kumar, A. Goswami,
Integration of EMT and cellular survival instincts in reprogramming of
hibitor (erlotinib) (PDB: 1M17) [78] were downloaded and obtained programmed cell death to anastasis, Cancer Metastasis Rev. 39 (2020) 1–14,
from the protein data bank. All water molecules were deleted then the https://doi.org/10.1007/s10555-020-09866-x.
active site was generated according to standard protocol and according [8] N. Shahar, S. Larisch, Inhibiting the inhibitors: targeting anti-apoptotic proteins in
cancer and therapy resistance, Drug Resist. Updat. 52 (2020) 100712, https://doi.
to our reported methods [16,17]. The structure of the newly designed
org/10.1016/j.drup.2020.100712.
and most promising compounds was drawn using ChemDraw14.0 then [9] T. Xu, M. Wang, L. Jiang, L. Ma, L. Wan, Q. Chen, C. Wei, Z. Wang, CircRNAs in
exported to MOE. The new structure was potentate 3D and minimized anticancer drug resistance: recent advances and future potential, Mol. Cancer. 19
energy using MMFF94x forcefield. The co-crystalized ligand was (2020) 1–20, https://doi.org/10.1186/s12943-020-01240-3.
[10] P. Gimenez-Bonafe, A. Tortosa, R. Perez-Tomas, Overcoming drug resistance by
exposed to validation process with RSMD 1.99 Å using Triangle Matcher enhancing apoptosis of tumor cells, Curr. Cancer Drug Targets 9 (2009) 320–340,
placement method and London dG as docking score energy. https://doi.org/10.2174/156800909788166600.

17
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

[11] Y. Wang, J. Teng, Increased multi-drug resistance and reduced apoptosis in [32] X. Zhu, J. Sun, S. Wang, W. Bu, M. Yao, K. Gao, Y. Song, J. Zhao, C. Lu, E. Zhang,
osteosarcoma side population cells are crucial factors for tumor recurrence, Exp. Synthesis, crystal structure, superoxide scavenging activity, anticancer and
Ther. Med. 12 (2016) 81–86, https://doi.org/10.3892/etm.2016.3303. docking studies of novel adamantyl nitroxide derivatives, J. Mol. Struct. 1108
[12] R.M. Mohammad, I. Muqbil, L. Lowe, C. Yedjou, H.-Y. Hsu, L.-T. Lin, M.D. Siegelin, (2016) 611–617, https://doi.org/10.1016/j.molstruc.2015.12.048.
C. Fimognari, N.B. Kumar, Q.P. Dou, Broad targeting of resistance to apoptosis in [33] M.I. Dawson, Z. Xia, G. Liu, M. Ye, J.A. Fontana, L. Farhana, B.B. Patel,
cancer, in, Elsevier, Semin. Cancer Biol. (2015) S78–S103, https://doi.org/ S. Arumugarajah, M. Bhuiyan, X.K. Zhang, An adamantyl-substituted retinoid-
10.1016/j.semcancer.2015.03.001. derived molecule that inhibits cancer cell growth and angiogenesis by inducing
[13] H.W. Findley, L. Gu, A.M. Yeager, M. Zhou, Expression and regulation of Bcl-2, Bcl- apoptosis and binds to small heterodimer partner nuclear receptor: effects of
xl, and Bax correlate With p53 status and sensitivity to apoptosis in childhood modifying its carboxylate group on apoptosis, proliferation, and protein-tyrosine
acute lymphoblastic leukemia, Blood 89 (1997) 2986–2993, https://doi.org/ phosphatase activity (Journal of Medicinal Chemistry (2007) 50, (2622)), J. Med.
10.1182/blood.V89.8.2986. Chem. 51 (2008), https://doi.org/10.1021/jm8010492.
[14] A.G. Maione, Y. Brudno, O. Stojadinovic, L.K. Park, A. Smith, A. Tellechea, E. [34] M.I. Dawson, Z. Xia, G. Liu, J.A. Fontana, L. Farhana, B.B. Patel, S. Arumugarajah,
C. Leal, C.J. Kearney, A. Veves, M. Tomic-Canic, Three-dimensional human tissue M. Bhuiyan, X.-K. Zhang, Y.-H. Han, An adamantyl-substituted retinoid-derived
models that incorporate diabetic foot ulcer-derived fibroblasts mimic in vivo molecule that inhibits cancer cell growth and angiogenesis by inducing apoptosis
features of chronic wounds, Tissue Eng. Part C Methods. 21 (2015) 499–508, and binds to small heterodimer partner nuclear receptor: effects of modifying its
https://doi.org/10.1089/ten.tec.2014.0414. carboxylate group on apoptosis, proliferation, and protein-tyrosine phosphatase
[15] E.A. Fayed, Y.A. Ammar, A. Ragab, N.A. Gohar, A.B.M. Mehany, A.M. Farrag, In activity, J. Med. Chem. 50 (2007) 2622–2639, https://doi.org/10.1021/
vitro cytotoxic activity of thiazole-indenoquinoxaline hybrids as apoptotic agents, jm0613323.
design, synthesis, physicochemical and pharmacokinetic studies, Bioorg. Chem. [35] A.Z. Mirza, H. Shamshad, Preparation and characterization of doxorubicin
100 (2020) 103951, https://doi.org/10.1016/j.bioorg.2020.103951. functionalized gold nanoparticles, Eur. J. Med. Chem. 46 (2011) 1857–1860,
[16] A.M.S. El-sharief, Y.A. Ammar, A. Belal, M.A.M.S. El-sharief, Y.A. Mohamed, A.B. https://doi.org/10.1016/j.ejmech.2011.02.048.
M. Mehany, G.A.M. Elhag, A. Ragab, Design, synthesis, molecular docking and [36] R.S. Schwab, A.C. England, D.C. Poskanzer, R.R. Young, Amantadine in the
biological activity evaluation of some novel indole derivatives as potent anticancer treatment of Parkinson’s disease, JAMA 208 (1969) 1168–1170, https://doi.org/
active agents and apoptosis inducers, Bioorg. Chem. 85 (2019) 399–412, https:// 10.1001/jama.1969.03160070046011.
doi.org/10.1016/j.bioorg.2019.01.016. [37] J.-J. Wang, Y.-T. Chern, Y.-F. Chang, T.-Y. Liu, C.-W. Chi,
[17] Y.A. Ammar, A.M. Sh El-Sharief, A. Belal, S.Y. Abbas, Y.A. Mohamed, A.B. Dimethyladamantylmaleimide-induced in vitro and in vivo growth inhibition of
M. Mehany, A. Ragab, Design, synthesis, antiproliferative activity, molecular human colon cancer Colo205 cells, Anticancer Drugs 13 (2002) 533–543.
docking and cell cycle analysis of some novel (morpholinosulfonyl) isatins with [38] J.-J. Wang, K.-T. Huang, Y.-T. Chern, Induction of growth inhibition and G1 arrest
potential EGFR inhibitory activity, Eur. J. Med. Chem. 156 (2018), https://doi.org/ in human cancer cell lines by relatively low-toxic diamantane derivatives,
10.1016/j.ejmech.2018.06.061. Anticancer Drugs 15 (2004) 277–286.
[18] Z.N. Oltval, C.L. Milliman, S.J. Korsmeyer, Bcl-2 heterodimerizes in vivo with a [39] L. Yan, K. Hu, X. Liu, Y. Li, X. Liu, Z. Jiang, Diiron ethane-1, 2-dithiolate complexes
conserved homolog, Bax, that accelerates programed cell death, Cell 74 (1993) with 1, 2, 3-thiadiazole moiety: synthesis, X-ray crystal structures,
609–619, https://doi.org/10.1016/0092-8674(93)90509-O. electrochemistry and fungicidal activity, Appl. Organomet. Chem. (2020) e6084,
[19] S.V. Sharma, D.W. Bell, J. Settleman, D.A. Haber, Epidermal growth factor receptor https://doi.org/10.1002/aoc.6084.
mutations in lung cancer, Nat. Rev. Cancer 7 (2007) 169–181, https://doi.org/ [40] A.M.M. Mohamed, M.F. Ismail, H.M.F. Madkour, A.F. Aly, M.S. Salem,
10.1038/nrc2088. Straightforward synthesis of 2-chloro-N-(5-(cyanomethyl)-1, 3, 4-thiadiazol-2-yl)
[20] A. Ayati, S. Emami, S. Moghimi, A. Foroumadi, Thiazole in the targeted anticancer benzamide as a precursor for synthesis of novel heterocyclic compounds with
drug discovery, Future Med. Chem. 11 (2019) 1929–1952, https://doi.org/ insecticidal activity, Synth. Commun. 50 (2020) 3424–3442, https://doi.org/
10.4155/fmc-2018-0416. 10.1080/00397911.2020.1802652.
[21] R. Rosell, T. Moran, C. Queralt, R. Porta, F. Cardenal, C. Camps, M. Majem, [41] M. Toolabi, M. Khoramjouy, A. Aghcheli, A. Ayati, S. Moghimi, L. Firoozpour,
G. Lopez-Vivanco, D. Isla, M. Provencio, Screening for epidermal growth factor S. Shahhosseini, R. Shojaei, A. Asadipour, K. Divsalar, Synthesis and radioligand-
receptor mutations in lung cancer, N. Engl. J. Med. 361 (2009) 958–967, https:// binding assay of 2, 5-disubstituted thiadiazoles and evaluation of their
doi.org/10.1056/NEJMoa0904554. anticonvulsant activities, Arch. Pharm. 353 (2020) 2000066, https://doi.org/
[22] A. Ayati, S. Moghimi, S. Salarinejad, M. Safavi, B. Pouramiri, A. Foroumadi, 10.1002/ardp.202000066.
A review on progression of epidermal growth factor receptor (EGFR) inhibitors as [42] S. Türk, Synthesis, characterization, antituberculosis activity and computational
an efficient approach in cancer targeted therapy, Bioorg. Chem. (2020) 103811, studies on novel Schiff bases of 1, 3, 4- thiadiazole derivatives, J. Res. Pharm. Res.
https://doi.org/10.1016/j.bioorg.2020.103811. 24 (2020) 793–800, https://doi.org/10.35333/jrp.2020.232.
[23] M. Fesatidou, P. Zagaliotis, C. Camoutsis, A. Petrou, P. Eleftheriou, C. Tratrat, [43] S. Vudhgiri, D. Koude, D.K. Veeragoni, S. Misra, R.B.N. Prasad, R.C.R. Jala,
M. Haroun, A. Geronikaki, A. Ciric, M. Sokovic, 5-Adamantan thiadiazole-based Synthesis and biological evaluation of 5-fatty-acylamido-1, 3, 4-thiadiazole-2-
thiazolidinones as antimicrobial agents. Design, synthesis, molecular docking and thioglycosides, Bioorg. Med. Chem. Lett. 27 (2017) 3370–3373, https://doi.org/
evaluation, Bioorg. Med. Chem. 26 (2018) 4664–4676, https://doi.org/10.1016/j. 10.1016/j.bmcl.2017.06.004.
bmc.2018.08.004. [44] G. Revelant, C. Gadais, V. Mathieu, G. Kirsch, S. Hesse, Synthesis and
[24] V.H. Pham, T.P.D. Phan, D.C. Phan, B.D. Vu, Synthesis and bioactivity of antiproliferative studies of 5-aryl-2-(3-thienylamino)-1, 3, 4-thiadiazoles, Bioorg.
thiosemicarbazones containing adamantane skeletons, Molecules 25 (2020) 324, Med. Chem. Lett. 24 (2014) 2724–2727, https://doi.org/10.1016/j.
https://doi.org/10.3390/molecules25020324. bmcl.2014.04.043.
[25] I. Stankova, K. Chuchkov, R. Chayrov, L. Mukova, A. Galabov, D. Marinkova, [45] G. Serban, Synthetic compounds with 2-amino-1, 3, 4-thiadiazole moiety against
D. Danalev, Adamantane derivatives containing thiazole moiety: synthesis, viral infections, Molecules 25 (2020) 942, https://doi.org/10.3390/
antiviral and antibacterial activity, Int. J. Pept. Res. Ther. 26 (2020) 1781–1787, molecules25040942.
https://doi.org/10.1007/s10989-019-09983-4. [46] A.A. Othman, M. Kihel, S. Amara, 1, 3, 4-Oxadiazole, 1, 3, 4-thiadiazole and 1, 2, 4-
[26] S. Liu, L.-Z. Niu, Y.-H. Shi, F.-X. Wan, L. Jiang, Design, Synthesis and Antifungal triazole derivatives as potential antibacterial agents, Arab. J. Chem. 12 (2019)
Activity of Novel 1-(Adamantan-1-yl) ethanone Oxime Esters, Lett. Drug Des. 1660–1675, https://doi.org/10.1016/j.arabjc.2014.09.003.
Discov. 17 (2020) 526–532, https://doi.org/10.2174/ [47] P.-L. Zhao, A.-N. Duan, M. Zou, H.-K. Yang, W.-W. You, S.-G. Wu, Synthesis and
1570180816666190329225307. cytotoxicity of 3, 4-disubstituted-5-(3, 4, 5-trimethoxyphenyl)-4H-1, 2, 4-triazoles
[27] M. Ms Wassel, W.M. Gamal Eldin, A. Ragab, G.A.M. Elhag Ali, Y.A. Ammar, and novel 5, 6-dihydro-[1, 2, 4] triazolo [3, 4-b][1, 3, 4] thiadiazole derivatives
Antiviral activity of adamantane-pyrazole derivatives against foot and mouth bearing 3, 4, 5-trimethoxyphenyl moiety, Bioorg. Med. Chem. Lett. 22 (2012)
disease virus infection in vivo and in vitro with molecular docking study, J. Appl. 4471–4474, https://doi.org/10.1016/j.bmcl.2012.03.023.
Vet. Sci. 5 (2020) 37–46, https://doi.org/10.21608/JAVS.2020.118001. [48] K. Ilango, P. Valentina, Facile synthesis and cytotoxic activity of 3, 6-disubstituted
[28] A. Orzeszko, B. Kamińska, G. Orzeszko, B.J. Starościak, Synthesis and antimicrobial 1, 2, 4-triazolo-[3, 4-b]-1, 3, 4-thiadiazoles, Eur. J. Chem. 1 (2010) 50–53, https://
activity of new adamantane derivatives, Farm. 55 (2000) 619–623, https://doi. doi.org/10.5155/eurjchem.1.1.50-53.4.
org/10.1016/S0014-827X(00)00075-6. [49] D. Chowrasia, C. Karthikeyan, L. Choure, M. Gupta, M. Arshad, P. Trivedi,
[29] M. Kritsanida, A. Mouroutsou, P. Marakos, N. Pouli, S. Papakonstantinou- Synthesis, characterization and anti cancer activity of some fluorinated 3, 6-diaryl-
Garoufalias, C. Pannecouque, M. Witvrouw, E. De Clercq, Synthesis and antiviral [1, 2, 4] triazolo [3, 4-b][1, 3, 4] thiadiazoles, Arab. J. Chem. 10 (2017)
activity evaluation of some new 6-substituted 3-(1-adamantyl)-1, 2, 4-triazolo [3, S2424–S2428, https://doi.org/10.1016/j.arabjc.2013.08.026.
4-b][1, 3, 4] thiadiazoles, Farm. 57 (2002) 253–257, https://doi.org/10.1016/ [50] O. Stanasel, S. Bota, 2-Amino-1,3,4-thiadiazole as a potential scaffold for promising
S0014-827X(01)01189-2. antimicrobial agents Georgeta, Drug Des. Devel. Ther. 12 (2018) 1545–1566,
[30] E.B. Villhauer, J.A. Brinkman, G.B. Naderi, B.F. Burkey, B.E. Dunning, K. Prasad, B. https://doi.org/10.2147/DDDT.S155958.
L. Mangold, M.E. Russell, T.E. Hughes, 1-[[(3-hydroxy-1-adamantyl) amino] [51] D. Kumar, B.R. Vaddula, K.-H. Chang, K. Shah, One-pot synthesis and anticancer
acetyl]-2-cyano-(S)-pyrrolidine: a potent, selective, and orally bioavailable studies of 2-arylamino-5-aryl-1, 3, 4-thiadiazoles, Bioorg. Med. Chem. Lett. 21
dipeptidyl peptidase IV inhibitor with antihyperglycemic properties, J. Med. Chem. (2011) 2320–2323, https://doi.org/10.1016/j.bmcl.2011.02.083.
46 (2003) 2774–2789, https://doi.org/10.1021/jm030091l. [52] K. Nepali, S. Sharma, M. Sharma, P.M.S. Bedi, K.L. Dhar, Rational approaches,
[31] M.M.S. Wassel, A. Ragab, G.A.M. Elhag Ali, A.B.M. Mehany, Y.A. Ammar, Novel design strategies, structure activity relationship and mechanistic insights for
adamantane-pyrazole and hydrazone hybridized: design, synthesis, cytotoxic anticancer hybrids, Eur. J. Med. Chem. 77 (2014) 422–487, https://doi.org/
evaluation, SAR study and molecular docking simulation as carbonic anhydrase 10.1016/j.ejmech.2014.03.018.
inhibitors, J. Mol. Struct. 1223 (2020), https://doi.org/10.1016/j. [53] S. de Castro, M.-J. Camarasa, Polypharmacology in HIV inhibition: can a drug with
molstruc.2020.128966. simultaneous action against two relevant targets be an alternative to combination

18
M.M.S. Wassel et al. Bioorganic Chemistry 110 (2021) 104794

therapy? Eur. J. Med. Chem. 150 (2018) 206–227, https://doi.org/10.1016/j. [69] A.A. Kadi, E.S. Al-Abdullah, I.A. Shehata, E.E. Habib, T.M. Ibrahim, A.A. El-Emam,
ejmech.2018.03.007. Synthesis, antimicrobial and anti-inflammatory activities of novel 5-(1-adamantyl)-
[54] E. Kucuksayan, T. Ozben, Hybrid compounds as multitarget directed anticancer 1, 3, 4-thiadiazole derivatives, Eur. J. Med. Chem. 45 (2010) 5006–5011, https://
agents, Curr. Top. Med. Chem. 17 (2017) 907–918. doi.org/10.1016/j.ejmech.2010.08.007.
[55] J. Dybal, E. Makrlík, P. Vaňura, J. Budka, Protonation of 25, 27-bis (1-octyloxy) [70] Y.A. Ammar, M.M. Aly, A.A.G. Al-Sehemi, M.A. Salem, M.S.A. El-Gaby,
calix [4] arene-crown-6 in the 1, 3-alternate conformation, Monatshefte Für Cyanoacetanilides intermediates in heterocyclic synthesis. Part 5: Preparation of
Chemie-Chemical Mon. 139 (2008) 1175–1178, https://doi.org/10.1007/s00706- hitherto unknown 5-aminopyrazole and pyrazolo [1, 5-a] pyrimidine derivatives
008-0910-8. containing sulfamoyl moiety, J. Chinese Chem. Soc. 56 (2009) 1064–1071, https://
[56] M.A. Salem, A. Ragab, A. El-Khalafawy, A.H. Makhlouf, A.A. Askar, Y.A. Ammar, doi.org/10.1002/jccs.200900154.
Design, synthesis, in vitro antimicrobial evaluation and molecular docking studies [71] Y.A. Ammar, M.S.A. El-Gaby, M.A. Salem, Cyanoacetanilides intermediates in
of indol-2-one tagged with morpholinosulfonyl moiety as DNA gyrase inhibitors, heterocyclic synthesis. Part 6: Preparation of some hitherto unknown 2-oxopyri­
Bioorg. Chem. 96 (2020) 103619, https://doi.org/10.1016/j.bioorg.2020.103619. dine, bipyridine, isoquinoline and chromeno[3,4-c]pyridine containing
[57] Y.A. Ammar, A.A. Farag, A.M. Ali, A. Ragab, A.A. Askar, D.M. Elsisi, A. Belal, sulfonamide moiety, Arab. J. Chem. 7 (2014) 615–622, https://doi.org/10.1016/j.
Design, synthesis, antimicrobial activity and molecular docking studies of some arabjc.2013.11.026.
novel di-substituted sulfonylquinoxaline derivatives, Bioorg. Chem. 104 (2020) [72] Y.A. Ammar, A.M. El-Sharief, Y.A. Mohamed, A.B. Mehany, A. Ragab, Synthesis,
104164, https://doi.org/10.1016/j.bioorg.2020.104164. spectral characterization and pharmacological evaluation of novel thiazole-
[58] M.A. Salem, A. Ragab, A.A. Askar, A. El-khalafawy, A.H. Makhlouf, European oxoindole hybrid compounds as potential anticancer agents, Al-Azhar Bull. Sci. 29
Journal of Medicinal Chemistry One-pot synthesis and molecular docking of some (2018) 25–37, https://doi.org/10.21608/absb.2018.33767.
new spiropyranindol-2-one derivatives as immunomodulatory agents and in vitro [73] V. Tomar, M. Mazumder, R. Chandra, J. Yang, M.K. Sakharkar, Small Molecule
antimicrobial potential with DNA gyrase inhibitor, Eur. J. Med. Chem. 188 (2020) Drug Design, in: S. Ranganathan, M. Gribskov, K. Nakai, C. Schönbach (Eds.),
111977, https://doi.org/10.1016/j.ejmech.2019.111977. Encycl. Bioinforma. Comput. Biol., Academic Press, Oxford, 2019, pp. 741–760.
[59] M.A.M.S. El-Sharief, S.Y. Abbas, M.A. Zahran, Y.A. Mohamed, A. Ragab, Y. https://doi.org/10.1016/B978-0-12-809633-8.20157-X.
A. Ammar, New 1,3-diaryl-5-thioxo-imidazolidin-2,4-dione derivatives: synthesis, [74] Y. Hao, J. Lyu, R. Qu, Y. Tong, D. Sun, F. Feng, L. Tong, T. Yang, Z. Zhao, L. Zhu,
reactions and evaluation of antibacterial and antifungal activities, Zeitschrift Fur J. Ding, Y. Xu, H. Xie, H. Li, Design, Synthesis, and Biological Evaluation of
Naturforsch. – Sect. B J. Chem. Sci. 71 (2016), https://doi.org/10.1515/znb-2016- Pyrimido[4,5-d]pyrimidine-2,4(1H,3H)-diones as Potent and Selective Epidermal
0054. Growth Factor Receptor (EGFR) Inhibitors against L858R/T790M Resistance
[60] T. Akhtar, K.A. Yasin, N.A. Al-Masoudi, P.G.G. Jones, S. Hameed, Synthesis, crystal Mutation, J. Med. Chem. 61 (2018) 5609–5622, https://doi.org/10.1021/acs.
structure and antiproliferative activity of 6-adamantyl-3-aryl [1, 2, 4] triazolo [3, jmedchem.8b00346.
4-b][1, 3, 4] thiadiazoles, Zeitschrift Für Naturforsch. B 65 (2010) 178–184, [75] C.K. Tang, Role of Epidermal Growth Factor Receptor in Breast Carcinoma, Handb.
https://doi.org/10.1515/znb-2010-0214. Immunohistochem. Situ Hybrid. Hum. Carcinomas Mol. Genet. Lung Breast
[61] A.G. Ali, M.F. Mohamed, A.O. Abdelhamid, M.S. Mohamed, A novel adamantane Carcinomas. 1 (2004) 415, https://doi.org/10.1016/S1874-5784(04)80052-7.
thiadiazole derivative induces mitochondria-mediated apoptosis in lung carcinoma [76] S. Elmore, Apoptosis: a review of programmed cell death, Toxicol. Pathol. 35
cell line, Bioorg. Med. Chem. 25 (2017) 241–253, https://doi.org/10.1016/j. (2007) 495–516, https://doi.org/10.1080/01926230701320337.
bmc.2016.10.040. [77] http://swissadme.ch/index.php (last access 4/5/2020).
[62] Y.A. Ammar, S.Y. Abbas, M.A.M.S. El-Sharief, M.A.E.-R. Salem, A.R. Mohamed, [78] https://www.rcsb.org/structure/1M17 (last access 4/5/2020).
Synthesis and characterization of new imidazolidineiminothione and bis- [79] A.V. Ivachtchenko, A.V. Khvat, V.V. Kobak, V.M. Kysil, C.T. Williams, A new
imidazolidineiminothione derivatives as potential antimicrobial agents, Eur. J. insight into the Pfitzinger reaction. A facile synthesis of 6-sulfamoylquinoline-4-
Chem. 8 (2017) 76–81, https://doi.org/10.5155/eurjchem.8.1.76-81.1542. carboxylic acids, Tetrahedron Lett. 45 (2004) 5473–5476, https://doi.org/
[63] H.F. Rizk, M.A. El-Borai, A. Ragab, S.A. Ibrahim, Design, synthesis, biological 10.1016/j.tetlet.2004.05.028.
evaluation and molecular docking study based on novel fused pyrazolothiazole [80] S.Y. Abbas, R.A.K. Al-Harbi, M.A.M.S. El-Sharief, Synthesis and anticancer activity
scaffold, J. Iran. Chem. Soc. (2020), https://doi.org/10.1007/s13738-020-01944- of thiourea derivatives bearing a benzodioxole moiety with EGFR inhibitory
9. activity, apoptosis assay and molecular docking study, Eur. J. Med. Chem. (2020)
[64] Y.A. Ammar, S. AM, M. El-Sharief, M.M. Ghorab, Y.A. Mohamed, A. Ragab, S. 112363, https://doi.org/10.1016/j.ejmech.2020.112363.
Y. Abbas, New imidazolidineiminothione, imidazolidin-2-one and [81] Y. Jia, C.M. Quinn, A.I. Gagnon, R. Talanian, Homogeneous time-resolved
imidazoquinoxaline derivatives: synthesis and evaluation of antibacterial and fluorescence and its applications for kinase assays in drug discovery, Anal.
antifungal activities, Curr. Org. Synth. 13 (2016) 466–475, https://doi.org/ Biochem. 356 (2006) 273–281, https://doi.org/10.1016/j.ab.2006.05.006.
10.2174/1570179412666150817221755. [82] J. Wang, M.J. Lenardo, Roles of caspases in apoptosis, development, and cytokine
[65] S. ElKalyoubi, E. Fayed, Synthesis and evaluation of antitumour activities of novel maturation revealed by homozygous gene deficiencies, J. Cell Sci. 113 (2000)
fused tri- and tetracyclic uracil derivatives, J. Chem. Res. 40 (2016) 771–777, 753–757.
https://doi.org/10.3184/174751916X14798125870610. [83] K.K.-W. Lo, T.K.-M. Lee, J.S.-Y. Lau, W.-L. Poon, S.-H. Cheng, Luminescent
[66] Y.A. Ammar, A.A. Farag, A.M. Ali, S.A. Hessein, A.A. Askar, E.A. Fayed, D.M. Elsisi, biological probes derived from ruthenium(II) estradiol polypyridine complexes,
A. Ragab, Antimicrobial evaluation of thiadiazino and thiazolo quinoxaline hybrids Inorg. Chem. 47 (2008) 200–208, https://doi.org/10.1021/ic701735q.
as potential DNA gyrase inhibitors; design, synthesis, characterization and [84] Ahmed, Sawsan, Ola, Entsar, Abeer, Ahmed, Yousry, Sulfaguanidine Hybrid with
morphological studies, Bioorg. Chem. 99 (2020) 103841, https://doi.org/10.1016/ Some New Pyridine-2-One Derivatives: Design, Synthesis, and Antimicrobial
j.bioorg.2020.103841. Activity against Multidrug-Resistant Bacteria as Dual DNA Gyrase and DHFR
[67] A.S. Hassan, A.A. Askar, A.M. Naglah, A.A. Almehizia, A. Ragab, Discovery of new Inhibitors, Antibiotics 10 (2) (2021) 162, https://doi.org/10.3390/
schiff bases tethered pyrazole moiety: design, synthesis, biological evaluation, and antibiotics10020162, https://www.mdpi.com/2079-6382/10/2/162.
molecular docking study as dual targeting DHFR/DNA gyrase inhibitors with [85] Sally I. Eissa, Amel M. Farrag, Samir Y. Abbas, Mohamed F. El Shehry,
immunomodulatory activity, Molecules 25 (2020), https://doi.org/10.3390/ Ahmed Ragab, Eman A. Fayed, Yousry A. Ammare, Novel Structural hybrids of
molecules25112593. quinolone and thiazole moieties: Synthesis and evaluation of antibacterial and
[68] Y.A. Ammar, E.A. Fayed, A.H. Bayoumi, R.R. Ezz, M.S. Alsaid, A.M. Soliman, M. antifungal activities with molecular modeling studies, Bioorganic Chemistry
M. Ghorab, New chalcones bearing isatin scaffold: synthesis, molecular modeling (2021), 104803, https://doi.org/10.1016/j.bioorg.2021.104803 https://www.sci
and biological evaluation as anticancer agents, Res. Chem. Intermed. 43 (2017) encedirect.com/science/article/abs/pii/S0045206821001802, (in press).
6765–6786, https://doi.org/10.1007/s11164-017-3019-z.

19

View publication stats

You might also like