Next Article in Journal
Pristine and Modified Porous Membranes for Zinc Slurry–Air Flow Battery
Next Article in Special Issue
Synthesis, Antibacterial and Antifungal Activity of New 3-Aryl-5H-pyrrolo[1,2-a]imidazole and 5H-Imidazo[1,2-a]azepine Quaternary Salts
Previous Article in Journal
Antioxidant Serine-(NSAID) Hybrids with Anti-Inflammatory and Hypolipidemic Potency
Previous Article in Special Issue
Densely Functionalized 2-Methylideneazetidines: Evaluation as Antibacterials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Exploration of the Antimicrobial Effects of Benzothiazolylthiazolidin-4-One and In Silico Mechanistic Investigation

by
Michelyne Haroun
1,*,
Christophe Tratrat
1,
Anthi Petrou
2,
Athina Geronikaki
2,*,
Marija Ivanov
3,
Ana Ćirić
3,
Marina Soković
3,
Sreeharsha Nagaraja
1,4,
Katharigatta Narayanaswamy Venugopala
1,5,
Anroop Balachandran Nair
1,
Heba S. Elsewedy
1 and
Hafedh Kochkar
6,7
1
Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
2
School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
3
Mycological Laboratory, Department of Plant Physiology, Institute for Biological Research, Siniša Stanković-National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
4
Department of Pharmaceutics, Vidya Siri College of Pharmacy, Off Sarjapura Road, Bengaluru 560 035, Karnataka, India
5
Department of Biotechnology and Food Technology, Faculty of Applied Sciences, Durban University of Technology, Durban 4001, South Africa
6
Department of Chemistry, College of Science, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
7
Basic & Applied Scientific Research Center, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
*
Authors to whom correspondence should be addressed.
Molecules 2021, 26(13), 4061; https://doi.org/10.3390/molecules26134061
Submission received: 26 May 2021 / Revised: 11 June 2021 / Accepted: 25 June 2021 / Published: 2 July 2021
(This article belongs to the Special Issue Biologically Active Heterocyclic Compounds)

Abstract

:
Background: Infectious diseases still affect large populations causing significant morbidity and mortality. Bacterial and fungal infections for centuries were the main factors of death and disability of millions of humans. Despite the progress in the control of infectious diseases, the appearance of resistance of microbes to existing drugs creates the need for the development of new effective antimicrobial agents. In an attempt to improve the antibacterial activity of previously synthesized compounds modifications to their structures were performed. Methods: Nineteen thiazolidinone derivatives with 6-Cl, 4-OMe, 6-CN, 6-adamantan, 4-Me, 6-adamantan substituents at benzothiazole ring were synthesized and evaluated against panel of four bacterial strains S. aureus, L. monocytogenes, E. coli and S. typhimirium and three resistant strains MRSA, E. coli and P. aeruginosa in order to improve activity of previously evaluated 6-OCF3-benzothiazole-based thiazolidinones. The evaluation of minimum inhibitory and minimum bactericidal concentration was determined by microdilution method. As reference compounds ampicillin and streptomycin were used. Results: All compounds showed antibacterial activity with MIC in range of 0.12–0.75 mg/mL and MBC at 0.25–>1.00 mg/mL The most active compound among all tested appeared to be compound 18, with MIC at 0.10 mg/mL and MBC at 0.12 mg/mL against P. aeruginosa. as well as against resistant strain P. aeruginosa with MIC at 0.06 mg/mL and MBC at 0.12 mg/mL almost equipotent with streptomycin and better than ampicillin. Docking studies predicted that the inhibition of LD-carboxypeptidase is probably the possible mechanism of antibacterial activity of tested compounds. Conclusion: The best improvement of antibacterial activity after modifications was achieved by replacement of 6-OCF3 substituent in benzothiazole moiety by 6-Cl against S. aureus, MRSA and resistant strain of E. coli by 2.5 folds, while against L. monocytogenes and S. typhimirium from 4 to 5 folds.

Graphical Abstract

1. Introduction

Amalgamations of drugs endowed with different medicinal activities have been dispensed to patients for decades. It is known that an adapted adjustment of different targets may offer an improved medicinal aspect and an advantageous side effect in contrast with the effect of a ligand that has a unique mode of action [1]. In comparison with drug combinations, there are several betterments emanating from drugs active on several receptors, including the more foreseeable pharmacokinetics and pharmacodynamics properties resulting of the treatment of a unique medicinal drug in addition to ameliorated patient acquiescence.
Characterization of new therapies for both antifungal and antibacterial disorders that can operate more efficaciously and that are exempt of the side effects related to the present medications continues to be a principal requirement in pharmaceutical research [2,3]. Applying several drugs to infective symptoms in correlation with inflammation is an impasse, particularly when patients suffer from defective hepatic or renal failure or in countering interaction between two drugs [4]. Furthermore, from the standpoint of medical financial efficiency, and pursuing a favorable patient safety, a dual antifungal/antibacterial drug having the slightest side effects in addition to improved safeness profile must be energetically advisable [5].
The initial step in investigating for dual-acting molecules is constituted by screening collections of drugs to design pharmacophores’ coupling [6]. This investigation may be accomplished in vitro and in silico. It ultimately grasps effective compounds on appropriate enzymes belonging to identical or similar enzyme families. Analysis of the framework of ligands effective on different targets or enzymes together with protein similarity study, dimensions, and aspects of their effective cores may be adopted to find possible targets for dual-acting drugs [7,8].
Computer-aided prediction of pharmacological activity spectra of compounds and drugs based on their structural formulas can be appraised by software Prediction of Activity Spectra for substances (PASS) [9] to perform investigations on novel antimicrobial. This strategy is built on the structure–activity relationship study in a heterogeneous data set. The set includes 989,000 various chemical compound families endowed with tremendous variety of biological potency. For the selection of compounds with predicted designed properties, PharmaExpert4 software was promoted. The version [10,11] is established from the literature data and furnished a single feasibility to look for compounds with potential multi-targeted activity. This study aims at discovering compounds endowed with anti-infective potency. It is worth mentioning that benzothiazole is a privileged heterocyclic scaffold with multiple applications and tremendous range of pharmacological activity. Benzothiazoles are recognized for their anti-inflammatory [12,13,14], antimicrobial [15,16,17,18,19], anesthetic [20], anticancer [21,22,23], anti-viral [24,25,26], analgesic [13,14], antipyretic [27], antidiabetic [28,29,30], antioxidant [12,16,20,21,31], carbonic anhydrize inhibitory [26,32,33], anticonvulsive [34,35], antifungal [33,36,37] and many other [38,39] activities. Additionally, the benzothiazole scaffold is present in three FDA approved drugs (Figure 1). They are quizartinb, a receptor tyrosine kinase inhibitor, the flutemetamol-diagnostic tool for Alzheimer disease and the drug riluzole for treatment of amyotrophic lateral sclerosis.
On the other hand, the thiazolidinone core attracted the interest of researchers owing to its various degrees of pharmacological and medicinal activities [18,40,41,42,43,44,45,46,47]. Herein, we explore the antimicrobial effects of benzothiazolylthiazolidin-4-one and their in silico mechanistic investigation.

2. Results and Discussion

Continuing our ongoing research in the field of antimicrobial agents [45,46,47] and based on results of our previous paper [47] we designed new series of compounds modifying the previously synthesized ones. We have replaced 6-trifluoromethoxy substituent of benzothiazole ring first by 6-CN, 6-Ad, 4-Me-6-Ad and after by 4-OCH3, and 6-Cl keeping the same substitution at position 4 of benzene ring mostly for 6-Cl, 4-OMe and in some cases of 6-CN (Figure 2). Thus, we synthesized 2-(2-(substituted phenyl)-4-oxothiazolidin-3-yl)benzo[d]thiazole-6-carbonitrile (15), 3-(6-(adamantan-1-yl)benzo[d]thiazol-2-yl)-2-(substituted phenyl) thiazolidin-4-one (6, 7), 3-(6-(adamantan-1-yl)-4-methylbenzo[d]thiazol-2-yl)-2-(substituted phenyl)thiazolidin-4-one (8, 9), 3-(4-methoxybenzo[d]thiazol-2-yl)-2-(4-substituted phenyl)thiazolidin-4-one (1014), 3-(6-chlorobenzo[d]thiazol-2-yl)-2-(4-substituted phenyl)thiazolidin-4-one (1519).

2.1. Chemistry

Compounds were synthesized according to Scheme 1 as published in our previous paper [47].
All products were obtained as racemates and synthesized compounds were characterized by TLC and spectroscopic methods (IR, 1H NMR, 13C-NMR and MS for some compounds).
In the IR spectra, characteristic strong absorption of the carbonyl vibration in the range of 1700 cm−1 and absorption in the 1600 and 1540 cm−1 corresponding to the C–C bond of the aromatic ring was observed. The C–H bond of the aromatic ring occurs close to 3000 cm−1, while the tertiary amine occurs at 2340–2360 cm−1. Finally, the chlorine atoms of phenyl appeared to be poorly absorbed at about 721 and 1100 cm−1, respectively.
In 1H-NMR spectra, signals at 7.00–8.12 ppm, 6.72–7.15 ppm N–CH–S and 3.85–4.13 ppm attributed to aromatic, N–CH–S and –CH2 protons, respectively, were observed. It is worth noting that the protons of the 5 position show two characteristic peaks, each one, double split. This is because these two protons are cleaved together as they are neither chemically nor magnetically equivalent. In cases of methoxy-substitution at the benzothiazole or benzene ring, a peak at 3.76–3.95 ppm was observed, while hydroxy-derivatives showed a wide peak at 5.32–5.35 ppm. Finally, the presence of adamantane as a substituent on the benzothiazole ring was confirmed by two peaks at 1.89–1.44 ppm. The first one corresponds to the protons of the three tertiary carbon atoms, while the second peak to the twelve protons of the six tertiary carbon atoms of adamantane.
The 13C-NMR peak attributed to the C=O group was observed at 170–171 ppm, while for the C-2 of the benzothiazole ring at 163–165 ppm and for C-2 and C-5 of thiazolidinone moiety at 60–63 ppm and at 31–34 ppm, respectively. The signal of adamantane’s carbon atoms were observed at 41–44, 32–36 and 27–28.5 ppm. Finally, peak attributed to the carbon atom of benzene attached to hydroxyl appeared at 156 ppm (see experimental).

2.2. Toxicity Prediction

Taking into account the importance of prediction of toxicity in drug design two software applications Protox and ToxPredict from Open Tox designed according to REACH legislation requirements were utilized in this study [48,49]. The data are presented in Table 1 and Table 2. These software predict probability of carcinogenicity and mutagenicity in various organisms using in silico models and the most accurate estimation of the mean lethal dose (LD50) administered to rodents. The reliable estimates are considered to be more than 0.025. All derivatives showed confidence from 0.026 to 0.041 and LD50 of 500–1000 mg/kg or higher belonging to group four according to Globally Harmonized System (GHS) of Labeling and Chemicals’ Classification [50] and considered safe for biological experiments. The results of the prediction are presented in Table 1 and Table 2. It is worth mentioning that the prediction accuracy augments as the confidence values increases. Specifically, reliable estimates are regarded to be more than 0.025.
According to Lasar model throughout OpenTox, all the compounds found to be at the category IV with LD50 between 500 and 1000 mg/kg and they are safe for use.

2.3. Prediction of Activity Spectra of Compounds by Program PASS

PASS prediction of antibacterial activity was performed for the whole set of designed molecules, which were chosen for synthesis and biological testing. Antibacterial activity for all compounds was predicted with the probability to be active Pa values ranging from 0.224 to 0.337 (Table S1). The mechanism of antibacterial activity was predicted as well. The prediction revealed that muramoyltetrapeptide carboxypeptidase inhibition is estimated with Pa between 0.314 and 0.607. The calculated Pa values for all compounds were less than 0.5, indicating their relative novelty compared to the structures of the compounds from the PASS training set [51]. Thus, it can be concluded that the studied compounds have some features different from those of well-known antibacterial agents, which may indicate their innovative potential.

2.4. Biological Evaluation

Compounds 19, derivatives of 6-CN, 6-Ad, 4-Me-6-Ad benzothiazole based thiazolidinones were evaluated for antibacterial activity, by microdilution method to determine the minimal inhibitory and bactericidal concentrations against the panel of five strains: two Gram positive (Streptococcus aureus and Listeria monocytogenes) and three Gram negative strains (Pseudomonas aeruginosa, Escherichia coli and Salmonella typhimurium). As reference compounds ampicillin and streptomycin were used. Antibacterial activity of tested compounds is shown in Table 3 with MIC values in the range of 0.10–0.75 mg/mL and MBC at 0.12–1.00 mg/mL. According to the order of activity which can be presented as: 8 > 2 > 4 > 6 > 7 > 1 > 9 > 5 > 3 the highest activity was achieved for compound 8 with MIC at 0.20–0.30 mg/mL and MBC at 0.25–0.50 mg/mL towards non-resistant bacterial strains. The lowest antibacterial potential was observed for compound 3 with MIC values in range of 0.20–0.50 mg/mL and MBC at 0.25–1.0 mg/mL. The most sensitive bacterium appeared to be E. coli (ATCC 35210), while S. typhimirium was the most resistant one. Four out of nine compounds (1, 2, 4, 5) showed very good activity against E. coli with MIC/MBC at 0.12/0.25 mg/mL almost equipotent with ampicillin, while compound 2 additionally, as well as compound 6, demonstrated the same good activity against S. aureus. As far as resistant strains are concern the most sensitive to compounds tested appeared to be P. aeruginosa and the most resistant MRSA. P. aeruginosa was found to be very sensitive to compounds 8, 4 and 5 with MIC/MBC at 0.06/0.12, 0.20/0.25 and 0.12/0.25 mg/mL, respectively with compound 8 exhibiting almost equipotent activity with streptomycin and higher than ampicillin and 5 being more potent than ampicillin. MRSA was more sensitive to compounds 2, 4 with MIC/MBC at 0.25/0.50.mg/mL and 3, 5 with MIC/MBC at 0.30/0.50 mg/mL, whereas E. coli demonstrated the same sensitivity to all compounds tested. It should be noticed that streptomycin showed only bacteriostatic activity against MRSA and not bactericidal, while ampicillin was totally inactive against MRSA, in comparison with tested compounds. Interestingly, our compounds manifested good efficiency against ampicillin resistant P. aeruginosa and E. coli as well as against streptomycin resistant MRSA.
According to the structure–activity relationship studies the presence of 4-CH3, 6-adamantyl substituents in benzothiazole ring in combination with 2,6-di-Cl substituents in benzene ring (8) seems to be beneficial for antibacterial activity of these group of compounds. Introducing CN group in position 6 of benzothiazole ring and 2-6-di-F substituents in benzene (2) decreased slightly the activity, while replacement of 2,6-di-F by 2,4-di-Cl substituents led to less active compound (4), which nevertheless is considered as active. On the other hand, introduction of 2-F,6-Cl substituents in benzene ring appeared to be detrimental. The analysis of structure–activity relationships revealed that antibacterial activity of these compounds depends on substituents on benzene ring as well as on benzothiazole one. Thus, in case of 6-CN substituted derivatives the most active is 2,6-di-F, followed by 2-4-di-Cl and the last is 2-F,6-Cl, while for 6-adamantane, 4-CH3 and 6-adamantyl substitution in benzothiazole moiety the presence of 2,6-di-Cl in benzene ring is beneficial.
The comparison, of obtained results on antibacterial activity with those of compounds with 6-OCF3 substituent in benzothiazole ring revealed that in the case of 4-F substitution in benzene ring the replacement of 6-OCF3 by 6-CN improved the activity only against resistant strain of P. aeruginosa, while the presence of the 4-NO2 group in the benzene ring was beneficial for the resistant strain of E. coli. Better improvement was observed in case of 2,6-di-F substitution, namely against S. aureus, MRSA, L. monocytogenes and resistant strain of E. coli up to 2, 2.7 fold, respectively The presence of 2-F,6-Cl substituent almost did not influence the activity, while 2,6-di-Cl substituent improved 2.5 fold the activity against E. coli. As for the adamantine moiety occupies the 6-position, introduction of 2,6-di-Cl substituent was beneficial for activity against S. aureus improving it two times; while for 4-Me-6-adamantane series, 2,6-di-Cl substituent induced a 2-fold increase in efficiency against resistant strain of P. aeruginosa. The same beneficial effect was observed against S. aureus with the presence of 2,6-di-F substitution in this series. Thus, it can be concluded that replacement of 6 OCF3 substituent by 6-CN improved the activity in most cases by 2–2.7 times.
In an attempt to improve more the antibacterial activity, we decided to introduce in position 4 and 6 of benzothiazole ring methoxy (1014) and chloro (1519) substituents, respectively.
Compounds 1019 were evaluated for their antibacterial activity against the same bacterial strains. The results are presented in Table 4 and MIC values are in the range of 0.06–0.75 mg/mL and MBC at 0.12–1.00 mg/mL. As already mentioned, all compounds showed antibacterial activity with the following order: 18 > 16 > 19 > 15 > 14 > 17 > 10 > 11 > 13 > 12. The best activity was achieved for compound 18 with MIC and MBC at 0.10–0.25 mg/mL and 0.12–0.5 mg/mL, respectively, while compound 12 showed the lowest one (MIC/MBC at 0.25–0.50/0.50–1.00 mg/mL) towards non-resistant strains tested. The most sensitive bacterium again was E. coli (ATCC 35210), followed by P. aeruginosa, while L. monocytogenes was the most resistant one. Compounds 15, 17 and 19 exhibited good activity against Gram negative bacterium P. aeruginosa with MIC/MBC at 0.12/0.25 mg/mL, while compound 18 demonstrated very good activity (MIC/MBC at 0.10/0.12 mg/mL) being all of them almost equipotent with streptomycin and twice more potent than ampicillin. Additionally, 18 showed good activity, better than both reference drugs, also against the most sensitive and most resistant bacterial strains (E. coli and L. monocytogenes) with MIC/MBC at 0.12/0.20 mg/mL. A little bit lower activity against E. coli was observed for 10 and 13 with MIC and MBC at 0.12 and 0.25 mg/mL, respectively. Good activity against S. aureus and S. typhimurium with MIC and MBC at 0.10 mg/mL and 0.12 exhibited compound 16. It should be mentioned that this compound appeared to be the most potent against L. monocytogenes with MIC/MBC at 0.06/0.12 mg/mL, followed by compound 19 being both more active than reference drugs. It was observed that in general compounds 1519 were found to be the most potent among all tested, with compound 18 to be equipotent with streptomycin against almost all bacteria strains tested except S. aureus and S. typhimurium.
These compounds were also tested against three resistant bacterial strains: MRSA, P. aeruginosa and E. coli. All of them exhibited activity against MRSA with MIC at 0.10–0.75 mg/mL and MBC in range of 0.12–>1.00 mg/mL. The best activity was shown by compound 16 (MIC/MBC at 0.10/0.12 mg/mL) followed by compound 18 with MIC and MBC at 0.20 mg/mL and 0.25 mg/mL, respectively, while compound 12 was the less potent. It should be mentioned that activity of compounds against MRSA was superior to reference drugs. Thus, streptomycin showed only bacteriostatic activity with MIC at 0.10 mg/mL, while ampicillin did not exhibit neither bacteriostatic nor bactericidal activities. As far as activity against resistant P. aeruginosa is concerned, compounds appeared to be very potent with MIC at 0.06–0.25 mg/mL and MBC in range of 0.12–0.50 mg/mL. It should be mentioned that even the less potent compounds 1014 appeared to be very potent against this bacterium strain with MIC ranging from 0.12–0.25 and MBC at 0.25–0.50 mg/mL. The most potent was found to be compound 18 (MIC/MBC at 0.06/0.12 mg/mL), followed by compounds 17 and 19 with MIC/MBC at 0.10/0.12 mg/mL. All compounds showed higher potential than ampicillin against P. aeruginosa with compound 18 being almost equipotent with streptomycin (MIC/MBC at 0.05/0.10 mg/mL) and 3-fold more active than ampicillin. Regarding resistant E. coli it was found that all compounds were more active than ampicillin (MIC at 0.20 mg/mL, without bactericidal activity). The best activity exhibited compounds 16 and 19 with MIC and MBC at 0.12 mg/mL and 0.25 mg/mL, respectively being almost equipotent with streptomycin.
In summary, all compounds were more potent than ampicillin against MRSA, while compounds 16, 19 and 18 appeared to be equipotent with streptomycin against resistant strains E. coli and P. aeruginosa respectively.
Compounds with the most promising antibacterial potential were studied for their effect on biofilm formation (Table 5). Despite that none of the tested compounds exhibited activity better than reference drugs in concentration of MIC, compound. 19 demonstrated the highest antibiofilm potency being, in concentration of half MIC better than streptomycin by 1.5 fold.
The structure–activity relationship revealed that the presence of 6-Cl substitution in benzothiazole ring is more beneficial than 4-OCH3 one. However, antibacterial activity of these compounds depends not only on substitution at the benzothiazole ring but on the combination of substituents at the benzothiazole moiety and benzene ring as well. Thus, the presence of 6-Cl substituent in combination with 4-OCH3 of benzene ring (18) appeared to be the most favorable. Replacement of 4-OCH3 by 4-NO2, led to a slightly less active compound 16. The third best compound was found to be 19 with 4-OH substitution in benzene ring, while the less active was compound 12 with 4-OCH3 of benzothiazole and 4-Cl substituent on benzene ring. It should be mentioned that the presence of 4-Cl substitution independent of 6-Cl or 4-OCH3 on benzothiazole ring was detrimental. The comparison of the activity of compounds 1519 and 1014 revealed that 4-OH substitution was beneficial in case of series with 4-OCH3 substitution in benzothiazole ring, while for compounds with 6-Cl at benzothiazole moiety compound 19 was third in order of activity. In general, it was observed the opposite activity of compounds with the same substituents in benzene ring, but different at benzothiazole ring. Thus, in case of 6-Cl benzothiazole derivatives activity can be presented as: 4-OCH3 > 4-NO2 > 4-OH > 4-F > 4-Cl, whereas in case of 4-OCH3 benzothiazole derivatives it is: OH > F > NO2 > OCH3 > Cl. The only common is that the presence of 4-Cl substituent in benzene ring is detrimental for antibacterial activity in both cases.
The comparison of obtained results on antibacterial activity with those of compounds with 6-OCF3 substituent in benzothiazole ring [47] revealed that in case of 4-F substitution on benzene ring the replacement of 6-OCF3 by 6-Cl improved twice the activity against P. aeruginosa and P. aeruginosa resistant, while the replacement by 4-OMe slightly improve only activity against E. coli (0.15 and 0.12 mg/mL). Against other species, these replacements did not improve the activity but decreased 3/2 times it in case of MRSA, L. monocytogenes and S typhimurium against resistant strain of E. coli respectively. Among 4-nitro derivatives the presence of 6-Cl substituent in benzothiazole ring appeared to be beneficial compared to 6-OCF3 and 4-OCH3, since activity against S. aureus, MRSA and resistant strain of E. coli increased 2.5 fold, while against L. monocytogenes and S. typhimurium from 4 to 5 fold. Better results (2–3 times) were obtained in case of the presence of 4-OMe and 4-OH substituents in benzene ring of 6-Cl-benzothiazole derivatives compared to the same substituents at the 4 position of benzene ring of 4-OCF3 derivatives.
From all mentioned above it is obvious that modifications performed (replacement of 6-OCF3 by 6-Cl in benzothiazole ring), improved the activity against some species from 2 to 5 fold.

2.5. Antifungal Activity

Compounds 19 were tested for their possible antifungal activity (Table 6), which was moderate to low and can be presented in following descending order: 6 > 9 > 1 = 7 > 8 > 2 = 3 = 4 > 5. Despite, in general these compounds were modestly effective; some of them demonstrated good activity against some fungi species. Thus, compounds 1 and 9 (MIC/MFC of 0.25/0.50 mg/mL) showed activity almost equal to ketoconazole (MIC/MFC of 0.20/0.50 mg/mL) against Aspergillus versicolor, while 6 against Penicillium funiculosum and Penicillium verrucosum var. cyclopium, at the same time exhibiting 3 fold higher activity than ketoconazole against Trichoderma viride. The same activity against this fungal was shown by compound 9.
Replacement of 6-CN group by 4-OMe of benzothiazole moiety did not improve much the antifungal activity, while the presence of 6-Cl substituent increased it but still being lower comparing with antibacterial. The order of activity can be presented as follows: 19 > 18 > 16 > 17 > 10 = 11 > 14 > 12 = 13 > 15. The best activity among compounds tested was achieved for compound 19 with MIC in range of 0.12–0.50 mg/mL and MFC at 0.25–1.00 mg/mL, while the lowest effect was observed for compound 15 (MIC from 0.25 to >1.00 mg/mL and MFC from 0.5 to >1.00 mg/mL). Compounds 18 and 19 exhibited the highest potency, twice better/equipotent than that of ketoconazole against A. versicolor and P.v.c., respectively with MIC at 0.12 mg.ml and MFC at 0.25 mg/mL, while compound 16 showed the same good activity against Aspergillus fumigatus. Furthermore, all compounds demonstrated good activity against T. viride being superior to ketoconazole with MIC/MFC at 1.0/1.5 mg/mL. Compounds 19, 10 and 14 were almost equipotent with ketoconazole against A. versicolor, while 19, additionally, against P. funiculosum. The most sensitive fungal appeared to be T. viride, while Aspergillus niger was the most resistant one.
The structure–activity relationships study showed that the presence of hydroxyl group at position 4 of benzene ring (19) is beneficial for antifungal activity. Replacement of hydroxyl by methoxy group led to a little less potent compound 18. Introduction of nitro or fluoro group at 4-position of benzene ring derivatives (16) or (15) had negative influence on antifungal activity, the latter being the less active compound. In case of compounds with methoxy group at position 4 of benzothiazole moiety, the presence of fluoro- and nitro substituents at position 4 of benzene ring demonstrated the same influence on antifungal activity, as compounds 1519. In group of 6-CN-benzothiazole based thiazolidinones, the presence of 4-F was detrimental, while nitro substituent showed the same behavior like in two groups mentioned above. The comparison between the results of these two series of compounds 1519 with 6-Cl substitution in benzothiazole moiety and 1014 revealed that antifungal activity of compounds depends not only on the nature of the substituent of benzene ring but also on the nature and position of substituent of benzothiazole ring.

2.6. In Silico Predictive Studies (Molecular Properties and Drug-Likeness)

Drug likeness is examined as an important part that provides the base for the molecules to be a powerful drug candidate. There are several rules, such. Lipinski [52], Ghose [53], Veber [54], Egan [55], and Muegge [56] can be used to measure drug-likeness of the candidate compounds according to some acute criterion. These criteria are a molecular weight, Log P, number of hydrogen bond acceptors and donors.
Molecular properties viz., bioavailability and membrane permeability are correlated with simple molecular descriptors such as partition coefficient log P, H-bond donors and acceptors in a molecule [56]. Lipinski’s rule [52] of 5 is employed to disclose “drugability” of molecules. Thus, only for two compounds 6 and 8 molecular weights was higher than 500. Violations to the above-revealed rules together with drug-likeness and oral bioavailability scores are represented in Table 7. Most of the compounds violated any rule and their bioavailability score was around 0.55. The absorption magnitude is given as an absorption percentage. Following the law %ABS = 109 − 0.345 PSA, the absorption percent was computed [57]. Polar surface area (PSA) was defined as the fragment-based increments described by Ertl and coworkers [58,59]. The existence of more than 10 hydrogen-bond acceptors, 5 hydrogen-bond donors, demonstrates poor absorption or permeation. All derivatives contain <10 hydrogen bond acceptors and <5 hydrogen bond donors (Table 7).
As depicted in figures of Table 8, curves with green color indicates non-drug-like behavior and blue color are considered as drug-like. Compounds with zero or negative value cannot be considered as drug-like. The drug-likeness score was found to be from −0.42 to 0.56 for the compounds under investigation. However, compounds 6 and 8 have two violations from Lipinski rule and cannot be treated as drug candidate; even they showed good antibacterial activity.

2.7. Docking Studies

Docking Studies to Antibacterial Targets

According to PASS prediction, our compounds found to be possible inhibitors of LD-carboxypeptidase (LdcA). As such, we included this enzyme to docking studies. LD-Carboxypeptidases acts by cleaving amide bonds between L- and D-amino acids in bacterial peptidoglycan. More specific, cleaving the link between meso-diaminopimelic acid and d-alanine and consequently reduce tetrapeptides to the corresponding tripeptides, which can then be reconverted into peptidoglycan building blocks by the attachment of preformed D-Ala-D-Ala dipeptides. Therefore, LD-carboxypeptidases are thought to play a critical role in peptidoglycan recycling [60]. Crystallographic studies revealed that LD-carboxypeptidase is a serine protease with that Ser115, His285, and Glu217 forming a functional catalytic triad [61].
The docking studies showed that the free energy of binding to E. coli DNA Gyrase, Thymidylate kinase, E. coli Primase and E. coli MurB were higher than that to LdcA, therefore it may be considered that inhibition of LdcA enzyme is probably the possible mechanism of action of the compounds (Table 9).
Docking studies revealed that the most active compound 18 binds to LdcA enzyme forming a favorable hydrogen bond interaction between the nitrogen atom of benzothiazole ring and the hydrogen of the side chain of Ser116 (distance 3.24 Å). The benzothiazole moiety interacts hydrophobically with the residues Val36, Arg86 and Gly87, while the thiazolidinone ring with the residues Gly88 and Val219 (Figure 3B). Furthermore, the benzene moiety is placed in a cavity that consists of the residues Tyr58, Gly88 and Tyr224, interacting hydrophobically. These interactions further stabilize the complex compound-enzyme contributing to inhibitory activity of the compound 18. Moreover, the hydrogen bond formation with the residue Ser115 is crucial for the inhibitory activity of this compound, as it is among the amino acids of the catalytic triad of the enzyme. Hydrogen bond interactions with the residues of the catalytic triad of the enzyme were also observed for compounds 16 (Figure 3B), 19, 2, 4, 6 and 8, explaining their higher inhibitory activity.
Docking studies to antifungal targets showed that only compounds 18 and 19 had a significant good estimated free energy of binding to CYP51ca enzyme with values −6.78 and −6.94 kcal/mol respectively. The rest of the compounds had values ranging from −2.10 to −6.21 kcal/mol.

3. Materials and Methods

The MEL-TEMP II device (LAB Devices, Holliston, MA, USA) was used to determine the melting points and are uncorrected. Infrared (IR) spectra were recorded in Nujol on the Perkin Elmer Spectrum BX dual-beam spectrometer. 1H NMR nuclear magnetic resonance spectra in DMSO-d6 or CDCl3 were obtained with an Agilent spectrometer at 500 MHz. Chemical shift values are given in parts per million (ppm/s), while tetramethylsilane (TMS, δTMS = 0) was used as the internal standard. The ESI-MS (Micromass ZMD Waters) spectrometer was used to obtain the mass spectra (MS). The progress of the reactions was checked by thin layer chromatography using F254 silica gel chromatography plates (Merck, Darmstadt, Germany). All reagents and solvents were purchased from Aldrich Chemie (Steinheim, Germany) and were of high analytical purity.

3.1. Chemistry

Method A. The appropriate (hetero) aromatic amine (1.0 mmol) and the appropriately substituted benzaldehyde (1.2 mmol) were refluxed in dry toluene followed by the addition of thioglycolic acid (2.0 mmol). Heating is continued for 4–39 h until the (hetero) aromatic amine complete reacted. At the end of the reaction the solvent was removed in vacuo and the residue was taken up in ethyl acetate. This is followed by successive washes of the organic layer with 5% aqueous citric acid, water and 5% aqueous sodium bicarbonate. The solvent was removed in vacuo and the organic layer was dried over sodium sulfate. A solid residue was obtained and washed with 95% ethanol. The final product was allowed to dry and recrystallized from 95% ethanol if necessary.
Method B. The reactions using microwave radiation were performed with the CEM-Discover Monomode instrument, with a frequency of 2.45 GHz and continuous irradiation with a maximum power of 100 W. The appropriate (hetero) aromatic amine (1 mmol) together with the appropriately substituted benzaldehyde (1.3 mmol) and thioglycolic acid (5 mmol) were placed in a special tube with a capacity of 10 mL. Add 2–3 mL of absolute ethanol, covered the tube with a special Teflon stopper and placed it in the instrument (CEM). The mixture was irradiated for 20–30 min at 80–100 °C using a maximum pressure of 250 psi. The reaction stirred continuously and after completion, the tube was cooled to ambient temperature. The solid product was filtered under reduced pressure, washed with methanol and allowed to dry.
  • Synthesis of 2-(2-(4-nitrophenyl)-4-oxothiazolidin-3-yl)benzo[d]thiazole-6-carbonitrile (1).
Method A: The reaction time was 13 h. Yield: 54.4% Method B (MW irradiation): The reaction was carried out at a temperature of 100 °C and the time required was 30 min. Yield: 82.5% M. p.: 166–167 °C. Rf: 0.62 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2364 (=Ν-), 2218 (-CN), 1706 (C=O). 1H-NMR (500 MHz, CHCl3-d): δ ppm 3.91 (d, J = 16.63 Hz, 1H, CH2), 4.11 (d, J = 16.63 Hz, 1H, CH2), 6.49 (s, 1H, N-CH-S), 7.48 (d, J = 7.83 Hz, 2H, Ar-C17, C21), 7.55–7.57 (m, 1H, Ar-C5), 7.77 (d, J = 7.86 Hz, 1H, Ar-C4), 8.20 (d, J = 7.81 Hz, 2H, Ar-C18, C20), 8.40 (d, J = 7.83 Hz, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 32.77 (1C, CH2), 62.57 (1C, N-CH-S), 103.88, 115.42 (2C, Ar), 118.80 (1C, -CN), 122.52, 124.30(2C), 126.41(2C), 133.25, 142.24, 147.80, 153.69, 157.25 (10C, Ar), 163.70 (1C, N=C-–N), 170.28 (1C, C=O).
  • Synthesis of 2-(2-(2,6-difluorophenyl)-4-oxothiazolidin-3-yl)benzo[d]thiazole-6-carbonitrile (2).
Reaction time: 10 h. Yield: 57%. M p.183–184°C. Rf: 0.71 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2361 (=Ν-), 2218 (-CN), 1711 (C=O), 1591 (C=C arom), 1001 (C-F). 1H-NMR (500 MHz, CHCl3-d): δ ppm 3.95 (d, J = 16.14 Hz, 1H, CH2) 4.11 (d, J = 16.14 Hz, 1H, CH2) 6.80 (s, 1H, N-CH-S) 7.31–7.40 (m, 2H, Ar-C18, C20) 7.42–7.49 (m, 1H, Ar-C19Ar-C18, C20) 7.53 (d, J = 8.31 Hz, 1H, Ar-C5) 7.92(d, J = 8.31 Hz, 1H, Ar-C4) 8.02 (d, J = 1.96 Hz, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 33.20 (1C, CH2), 60.11 (1C, N-CH-S), 104.43, 111.07(2C), 112.85, 117.65 (5C, Ar), 118.25 (1C, -CN), 126.72, 128.71, 129.47, 130.48, 145.72, 158.02 (7C, Ar), 163.44 (2C, C-F), 163.59 (1C, N=C-N), 171.01 (1C, C=O).
  • Synthesis of 2-(2-(2-chloro-6-fluorophenyl)-4-oxothiazolidin-3-yl)benzo[d]thiazole-6-carbonitrile (3).
Reaction time: 16 h (A), 30 min (B) respectively. Yield: 41.5%(A), 88.5% (B), respectively. M p.176–177 °C. Rf: 0.76 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2361 (=Ν-), 2218 (-CN), 1709 (C=O), 1602 (C=C arom), 1191 (C-F), 721 (C-Cl). 1H-NMR (500 MHz, CHCl3-d): δ ppm 4.04 (d, J = 16.63 Hz, 1H, CH2), 4.28 (d, J = 16.63 Hz, 1H, CH2), 6.84 (s, 1H, N-CH-S), 7.09 (t, J = 8.43 Hz, 1H, Ar-C19), 7.59–7.65 (m, 2H, Ar-C18, C20), 7.72 (d, J = 7.32 Hz, 1H, Ar-C5), 7.93 (d, J = 7.32 Hz, 1H, Ar-C4), 8.37 (d, J = 8.32 Hz, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 33.72 (1C, CH2), 60.28 (1C, N-CH-S), 104.49, 113.67, 117.57 (2C, Ar), 118.19 (1C, -CN), 125.31, 126.87, 128.79, 129.85, 130.81, 131.47, 135.29, 157.02 161.18 (9C, Ar), 163.74 (1C, N=C-N), 171.23 (1C, C=O).
  • Synthesis of 2-(2-(2,6-dichlorophenyl)-4-oxothiazolidin-3-yl)benzo[d]thiazole-6-carbonitrile (4).
Reaction time: 18 h (A), 30 min (B) respectively. Yield: 29.1%(A), 83.4% (B), respectively. M p.198–200 °C. Rf: 0.63 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 1706 (C=O), 1572, 1230, 1213. 1H-NMR (500 MHz, CHCl3-d): 3.76 (s, 3H, O-CH3), 3.91 (d, J = 16.38 Hz, 1H, CH2), 4.10 (d, J = 16.63 Hz, 1H, CH2), 6.73 (s, 1H, N-CH-S), 6.84 (d, J = 8.80 Hz, 2H, Ar-C18, C20), 7.12 (t, J = 8.56 Hz, 1H, Ar-C5), 7.23–7.35 (m, 1H, Ar-C4), 7.46–7.55 (m, 2H, Ar-C17, C21), 7.58–7.79 (m, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 33.72 (1C, CH2), 58.01, 63.08, 109.98, 113.89, 114.13, 117.94, 129.49, 131.98, 132.35, 137.38, 146.73, 156.1, 158.01, 163.51, 171.03 (1C, C=O).
  • Synthesis of 2-(2-(4-fluorophenyl)-4-oxothiazolidin-3-yl)benzo[d]thiazole-6-carbonitrile (5).
Reaction time: 25 h (A), 30 min (B) respectively. Yield: 52.4%(A), 87.1% (B), respectively. M p.185–186 °C. Rf: 0.68 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2364 (=Ν-), 2218 (-CN), 1709 (C=O). 1H-NMR (500 MHz, CHCl3-d): 3.94 (d, J = 16.14 Hz, 1H, CH2), 4.10 (d, J = 16.14 Hz, 1H, CH2), 6.82 (s, 1H, N-CH-S), 7.12–7.22 (m, 4H, Ar-C17, C18, C20, C21), 7.81 (d, J = 8.31 Hz, 1H, Ar-C5), 7.92 (d, J = 8.31 Hz, 1H, Ar-C4) 8.16 (d, J = 1.96 Hz, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 33.21 (1C, CH2), 64.14, 104.41, 115.45, 117.65, 118.23, 126.72, 129.47, 130.55, 131.09, 134.18, 158.15, 161.25, 164.69, 170.92 (1C, C=O).
  • Synthesis of 3-(6-(adamantan-1-yl)benzo[d]thiazol-2-yl)-2-(2,6-dichlorophenyl)thiazolidin-4-one (6).
Reaction time: 21 h (A), 30 min (B) respectively. Yield: 88.3% (A), 89.7% (B), respectively. M p.184–185 °C. Rf: 0.45 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2974, 2879, 2840, (C-H adamant), 2347 (=Ν-), 1700 (C=O), 1532 (C=C arom), 721 (C-Cl). 1H-NMR (500 MHz, CHCl3-d): δ ppm 1.75–1.82 (m, 3H, Ad), 1.85–1.93 (m, 3H, Ad), 1.95–2.01 (m, 3H, Ad), 3.91 (br d, J = 16.14 Hz, 1H, CH2), 4.13 (d, J = 16.14 Hz, 1H, CH2), 6.80 (s, 1H, N-CH-S), 7.45–7.48 (m, 3H, Ar-C18, C19, C20), 7.93 (d, J = 8.31 Hz, 1H, Ar-C4), 8.04 (d, J = 1.96 Hz, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 28.93 (3C, Ad), 32.75 (1C, CH2), 36.46(1C, Ad), 36.80(3C, Ad), 43.42(3C, Ad), 62.15 (1C, N-CH-S), 119.58, 121.05, 124.11, 128.39(2C), 129.55, 130.98, 135.42(2C), 139.11, 142.03, 150.54 (12C, Ar), 164.13 (1C, N=C-N), 171.10 (1C, C=O).
  • Synthesis of 3-(6-(adamantan-1-yl)benzo[d]thiazol-2-yl)-2-(2-chloro-6-fluorophenyl) thiazolidin-4-one (7).
Reaction time: 15 h (A). Yield: 73.4%(A). M p.183–184 °C. Rf: 0.59 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2843 (C-H Ad), 2358 (=Ν-), 1653 (C=O), 1534 (C=C arom), 1099 (C-F), 724 (C-Cl). 1H-NMR (500 MHz, CHCl3-d): δ ppm 1.75–1.82 (m, 3H, Ad), 1.93 (d, J = 2.45 Hz, 3H, Ad) 2.11 (br s, 3H, Ad) 3.91 (br d, J = 16.14 Hz, 1H, CH2), 4.27 (d, J = 16.14 Hz, 1H, CH2), 6.80–6.90 (m, 2H, Ar-C18, C20), 7.10 (s, 1H, N-CH-S), 7.15–7.24 (m, 1H, Ar-C19) 7.42 (dd, J = 8.80, 1.96 Hz, 1H, Ar-C5), 7.65 (d, J = 8.31 Hz, 1H, Ar-C4), 7.74 (d, J = 1.96 Hz, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 28.92 (2C, Ad), 29.68 (1C, Ad), 33.82 (1C, CH2), 36.45(1C, Ad), 36.69(3C, Ad), 43.41(3C, Ad), 53.99 (1C, N-CH-S), 111.71, 111.88, 111,90, 117.13, 121.38, 123.51, 128.92(2C), 129.97, 132.14, 146.10, 148.11 (12C, Ar), 155.37 (1C, N=C-N), 170.65 (1C, C=O).
  • Synthesis of 3-(6-(adamantan-1-yl)-4-methylbenzo[d]thiazol-2-yl)-2-(2,6-dichlorophenyl)thiazolidin-4-one (8).
Reaction time: 17 h (A). Yield: 81.8% (A). M p.191–192 °C. Rf: 0.43 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2856 (C-H Ad), 2364 (=Ν-), 1700 (C=O), 1381 (-CH3), 721 (C-Cl). 1H-NMR (500 MHz, CHCl3-d): δ ppm 1.73–1.82 (m, 3H, Ad), 1.85–1.93 (m, 3H, Ad), 1.97–2.02 (m, 3H, Ad), 2.55 (s, 3H, -CH3), 3.95 (d, J = 16.14 Hz, 1H, CH2), 4.12 (d, J = 16.14 Hz, 1H, CH2), 6.81 (s, 1H, N-CH-S), 7.24 (s, 1H, Ar-C5), 7.40–7.53 (m, 3H, Ar-C18, C19, C20), 7.95 (d, J = 1.96 Hz, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 16.75(1C, -CH3) 28.92 (3C, Ad), 32.74 (1C, CH2), 36.44(1C, Ad), 36.80(3C, Ad), 43.42(3C, Ad), 62.47 (1C, N-CH-S), 115.53, 124.17, 124.98, 128.16(2C), 129.17, 131.01, 135.44(2C), 139.12, 142.05, 145.00 (12C, Ar), 164.13 (1C, N=C-N), 171.11 (1C, C=O).
  • Synthesis of 3-(6-(adamantan-1-yl)-4-methylbenzo[d]thiazol-2-yl)-2-(2,6-difluorophenyl)thiazolidin-4-one (9).
Reaction time: 22 h (A), 30 min (B) respectively. Yield: 87.3% (A), 89.6% (B), respectively. M p.188–189 °C. Rf: 0.59 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2842 (C-H Ad), 2352 (=Ν-), 1709 (C=O), 1535 (C=C arom), 1383 (-CH3), 1099 (C-F). 1H-NMR (500 MHz, CHCl3-d): δ ppm 1.73–1.84 (m, 3H, Ad), 1.86–1.93 (m, 3H, Ad), 1.95–2.03 (m, 3H, Ad), 2.54 (s, 3H, -CH3), 3.86 (d, J = 16.14 Hz, 1H, CH2), 4.14 (d, J = 16.14 Hz, 1H, CH2), 6.85 (s, 1H, N-CH-S), 7.25–7.39 (m, 3H, Ar-C5, C18, C20), 7.59 (br t, J = 8.80, 1H, Ar-C19), 7.95 (d, J = 1.96 Hz, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 16.75(1C, -CH3) 28.91 (3C, Ad), 32.74 (1C, CH2), 36.41(1C, Ad), 36.92(3C, Ad), 43.41(3C, Ad), 58.86 (1C, N-CH-S), 111.51(2C), 112.74, 115.91, 124.12, 124.76, 128.49(2C), 131.01, 142.33 (10C, Ar), 162.85(2C, C-F), 164.11 (1C, N=C-N), 171.08 (1C, C=O). (MS): (m/z) 497 (M+, 12%), 482 (7%), 463 (5%), 423 (78%), 244 (100%), 198 (10%), 171 (11%), 152 (18%).
  • Synthesis of 2-(4-fluorophenyl) -3-(4-methoxybenzo [d] thiazol-2-yl) thiazolidin-4-one (10).
Reaction time: 6h (A). Yield:51.4%. M p. 251–252 °C.: Rf: 0.73 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2364 (=Ν-), 1709 (C=O), 1587 (C=C arom), 1272 (-OCH3), 1113 (C-F, benz). 1H-NMR (500 MHz, DMSO-d6): δ ppm 3.68–3.86 (m, 3H, O-CH3), 3.91–4.09 (m, 1H, CH2), 4.25 (br d, J = 9.78 Hz, 1H, CH2), 6.88 (s, 1H, N-CH-S), 7.06 (br d, J = 10.27 Hz, 1H, Ar-C5), 7.26 (br dd, J = 10.03, 8.07 Hz, 1H, Ar-C6), 7.55 (br d, J = 10.27 Hz, 1H, Ar-C7), 7.61–7.76 (m, 2H, Ar-C17, C21), 8.08–8.26 (m, 2H, Ar-C18, C20). 13C-NMR (500 MHz, DMSO-d6): δ ppm 32.86 (1C, CH2), 55.82 (1C, O-CH3), 63.18 (1C, N-CH-S), 108.26, 114.11, 115.43(2C), 121.18, 130.33(2C), 131.99, 135.59, 142.31, 150.23, 161.51 (12C, Ar), 163.91 (1C, N=C-N), 171.08 (1C, C=O). (MS): (m/z) 361 (M+, 100%), 343 (42%), 321 (36%), 290 (72%), 288 (31%), 279 (39%), 225 (24%).
  • Synthesis of 3-(4-methoxybenzo[d]thiazol-2-yl)-2-(4-nitrophenyl)thiazolidin-4-one (11).
Reaction time: 10 h (A). Yield: 63.5%.M p: 298–299 °C. Rf: 0.48 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2364 (=Ν-), 1709 (C=O), 1585 (C=C arom), 1272 (-OCH3). 1H-NMR (500 MHz, DMSO-d6): δ ppm 3.77 (s, 3H, O-CH3), 4.03 (d, J = 17.12 Hz, 1H, CH2), 4.25 (d, J = 17.12 Hz, 1H, CH2), 6.93 (br d, J = 10.27 Hz, 1H, Ar-C5), 7.04 (s, 1H, N-CH-S), 7.26 (s, 1H, Ar-C6) 7.56 (d, J = 7.83 Hz, 1H, Ar-C17) 7.67 (d, J = 8.80 Hz, 2H, Ar-C7, C21) 8.17 (d, J = 8.32 Hz, 2H, Ar-C18, C20). 13C-NMR (500 MHz, DMSO-d6): δ ppm 32.85 (1C, CH2), 55.86 (1C, O-CH3), 63.21 (1C, N-CH-S), 108.25, 114.12, 121.18, 123.84(2C), 129.66(2C), 131.97, 142.31, 145.26, 146.32, 150.21(12C, Ar), 163.95 (1C, N=C-N), 171.02 (1C, C=O). (MS): (m/z) 388 (M+, 100%), 288 (73%), 272 (3%), 225 (42%), 184 (76%), 136 (42%), 115 (31%), 73 (28%).
  • Synthesis of 2-(4-chlorophenyl)-3-(4-methoxybenzo[d]thiazol-2-yl)thiazolidin-4-one (12).
Reaction time 8 h (A) Yield: 43.2%. (B) reaction time 30 min. Yield: 89.4%. M p: 218–219 °C. Rf: 0.53 (petroleum ether/ethyl acetate:8/2). IR: (cm−1, Nujol): 2364 (=Ν-), 1706 (C=O), 1594 (C=C arom), 1275 (-OCH3), 721 (C-Cl benz). 1H-NMR (500 MHz, DMSO-d6): δ ppm 3.80 (s, 3H, O-CH3), 3.99 (br d, J = 17.12 Hz, 1H, CH2), 4.23 (br d, J = 16.63 Hz, 1H, CH2), 6.93 (s, 1H, N-CH-S), 6.91–6.97 (m, 1H, Ar-C5), 7.26 (br t, J = 8.07 Hz, 1H, Ar-C4), 7.39 (br d, J = 9.29 Hz, 4H, Ar-C17, C18, C20, C21), 7.54 (br d, J = 7.83 Hz, 1H, Ar-C7). 13C-NMR (500 MHz, DMSO-d6): δ ppm 32.82 (1C, CH2), 55.88 (1C, O-CH3), 63.27 (1C, N-CH-S), 108.25, 114.11, 121.32, 128.73(2C), 130.12(2C), 131.93, 132.75, 137.36, 142.32, 150.22(12C, Ar), 164.03 (1C, N=C-N), 171.00 (1C, C=O).
  • Synthesis of 3-(4-methoxybenzo[d]thiazol-2-yl)-2-(4-methoxyphenyl)thiazolidin-4-one (13).
Reaction time 15 h. Yield: 52.1% (A). Reaction time: 30 min. Yield: 78.5%. (B) Mp: 196–197 °C. Rf: 0.63 petroleum ether/ethyl acetate (8/2). IR: (cm−1, Nujol): 2364 (=Ν-), 1703 (C=O), 1569 (C=C arom), 1269 (-OCH3). 1H-NMR (500 MHz, CHCl3-d): δ ppm 3.76 (s, 3H, O-CH3benz), 3.94 (d, J = 16.63 Hz, 1H, CH2), 3.95 (s, 3H, O-CH3), 4.13 (d, J = 16.63 Hz, 1H, CH2), 6.81–6.85 (m, 2H, Ar-C18, C20), 7.93 (s, 1H, N-CH-S), 7.25 (br d, J = 9.29 Hz, 3H, Ar-C5, C6, C7), 7.39 (br d, J = 7.83 Hz, 2H, Ar-C17, C21). 13C-NMR (500 MHz, CHCl3-d): δ ppm 32.86 (1C, CH2), 55.83 (1C, O-CH3), 63.35 (1C, N-CH-S), 108.25, 114.11, 114.76(2C), 121.82, 129.75(2C), 131.55, 131.96, 142.34, 150.23, 160.82(12C, Ar), 164.06 (1C, N=C-N), 171.11 (1C, C=O). (MS): (m/z) 373 (M+, 76%), 272 (33%), 243 (56%), 244 (100%), 242 (8%), 165 (5%), 164 (7%), 135 (11%).
  • Synthesis of 2-(4-hydroxyphenyl)-3-(4-methoxybenzo[d]thiazol-2-yl)thiazolidin-4-one (14).
Reaction time: 26 h. Yield: 29.0% (A), reaction time: 30 min. Yield: 72.8%. (B). M p: 261–262 °C. Rf: 0.60 petroleum ether/ethyl acetate (8/2). IR: (cm−1, Nujol): 3193 (-OH), 1703 (C=O), 1572 (C=C arom), 1271 (-OCH3). 1H-NMR (500 MHz, DMSO-d6): δ ppm 3.77 (s, 3H, O-CH3), 4.03 (d, J = 17.12 Hz, 1H, CH2), 4.20 (d, J = 17.12 Hz, 1H, CH2), 5.32 (br s, 1H, -OH), 6.93 (d, J = 7.83 Hz, 2H, Ar-C18, C20), 7.05 (s, 1H, N-CH-S), 7.26 (br t, J = 8.56 Hz, 1H, Ar-C5), 7.56 (d, J = 7.83 Hz, 1H, Ar-C6), 7.67 (d, J = 8.80 Hz, 1H, Ar-C7), 8.17 (d, J = 8.32 Hz, 2H, Ar-C17, C21). 13C-NMR (500 MHz, CHCl3-d): δ ppm 32.88 (1C, CH2), 55.82 (1C, O-CH3), 63.33 (1C, N-CH-S), 108.21, 114.13, 115.81(2C), 121.73, 130.13(2C), 131.52, 131.96, 142.31, 150.22, 156.95(12C, Ar), 164.13 (1C, N=C-N), 171.04 (1C, C=O).
  • Synthesis of 3-(6-chlorobenzo[d]thiazol-2-yl)-2-(4-fluorophenyl)thiazolidin-4-one (15).
Reaction time: 8 h (A). Yield: 51.3%. M p.: 196–197 °C. Rf: 0.76(petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2364 (=Ν-), 1681 (C=O), 1571 (C=C arom), 1101 (C-F Bz). 1H-NMR (500 MHz, CHCl3-d): δ ppm 3.85 (d, J = 16.38 Hz, 1H, CH2), 4.11 (d, J = 16.63 Hz, 1H, CH2), 6.75 (s, 1H, N-CH-S), 7.00–7.03 (m, 2H, Ar-C18, C20), 7.30–7.34 (m, 3H, Ar-C5, C17, C21), 7.60 (d, J = 8.02 Hz, 1H, Ar-C4), 7.76 (s, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 32.79 (1C, CH2), 63.09 (1C, N-CH-S), 115.77, 115.94, 120.83, 122.70, 126.89, 127.49, 127.55, 133.32, 136.12, 146.77, 156.13 (12C, Ar), 163.52 (1C, N=C-N), 170.87 (1C, C=O). (MS): (m/z) 364 (M+, 100%), 343 (10%), 299 (10%), 290 (100%), 279 (9%), 184 (11%), 115 (80%), 88 (9%), 73 (94%).
  • Synthesis of 3-(6-chlorobenzo[d]thiazol-2-yl)-2-(4-nitrophenyl)thiazolidin-4-one (16).
Reaction time: 12 h (A), 30 min, 100 °C (B), respectively. Yield: 52.7% (A), 88.3% (B) respectively. M p.: 208–209 °C. Rf: 0.62(petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2364 (=Ν-), 1704 (C=O), 1591 (C=C arom). 1H-NMR (500 MHz, CHCl3-d): δ ppm 3.90 (d, J = 16.63 Hz, 1H, CH2), 4.12 (d, J = 16.63 Hz, 1H, CH2), 6.78 (s, 1H, N-CH-S), 7.21–7.30(d, J = 7.34 Hz, 2H, Ar-C17, C21), 7.53 (br d, J = 8.80 Hz, 1H, Ar-C5), 7.80 (br d, J = 8.80 Hz, 1H, Ar-C4), 8.20 (d, J = 8.31 Hz, 3H, Ar-C7, C18, C20). 13C-NMR (500 MHz, CHCl3-d): δ ppm 32.86 (1C, CH2), 62.55 (1C, N-CH-S), 121.09, 122.76, 124.42 (2C), 126.37, 127.20(2C), 130.93, 133.31, 145.21 146.85, 151.28 (12C, Ar), 163.55 (1C, N=C-N), 170.48 (1C, C=O).
  • Synthesis of 3-(6-chlorobenzo[d]thiazol-2-yl)-2-(4-chlorophenyl)thiazolidin-4-one (17).
Reaction time: 28 h (A), 30 min, 100 °C (B), respectively. Yield: 44.2% (A), 72.1% (B) respectively. M p.: 188–189 °C. Rf: 0.71 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2358(=Ν-), 1709 (C=O), 1591 (C=C arom), 720 (C-Cl bz). 1H-NMR (500 MHz, CHCl3-d): δ ppm 3.86 (d, J = 16.38 Hz, 1H, CH2), 4.10 (d, J = 16.63 Hz, 1H, CH2), 6.73 (s, 1H, N-CH-S), 7.26–7.40 (m, 4H, Ar-C17, C18, C20, C21), 7.56 (br d, J = 7.83 Hz, 1H, Ar-C5), 7.76 (br d, J = 8.80 Hz, 1H, Ar-C4), 8.13 (s, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 32.84 (1C, CH2), 63.11 (1C, N-CH-S), 119.02, 120.82, 122.76, 127.98(2C), 129.74(2C), 131.97, 133.30, 135.46, 138.85, 148.06 (12C, Ar), 158.04 (1C, N=C-N), 171.19 (1C, C=O).
  • Synthesis of 3-(6-chlorobenzo[d]thiazol-2-yl)-2-(4-methoxyphenyl)thiazolidin-4-one (18).
Reaction time: 13 h (A). Yield: 62.3%. M p. 220–221 °C: Rf: 0.62 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 2364 (=Ν-), 1709 (C=O), 1571 (C=C arom), 1222 (-OCH3). 1H-NMR (500 MHz, CHCl3-d): δ ppm 3.76 (s, 3H, O-CH3), 3.91 (d, J=16.38 Hz, 1H, CH2), 4.10 (d, J = 16.63 Hz, 1H, CH2), 6.72 (s, 1H, N-CH-S), 6.82 (d, J = 8.80 Hz, 2H, Ar-C18, C20), 7.12 (t, J = 8.56 Hz, 1H, Ar-C5), 7.23–7.38 (m, 1H, Ar-C4), 7.46–7.53 (m, 2H, Ar-C17, C21), 8.08 (s, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d): δ ppm 32.88 (1C, CH2), 55.27 (1C, O-CH3), 63.47 (1C, N-CH-S), 114.20(2C), 120.79, 126.76, 126.79(2C), 127.73, 129.92, 132.28, 133.36, 147.44, 158.89 (12C, Ar), 163.59 (1C, N=C-N), 171.01 (1C, C=O).
  • Synthesis of (3-(6-chlorobenzo[d]thiazol-2-yl)-2-(4-hydroxyphenyl)thiazolidin-4-one (19).
Reaction time 18 h (A), 30 min, 100 °C (B), respectively. Yield: 24.1% (A), 72.5% (B) respectively. Mp. 236–237°C °C: Rf: 0.40 (petroleum ether/ethyl acetate: 8/2). IR: (cm−1, Nujol): 3198 (-OH), 2358 (=Ν-), 1709 (C=O), 1573 (C=C arom). 1H-NMR (500 MHz, CHCl3-d): δ ppm 3.86 (d, J = 16.38 Hz, 1H, CH2), 4.10 (d, J = 16.63 Hz, 1H, CH2), 5.35 (br s, 1H, -OH), 6.73 (s, 1H, N-CH-S), 7.26–7.43 (m, 5H, Ar-C5, C17, C18, C20, C21), 7.76 (br d, J = 8.80 Hz, 1H, Ar-C4), 8.13 (s, 1H, Ar-C7). 13C-NMR (500 MHz, CHCl3-d) δ ppm 33.12 (1C, CH2), 63.71 (1C, N-CH-S), 115.75 (2C), 118.23, 121.55, 125.14, 129.02, 130.01(2C), 131.52, 132.87, 151.68, 156.71 (12C, Ar), 163.77 (1C, N=C-N), 171.12 (1C, C=O). (MS): (m/z) 363 (M+, 100%), 288 (88%), 272 (9%), 225 (33%), 184 (48%), 146 (7%), 136 (21%), 115 (18%), 73 (17%).

3.2. Biological Evaluation

3.2.1. Antibacterial Action

Bacterial strains utilized include Gram-negative: Salmonella typhimurium, (ATCC 13311).
Pseudomonas aeruginosa (ATCC 27853), Escherichia coli (ATCC 35210), and Gram-positive bacteria: Staphylococcus aureus (ATCC 6538) and Listeria monocytogenes (NCTC 7973) bacteria. Pathogens were provided from the Mycological Laboratory, Institute for Biological Research “Siniša Stankovic” Belgrade. Resistant strains used were MRSA IBRS MRSA 011, E. coli IBRS E003 and P. aeruginosa IBRS P001 obtained as described in Kartsev et al. [62]. The MIC/MBC were effectuated utilizing microdilution assay as previous described [63,64].

E. coli

Sensitivity studies of E. coli strain were tested by the disc diffusion method on Mueller Hinton agar with the use of antibiogram discs (Bioanalyse) and tablets (Torlak, Serbia) for the following antibiotics: penicillin, amoxicillin, tetracycline, neomicin, gentamicin, colistin, ceftriaxon, sulfamethaxasole with trimetoprim, enrofloxacin and florfenicol. E. coli strain was resistant to all tested antibiotics with the exception of enrofloxacin, colistin and florfenicol [65]. It is described in detail in our previous paper [46].

Pseudomonas aeruginosa

Sensitivity studies of P. aeruginosa strain were tested by the disc diffusion method on Mueller Hinton agar with the use of antibiogram discs (Bioanalyse) and tablets (Torlak, Serbia) for the following antibiotics: penicillin, amoxicillin, tetracycline, neomicin, gentamicin, ceftriaxon, sulfamethaxasole with trimetoprim, enrofloxacin and florfenicol. P. aeruginosa strain was resistant to all tested antibiotics with the exception of enrofloxacin, and florfenicol [65,66].

3.2.2. Inhibition of Biofilm Formation

This method was performed as described previously [48,67] with some modifications. The percentage of inhibition of biofilm formation was calculated by the following formula:
[(A620 control − A620 sample)/A620 control] × 100

3.2.3. Antifungal Activity

For the antifungal bioassays, six fungi were used: Aspergillus niger (ATCC 6275), Aspergillus fumigatus (human isolate), Aspergillus versicolor (ATCC 11730), Penicillium funiculosum (ATCC 36839), Trichoderma viride (IAM 5061), Penicillium verrucosum var. cyclopium (food isolate). The organisms were obtained from the Mycological Laboratory, Department of Plant Physiology, Institute for Biological Research ‘Siniša Stankovic’, Belgrade, Serbia. All experiments were performed in duplicate and repeated three times [68,69].

3.3. Statistical Analysis

All tests were performed three times and the values were determined as standard deviation (SD) and mean values. One-way ANOVA test was allowed to determine variance analysis with Tukey HSD Test (0.05 levels). Analysis was executed with the help of SPSS statistics software (version 18).

3.4. Drug-Likeness

The targeted molecules were appraised for predicting the Drug-likeness based on 5 separate filters namely Egan [55], Ghose [53], Muegge [56], Veber [54] and Lipinski [52] rules accompanying bioavailability and Drug-likeness scores using the Molsoft software and SwissADME program (http://swissadme.ch, accessed on 28 June 2021) using the ChemAxon’s Marvin JS structure drawing tool.

3.5. Docking Studies

Protein Preparation: X-ray crystal structures of E. coli DNA GyrB, Thymidylate kinase, E. coli primase, E. coli MurB, LD-carboxypeptidase, (PDB code: 1KZN, AQGG, 1DDE, 2Q85, 1ZRS, respectively) were retrieved from Brookhaven Protein Data Bank (PDB). The pdb files of proteins were submitted to “Build/check/repair model” to the session “Prepare PDB file for docking programs” and missing side chains were modeled in; water positions and symmetry were corrected, and hydrogen atoms were added. Only chain A of each enzyme of the repaired pdb file was evaluated and passed to AutodockTools (ADT ver. 1.5.6) for pdbqt file preparation. ADT assigned polar hydrogens, water molecules and non-standard residues were removed, only polar hydrogens were maintained, and Gasteiger charges were computed for protein atoms. AutoDock saved the prepared file in PDBQT format.
Ligand Preparation: All the molecules were sketched in chemdraw12.0 program. The geometry of built compounds was optimized using the molecular mechanical force fields 94 (MMFF94) energy via program LigandScout, partial charges were also calculated, comformers of each ligand were generated and the best one was maintained and saved as mol2 files that were passed to ADT for pdbqt file preparation. Polar hydrogens were added to each structure, followed by computing Gasteiger and Kollman charges, and the torsions.
Docking Procedure: Autodock 4 (ver. 4.2.6) was employed for docking simulations. The region of interest, used by Autodock4 for docking runs and by Autogrid4 for affinity grid maps preparation, was defined in such a way to comprise the whole catalytic binding site using a grid of 50 × 50 × 50 points with a grid space of 0.375 Å. All parameters used in docking were default. The translation, quaternion and torsions steps were taken from default values in AutoDock. The Lamarckian genetic algorithm and the pseudo-Solis and Wets methods were applied for minimization using default parameters. The number of docking runs was 100. After docking, the 100 solutions were clustered into groups with RMS lower than 1.0 Ε. The clusters were ranked by the lowest energy representative of each cluster. Upon completion of docking, the best poses were screened by examination of binding energy (ΔGbinding, kcal/mol) and number in cluster. A preliminary blind docking was performed in order to validate the protocol. The RMSD values were predicted by superimposing each docked co-ligand on its original crystallographic bound conformation. The RMSD of all enzymes were in range of 0.85 to 1.43, which are acceptable.
In order to describe the ligand-binding pocket interactions, the top ranked binding mode found by AutoDock in complex with the binding pocket of each enzyme was selected. The resulting poses and potential interactions were visualized using the Discovery studio visualizer version 4.0 (BIOVIA, San Diego, CA, USA) and ligPlot+ (ver. 2.2).

4. Conclusions

With the purpose to improve the antibacterial activity of previously synthesized compounds, modifications were performed to the initial structure and nineteen new derivatives with 6-Cl, 4-OMe, 6-CN, 6-adamantan, 4-Me,6-adamantan substituents at benzothiazole ring were synthesized and evaluated in silico and experimentally for their antimicrobial activity against panel of four bacterial strains S. aureus, L. monocytogenes, E. coli and S. Typhimirium and three resistant strains MRSA, E. coli and P. aeruginosa.
It was observed that among all compounds tested the derivatives of 6-chlorobenzothiazole-based thiazolidinone exhibited higher activity, with compound 18 being the most promising (MIC and MBC at 0.10–0.25 mg/mL and 0.12–0.5 mg/mL, respectively). It should be mentioned that some of compounds exhibited superior/equal activity mostly against P. aeruginosa resistant and non-resistant, E. coli and two of them against the most resistant L. monocytogenes than reference drugs, ampicillin and streptomycin. The most sensitive bacteria appeared to be E. coli.
Compounds with the most promising antibacterial potential were studied for their effect on biofilm formation. It was found that the activity of tested compounds in concentration of MIC did not exceed the activity of reference drugs. On the other hand, in concentration of 0.5 MIC compound 19 exhibited higher antibiofilm activity than streptomycin. The comparison of obtained results on antibacterial activity with those of compounds with 6-OCF3 substituent in benzothiazole ring revealed that activity depends on substitutients not only at benzothiazole moiety but also of benzene ring. Thus, in case of 4-F substitution at benzene ring the replacement of 6-OCF3 by 6-Cl improved twice the activity against P. aeruginosa and P. aeruginosa resistant, while the replacement by 4-OMe slightly improve only activity against E. coli (0.15 mg/mL and 0.12mg/mL). Among 4-nitro derivatives the presence of 6-Cl as well as 6-CN substituent at benzothiazole ring appeared to be beneficial compared to 6-OCF3 and 4-OCH3, since activity against S. aureus, MRSA and resistant strain of E. coli increased 2.5 folds, while against L. monocytogenes and S. typhimurium from 4- to 5-folds better results (2–3 times) were obtained in case of the presence of 4-OMe and 4-OH substituents in benzene ring of 6-Cl-benzothiazole derivatives compared to the same substituents at the 4 position of benzene ring of 6-OCF3 derivatives.
Docking analysis to DNA Gyrase, thymidylate kinase, E. coli primase, E. coli MurB and E. coli LD carboxypeptidase indicate the probable involvement of the last enzyme in the mechanism of the antibacterial activity of the tested compounds.
Antifungal activity was moderate to low, lower than antibacterial. Finally, it can be concluded that in general compounds 1519 and especially 18 are promising for further modifications in order to develop new more active antibacterial agents.

Supplementary Materials

The following are available online, Table S1: PASS prediction results.

Author Contributions

Conceptualization—A.G.; methodology—M.I. and A.P.; software—A.P.; validation—A.P. and C.T.; formal analysis—K.N.V., A.B.N., H.K. and H.S.E.; investigation—A.P., M.I., A.Ć. and M.S.; data curation—A.G. and M.I.; writing—original draft preparation—A.G.; writing—review editing—A.G. and M.H.; supervision—A.G.; funding—M.H. and C.T.; resources—S.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Deanship of Scientific Research of King Faisal University, Saudi Arabia (Grant number: 1811019) and the Serbian Ministry of Education, Science and Technological Development [Contract No. 451-03-9/2021-14/200007].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Authors acknowledge the offer of PASS program by Poroikov V.V.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not applicable.

References

  1. Bruder, M.; Polo, G.; Trivella, D.B.B. Natural allosteric modulators and their biological targets: Molecular signatures and mechanisms. Nat. Prod. Rep. 2020, 37, 488–514. [Google Scholar] [CrossRef]
  2. Roemer, T.; Krysan, D.J. Antifungal drug development: Challenges, unmet clinical needs, and new approaches. Cold Spring Harb. Perspect. Med. 2014, 4, a019703. [Google Scholar] [CrossRef] [PubMed]
  3. Chandna, N.; Kapoor, J.K.; Grover, J.; Bairwa, K.; Goyal, V.; Jachak, S.M. Pyrazolylbenzyltriazoles as cyclooxygenase inhibitors: Synthesis and biological evaluation as dual anti-inflammatory and antimicrobial agents. New J. Chem. 2014, 38, 3662–3672. [Google Scholar] [CrossRef]
  4. Moore, N.; Pollack, C.; Butkerait, P. Adverse drug reactions and drug-drug interactions with over-the-counter NSAIDs. Ther. Clin. Risk Manag. 2015, 11, 1061–1075. [Google Scholar] [PubMed] [Green Version]
  5. Bersani, I.; Piersigilli, F.; Goffredo, B.M.; Santisi, A.; Cairoli, S.; Ronchetti, M.P.; Auriti, C. Antifungal Drugs for Invasive Candida Infections (ICI) in Neonates: Future Perspectives. Front. Ped. 2019, 7, 375–384. [Google Scholar] [CrossRef] [Green Version]
  6. Ferreira, L.G.; Dos Santos, R.N.; Oliva, G.; Andricopulo, A.D. Molecular docking and structure-based drug design strategies. Molecules 2015, 20, 13384–13421. [Google Scholar] [CrossRef]
  7. Ramsay, R.R.; Popovic-Nikolic, M.R.; Nikolic, K.; Uliassi, E.; Bolognesi, M.L. A perspective on multi-target drug discovery and design for complex diseases. Clin. Transl. Med. 2018, 7, 3–16. [Google Scholar] [CrossRef] [Green Version]
  8. Vázquez, J.; López, M.; Gibert, E.; Herrero, E.; Luque, F.J. Merging Ligand-Based and Structure-Based Methods in Drug Discovery: An Overview of Combined Virtual Screening Approaches. Molecules 2020, 25, 4723. [Google Scholar] [CrossRef]
  9. Stepanchikova, A.V.; Lagunin, A.; Filimonov, D.A.; Poroikov, V. Prediction of Biological Activity Spectra for Substances: Evaluation on the Diverse Sets of Drug-Like Structures. Curr. Med. Chem. 2003, 10, 225–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Filimonov, D.A.; Rudik, A.V.; Dmitriev, A.V.; Poroikov, V.V. Computer-Aided Estimation of Biological Activity Profiles of Drug-Like Compounds Taking into Account Their Metabolism in Human Body. Int. J. Mol. Sci. 2020, 21, 7492. [Google Scholar] [CrossRef]
  11. Lagunin, A.; Filimonov, D.; Poroikov, V. Multi-targeted natural products evaluation based on biological activity prediction with PASS. Curr. Pharm. Des. 2010, 16, 1703–1717. [Google Scholar] [CrossRef] [Green Version]
  12. Zheng, X.-J.; Li, C.-S.; Cui, M.-Y.; Song, Z.-W.; Bai, X.-Q.; Liang, C.-W.; Wang, H.-Y.; Zhang, T.-Y. Synthesis, biological evaluation of benzothiazole derivatives bearing a 1,3,4-oxadiazole moiety as potential anti-oxidant and anti-inflammatory agents. Bioorg. Med. Chem. Lett. 2020, 30, 127237–127244. [Google Scholar] [CrossRef]
  13. Kumar, G.; Singh, N.P. Synthesis, anti-inflammatory and analgesic evaluation of thiazole/oxazole substituted benzothiazole derivatives. Bioorg. Chem. 2021, 107, 104608–104620. [Google Scholar] [CrossRef]
  14. Ugwu, D.I.; Okoro, U.C.; Ukoha, P.O.; Gupta, A.; Okafor, S.N. Novel anti-inflammatory and analgesic agents: Synthesis, molecular docking and in vivo studies. J. Enzym. Inhib. Med. Chem. 2018, 33, 405–415. [Google Scholar] [CrossRef] [Green Version]
  15. Morsy, M.A.; Ali, E.M.; Kandeel, M.; Venugopala, K.N.; Nair, A.B.; Greish, K.; El-Daly, M. Screening and Molecular Docking of Novel Benzothiazole Derivatives as Potential Antimicrobial Agents. Antibiotics 2020, 9, 221. [Google Scholar] [CrossRef]
  16. Nishad, R.K.; Shukla, K.S.; Chawla, P.A. Design and Synthesis of 2-Substituted Benzothiazole Derivatives as Antioxidant and Antimicrobial Agents. Curr. Bioact. Comp. 2020, 16, 7. [Google Scholar] [CrossRef]
  17. Kaushik, C.P.; Chahal, M. Synthesis and antibacterial activity of benzothiazole and benzoxazole-appended substituted 1,2,3-triazoles. J. Chem. Sci. 2020, 132, 142–151. [Google Scholar] [CrossRef]
  18. Haroun, M.; Tratrat, C.; Kositsi, K.; Tsolaki, E.; Petrou, A.; Al-Dhubiab, B.; Attimarad, M.; Harsha, S.; Elsewedy, H.; Gavalas, A.; et al. New benzothiazole-based thiazolidinones as potent dual anti-inflammatory /antimicrobial agents. Design, synthesis and biological evaluation. Curr. Top. Med. Chem. 2018, 18, 75–87. [Google Scholar] [CrossRef] [PubMed]
  19. Padalka, V.S.; Borse, B.N.; Gupta, V.D.; Phatangare, K.R.; Patil, V.S.; Umape, P.G.; Sekar, N.N. Synthesis and antimicrobial activity of novel 2-substituted benzimidazole, benzoxazole and benzothiazole derivatives. Arab. J. Chem. 2016, 9, S1125–S1130. [Google Scholar] [CrossRef] [Green Version]
  20. Gulati, S.; Wakode, S.; Kaur, A.; Anand, K. Synthesis, biological activity and recent advancement of benzothiazoles: A classical review. WJPPS 2017, 6, 1842–1869. [Google Scholar]
  21. Uremis, N.; Uremis, M.M.; Tolun, F.I.; Ceylan, M.; Doganer, A.; Kurt, A.H. Synthesis of 2-Substituted Benzothiazole Derivatives and Their In Vitro Anticancer Effects and Antioxidant Activities Against Pancreatic Cancer Cells. Anticancer Res. 2017, 37, 6381–6389. [Google Scholar] [PubMed]
  22. Irfan, A.; Batool, F.; Zahra Naqvi, S.A.; Islam, A.; Osman, S.M.; Nocentini, A.; Alissa, S.A.; Supuran, C.T. Benzothiazole derivatives as anticancer agents. J. Enzym. Inhib. Med. Chem. 2020, 35, 265–279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Osmaniye, D.; Levent, S.; Karaduman, A.B.; Ilgın, S.; Özkay, Y.; Kaplancıklı, I.Z. Synthesis of New Benzothiazole Acylhydrazonesas Anticancer Agents. Molecules 2018, 23, 1054. [Google Scholar] [CrossRef] [Green Version]
  24. Asiri, Y.I.; Alsayari, A.; Muhsinah, A.B.; Mabkhot, Y.N.; Hassan, M.Z. Benzothiazoles as potential antiviral agents. J. Pharm. Pharmacol. 2020, 72, 1459–1480. [Google Scholar] [CrossRef] [PubMed]
  25. Azzam, R.A.; Elboshi, H.A.; Elgemeie, G.H. Novel Synthesis and Antiviral Evaluation of New Benzothiazole-Bearing N-Sulfonamide 2-Pyridone Derivatives as USP7 Enzyme Inhibitors. ACS Omega 2020, 5, 30023–30036. [Google Scholar] [CrossRef]
  26. Al-Masoudi, N.A.; Jafar, N.A.; Abbas, L.J.; Baqir, S.J.; Pannecouque, C. Synthesis and anti-HIV Activity of New Benzimidazole, Benzothiazole and Carbohyrazide Derivatives of the anti-Inflammatory Drug Indomethacin. Z. Naturforsch. 2011, 66b, 953–960. [Google Scholar] [CrossRef]
  27. Sumit Arvind, K.; Arun Kumar, M. Advancement in Pharmacological Activities of Benzothiazole and its Derivatives: An Up to Date Review. Mini Rev. Med. Chem. 2021, 21, 314–335. [Google Scholar] [CrossRef]
  28. Haroun, M. Novel Hybrids of Pyrazolidinedione and Benzothiazole as TZD Analogues. Rationale Design, Synthesis and In Vivo Anti-Diabetic Evaluation. Med. Chem. 2019, 15, 624–633. [Google Scholar] [CrossRef]
  29. Gollapalli, M.; Taha, M.; Javid, M.T.; Almandil, N.B.; Rahim, F.; Wadood, A.; Mosaddik, A.; Ibrahim, M.; Alqahtani, M.A.; Bamarouf, Y.A. Synthesis of benzothiazole derivatives as a potent α-glucosidase inhibitor. Bioorg. Chem. 2019, 85, 33–48. [Google Scholar] [CrossRef]
  30. Kumar, S.; Mittal, A.; Pathak, A.; Sahu, S.K. Biological assessments of substituted benzothiazole derivatives in streptozocin induced diabetes rats. Plant. Arch. 2020, 20, 3250–3253. [Google Scholar]
  31. Cabrera-Péreza, L.C.; Padilla-Martínez, I.I.; Cruzb, A.; Mendieta-Wejebea, J.E.; Tamay-Cach, F.; Rosales-Hernández, M.C. Evaluation of a new benzothiazole derivative with antioxidant activity in the initial phase of acetaminophen toxicity. Arab. J. Chem. 2019, 12, 3871–3882. [Google Scholar] [CrossRef]
  32. Küçükbay, F.Z.; Buğday, N.; Küçükbay, H.; Tanc, M.; Supuran, C.T. Synthesis, characterization and carbonic anhydrase inhibitory activity of novel benzothiazole derivatives. J. Enzym. Inhib. Med. Chem. 2016, 31, 1221–1225. [Google Scholar] [CrossRef]
  33. Güzel-Akdemir, Ö.; Carradori, S.; Grande, R.; Demir-Yazıcı, K.; Angeli, A.; Supuran, C.T.; Akdemir, A. Development of Thiazolidinones as Fungal Carbonic Anhydrase Inhibitors. Int. J. Mol. Sci. 2020, 21, 2960. [Google Scholar] [CrossRef] [Green Version]
  34. Gagoria, J.; Verma, P.K.; Khatkar, A. Anticonvulsant and neurological profile of benzothiazoles: A mini-review. Cent. Nerv. Syst. Agents Med. Chem. 2015, 15, 11–60. [Google Scholar] [CrossRef]
  35. Liu, D.C.; Zhang, H.; Jin, C.-M.; Quan, Z.-C. Synthesis and biological evaluation of novel benzothiazole derivatives as potential anticonvulsant agents. Molecules 2016, 21, 164. [Google Scholar] [CrossRef] [Green Version]
  36. Ballari, M.S.; Herrera Cano, N.; Wunderlin, D.A.; Feresin, G.E.; Santiago, A.N. One-pot sequential synthesis and antifungal activity of 2-(benzylsulfonyl)benzothiazole derivatives. RSC Adv. 2019, 9, 29405–29413. [Google Scholar] [CrossRef] [Green Version]
  37. Zhao, S.; Zhao, L.; Zhang, X.; Liu, C.; Hao, C.; Xie, H.; Sun, B.; Zhao, D.; Cheng, M. Design, synthesis, and structure-activity relationship studies of benzothiazole derivatives as antifungal agents. Eur. J. Med. Chem. 2016, 123, 514–522. [Google Scholar] [CrossRef]
  38. Hout, S.; Azas, N.; Darque, A.; Robin, M.; Di Giorgio, C.; Gasquet, M.; Galy, J.; Timon-David, P. Activity of benzothiazoles and chemical derivatives on Plasmodium falciparum. Parasitology 2004, 129, 525–542. [Google Scholar] [CrossRef]
  39. Sharma, P.S.; Sinhmar, A.; Sharma, A.; Rajak, H.; Patnak, D.P. Medicinal significance of benzothiazole scaffold: Insight view. J. Enzym. Inhib. Med. Chem. 2013, 28, 240–266. [Google Scholar] [CrossRef] [PubMed]
  40. Agrawal, N. Synthetic and therapeutic potential of 4-thiazolidinone and its analogs. Curr. Chem. Lett. 2021, 10, 119–138. [Google Scholar] [CrossRef]
  41. Dincel, E.D.; Ulusoy-Güzeldemirci, N.; Şatana, D.; Küçükbasmacı, Ö. Design, synthesis, characterization and antimicrobial evaluation of some novel hydrazinecarbothioamide, 4-thiazolidinone and 1,2,4-triazole-3-thione derivatives. J. Heterocycl. Chem. 2021, 58, 195–205. [Google Scholar] [CrossRef]
  42. Desai, N.; Jadeja, K.; Jadeja, D.; Khedkar, V.; Jha, P. Design, synthesis, antimicrobial evaluation, and molecular docking study of some 4-thiazolidinone derivatives containing pyridine and quinazoline moiety. Synth. Commun. 2020, 51, 1–12. [Google Scholar] [CrossRef]
  43. Zhang, H.; Zhang, J.; Qu, W.; Xie, S.; Huang, L.; Chen, D.; Tao, Y.; Liu, Z.; Pan, Y.; Yuan, Z. Design, Synthesis, and Biological Evaluation of Novel Thiazolidinone-Containing Quinoxaline-1,4-di-N-oxides as Antimycobacterial and Antifungal Agents. Front. Chem. 2020, 8, 598–617. [Google Scholar] [CrossRef] [PubMed]
  44. Da Rosa Monte Machado, G.; Diedrich, D.; Ruaro, T.C.; Zimmer, A.R.; Lettieri Teixeira, M.; de Oliveira, L.F.; Jean, M.; Van de Weghe, P.; de Andrade, S.F.; Baggio Gnoatto, S.C.; et al. Quinolines derivatives as promising new antifungal candidates for the treatment of candidiasis and dermatophytosis. Braz. J. Microbiol. 2020, 51, 1691–1701. [Google Scholar] [CrossRef]
  45. Fesatidou, M.; Zagaliotis, P.; Camoutsis, C.; Petrou, A.; Eleftheriou, P.; Tratrat, C.; Haroun, M.; Geronikaki, A.; Ciric, A.; Sokovic, M. 5-Adamantan thiadiazole-based thiazolidinones as antimicrobial agents. Design, synthesis, molecular docking and evaluation. Bioorg. Med. Chem. 2018, 26, 4664–4676. [Google Scholar] [CrossRef] [PubMed]
  46. Haroun, M.; Tratrat, C.; Kolokotroni, A.; Petrou, A.; Geronikaki, A.; Ivanov, M.; Kostic, M.; Sokovic, M.; Carazo, A.; Mladěnka, P.; et al. 5-Benzyliden-2-(5-methylthiazol-2-ylimino)thiazolidin-4-ones as Antimicrobial Agents. Design, Synthesis, Biological Evaluation and Molecular Docking Studies. Antibiotics 2021, 10, 309. [Google Scholar] [CrossRef] [PubMed]
  47. Haroun, M.; Tratrat, C.; Petrou, A.; Geronikaki, A.; Ivanov, M.; Ciric, A.; Sokovic, M. 2-Aryl-3-(6-trifluoromethoxy)benzo[d]thiazole-based thiazolidinone hybrids as potential anti-infective agents: Synthesis, biological evaluation and molecular docking studies. Bioorg. Med. Chem. Lett. 2021, 32, 127718–127725. [Google Scholar] [CrossRef]
  48. ToxPredict. Available online: https://apps.ideaconsult.net/ToxPredict (accessed on 11 May 2018).
  49. Prediction of TOXicity of Chemicals. Available online: http://tox.charite.de/tox (accessed on 11 May 2018).
  50. Miyagawa, M. Globally harmonized system of classification and labelling of chemicals (GHS) and its implementation in Japan. Nihon Eiseigaku Zasshi 2010, 65, 5–13. [Google Scholar] [CrossRef]
  51. Filimonov, D.A.; Lagunin, A.A.; Gloriozova, T.A.; Rudik, A.V.; Druzhilovskii, D.S.; Pogodin, P.V.; Poroikov, V.V. Prediction of the Biological Activity Spectra of Organic Compounds Using the Pass Online Web Resource. Chem. Heterocycl. Compd. 2014, 50, 444–457. [Google Scholar] [CrossRef]
  52. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug. Deliv. Rev. 2001, 46, 3–26. [Google Scholar] [CrossRef]
  53. Ghose, A.K.; Viswanadhan, V.N.; Wendoloski, J.J. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J. Comb. Chem. 1999, 1, 55–68. [Google Scholar] [CrossRef]
  54. Veber, D.F.; Johnson, S.R.; Cheng, H.Y.; Smith, B.R.; Wars, K.W.; Kopple, K.D. Molecular Properties That Influence the Oral Bioavailability of Drug Candidates. J. Med. Chem. 2002, 45, 2615–2623. [Google Scholar] [CrossRef]
  55. Egan, W.J.; Merz, K.M.; Baldwin, J.J. Prediction of Drug Absorption Using Multivariate Statistics. J. Med. Chem. 2000, 43, 3867–3877. [Google Scholar] [CrossRef] [PubMed]
  56. Muegge, I.; Heald, S.L.; Brittelli, D. Simple selection criteria for drug-like chemical matter. J. Med. Chem. 2001, 44, 1841–1846. [Google Scholar] [CrossRef]
  57. Wang, R.X.; Fu, Y.; Lai, L.H. A New Atom-Additive Method for Calculating Partition Coefficients. J. Chem. Inf. Comput. Sci. 1997, 37, 615–621. [Google Scholar] [CrossRef]
  58. Ertl, P.; Rohde, B.; Selzer, P. Fast calculation of molecular polar surface area as a sum of fragment-based contributions and its application to the prediction of drug transport properties. J. Med. Chem. 2000, 43, 3714–3717. [Google Scholar] [CrossRef]
  59. Bakht, M.A.; Yar, M.S.; Abdel-Hamid, S.G.; Al Qasoumi, S.I.; Samad, A. Molecular properties prediction, synthesis and antimicrobial activity of some newer oxadiazole derivatives. Eur. J. Med. Chem. 2010, 45, 5862–5869. [Google Scholar] [CrossRef]
  60. Templin, M.F.; Ursinus, A.; Höltje, J.V. A defect in cell wall recycling triggers autolysis during the stationary growth phase of Escherichia coli. EMBO J. 1999, 18, 4108–4117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Korza, H.J.; Bochtler, M. Pseudomonas aeruginosa LD-carboxypeptidase, a serine peptidase with a Ser-His-Glu triad and a nucleophilic elbow. J. Biol. Chem. 2005, 280, 40802–40812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Kartsev, V.; Lichitsky, B.; Geronikaki, A.; Petrou, A.; Smiljkovic, M.; Kostic, M.; Radanovic, O.; Soković, M. Design, synthesis and antimicrobial activity of usnic acid derivatives. Med. Chem. Commun. 2018, 9, 870–882. [Google Scholar] [CrossRef] [Green Version]
  63. Kostić, M.; Smiljković, M.; Petrović, J.; Glamočilija, J.; Barros, L.; Ferreira, I.C.F.R.; Ćirić, A.; Soković, M. Chemical, nutritive composition and a wide range of bioactive properties of honey mushroom Armillariamellea (Vahl: Fr.) Kummer. Food Funct. 2017, 8, 3239–3249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kritsi, E.; Matsoukas, M.T.; Potamitis, C.; Detsi, A.; Ivanov, M.; Sokovic, M.; Zoumpoulakis, P. Novel Hit Compounds as Putative Antifungals: The Case of Aspergillus fumigatus. Molecules 2019, 24, 3853. [Google Scholar] [CrossRef] [Green Version]
  65. Quinn, P.J.; Markey, B.K.; Leonard, F.C.; FitzPatrick, E.S.; Fanning, S.; Hartigan, P.J. Veterinary Microbiology and Microbial Disease, 2nd ed.; Wiley-Blackwell: Hoboken, NJ, USA, 2011. [Google Scholar]
  66. European Committee on Antimicrobial Susceptibility Testing. Disk Diffusion Method for Antimicrobial Susceptibility Testing-EUCAST Disk Diffusion Method Version 4.0. June 2014. Available online: www.eucast.org (accessed on 24 March 2021).
  67. Cady, N.C.; McKean, K.A.; Behnke, J.; Kubec, R.; Mosier, A.P.; Kasper, S.H.; Burz, D.S.; Musah, R.A. Inhibition of biofilm formation, quorum sensing and infection in Pseudomonas aeruginosa by natural products-inspired organosulfur compounds. PLoS ONE 2012, 7, e38492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Aleksić, M.; Stanisavljević, D.; Smiljković, M.; Vasiljević, P.; Stevanović, M.; Soković, M.; Stojković, D. Pyrimethanil: Between efficient fungicide against Aspergillus rot on cherry tomato and cytotoxic agent on human cell lines. Ann. Appl. Biol. 2019, 175, 228–235. [Google Scholar] [CrossRef]
  69. Horishny, V.; Kartsev, V.; Matiychuk, V.; Geronikaki, A.; Anthi, P.; Pogodin, P.; Poroikov, V.; Ivanov, M.; Kostic, M.; Soković, M.D.; et al. 3-Amino-5-(indol-3-yl)methylene-4-oxo-2-thioxothiazolidine Derivatives as Antimicrobial Agents: Synthesis, Computational and Biological Evaluation. Pharmaceuticals 2020, 13, 229. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Drugs approved by FDA.
Figure 1. Drugs approved by FDA.
Molecules 26 04061 g001
Figure 2. The structural modifications.
Figure 2. The structural modifications.
Molecules 26 04061 g002
Scheme 1. Synthesis of the title compounds.
Scheme 1. Synthesis of the title compounds.
Molecules 26 04061 sch001
Figure 3. Docked conformations of the two most active compounds: A-16 and B-18 in LdcA enzyme and 2D diagrams of ligand–enzyme interactions. Green dotted lines represent hydrogen bond.
Figure 3. Docked conformations of the two most active compounds: A-16 and B-18 in LdcA enzyme and 2D diagrams of ligand–enzyme interactions. Green dotted lines represent hydrogen bond.
Molecules 26 04061 g003
Table 1. Predicted toxicity with program PROTOX.
Table 1. Predicted toxicity with program PROTOX.
No.Predicted LD50Predicted Toxicity ClassHepatotoxicityCarcinogenicityImmunotoxicityMutagenicityCytotoxicity
1500 mg/kg4Inactive 0.54Inactive 0.57Inactive 0.99Inactive 0.67Inactive 0.76
21000 mg/kg4Inactive 0.52Inactive 0.63Inactive 0.97Inactive 0.66Inactive 0.62
3500 mg/kg4Inactive 0.56Inactive 0.65Inactive 0.99Inactive 0.67Inactive 0.78
4500 mg/kg4Inactive 0.54Inactive 0.57Inactive 0.99Inactive 0.67Inactive 0.76
5500 mg/kg4Inactive 0.58Inactive 0.56Inactive 0.99Inactive 0.66Inactive 0.77
6500 mg/kg4Inactive 0.50Inactive 0.55Inactive 0.94Inactive 0.64Inactive 0.75
7500 mg/kg4Inactive 0.52Inactive 0.55Inactive 0.79Inactive 0.64Inactive 0.74
8500 mg/kg4Inactive 0.52Inactive 0.56Inactive 0.96Inactive 0.63Inactive 0.75
9500 mg/kg4Inactive 0.54Inactive 0.56Inactive 0.94Inactive 0.63Inactive 0.77
10500 mg/kg4Inactive 0.50Inactive 0.62Inactive 0.98Inactive 0.61Inactive 0.63
11500 mg/kg4Inactive 0.52Inactive 0.64Inactive 0.99Inactive 0.60Inactive 0.63
12500 mg/kg4Inactive 0.59Inactive 0.60Inactive 0.98Inactive 0.66Inactive 0.66
131000 mg/kg4Inactive 0.52Inactive 0.65Inactive 0.99Inactive 0.66Inactive 0.74
14500 mg/kg4Inactive 0.56Inactive 0.60Inactive 0.98Inactive 0.67Inactive 0.65
15500 mg/kg4Inactive 0.57Inactive 0.55Inactive 0.99Inactive 0.67Inactive 0.81
16500 mg/kg4Inactive 0.60Inactive 0.58Inactive 0.99Inactive 0.68Inactive 0.68
17500 mg/kg4Inactive 0.57Inactive 0.55Inactive 0.99Inactive 0.67Inactive 0.81
181000 mg/kg4Inactive 0.52Inactive 0.65Inactive 0.99Active 0.91Inactive 0.74
19500 mg/kg4Inactive 0.54Inactive 0.55Inactive 0.99Inactive 0.68Inactive 0.82
Table 2. Prediction of carcinogenicity and mutagenesis of compounds by ToxPredict application.
Table 2. Prediction of carcinogenicity and mutagenesis of compounds by ToxPredict application.
NCarcinogenicity (Rodents (Multiple Species/Sites))Carcinogenicity (Rat)Carcinogenicity (Mouse)Mutagenicity (Salmonella typhimurium)
1Inactive Conf.: 0.026Inactive Conf.: 0.026Inactive Conf.: 0.026Inactive Conf.: 0.026
2Inactive Conf.: 0.027Inactive Conf.: 0.027Inactive Conf.: 0.026Inactive Conf.: 0.027
3Inactive Conf.: 0.024Inactive Conf.: 0.027Inactive Conf.: 0.033Inactive Conf.: 0.036
4Inactive Conf.: 0.031Inactive Conf.: 0.028Inactive Conf.: 0.041Inactive Conf.: 0.028
5Inactive Conf.: 0.028Inactive Conf.: 0.028Inactive Conf.: 0.041Inactive Conf.: 0.026
6Inactive Conf.: 0.026Inactive Conf.: 0.027Inactive Conf.: 0.029Inactive Conf.: 0.026
7Inactive Conf.: 0.031Inactive Conf.: 0.027Inactive Conf.: 0.031Inactive Conf.: 0.028
8Inactive Conf.: 0.025Inactive Conf.: 0.028Inactive Conf.: 0.027Inactive Conf.: 0.028
9Inactive Conf.: 0.028Inactive Conf.: 0.026Inactive Conf.: 0.049Inactive Conf.: 0.041
10Inactive Conf.: 0.033Inactive Conf.: 0.028Inactive Conf.: 0.041Inactive Conf.: 0.031
11Inactive Conf.: 0.032Inactive Conf.: 0.026Inactive Conf.: 0.041Inactive Conf.: 0.033
12Inactive Conf.: 0.028Inactive Conf.: 0.029Inactive Conf.: 0.028Inactive Conf.: 0.028
13Inactive Conf.: 0.031Inactive Conf.: 0.028Inactive Conf.: 0.041Inactive Conf.: 0.039
14Inactive Conf.: 0.034Inactive Conf.: 0.028Inactive Conf.: 0.041Inactive Conf.: 0.038
15Inactive Conf.: 0.026Inactive Conf.: 0.026Inactive Conf.: 0.026Inactive Conf.: 0.028
16Inactive Conf.: 0.026Inactive Conf.: 0.026Inactive Conf.: 0.026Inactive Conf.: 0.026
17Inactive Conf.: 0.028Inactive Conf.: 0.028Inactive Conf.: 0.029Inactive Conf.: 0.027
18Inactive Conf.: 0.026Inactive Conf.: 0.027Inactive Conf.: 0.033Inactive Conf.: 0.035
19Inactive Conf.: 0.033Inactive Conf.: 0.027Inactive Conf.: 0.041Inactive Conf.: 0.027
Table 3. Antibacterial activity of compounds (MIC and MBC in mg/mL).
Table 3. Antibacterial activity of compounds (MIC and MBC in mg/mL).
CompoundsS.a.MRSAL.m.P.a.P.a. ResE. coliE. coli ResS. ty.
1MIC0.30 ± 0.070.50 ± 0.000.50 ± 0.000.25 ± 0.000.25 ± 0.110.12 ± 0.000.25 ± 0.110.50 ± 0.00
MBC0.50 ± 0.001.00 ± 0.000.83 ± 0.110.58 ± 0.110.50 ± 0.000.25 ± 0.000.50 ± 0.000.75 ± 0.05
2MIC0.12 ± 0.000.25 ± 0.000.25 ± 0.000.30 ± 0.070.25 ± 0.040.12 ± 0.000.25 ± 0.000.25 ± 0.04
MBC0.25 ± 0.000.50 ± 0.000.75 ± 0.000.50 ± 0.000.50 ± 0.000.25 ± 0.000.50 ± 0.000.50 ± 0.00
3MIC0.50 ± 0.000.30 ± 0.070.50 ± 0.000.25 ± 0.040.20 ± 0.010.20 ± 0.000.25 ± 0.000.30 ± 0.07
MBC1.00 ± 0.000.50 ± 0.001.00 ± 0.000.50 ± 0.000.25 ± 0.040.25 ± 0.040.50 ± 0.000.50 ± 0.00
4MIC0.25 ± 0.040.25 ± 0.000.25 ± 0.000.25 ± 0.000.20 ± 0.010.10 ± 0.000.25 ± 0.040.25 ± 0.00
MBC0.50 ± 0.000.50 ± 0.000.50 ± 0.000.50 ± 0.000.25 ± 0.040.12 ± 0.000.50 ± 0.000.83 ± 0.11
5MIC0.25 ± 0.000.30 ± 0.070.50 ± 0.000.25 ± 0.040.12 ± 0.000.12 ± 0.000.25 ± 0.000.50 ± 0.00
MBC0.50 ± 0.000.50 ± 0.001.00 ± 0.000.50 ± 0.000.25 ± 0.000.25 ± 0.000.50 ± 0.001.00 ± 0.00
6MIC0.12 ± 0.000.25 ± 0.000.25 ± 0.000.25 ± 0.040.20 ± 0.000.25 ± 0.000.25 ± 0.000.50 ± 0.00
MBC0.25 ± 0.040.50 ± 0.000.50 ± 0.000.50 ± 0.000.25 ± 0.040.50 ± 0.000.50 ± 0.000.83 ± 0.11
7MIC0.20 ± 0.000.50 ± 0.000.75 ± 0.000.25 ± 0.040.25 ± 0.000.20 ± 0.000.25 ± 0.000.50 ± 0.00
MBC0.25 ± 0.041.00 ± 0.001.00 ± 0.000.58 ± 0.110.50 ± 0.000.25 ± 0.040.50 ± 0.000.83 ± 0.11
8MIC0.20 ± 0.000.50 ± 0.000.30 ± 0.070.25 ± 0.000.06 ± 0.000.20 ± 0.000.25 ± 0.000.30 ± 0.07
MBC0.25 ± 0.041.00 ± 0.000.50 ± 0.000.50 ± 0.000.12 ± 0.000.25 ± 0.0040.50 ± 0.000.50 ± 0.00
9MIC0.12 ± 0.000.58 ± 0.110.50 ± 0.000.25 ± 0.040.30 ± 0.070.20 ± 0.000.25 ± 0.000.50 ± 0.00
MBC0.25 ± 0.041.00 ± 0.001.00 ± 0.000.50 ± 0.000.50 ± 0.000.25 ± 0.040.50 ± 0.001.00 ± 0.00
StreptomycinMIC0.10 ± 0.000.10 ± 0.000.15 ± 0.000.10 ± 0.000.05 ± 0.000.10 ± 0.000.10 ± 0.000.10 ± 0.00
MBC0.20 ± 0.01 0.30 ± 0.010.20 ± 0.010.10 ± 0.000.20 ± 0.000.20 ± 0.010.20 ± 0.01
AmpicillinMIC0.10 ± 0.00 0.15 ± 0.000.30 ± 0.010.2 ± 0.010.15 ± 0.000.20 ± 0.010.10 ± 0.00
MBC0.15 ± 0.00 0.30 ± 0.020.50 ± 0,01 0.20 ± 0.01 0.20 ± 0.00
MIC—minimal inhibitory concentration, MBC—minimal bactericidal concentration, S.a.—S. aurues (ATCC 6538), l.m.—L. monocytogenes (NCTC 7973), P.a.—P. aeruginosa (ATCC 27853), S. ty.—S. typhimirium (ATCC 13311), MRSA—methicillin resistant S. aureus, (IBRS MRSA 011), E. coli res—resistant E. coli (IBRS E003), P.a. res—resistant P. aeruginosa (IBRS P001).
Table 4. Antibacterial activity of compounds 1019 (MIC and MBC in mg/mL).
Table 4. Antibacterial activity of compounds 1019 (MIC and MBC in mg/mL).
CompoundsS.a.MRSAL.m.P.a.P.a. ResE. coliE. coli ResS. ty.
10MIC0.50 ± 0.000.75 ± 0.000.75 ± 0.020.20 ± 0.000.12 ± 0.000.12 ± 0.000.50 ± 0.030.50 ± 0.02
MBC1.00 ± 0.081.00 ± 0.00>1.000.25 ± 0.000.25 ± 0.000.25 ± 0.001.00 ± 0.041.00 ± 0.04
11MIC0.50 ± 0.020.75 ± 0.030.50 ± 0.020.25 ± 0.000.15 ± 0.000.25 ± 0.000.50 ± 0.030.50 ± 0.02
MBC0.75 ± 0.041.00 ± 0.061.00 ± 0.060.50 ± 0.030.25 ± 0.010.50 ± 0.030.75 ± 0.021.00 ± 0.04
12MIC0.50 ± 0.020.75 ± 0.030.50 ± 0.030.25 ± 0.010.25 ± 0.000.25 ± 0.010.50 ± 0.030.50 ± 0.01
MBC1.00 ± 0.06>1.00>1.000.50 ± 0.030.50 ± 0.020.50 ± 0.021.00 ± 0.081.00 ± 0.06
13MIC0.50 ± 0.020.25 ± 0.000.75 ± 0.040.30 ± 0.010.15 ± 0.000.12 ± 0.000.25 ± 0.000.50 ± 0.03
MBC0.75 ± 0.040.50 ± 0.001.00 ± 0.080.50 ± 0.020.25 ± 0.010.25 ± 0.010.50 ± 0.031.00 ± 0.04
14MIC0.25 ± 0.010.75 ± 0.030.50 ± 0.020.25 ± 0.010.15 ± 0.000.12 ± 0.000.25 ± 0.000.12 ± 0.00
MBC0.50 ± 0.021.00 ± 0.061.00 ± 0.080.50 ± 0.030.25 ± 0.010.25 ± 0.010.50 ± 0.030.50 ± 0.02
15MIC0.30 ± 0.010.50 ± 0.030.25 ± 0.010.12 ± 0.000.12 ± 0.000.25 ± 0.010.25 ± 0.000.50 ± 0.03
MBC0.50 ± 0.030.75 ± 0.000.50 ± 0.030.25 ± 0.000.25 ± 0.000.50 ± 0.020.50 ± 0.000.75 ± 0.03
16MIC0.10 ± 0.000.10 ± 0.000.06 ± 0.000.30 ± 0.000.25 ± 0.010.30 ± 0.010.12 ± 0.000.10 ± 0.00
MBC0.12 ± 0.000.12 ± 0.000.12 ± 0.000.60 ± 0.000.50 ± 0.030.60 ± 0.030.25 ± 0.000.12 ± 0.00
17MIC0.50 ± 0.020.50 ± 0.020.25 ± 0.000.12 ± 0.000.10 ± 0.000.25 ± 0.010.25 ± 0.000.30 ± 0.00
MBC0.75 ± 0.030.75 ± 0.040.50 ± 0.020.25 ± 0.010.12 ± 0.000.50 ± 0.020.50 ± 0.030.50 ± 0.03
18MIC0.20 ± 0.000.20 ± 0.000.12 ± 0.000.10 ± 0.000.06 ± 0.000.12 ± 0.000.25 ± 0.010.25 ± 0.00
MBC0.25 ± 0.010.25 ± 0.000.20 ± 0.000.12 ± 0.000.12 ± 0.000.20 ± 0.010.50 ± 0.020.50 ± 0.02
19MIC0.25 ± 0.010.25 ± 0.000.12 ± 0.000.12 ± 0.000.10 ± 0.000.12 ± 0.000.12 ± 0.000.25 ± 0.00
MBC0.50 ± 0.020.50 ± 0.020.25 ± 0.000.25 ± 0.000.12 ± 0.000.25 ± 0.010.25 ± 0.000.50 ± 0.02
StreptomycinMIC0.10 ± 0.000.10 ± 0.000.15 ± 0.000.10 ± 0.000.05 ± 0.000.10 ± 0.000.10 ± 0.000.10 ± 0.00
MBC0.20 ± 0.01 0.30 ± 0.010.20 ± 0.010.10 ± 0.000.20 ± 0.000.20 ± 0.010.20 ± 0.01
AmpicillinMIC0.10 ± 0.00 0.15 ± 0.000.30 ± 0.010.2 ± 0.010.15 ± 0.000.20 ± 0.010.10 ± 0.00
MBC0.15 ± 0.00 0.30 ± 0.020.50 ± 0,01 0.20 ± 0.01 0.20 ± 0.00
MIC—minimal inhibitory concentration, MBC—minimal bactericidal concentration, S.a.—S. aurues (ATCC 6538), L.m.—L. monocytogenes (NCTC 7973), P.a.—P. aeruginosa (ATCC 27853), S. ty.—S. typhimurium (ATCC 13311), MRSA—Methicillin resistant S. aureus (IBRS MRSA 011), E. coli res—resistant E. coli (IBRS E003), P.a. res—P. aeruginosa (IBRS P001).
Table 5. Inhibition of P. aeruginosa biofilm formation (%).
Table 5. Inhibition of P. aeruginosa biofilm formation (%).
CompoundMIC0.5 MIC
1649.46 ± 5.6735.55 ± 2.30
18NENE
1951.46 ± 8.7641.76 ± 9.99
8NENE
Ampicillin70.00 ± 10.2352.36 ± 3.67
Streptomycin63.56 ± 8.2829.12 ± 1.22
NE—no effect.
Table 6. Antifungal activity of tested compounds (MIC and MBC in mg/mL).
Table 6. Antifungal activity of tested compounds (MIC and MBC in mg/mL).
CompoundsA.f.A.n.A.v.P.f.T.v.P.v.c.
1MIC>1.000.50 ± 0.000.50 ± 0.000.50 ± 0.020.50 ± 0.000.50 ± 0.02
MFC>1.001.00 ± 0.001.00 ± 0.001.00 ± 0.001.00 ± 0.001.00 ± 0.00
2MIC>1.000.50 ± 0.001.00 ± 0.000.50 ± 0.000.50 ± 0.001.00 ± 0.00
MFC>1.001.00 ± 0.00>1.001.00 ± 0.001.00 ± 0.00>1.00
3MIC>1.001.00 ± 0.001.00 ± 0.000.50 ± 0.000.50 ± 0.020.50 ± 0.00
MFC>1.00>1.00>1.001.00 ± 0.001.00 ± 0.001.00 ± 0.00
4MIC1.00 ± 0.000.50 ± 0.020.50 ± 0.020.50 ± 0.021.00 ± 0.00>1.00
MFC>1.001.00 ± 0.001.00 ± 0.001.00 ± 0.00>1.00>1.00
5MIC>1.00>1.00>1.00>1.00>1.00>1.00
MFC>1.00>1.00>1.00>1.00>1.00>1.00
6MIC>1.00>1.000.25 ± 0.000.25 ± 0.040.25 ± 0.040.25 ± 0.04
MFC>1.00>1.000.50 ± 0.020.50 ± 0.020.50 ± 0.020.50 ± 0.02
7MIC0.50 ± 0.001.00 ± 0.000.25 ± 0.040.50 ± 0.000.50 ± 0.000.50 ± 0.00
MFC1.00 ± 0.00>1.000.50 ± 0.001.00 ± 0.001.00 ± 0.001.00 ± 0.00
8MIC>1.00>1.000.50 ± 0.000.50 ± 0.000.50 ± 0.000.50 ± 0.00
MFC>1.00>1.001.00 ± 0.001.00 ± 0.001.00 ± 0.001.00 ± 0.00
9MIC0.50 ± 0.021.00 ± 0.000.25 ± 0.000.50 ± 0.000.25 ± 0.000.50 ± 0.00
MFC1.00 ± 0.00>1.000.50 ± 0.001.00 ± 0.000.50 ± 0.000.83 ± 0.11
10MIC0.50 ± 0.021.00 ± 0.000.25 ± 0.001.00 ± 0.000.50 ± 0.020.50 ± 0.02
MBC1.00 ± 0.00>1.000.50 ± 0.03>1.001.00 ± 0.001.00 ± 0.00
11MIC1.00 ± 0.00>1.000.50 ± 0.010.50 ± 0.030.25 ± 0.000.50 ± 0.00
MBC>1.00>1.001.00 ± 0.001.00 ± 0.000.50 ± 0.021.00 ± 0.00
12MIC0.50 ± 0.021.00 ± 0.000.50 ± 0.021.00 ± 0.000.50 ± 0.020.50 ± 0.02
MBC1.00 ± 0.00>1.001.00 ± 0.00>1.001.00 ± 0.001.00 ± 0.00
13MIC1.00 ± 0.001.00 ± 0.000.50 ± 0.020.50 ± 0.020.50 ± 0.030.50 ± 0.03
MBC>1.00>1.001.00 ± 0.001.00 ± 0.001.00 ± 0.001.00 ± 0.00
14MIC1.00 ± 0.000.50 ± 0.030.25 ± 0.000.50 ± 0.030.50 ± 0.030.50 ± 0.02
MBC>1.001.00 ± 0.000.50 ± 0.001.00 ± 0.001.00 ± 0.001.00 ± 0.00
15MIC1.00 ± 0.00>1.001.00 ± 0.000.50 ± 0.000.50 ± 0.020.25 ± 0.00
MBC>1.00>1.00>1.001.00 ± 0.001.00 ± 0.000.50 ± 0.02
16MIC0.12 ± 0.001.00 ± 0.000.50 ± 0.000.50 ± 0.020.50 ± 0.021.00 ± 0.00
MBC0.25 ± 0.00>1.001.00 ± 0.001.00 ± 0.001.00 ± 0.00>1.00
17MIC1.00 ± 0.00>1.000.50 ± 0.020.50 ± 0.020.25 ± 0.000.25 ± 0.00
MBC>1.00>1.001.00 ± 0.001.00 ± 0.000.50 ± 0.020.50 ± 0.00
18MIC0.50 ± 0.010.50 ± 0.020.12 ± 0.000.50 ± 0.020.25 ± 0.000.25 ± 0.04
MBC1.00 ± 0.001.00 ± 0.000.25 ± 0.001.00 ± 0.000.50 ± 0.020.50 ± 0.02
19MIC0.50 ± 0.030.50 ± 0.000.25 ± 0.040.25 ± 0.000.25 ± 0.000.12 ± 0.00
MBC1.00 ± 0.001.00 ± 0.000.50 ± 0.020.50 ± 0.020.50 ± 0.020.25 ± 0.01
BifonazoleMIC1.00 ± 0.00>1.001.00 ± 0.000.50 ± 0.000.50 ± 0.020.25 ± 0.00
MFC0.20 ± 0.0000.20 ± 0.0000.20 ± 0.010.25 ± 0.010.20 ± 0.010.20 ± 0.00
KetoconazoleMIC0.20 ± 0.010.20 ± 0.010.20 ± 0.000.20 ± 0.001.00 ± 0.010.20 ± 0.01
MFC0.50 ± 0.030.50 ± 0.030.50 ± 0.020.50 ± 0.031.50 ± 0.020.30 ± 0.01
A.v.—A. versicolor (ATCC 11730), T.v.—T. viride (IAM 5061), A.n.—A. niger (ATCC 6275), P.v.c.—Penicillium verrucosum var. cyclopium (food isolate). P.f.—P. funiculosum (ATCC 36839), A.f.—A. fumigatus (human isolate).
Table 7. Drug likeness predictions and physicochemical-pharmacokinetic/ADME properties of tested compounds.
Table 7. Drug likeness predictions and physicochemical-pharmacokinetic/ADME properties of tested compounds.
No.MWNumber of HBA aNumber of HBD bLog Po/w cLog S dTPSA eAbsorption% fLipinski, Ghose, Veber, Egan, and Muegge ViolationsBioavailability ScoreDrug-Likeness Model Score
1382.42502.09Moderately soluble156.3568.900.55−0.45
2373.4502.7Poorly soluble110.5355.100.55−0.49
3389.85402.79Poorly soluble110.5370.900.55−0.42
4406.31302.87Poorly soluble110.5370.900.55−0.39
5355.41402.62Moderately soluble110.5370.900.55−0.07
6515.52204.3Poorly soluble86.7479.120.170.22
7499.06304.24Poorly soluble86.7479.110.550.2
8529.54204.55Poorly soluble86.7479.120.170.56
9496.63404.34Poorly soluble86.7479.110.550.45
10360.43403.02Moderately soluble95.9776.400.550.28
11387.43502.49Moderately soluble141.7959.100.55−0.17
12376.88303.1Poorly soluble95.9776.400.550.39
13372.46403.45Moderately soluble105.272.700.55−0.06
14358.43412.56Moderately soluble116.2No00.550.13
15364.84303.25Poorly soluble86.7479.100.55−0.08
16391.85402.38Moderately soluble132.5663.300.55−0.13
17381.3203.42Poorly soluble86.7479.100.55−0.08
18376.88303.37Poorly soluble95.9776.400.550.18
19362.85312.73Moderately soluble106.9772.100.550.1
a number of hydrogen bond acceptors; b number of hydrogen bond donors; c lipophilicity; d Water solubility (SILICOS-IT (S = soluble)); e topological polar surface area (Å2); f blood brain barrier permeant.
Table 8. Drug-likeness modes of tested compounds.
Table 8. Drug-likeness modes of tested compounds.
No.Drug-Likeness ModelNo.Drug-Likeness ModelNo.Drug-Likeness Model
1 Molecules 26 04061 i0018 Molecules 26 04061 i00214 Molecules 26 04061 i003
2 Molecules 26 04061 i0049 Molecules 26 04061 i00515 Molecules 26 04061 i006
3 Molecules 26 04061 i00710 Molecules 26 04061 i00816 Molecules 26 04061 i009
4 Molecules 26 04061 i01011 Molecules 26 04061 i01117 Molecules 26 04061 i012
5 Molecules 26 04061 i01312 Molecules 26 04061 i01418 Molecules 26 04061 i015
6 Molecules 26 04061 i01613 Molecules 26 04061 i01719 Molecules 26 04061 i018
7 Molecules 26 04061 i019
Table 9. Molecular docking estimated free energy of binding to antibacterial targets.
Table 9. Molecular docking estimated free energy of binding to antibacterial targets.
No.Est. Binding Energy (kcal/mol)I-H
LdcA
Residues Involved in H-Bond Formation
LdcA
E. coli Gyrase
1KZN
Thymidylate Kinase
4QGG
E. coli Primase
1DDE
E. coli MurB 2Q85E. coli LdcA 1ZRS (R+)E. coli LdcA 1ZRS (S−)
1-5.73(R+)−2.41(S−) −5.12(R+)−5.81−4.63
2−5.01(S−)−1.24(S−)−1.88(R+)−5.96(R+)−7.53−7.232Tyr138, His285
3−4.12(R+)−3.32(R+) −5.23(S−)−5.34−4.10
4−4.51(R+) −4.14(S−)−7.31−6.451Ser115, Tyr224
5−4.96(R+) −1.53(S−)−4.76(R+)−5.52−5.14
6−3.41(R+) −5.66(S−)−7.10−6.802Ser115
7−4.40(R+)−2.33(S−) −4.71(R+)−5.82−5.62
8−4.56(S−) −1.95(S−)−5.92(R+)−7.80−7.531His285
9−5.67(R+) −6.11(R+)−6.01−5.16
10−3.35(S−) −2.58(R+)−4.22(S−)−4.76−4.24
11−2.14(S−)−1.28(S−)−2.10(S−)−3.01(S−)−4.16−3.28
12−3.45(R+) −3.61(S−)−4.50−3.95
13−4.55(R+) −1.42(S−)−3.34(R+)−6.02−5.121Tyr138
14−2.03(R+) −1.22(S−)−6.13−5.101Tyr224
15−5.01(R+)−3.17(R+)−2.99(R+)−4.86(R+)−6.14−6.111Tyr224
16−4.17(S−)−1.09(R+) −6.33(R+)−8.63−7.582Ser115, Tyr224
17−2.92(R+) −4.18(S−)−6.11−5.131Tyr224
18−5.21(S−)−1.78(S−)−2.86(R+)−6.53(R+)−8.52−7.641Ser115
19−4.69(R+)−1.62(S−) −6.71(R+)−8.11−7.953Tyr138, His285, Gly286
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Haroun, M.; Tratrat, C.; Petrou, A.; Geronikaki, A.; Ivanov, M.; Ćirić, A.; Soković, M.; Nagaraja, S.; Venugopala, K.N.; Balachandran Nair, A.; et al. Exploration of the Antimicrobial Effects of Benzothiazolylthiazolidin-4-One and In Silico Mechanistic Investigation. Molecules 2021, 26, 4061. https://doi.org/10.3390/molecules26134061

AMA Style

Haroun M, Tratrat C, Petrou A, Geronikaki A, Ivanov M, Ćirić A, Soković M, Nagaraja S, Venugopala KN, Balachandran Nair A, et al. Exploration of the Antimicrobial Effects of Benzothiazolylthiazolidin-4-One and In Silico Mechanistic Investigation. Molecules. 2021; 26(13):4061. https://doi.org/10.3390/molecules26134061

Chicago/Turabian Style

Haroun, Michelyne, Christophe Tratrat, Anthi Petrou, Athina Geronikaki, Marija Ivanov, Ana Ćirić, Marina Soković, Sreeharsha Nagaraja, Katharigatta Narayanaswamy Venugopala, Anroop Balachandran Nair, and et al. 2021. "Exploration of the Antimicrobial Effects of Benzothiazolylthiazolidin-4-One and In Silico Mechanistic Investigation" Molecules 26, no. 13: 4061. https://doi.org/10.3390/molecules26134061

Article Metrics

Back to TopTop