Next Article in Journal
Exploration of the Antioxidant and Anti-inflammatory Potential of Cassia sieberiana DC and Piliostigma thonningii (Schumach.) Milne-Redh, Traditionally Used in the Treatment of Hepatitis in the Hauts-Bassins Region of Burkina Faso
Next Article in Special Issue
Novel Antibacterial Agents 2022
Previous Article in Journal
Protective Effect of Vitamin D Supplementation on COVID-19-Related Intensive Care Hospitalization and Mortality: Definitive Evidence from Meta-Analysis and Trial Sequential Analysis
Previous Article in Special Issue
Recent Developments in Electrochemical Sensors for the Detection of Antibiotic-Resistant Bacteria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

N-Derivatives of (Z)-Methyl 3-(4-Oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylates as Antimicrobial Agents—In Silico and In Vitro Evaluation

by
Anthi Petrou
1,
Athina Geronikaki
1,*,
Victor Kartsev
2,
Antonios Kousaxidis
1,
Aliki Papadimitriou-Tsantarliotou
3,
Marina Kostic
4,
Marija Ivanov
4,
Marina Sokovic
4,
Ioannis Nicolaou
1 and
Ioannis S. Vizirianakis
3,5
1
Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
2
InterBioScreen, 142432 Chernogolovka, Russia
3
Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
4
Mycological Laboratory, Department of Plant Physiology, Institute for Biological Research “Siniša Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
5
Department of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(1), 131; https://doi.org/10.3390/ph16010131
Submission received: 5 December 2022 / Revised: 9 January 2023 / Accepted: 10 January 2023 / Published: 16 January 2023
(This article belongs to the Special Issue Novel Antibacterial Agents 2022)

Abstract

:
Herein, we report the experimental evaluation of the antimicrobial activity of seventeen new (Z)-methyl 3-(4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylate derivatives. All tested compounds exhibited antibacterial activity against eight Gram-positive and Gram-negative bacteria. Their activity exceeded those of ampicillin as well as streptomycin by 10–50 fold. The most sensitive bacterium was En. Cloacae, while E. coli was the most resistant one, followed by M. flavus. The most active compound appeared to be compound 8 with MIC at 0.004–0.03 mg/mL and MBC at 0.008–0.06 mg/mL. The antifungal activity of tested compounds was good to excellent with MIC in the range of 0.004–0.06 mg/mL, with compound 15 being the most potent. T. viride was the most sensitive fungal, while A. fumigatus was the most resistant one. Docking studies revealed that the inhibition of E. coli MurB is probably responsible for their antibacterial activity, while 14a–lanosterol demethylase of CYP51Ca is involved in the mechanism of antifungal activity. Furthermore, drug-likeness and ADMET profile prediction were performed. Finally, the cytotoxicity studies were performed for the most active compounds using MTT assay against normal MRC5 cells.

1. Introduction

Infectious diseases symbolize a consequential global strain on public health security and impact socio-economic stability all over the world. They have monopolized for centuries the dominant factors of death and disability of millions of humans.
Since their discovery, antimicrobial agents have been a reliable weapon in fighting life-threatening infections. Despite the availability of effective antibiotics for the most common infections, the emergence of multidrug-resistant bacteria pathogens and the spread of new infectious diseases threaten to weaken the efficiency of the drugs currently approved for infectious disease therapy [1,2].
On the other hand, infections evoked by several pathogenic fungi species result in 1.5 million deaths worldwide every year [3]. The therapeutic approach to these diseases is still challenging due to the continuous increase of resistance to antifungal drugs in sufferers, especially in immuno-suppressed patients (cancer, transplants, HIV) and those who are frequently treated with antimitotic therapy. Therefore, there is an urgent necessity to put efforts into discovering novel agents against invasive microbial infections.
Aromatic heterocycles have been used as structural scaffolds to obtain a variety of bioactive compounds possessing antibacterial, antifungal, antiviral, and anti-parasitic activities [4]. Over the past decades, thiazolidine-2,4-diones (glitazones) have emerged as the most successful member of the 4-thiazolidinone compounds [5]. There are several drugs from this class of heterocycles, such as Lobeglitazone, Pioglitazone, Rosiglitazone, and Epalrestat, used broadly as oral anti-hyperglycemic agents for diabetes mellitus type 2 therapies, and recently, Ponesimod was approved for healing multiple sclerosis and psoriasis (Figure 1).
In addition, 2 hodamine derivatives demonstrate a broad spectrum of pharmacological activities, such as antimicrobial [6,7,8,9], anticancer [10,11,12], anti-HIV [13,14], anti-diabetic [15,16], anti-tubercular [17], and immunoproteasome inhibitory activity [18], suggesting that this scaffold occupies a vital position in drug discovery.
Another promising scaffold is the indole ring attracting considerable attention because of its presence in proteins, amino acids, and bioactive alkaloids [19], and the wide range of biological activities of its derivatives [20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35], among which is antimicrobial activity [36,37,38,39,40,41]. Furthermore, indole ring is present in many approved drugs (Figure 2).
In recent years, the concept of designing hybrid molecules that contain two or more pharmacophore groups bound together covalently into one molecular framework is gaining ground. There are some publications regarding the importance of co-operative hydrogen bonding in antibiotics and the molecular hybridization approach of sugar-fused indoles [42,43].
Since the molecular hybridization approach may result in compounds that inhibit more than one target, we set up our investigation to discover novel antibacterial and antifungal agents based on this rationale [44].
So far, many indole-based rhodanine derivatives (Figure 3) have been proposed [6,41,45,46,47,48,49] as antimicrobial agents, some of which possess high efficacy in treating multi-drug resistant pathogens [6,41,47,48,49]. Therefore, it is noteworthy that the combination of indole and rhodanine scaffolds into new chemical entities could be a promising strategy for antimicrobial therapies. Inspired by this strategy, we have described in our previous paper [6,41] the design and synthesis of potential antimicrobial agents by incorporating indole and rhodanine moieties into new molecules. Herein, we report the synthesis, the evaluation of antimicrobial activity, molecular docking studies, and the prediction of pharmacokinetic and toxicity profiles of our compounds.

2. Results and Discussion

2.1. Prediction of Toxicity

The prediction of compound toxicities is a key part of the drug development pipeline. Toxicity assessments in silico are not only faster than the determination of toxic doses in animals but can also facilitate the lessening of the number of experiments in vivo. In our case, a computational toxicity study was carried out prior to the in vitro evaluation of compounds 117 with the aim to identify and remove potentially toxic structures. ProTox-II web server was used for the prediction of various toxicity endpoints, such as rat acute toxicity, hepatotoxicity, cytotoxicity, carcinogenicity, mutagenicity, immunotoxicity, and adverse outcome (Tox21) pathways (Tables S1 and S2). In addition, ADMET Predictor 10.4 (Simulation Plus) was used to investigate other critical toxicity issues such as phospholipidosis and hERG-related cardiotoxicity (Table S1).
It is known that toxic doses are often given as LD50 values in mg/kg body weight, where LD50 represents the median lethal dose upon exposure to a compound. Toxicity classes are defined according to the globally harmonized system of classification of the labeling of chemicals (GHS), that is, Class I: fatal if swallowed (LD50 ≤ 5); Class II: fatal if swallowed (5 < LD50 ≤ 50); Class III: toxic if swallowed (50 < LD50 ≤ 300); Class IV: harmful if swallowed (300 < LD50 ≤ 2000); Class V: may be harmful if swallowed (2000 < LD50 ≤ 5000); and Class VI: non-toxic (LD50 > 5000). In our preliminary investigation, it was estimated (54–66% accuracy) that all examined compounds are classified in category IV, indicating a promising profile towards oral rat acute toxicity. Furthermore, our compounds did not predict to demonstrate adverse outcomes (Table S2).

2.2. Chemistry

Compounds were synthesized according to the process described in detail in our previous paper [41] and presented in Scheme 1. The structures of all synthesized compounds are illustrated in Table 1 (Table S1) and confirmed by 1H and 13C NMR spectroscopy and elemental analysis, which are described in the experimental part.

2.3. Biological Evaluation

2.3.1. Antimicrobial Activity

The synthesized compounds (Table 1) were evaluated for their antibacterial activity against a panel of eight species using a microdilution method with ampicillin and streptomycin as reference drugs. The antibacterial activity of tested compounds was good to very good with MIC/MBC values ranging from 0.004 mg/mL to 0.045 mg/mL and 0.008 mg/mL to 1.2 mg/mL, respectively, presented in Table 2. The order of activity can be presented as: 8 > 11 > 2 > 1 > 12 > 3 > 17 > 7 > 5 > 13 > 14 > 16 > 9 > 4 = 6 > 15 > 10. The best activity was expressed by compound 8 with MIC at 0.004–0.03 mg/mL and MBC at 0.008–0.06 mg/mL, while compound 10 was the least active one. Compounds 1, 2, and 3 exhibited good activity against B. cereus with MIC at 0.015 mg/mL. The same good activity was observed for compounds 2, 3, 5, 6, and 7 against S. aureus; 8 and 12 against L. monocytogenes; and compounds 26 and 12 against En. Cloacae. Compound 12, together with compounds 1, 3, 7, and 11 (MIC 0.015 mg/mL), showed good activity against S. typhimurium, while derivatives 1 and 11 showed good activity against E. coli. On the other hand, compounds 8 and 12 showed excellent activity with MIC at 0.004 mg/mL against En. cloacae and E. coli, respectively. Very good activity (MIC 0.008 mg/mL) was observed for compounds 11 and 17 against B. cereus and S. aureus, respectively. At the same time, compound 11 exhibited good activity with MIC at 0.011 mg/mL against En. cloacae and P. aeruginosa. It should be mentioned that all compounds were more potent than both reference drugs against all bacteria tested. The most sensitive bacteria appeared to be En. cloacae, while E. coli was the most resistant one, followed by M. flavus.
The structure–activity relationship revealed that the presence of 3-methylbutanoic acid as a substituent on the nitrogen of the 2-thioxothiazolidin-4-one moiety of (Z)-2-(5-((5-fluoro-2-(methoxycarbonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-3-methylbutanoic acid, as well as methylformate in the indole ring (8), is beneficial for antibacterial activity. The replacement of 3-methylbutanoic acid by an acetic acid substituent on the nitrogen of 2-thioxothiazolidin-4-one ring, as well as the removal of the F-substituent at position 5 of the indole ring, decreased a little in activity (11), while the presence of benzoic acid at the same position led to a slightly less active compound 2. The removal of F-substituent at position 5 on the indole ring from compound 8 led to compound 12, which is fifth in the order of activity, while the presence of 4-hydroxybenzene substituent on the nitrogen of 2-thioxothiazolidin-4-one moiety appeared to be detrimental for antibacterial activity. Thus, the structure–activity relationship studies revealed that the activity of compounds depends not only on the substituent at the 2-thioxothiazolidin-4-one ring but as well as at the indole ring.

2.3.2. Antifungal Activity

The antifungal activity of compounds was tested against eight fungal species using the drugs ketoconazole and bifonazole as reference. The antifungal activity of tested compounds was good to excellent with MIC in the range of 0.004–0.06 mg/mL and MFC at 0.008–0.12 mg/mL, presented in Table 3.
The order of activity of tested compounds can be presented as follows: 15 > 3 > 16 > 10 > 7 > 6 > 2 > 5 > 11 > 9 > 13 > 4 > 12 > 17 > 14 > 1 > 8. Thus, the best activity was achieved by compound 15 with MIC/MFC at 0.008–0.015/0.015–0.03 mg/mL, respectively, while compound 8 was the least active. The opposite was observed in the case of antibacterial activity, where compound 8 was the most active and 15 was one of the least active. Some compounds showed excellent activity against some fungal species being more potent than both reference drugs. Thus, compound 1 expressed activity with MIC 0.004 mg/mL against P. ochramensis, while 3, 5, 7, and 10 expressed activity against T. viride. At the same time, compounds 10 and 9 showed the same good activity against A. ochraceus. Most of the compounds demonstrated very good activity against A. ochraceus with MIC at 0.008 mg/mL (1, 2, 48, 11), while compounds 4, 6, 9, 11, 12, 15, and 16 were also very good against T. viride. Furthermore, compounds 6, 7, 10, and 15 were also very potent against P. ochramensis, whereas 3 7, 10, and 15 also demonstrated very good activity against A. niger and P. funiculosum, respectively. Finally, compound 15 appeared to be very active also against P. cyclpoium var verucosum after A. fumigatus, the most resistant fungi. T. viride was found to be the most sensitive one.
The structure–activity relationship studies revealed that the presence of the methyl group substituent on the nitrogen of 2-thioxothiazolidin-4-one (15) moiety of (Z)-methyl 5-fluoro-3-((3-methyl-4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylate is favorable for antifungal activity. The replacement of the methyl group by 4-hydroxybenzene (3) and the removal of the F-substituent of the indole ring slightly decreased the activity. The introduction of morpholine as a substituent instead of methyl in compound 15 led to a less active compound (16), nevertheless being among the third most active compounds, while the removal of the fluorine atom from the indole ring resulted in compound 6 being sixth in order of activity.
Finally, the presence of 3-methylbutanoic acid as a substituent at the nitrogen of 2-thioxothiazolidin-4-one moiety (8) was detrimental to antifungal activity, while showing the best antibacterial activity among all tested compounds. Thus, the antifungal activity of compounds, as in the case of antibacterial, depends not only on substituent at 2-thioxothiazolidin-4-one but also at the indole ring as well.

2.4. Docking Studies

2.4.1. Docking to Antibacterial Targets

In order to predict the possible mechanism of activity of synthesized compounds, docking studies in different targets were carried out. In this direction, for docking studies, we used enzymes responsible for the most common mechanisms of activity of antibacterial agents, such as E. coli DNA gyrase, thymidylate kinase, E. coli primase, and E. coli MurA and E. coli MurB enzymes.
A low Free Energy of Binding represents a strong binding of ligand to the enzyme. Taking this into account, docking studies revealed that the Free Energy of Binding of all tested compounds to E. coli DNA gyrase, thymidylate kinase, E. coli primase, and E. coli MurA enzymes was higher than that of E. coli MurB (−7.54–10.88 kcal/mol). Therefore, it may be suggested that the inhibition of E. coli MurB is probably the most suitable mechanism of action of the compounds where binding scores were consistent with biological activity (Table 4).
The docking pose of the most active compound 8 in the E. coli MurB enzyme showed two favorable hydrogen bond interactions. The first one is between the oxygen atom of the carbonyl group of the compound and the hydrogen of the side chain of Ser229 (distance 3.11 Å), and the other one is between the S atom of thiazolidinone moiety and Ser50 residue (distance 3.64 Å). Moreover, hydrophobic interactions between residues Val52, Arg159, Ile110, and the compound were detected, contributing to the stability of the complex ligand–enzyme (Figure 4). It is noteworthy to highlight that the hydrogen bond with the residue Ser229 is crucial for the inhibitory action of this compound because this residue takes part in the proton transfer at the second stage of peptidoglycan synthesis [50]. Hydrogen bond interactions with the residue Ser229 were also observed for most of the compounds (Table 4).
Moreover, detailed analysis of the docking pose of the most active compounds showed that they bind MurB in a similar way to FAD and they fit into the binding center of the enzyme in the same way, interacting with the same residues, such as Ser50, Arg213, Arg158, and Ser229 (Figure 4A). This is probably the reason why these compounds showed good inhibitory activity comparable to that of ampicillin.

2.4.2. Docking to Antifungal Targets

All the synthesized compounds and the reference drug ketoconazole were docked to lanosterol 14α-demethylase of C. albicans and DNA topoisomerase IV (Table 5) in order to explore the possible mechanism of antifungal activity of compounds.
Docking studies showed that all compounds can bind the CYP51Ca enzyme in an analogous mode to that of ketoconazole. Compound 15 is placed inside the enzyme by the side of the heme group, interacting hydrophobically aromatically and binding to the Fe of the heme. Furthermore, compound 15 forms one H-bond between the oxygen atom of the CO2 group and the side-chain hydrogen of Thr311. Hydrophobic interactions were detected between residues Ile304, Leu300, and Ile131 and the benzene ring of the compound 15 (Figure 5B). Interaction with the heme group was also observed with the benzene ring of ketoconazole, which forms aromatic and hydrophobic interactions (Figure 5). However, compound 15 formed a more stable complex of the ligand with the enzyme, indicating their interaction with the Fe of heme. This is probably the reason why compound 15 (Figure 5A) has better antifungal activity than ketoconazole (Figure 6).

2.5. Drug-Likeness and Bioavailability

The drug-likeness and bioavailability scores of all tested compounds are shown in Table 6. According to prediction, the bioavailability score of most of the compounds was about 0.55, except for compounds 2, 9, 12, 14, and 17 with a value of 0.11. According to the BOILED-Egg illustration (Figure 7A), all compounds are predicted to demonstrate moderate to high gastrointestinal (GI) absorption. Especially, compounds 5, 6, 7, 15, and 16 are predicted to be passively absorbed by the gastrointestinal tract. In addition, none of the examined compounds are predicted to passively permeate the blood–brain barrier. All compounds showed no violation of Lipinski’s rule of five. Half of the compounds demonstrated a TPSA value < 140 Å, thus indicating their good oral bioavailability. Furthermore, all compounds displayed moderate drug-likeness scores ranging from −0.95 to 0.38, probably due to the enhanced TPSA values and the absence of a basic moiety on rhodanine’s nitrogen. The best in silico prediction was achieved for compound 5, bearing the basic propyl-morpholine group with a drug-likeness score of 0.24 (Figure 7, Table 6).

2.6. ADMET Properties

All compounds have been assessed for their ADMET profile in ADMET Predictor version 10.4 provided by the Simulation Plus software package [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. We have found that compounds 1, 8, 9, 11, 12, 14, and 17 demonstrate optimal properties regarding distribution and, especially, absorption (Table 7). Although these compounds have moderate permeability indices due to their carboxylate moiety, they showed low potential to penetrate the blood–brain barrier (decreased logBB values) as well as acceptable water and salt solubility at a normal blood pH value of 7.4. Moreover, we believe that these compounds could be well absorbed orally after food in the small intestine based on the favorable solubility values in fed state simulated intestinal fluid (FeSSIF). On the other hand, they exhibit low fraction unbound values (less than 6.0%) in comparison with other derivatives in this series (e.g., neutral or basic analogs). Nevertheless, the values of volume of distribution (Vd) and blood-to-plasma ratio (RBP) in humans were predicted to be satisfactory for all compounds in this series.
Furthermore, it has been predicted that the clearance mechanism of all compounds is metabolism (possibility of 74–99%) rather than hepatic uptake or renal elimination (99% both). In view of this fact, a comprehensive in silico analysis of cytochrome P450-mediated metabolism of our compounds was executed (Table 8). First of all, we have found that the different groups incorporated on the nitrogen atom of the rhodanine scaffold can alter compounds’ metabolism. Among the most well-absorbed compounds, 8 and 12 have been assessed to show high clearance through CYP2C9 isoenzyme; thus, they exhibit a CYP risk greater than zero. In addition, compounds 1, 9, 11, 14, and 17 display zero possibilities of CYP risks. Therefore, they show optimal metabolism characteristics for further studies. We have also estimated that compounds 1, 2, 812, 14, and 17 are not CYP2E1 substrates (67–87%), whereas compounds 37, 13, 15, and 16 are predicted to be good CYP2E1 substrates. Moreover, the majority of compounds may inhibit CYP3A4 isoenzyme (33–80%) except for compounds 1, 2, 8, 11, and 12 (71–75%). Despite their differences in metabolism characteristics, all compounds share some common characteristics. Particularly, all compounds may inhibit CYP1A2 (68–97%) but not the isoenzymes of CYP2C9 (62–95%), CYP2C19 (94–99%), and CYP2D6 (95%). As illustrated in Table 8, it has been estimated that the major metabolizing enzymes for compounds 1, 9, 11, 14, and 17 are CYP2C9 and CYP2C19 (over 90% both). On the other hand, all compounds may be also metabolized through CYP2C8 (70–91%) rather than CYP2A6 (67–99%) and CYP2B6 (57–98%). In addition, these compounds may be subjected to glucuronidation by UDP-glucuronosyltransferases 1A3 and 1A9. Last but not least, we suggest that compounds 1, 9, 11, 14, and 17 may exhibit the most promising metabolism profiles according to the low intrinsic clearance values concerning cytochrome P450 metabolism (CYP-CLint) as well as molecule-level hepatic clearance in humans (HEP-CLint).
We have also predicted the metabolic pathways that will take place for the most potent compounds with optimal metabolism profiles, e.g., compounds 1 (Figure 8) and 11 (Figure 9). For both compounds, two main routes of oxidation and demethylation have been found. In particular, demethylated carboxylic acid derivatives account for 10% and 6% of the metabolisms of compounds 1 and 11, respectively. It is obvious that CYP2C19 and, even more, CYP2C9 are converting the free esters to carboxylic acid derivatives with similar yields. During oxidation, the 6-hydroxy-indole derivatives are also formed in almost equal amounts of 27% and 22% of the metabolic pathway of compounds 1 and 11, respectively. In addition, it has been shown that CYP2C9 is the major isoenzyme that may be responsible for the sulfone formation of these compounds. Thus, M4 is formed in 27% of the metabolism of compound 1 with regard to M2, which occurs in 18% of compound 11. Furthermore, it was also assumed that the main metabolites of these compounds could be the 2,4-thiazolidinones, namely 1-M3 and 11-M1, with surprising values of 36% and 53%, respectively. On the other hand, CYP2C8 has also an impact on the metabolism of compound 1 since it may cleave the acidic tail from the nitrogen atom of the rhodanine group, leaving a second metabolite of 6-oxohexanoic acid to be formed. However, this procedure has been established to occur only in compound 1 and not in the case of compound 11, which possesses the short acetic acid chain. Last but not least, it is worth mentioning that all metabolites of compounds 1 and 11 which have been estimated during at least three metabolic cycles have been found to be non-toxic.
We further studied in detail the possibility of our compounds being substrates or inhibitors against selected human transporters, which play a critical role in pharmacokinetic properties, especially distribution and excretion (Table 9). The ADMET Predictor software revealed that all compounds are both good P-glycoprotein (90–99%) substrates and also breast cancer receptor protein (BCRP) (62–95%) substrates except compound 12. It has been also assessed that all compounds may not inhibit BCRP (69–97%). Moreover, most of the examined compounds are non-P-glycoprotein inhibitors except compounds 27, 10, 13, and 16. In addition, the bile salt export pump (BSEP) which is almost exclusively expressed in the liver, is of great relevance to hepatotoxicity. BSEP inhibition can result in the accumulation of bile salts in the liver, which can lead to cholestasis and drug-induced liver injury (DILI) [54]. In our case, the majority of compounds exhibit no possibilities (≥52%) to inhibit BSEP and thus induce no potential hepatotoxicity.

2.7. Cytotoxicity

To evaluate cytotoxicity, compounds 1, 2, 8, and 11 were chosen to be tested in the human normal fetal lung fibroblast MRC-5 cell line within the following range of three concentrations for each substance: 1 × 10−7 M (0.1 μM), 1 × 10−6 Μ (1 μM), and 1 × 10−5 M (10 μΜ). As shown in Figure 10, after 48 h of exposure in culture, all three compounds exhibited no substantial cytotoxicity within the range of concentration tested since viability was ≥91% compared to control untreated culture. By comparing the three agents, only compound 11 exhibited a very slight inhibition in cellular viability at concentrations of 1 μM and 10 μM. More specifically, under this exposure to compound 11, the cellular viability was measured to the level of 91.0%. The other compounds, 1, 2, and 8, showed no statistically significant change in cellular viability compared to control cells. Overall, these data propose that the tested compounds at the level of concentrations investigated are not cytotoxic in human normal MRC-5 cells.

3. Materials and Methods

3.1. Prediction of Toxicity

The prediction of toxicity was performed using Protox II webserver [55].

3.2. Chemistry

NMR spectra were determined with Varian Mercury VX-400″ (Varian Co., Palo Alto, CA, USA) and AM-300 Bruker 300 MHz. spectrometers in DMSO-d6, and spectra are in Table S1.
Chemical shifts of nuclei 1H were measured relatively in the residual signals of deuteron solvent (δ = 2.50 ppm). Coupling constants (J) are reported in Hz. Τhe assignment was based on 2D NMR techniques. Melting points were determined using the Fisher-Johns Melting Point Apparatus (Fisher Scientific, Hampton, NH, USA) and are uncorrected. Elemental analysis was performed by the classical method of microanalysis.
All compounds were synthesized analogous to the process described in our previous paper [6].
A mixture of a corresponding amino acid (50 mmol), a cooled solution of KOH in water (20 mL) (150 mmol in case of dicarbonic acids), and CS2 (mmol) were stirred in a flat-bottomed flask until a solution was formed. A solution of monochloracetic acid (55 mmol) was added with stirring, pre-neutralized with sodium bicarbonate (55 mmol) in water (25 mL), and left at room temperature for 2 days.
Then, to the formed solution, a 6N HCl solution (20 mL) was added and heated to boiling and kept at a slow boil for 1 h. After cooling, the precipitate formed was filtered off, dried, and recrystallized, alternately, from diluted acetic acid, ethanol, and toluene.
In a round-bottom flask equipped with a reflux condenser, 2.5 mmol of methyl 3-formyl-5,6-disabstituted-1H-indole-2-carboxylate, 3.3 mmol of 3-substituted-2-thioxothiazolidin-4-one, 2.5 mmol of ammonium acetate, and 5 mL of acetic acid were placed. The reaction mixture was boiled for 2 h and cooled. Then, the precipitate was filtered off, washed with acetic acid and water, dried, and recrystallized.
  • (Z)-6-(5-((2-(methoxycarbonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)hexanoic acid (1). m.p. 135–138 °C. 1H NMR (300 MHz, DMSO-d6) δ 12.06 (s, 2H,NH, OH), 8.49 (s, 1H), 7.79 (s, 1H), 7.58 (s, 1H), 7.35 (s, 1H), 7.24 (s, 1H), 4.09 (s, 2H), 4.00 (s, 3H, CH3), 2.21 (s, 2H), 1.75 (s, 2H), 1.65 (s, 2H), 1.45 (s, 2H). Anal. Calcd. For C20H20N2O5S2 (%): C, 55.54; H, 4.66; N, 6.48; O, 18.50. Found (%): C, 55.50; H, 4.69; N, 6.52; O, 18.47.
  • (Z)-4-(5-((2-(methoxycarbonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)benzoic acid (2). m.p. 294–295 °C. 1H NMR (300 MHz, DMSO-d6) δ 12.69 (s, 1H, OH), 8.51 (s, 1H), 8.16 (d, J = 8.1 Hz, 2H), 7.87 (d, J = 8.2 Hz, 1H), 7.60 (d, J = 8.1 Hz, 1H), 7.47 (d, J = 8.2 Hz, 2H), 7.37 (t, J = 7.6 Hz, 1H), 7.28 (t, J = 7.5 Hz, 1H), 4.00 (s, 3H, CH3). Anal. Calcd. For C21H14N2O5S2 (%): C, 57.52; H, 3.22; N, 6.39; O, 18.24. Found (%): C, 57.54; H, 3.20; N, 6.42; O, 18.25.
  • (Z)-methyl 3-((3-(3-hydroxyphenyl)-4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylate (3). m.p. 245–247 °C. 1H NMR (300 MHz, DMSO-d6) δ 12.66 (s, 1H, OH), 9.61 (s, 1H, NH), 8.48 (s, 1H), 7.87 (d, J = 8.2 Hz, 1H), 7.59 (d, J = 8.3 Hz, 1H), 7.31 (dq, J = 7.2, 22.3 Hz, 3H), 6.90 (d, J = 8.4 Hz, 1H), 6.71 (d, J = 5.2 Hz, 2H), 4.00 (s, 3H,CH3). Anal. Calcd. For C20H14N2O4S2 (%): C, 58.52; H, 3.44; N, 6.82; O, 15.59. Found (%): C, 58.50; H, 3.43; N, 6.87; O, 15.53.
  • (Z)-methyl 3-((3-(4-hydroxyphenyl)-4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylate (4). m.p. 282–284 °C. 1H NMR (300 MHz, DMSO-d6) δ 12.64 (s, 1H, OH), 9.62 (s, 1H, NH), 8.47 (s, 1H), 7.86 (d, J = 8.2 Hz, 1H), 7.59 (d, J = 8.3 Hz, 1H), 7.36 (t, J = 7.7 Hz, 1H), 7.26 (t, J = 7.6 Hz, 1H), 7.08 (d, J = 8.3 Hz, 2H), 6.90 (d, J = 8.3 Hz, 2H), 4.00 (s, 3H, CH3). Anal. Calcd. For C20H14N2O4S2 (%): C, 58.52; H, 3.44; N, 6.82; O, 15.59. Found (%): C, 58.50; H, 3.47; N, 6.86; O, 15.55.
  • (Z)-methyl 3-((3-(3-morpholinopropyl)-4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylate (5). m.p. 213–214 °C. 1H NMR (300 MHz, DMSO-d6) δ 12.64 (s, 1H, NH), 8.47 (s, 1H), 7.78 (d, J = 8.2 Hz, 1H), 7.58 (d, J = 8.3 Hz, 1H), 7.35 (t, J = 7.6 Hz, 1H), 7.24 (t, J = 7.6 Hz, 1H), 4.18 (t, J = 7.0 Hz, 2H), 4.00 (s, 2H), 3.55 (t, J = 4.6 Hz, 4H, CH3), 2.58–2.29 (m, 8H), 1.89 (p, J = 6.8 Hz, 2H). Anal. Calcd. For C21H23N3O4S2 (%): C, 56.61; H, 5.20; N, 9.43; O, 14.36. Found (%): C, 56.64; H, 5.18; N, 9.45; O, 14.30.
  • (Z)-methyl 3-((3-morpholino-4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylate (6). m.p. 278–280 °C. 1H NMR (300 MHz) δ 12.65 (s, 1H, NH), 8.42 (s, 1H), 7.78 (d, J = 8.1 Hz, 1H), 7.57 (d, J = 8.3 Hz, 1H), 7.34 (t, J = 7.7 Hz, 1H), 7.23 (t, J = 7.6 Hz, 1H), 4.00 (s, 3H, CH3), 3.05 (d, J = 22.9 Hz, 4H), 2.50 (s, 3H). Anal. Calcd. For C18H17N3O4S2 (%): C, 53.58; H, 4.25; N, 10.41; O, 15.86. Found (%): C, 53.60; H, 4.21; N, 10.46; O, 15.83.
  • (Z)-methyl 3-((3-(furan-2-ylmethyl)-4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylate (7). 1H NMR (300 MHz, DMSO-d6) δ 12.65 (s, 1H, NH), 8.52 (s, 1H), 7.79 (d, J = 8.2 Hz, 1H), 7.57 (d, J = 8.2 Hz, 1H), 7.47 (s, 1H), 7.34 (t, J = 7.7 Hz, 1H), 7.23 (t, J = 7.6 Hz, 1H), 6.41 (s, 1H), 6.36 (s, 1H), 5.26 (s, 2H), 4.00 (s, 3H, CH3). m.p. 212–213 °C. Anal. Calcd. For C19H14N2O4S2 (%): C, 57.27; H, 3.54; N, 7.03; O, 16.06. Found (%): C, 57.31; H, 3.50; N, 7.08; O, 16.02.
  • (Z)-2-(5-((5-fluoro-2-(methoxycarbonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-3-methylbutanoic acid (8). 1H NMR (300 MHz, DMSO-d6) δ 8.44 (s, 1H, NH), 7.58 (dd, J = 4.7, 9.0 Hz, 1H), 7.48 (d, J = 9.7 Hz, 1H), 7.15 (t, J = 9.2 Hz, 1H), 5.12 (d, J = 9.6 Hz, 1H), 4.00 (s, 3H, O-CH3), 2.80 (s, 1H, CH-(CH3)2), 1.28 (d, J = 6.2 Hz, 3H, CH-CH3), 0.84 (d, J = 6.7 Hz, 3H, CH-CH3). m.p. 234–235 °C. Anal. Calcd. For C19H17FN2O5S2 (%): C, 52.28; H, 3.93; F, 4.35; N, 6.42; O, 18.33. Found (%): C, 52.23; H, 3.90; F, 4.28; N, 6.54; O, 18.29.
  • (Z)-4-(5-((5-fluoro-2-(methoxycarbonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)butanoic acid (9). m.p. 192–193 °C. 1H NMR (300 MHz, DMSO-d6) δ 12.72 (s, 1H, NH), 11.90 (s, 1H, OH), 8.38 (s, 1H), 7.57 (dd, J = 4.7, 9.0 Hz, 1H), 7.46 (dd, J = 2.3, 9.9 Hz, 1H), 7.14 (td, J = 2.5, 9.1 Hz, 1H), 4.14 (t, J = 7.0 Hz, 2H), 3.99 (s, 3H, CH3), 2.32 (t, J = 7.4 Hz, 2H), 1.98 (p, J = 7.4 Hz, 2H). Anal. Calcd. For C18H15FN2O5S2 (%): C, 51.18; H, 3.58; F, 4.50; N, 6.63; O, 18.94. Found (%): C, 51.23; H, 3.55; F, 4.48; N, 6.69; O, 18.90.
  • (Z)-methyl 5-fluoro-3-((3-(4-hydroxyphenyl)-4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylate (10). m.p. 267–268 °C. 1H NMR (300 MHz, DMSO-d6) δ 12.74 (s, 1H, OH), 9.61 (s, 1H, NH), 8.37 (s, 1H), 7.64 – 7.47 (m, 2H), 7.15 (t, J = 8.7 Hz, 1H), 7.07 (d, J = 8.4 Hz, 2H), 6.90 (d, J = 8.4 Hz, 2H), 3.99 (s, 3H, O-CH3). Anal. Calcd. For C20H13FN2O4S2 (%): C, 56.06; H, 3.06; F, 4.43; N, 6.54; O, 14.94. Found (%): C, 56.12; H, 3.02; F, 4.39; N, 6.58; O, 14.87.
  • (Z)-2-(5-((2-(methoxycarbonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)acetic acid (11). m.p. 292–294 °C. 1H NMR (300 MHz, DMSO-d6) δ 8.51 (s, 1H), 7.80 (d, J = 8.3 Hz, 1H), 7.57 (d, J = 8.3 Hz, 1H), 7.34 (t, J = 7.6 Hz, 1H), 7.24 (t, J = 7.6 Hz, 1H), 4.65 (s, 2H), 3.99 (s, 3H,O-CH3). Anal. Calcd. For C16H12N2O5S2 (%): C, 51.05; H, 3.21; N, 7.44; O, 21.25. Found (%): C, 51.11; H, 3.27; N, 7.33; O, 21.20.
  • (Z)-2-(5-((2-(methoxycarbonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-3-methylbutanoic acid (12). m.p. 256–258 °C. 1H NMR (300 MHz, DMSO-d6) δ 8.51 (s, 1H), 7.82 (d, J = 8.2 Hz, 1H), 7.58 (d, J = 8.2 Hz, 1H), 7.34 (t, J = 7.7 Hz, 1H), 7.24 (t, J = 7.6 Hz, 1H), 5.13 (d, J = 9.2 Hz, 1H), 4.00 (s, 3H), 2.79 (s, 1H, CH-(CH3)2), 1.28 (d, J = 6.3 Hz, 3H, CH-CH3), 0.84 (d, J = 6.8 Hz, 3H, CH-CH3). Anal. Calcd. For C19H18N2O5S2 (%): C, 54.53; H, 4.34; N, 6.69; O, 19.12. Found (%): C, 54.49; H, 4.37; N, 6.71; O, 19.10.
  • (Z)-methyl 3-((3-(3-fluorophenyl)-4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-6-methoxy-1H-indole-2-carboxylate (13). m.p. 238–240 °C. 1H NMR (300 MHz, DMSO-d6) δ 12.65 (s, 1H, NH), 8.52 (s, 1H), 7.79 (d, J = 8.2 Hz, 1H), 7.57 (d, J = 8.2 Hz, 1H), 7.47 (s, 1H), 7.34 (t, J = 7.7 Hz, 1H), 7.23 (t, J = 7.6 Hz, 1H), 6.41 (s, 1H), 6.36 (s, 1H), 5.26(s, 3H, O-CH3), 4.00 (s, 3H, O-CH3). Anal. Calcd. For C21H15FN2O4S2 (%): C, 57.00; H, 3.42; F, 4.29; N, 6.33; O, 14.46. Found (%): C, 57.12; H, 3.39; F, 4.32; N, 6.38; O, 14.51.
  • (Z)-3-(5-((5-fluoro-2-(methoxycarbonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)propanoic acid (14). m.p. 275–276 °C. 1H NMR (300 MHz, DMSO-d6) δ 12.76 (s, 1H, OH), 12.30 (s, 1H, NH), 8.39 (s, 1H), 7.57 (dd, J = 4.6, 9.0 Hz, 1H), 7.44 (d, J = 9.8 Hz, 1H), 7.15 (t, J = 8.9 Hz, 1H), 4.29 (t, J = 7.9 Hz, 2H), 3.99 (s, 3H, CH3), 2.64 (t, J = 8.1 Hz, 2H). Anal. Calcd. For C17H13FN2O5S2 (%): C, 49.99; H, 3.21; F, 4.65; N, 6.86; O, 19.59. Found (%): C, 49.92; H, 3.25; F, 4.60; N, 6.90; O, 19.62.
  • (Z)-methyl 5-fluoro-3-((3-methyl-4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylate (15). m.p. 234–244 °C. 1H NMR (300 MHz) δ 12.75 (s, 1H, NH), 8.40 (s, 1H), 7.57 (dd, J = 4.7, 9.0 Hz, 1H), 7.43 (d, J = 9.7 Hz, 1H), 7.15 (t, J = 9.0 Hz, 1H), 3.99 (s, 3H, O-CH3), 3.48 (s, 3H, N-CH3). Anal. Calcd. For C15H11FN2O3S2 (%): C, 51.42; H, 3.16; F, 5.42; N, 7.99; O, 13.70. Found (%): C, 51.40; H, 3.21; F, 5.45; N, 7.93; O, 13.75.
  • (Z)-methyl 5-fluoro-3-((3-morpholino-4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylate (16). m.p. 273–274 °C. 1H NMR (300 MHz, DMSO-d6) δ 12.76 (s, 1H, NH), 8.32 (s, 1H), 7.57 (dd, J = 4.6, 9.1 Hz, 1H), 7.46 (d, J = 9.6 Hz, 1H), 7.15 (t, J = 9.1 Hz, 1H), 4.00 (s, 3H, CH3), 3.77 (dd, J = 16.8, 29.8 Hz, 6H), 3.05 (d, J = 26.1 Hz, 3H). Anal. Calcd. For C18H16FN3O4S2 (%): C, 51.30; H, 3.83; F, 4.51; N, 9.97; O, 15.18. Found (%): C, 51.36; H, 3.79; F, 4.54; N, 9.93; O, 15.21.
  • (Z)-3-(5-((2-(methoxycarbonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)propanoic acid (17). m.p. 265–266 °C. 1H NMR (300 MHz, DMSO-d6) δ 12.65 (s, 1H, NH), 12.30 (s, 1H, OH), 8.49 (s, 1H), 7.78 (d, J = 8.2 Hz, 1H), 7.57 (d, J = 8.3 Hz, 1H), 7.35 (t, J = 7.7 Hz, 1H), 7.24 (t, J = 7.4 Hz, 1H), 4.29 (t, J = 7.9 Hz, 2H), 4.00 (s, 3H, O-CH3), 2.64 (t, J = 8.1 Hz, 2H). Anal. Calcd. For C17H14N2O5S2 (%): C, 52.30; H, 3.61; N, 7.17; O, 20.49. Found (%): C, 52.28; H, 3.65; N, 7.21; O, 20.52.

3.3. Biological Evaluation

3.3.1. Antibacterial Activity

The following Gram-negative bacteria: Escherichia coli (ATCC 35210), Enterobacter cloacae (clinical isolate), Salmonella typhimurium (ATCC 13311), as well as Gram-positive bacteria: Listeria monocytogenes (NCTC 7973), Bacillus cereus (clinical isolate), and Staphylococcus aureus (ATCC 6538) were used. The organisms were obtained from the Mycological Laboratory, Department of Plant Physiology, Institute for Biological Research “Siniša Stankovic”, Belgrade, Serbia. The minimum inhibitory (MIC) and minimum bactericidal (MBC) concentrations were determined by the modified microdilution method, as previously reported [6,41].

3.3.2. Antifungal Activity

For the antifungal bioassays, six fungi were used: Aspergillus niger (ATCC 6275), Aspergillus fumigatus (ATCC 1022), Aspergillus versicolor (ATCC 11730), Penicillium funiculosum (ATCC 36839), Trichoderma viride (IAM 5061), and 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 [56,57].

3.4. Docking Studies

AutoDock 4.2® software was used for the in silico studies, and a detailed procedure is reported in our previous paper [58].

3.5. Drug Likeness

Five filters were used to predict drug-likeness [59] by the Molsoft software and SwissADME program (http://swissadme.ch, accessed on 25 October 2022) via the ChemAxon’s Marvin JS structure drawing tool.

3.6. ADMET

The prediction of the pharmacokinetic and toxicity (phospholipidosis, hERG-cardiotoxicity) profile of compounds was executed by ADMET Predictor 10.4 [51,52,53].

3.7. Assessment of Cytotoxicity

The normal human fetal lung fibroblast MRC-5 cell line was maintained and used in our laboratory (Dr. I.s.Vizirianakis, Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Greece) (passage 15 < 40), as previously published [60]. Cells were grown in culture under the following conditions: 37 °C, humidified atmosphere containing 5% v/v CO2, in DMEM (Dulbecco’s modified Eagle Medium) enriched with 10% FBS and 1% PS. The compounds tested were dissolved in DMSO and stored at 4 °C. For the assessment of cytotoxicity, the cells were cultivated in a 96-well plate at an initial concentration of 5 × 104 cells/mL and allowed to attach for 20 h before the addition of the compounds at concentrations of 1 × 10−5 M (10 μM), 1 × 10−6 M(1 μM), and 1 × 10−7 M (0.1 μM). It is worth noting that the concentration of DMSO in the culture was 0.02% v/v, in which no detectable effect on cell toxicity was observed. To assess the cytotoxicity of each compound, the cells were allowed to grow for 48 h under the effect of each substance. Subsequently, Cell Counting Kit-8 (CCK8, St. Louis, MO, USA, Sigma-Aldrich) reagent was added to each well and incubated for 4 h at 37 °C. After the incubation, the OD for each well was determined at 450 nm in a multifunction microplate reader. Wells containing only the CCK-8 reagent were used as blank control. Data are presented as mean ± standard deviation (SD) of triplicate incubations. Statistical t-test and one-way analysis of variance (ANOVA) were performed via the use of the SPSS program and the significance level was determined at p < 0.05.

4. Conclusions

Seventeen (Z)-methyl-3-(4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylates 117 were designed, synthesized, and evaluated in silico and experimentally for their antimicrobial actions against a panel of Gram-positive and Gram-negative bacteria and fungi.
The evaluation revealed that all compounds were more potent than both reference drugs, ampicillin, and streptomycin against all bacteria tested. En. cloacae appeared to be the most sensitive bacterial strain towards our derivatives, whereas E. coli was the most resistant one, followed by M. flavus.
Concerning antifungal action, the tested compounds exhibited very good to excellent activity against all the fungal species tested, being more active than ketoconazole and bifonazole. Most of the compounds appeared to be very potent against A. ochraceus and T. viride with the last one being the most sensitive to tested compounds. Filamentous A. fumigatus was the most resistant strain.
It can be observed that the growth of both Gram-negative and Gram-positive bacteria and fungi responded differently to the tested compounds, suggesting that different substituents may lead to different modes of action or that the metabolism of some bacteria/fungi was able to overcome the effect of the compounds or adapt to it.
Docking analysis to DNA Gyrase, Thymidylate kinase, and E. coli MurB indicated MurB inhibition as a putative antibacterial mechanism of compounds tested, while docking analysis to 14α-lanosterol demethylase (CYP51) and tetrahydrofolate reductase of Candida albicans pointed out a probable implication of CYP51 reductase in the antifungal activity of the compounds.
Even though compounds displayed moderate to good drug-likeness scores (−0.89 to +0.24), no violation of the Lipinski rule was observed. Furthermore, according to predicted results, six out of seventeen compounds can be orally absorbed since their TPSA are less than 140 Å. All compounds have been assessed for their ADMET profile in ADMET Predictor version 10.4, provided by the Simulation Plus software package. We have found that compounds 1, 8, 9, 11, 12, 14, and 17 demonstrate optimal properties regarding distribution and especially absorption.
The prediction of metabolic pathways that could take place for the potent compounds 1 and 11 with optimal metabolism profiles indicated the transformation of rhodanine to thiazolidinone and indole ring hydroxylation as the two main routes. The evaluation of the cytotoxicity of compounds on normal human fetal lung fibroblast MRC-5 cell lines revealed that compounds are not toxic.
Thus, these derivatives can be considered as lead compounds for the development of novel potent and safe antimicrobial agents.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph16010131/s1, Table S1. Prediction of organ toxicity and toxicity end points for compounds 117, Table S2. Prediction of adverse outcomes according to TOX21 of compounds 117, 1H-NMR and 13C-NMR spectra of compounds.

Author Contributions

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

Funding

This research was supported by the Serbian Ministry of Education, Science and Technological Development for financial support (project No. 451-03-68/2020-14/200007).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We acknowledge the help of Volodimr Horishny from the Department of Chemistry, Danylo Halytsky Lviv National Medical University, Ukraine, with the synthesis of compounds.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chopra, I.; Schofield, C.; Everett, M.; O’Neill, A.; Miller, K.; Wilcox, M.; Frere, J.-M.; Dawson, M.; Czaplewsky, L.; Urleb, U.; et al. Treatment of Health-Care-Associated Infections Caused by Gram-Negative Bacteria: A Consensus Statement. Lancet Infect. Dis. 2008, 8, 133–139. [Google Scholar] [CrossRef] [PubMed]
  2. Overbye, K.M.; Barrett, J.F. Antibiotics: Where Did We Go Wrong? Drug Discov. Today 2005, 10, 45–52. [Google Scholar] [CrossRef] [PubMed]
  3. Baker, A.W.; Maziarz, E.K.; Arnold, C.J.; Johnson, M.D.; Workman, A.D.; Reynolds, J.M.; Perfect, J.R.; Alexander, B.D. Invasive fungal infection after lung transplantation: Epidemiology in the setting of antifungal prophylaxis. Clin. Infect. Dis. 2020, 70, 30–39. [Google Scholar] [CrossRef] [PubMed]
  4. Sayed, M.; Kamal El-Dean, A.M.K.; Ahmed, M.; Hassanien, R. Synthesis of some heterocyclic compounds derived from indole as antimicrobial agents. Synth. Commun. 2018, 48, 413–421. [Google Scholar] [CrossRef]
  5. Roszczenko, P.; Holota, S.; Szewczyk, O.K.; Dudchak, R.; Bielawski, K.; Bielawska, A.; Lesyk, R. 4-Thiazolidinone-Bearing Hybrid Molecules in Anticancer Drug Design. Int. J. Mol. Sci. 2022, 23, 13135. [Google Scholar] [CrossRef]
  6. Horishny, V.; Kartsev, V.; Matiychuk, V.; Geronikaki, A.; Petrou, A.; 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]
  7. Barakat, A.; Al-Najjar, H.J.; Al-Majid, A.M.; Soliman, S.M.; Mabkhot, Y.N.; Al-Agamy, M.H.M.; Ghabbour, H.A.; Fun, H.K. Synthesis, molecular structure investigations and antimicrobial activity of 2- thioxothiazolidin-4-one derivatives. J. Mol. Struct. 2015, 1081, 519–529. [Google Scholar] [CrossRef]
  8. Cebeci, Y.U.; Karaoğlu, Ş.A. Quinolone-Rhodanine Hybrid Compounds: Synthesis and Biological Evaluation as Anti-Bacterial Agents. ChemistrySelect 2022, 7, e202201007. [Google Scholar] [CrossRef]
  9. Abusetta, A.; Alumairi, J.; Alkaabi, M.Y.; Al Ajeil, R.; Shkaidim, A.A.; Akram, D.; Pajak, J. Design, Synthesis, in Vitro Antibacterial Activity, and Docking Studies of New Rhodanine Derivatives. Open J. Med. Chem. 2020, 10, 15–34. [Google Scholar]
  10. Kumar, A.S.; Kumar, R.A.; Reddy, E.P.; Satyanarayanaa, V.; Kashannab, J.; Reddy, B.J.M.; Reddy, B.V.S.; Yadav, J.S. Synthesis of Novel 2-Thioxothiazolidin-4-one and Thiazolidine-2, 4-dione.Derivatives as Potential Anticancer Agents. Nat. Prod. Commun. 2018, 13, 589–591. [Google Scholar]
  11. Manikala, V.; Rao, M.V. Synthesis, Molecular Docking and Anticancer Activity of Novel (E)-5-((1-phenyl-1H-1,2,3-triazol-4-yl)methylene)-2-thioxothiazolidin-4-one Analogues. Iran. J. Chem. Chem. Eng. 2021, 40, 1793–1799. [Google Scholar]
  12. Battula, H.; Bommi, S.; Bobde, Y.; Tarun Patel, T.; Balaram Ghosh, B.; Jayanty, S. Distinct rhodamine B derivatives exhibiting dual effect of anticancer activity and fluorescence property. J. Photochem. Photobiol. 2021, 6, 100026. [Google Scholar] [CrossRef]
  13. Tintori, C.; Iovenitt, G.; Ceresola, E.R.; Ferrarese, R.; Zamperini, C.; Brai, A.; Giulio Poli, G.; Dreassi, E.; Cagno, V.; Lembo, D.; et al. Rhodanine derivatives as potent anti-HIV and anti-HSV microbicides. PLoS ONE 2018, 13, e0198478. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, Y.; Xia, L.; Yuan, Y.; Li, Q.; Han, L.; Yang, G.; Hu, H. Rhodanine derivative LJ001 inhibits TGEV and PDCoV replication in vitro. Virus Res. 2020, 289, 198167. [Google Scholar] [CrossRef]
  15. Khairul, A.; Kamar, D.A.; Yin, L.J.; Liang, C.T.; Fung, G.T.; Avupati, V.R. Rhodanine scaffold: A review of antidiabetic potential and structure–activity relationships (SAR). Med. Drug Dis. 2022, 15, 100131. [Google Scholar]
  16. Toumi, A.; Boudriga, S.; Hamden, K.; Sobeh, M.; Cheurfa, M.; Askri, M.; Knorr, M.; Strohmann, C.; Brieger, L. Synthesis, antidiabetic activity and molecular docking study of rhodanine-substitued spirooxindole pyrrolidine derivatives as novel α-amylase inhibitors. Bioorg. Chem. 2021, 106, 104507. [Google Scholar] [CrossRef]
  17. Xu, J.; Wang, T.T.; Yuan, Q.; Duan, Y.T.; Xu, Y.J.; Lv, P.C.; Wang, X.M.; Yang, Y.S.; Zhu, H.L. Discovery and development of novel rhodanine derivatives targeting enoyl-acyl carrier protein reductase. Bioorg. Med. Chem. 2019, 27, 1509–1516. [Google Scholar] [CrossRef] [PubMed]
  18. Previti, S.; Grasso, S.; Zappalà, M.; Ottanà, R. Identification of 2-thioxoimidazolidin-4-one derivatives as novel noncovalent proteasome and immunoproteasome inhibitors. Bioorg. Med. Chem. Lett. 2018, 28, 278–283. [Google Scholar]
  19. Vitaku, D.T.; Smith, J.T.N. Analysis of the structural diversity, substitution patterns, and frequency of nitrogen heterocycles among U.S. FDA approved pharmaceuticals. J. Med. Chem. 2014, 57, 10257–10274. [Google Scholar] [CrossRef]
  20. Dhiman, A.; Sharma, R.; Singh, R.K. Target-based anticancer indole derivatives and insight into structure—Activity relationship: A mechanistic review update (2018–2021). Acta Pharm. Sin. B 2022, 12, 3006–3027. [Google Scholar] [CrossRef]
  21. Devi, N.; Kaur, K.; Biharee, A.; Vikas Jaitak, V. Recent Development in Indole Derivatives as Anticancer Agent: A Mechanistic Approach. Anti-Cancer Agents Med. Chem. 2021, 21, 1802–1824. [Google Scholar] [CrossRef] [PubMed]
  22. Jia, Y.; Wen, X.; Gong, Y.; Wang, X. Current scenario of indole derivatives with potential anti-drug-resistant cancer activity. Eur. J. Med. Chem. 2020, 200, 112359. [Google Scholar] [CrossRef] [PubMed]
  23. Bhat, M.A.; Al-Omar, M.A.; Raish, M.; Ansari, M.A.; Abuelizz, H.A.; Bakheit, A.H.; Naglah, A.M. Indole Derivatives as Cyclooxygenase Inhibitors: Synthesis, Biological Evaluation and Docking Studies. Molecules 2018, 23, 1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Kumari, A.; Singh, R.K. Synthesis, Molecular Docking and Biological Evaluation of N-Substituted, Indole Derivatives as Potential Anti-Inflammatory and Antioxidant Agents. Chem. Biodivers. 2022, 19, e202200290. [Google Scholar] [CrossRef] [PubMed]
  25. Ji, J.; He, H.; Zhang, X.; Wu, R.; Gan, L.; Li, D.; Lu, Y.; Wu, P.; Wong, K. The in vitro and in vivo study of oleanolic acid indole derivatives as novel anti-inflammatory agents: Synthesis, biological evaluation, and mechanistic analysis. Bioorg. Chem. 2021, 113, 104981. [Google Scholar]
  26. Zhu, Y.; Zhao, J.; Luo, L.; Gao, Y.; Bao, H.; Li, P.; Zhang, H. Research progress of indole compound with potential antidiabetic activity. Eur. J. Med. Chem. 2021, 223, 113665. [Google Scholar] [CrossRef]
  27. Taha, M.; Alrashedy, A.S.; Almandil, N.B.; Iqbal, N.; Anouar, E.H.; Nawaz, M.; Uddin, N.; Chigurupati, S.; Wadood, A.; Rahim, F.; et al. Synthesis of indole derivatives as diabetics II inhibitors and enzymatic kinetics study of α-glucosidase and α-amylase along with their in-silico study. Int. J. Biol. Macromol. 2021, 190, 301–318. [Google Scholar] [CrossRef]
  28. Dorababu, A. Indole—A promising pharmacophore in recent antiviral drug discovery. RSC Med. Chem. 2020, 11, 1335–1353. [Google Scholar] [CrossRef]
  29. Sahin, K. Investigation of novel indole-based HIV-1 protease inhibitors using virtual screening and text mining. Investigation of novel indole-based HIV-1 protease inhibitors using virtual screening and text mining. J. Biomol. Struct. Dyn. 2021, 39, 3638–3648. [Google Scholar] [CrossRef]
  30. Cihan-Üstündağ, G.; Naesens, L.; Şatana, D.; Gonca Erköse-Genç, G.; Mataracı-Kara, E.; Çapan, G. Design, synthesis, antitubercular and antiviral properties of new spirocyclic indole derivatives. Mon. Chem. 2019, 150, 1533–1544. [Google Scholar] [CrossRef] [Green Version]
  31. Bajad, N.G.; Singh, S.K.; Singh, S.K.; Singh, T.D.; Singh, M. Indole: A promising scaffold for the discovery and development of potential anti-tubercular agents. Curr. Res. Pharmacol. Drug Discov. 2022, 3, 100119. [Google Scholar] [CrossRef] [PubMed]
  32. Porwal, S.; Gupta, S.; Chauhan, P.M.S. gem-Dithioacetylated indole derivatives as novel antileishmanial agents. Bioorg. Med. Chem. Lett. 2017, 27, 4643–4646. [Google Scholar] [CrossRef] [PubMed]
  33. Tiwari, S.; Kirar, S.; Banerjee, U.C.; Neerupudi, K.B.; Singh, S.; Abdullah, A.; Prasad, V.; Bharatam, V.; Singh, I.P. Synthesis of N-substituted indole derivatives as potential antimicrobial and antileishmanial agents. Bioorg. Chem. 2020, 99, 103787. [Google Scholar] [CrossRef] [PubMed]
  34. Elkamhawy, A.; Woo, J.; Nada, H.; Angeli, A.; Bedair, T.M.; Supuran, C.T.; Lee, K. Identification of Novel and Potent Indole-Based Benzenesulfonamides as Selective Human Carbonic Anhydrase II Inhibitors: Design, Synthesis, In Vitro, and In Silico Studies. Int. J. Mol. Sci. 2022, 23, 2540. [Google Scholar] [CrossRef] [PubMed]
  35. Asati, V.; Bhupal, R.; Bhattacharya, S.; Kaur, K.; Gupta, G.D.; Pathak, A.; Mahapatra, D.K. Recent updates of Indole derivatives as kinase inhibitors in the treatment of cancer. Anti-Cancer Agents Med. Chem. 2022, 23, 404–416. [Google Scholar] [CrossRef]
  36. Yuan, W.; Yu, Z.; Song, W.; Li, Y.; Fang, Z.; Zhu, B.; Li, X.; Wang, H.; Hong, W.; Sun, N. Indole-core-based novel antibacterial agent targeting FtsZ. Infect. Drug Resist. 2019, 12, 2283–2296. [Google Scholar] [CrossRef]
  37. Al-Wabli, R.I.; Alsulami, M.A.; Bukhari, S.I.; Moubaye, N.M.S.; Al-Mutairi, M.S.; Attia, M.I. Design, Synthesis, and Antimicrobial Activity of Certain New Indole-1,2,4 Triazole Conjugates. Molecules 2021, 26, 2292. [Google Scholar] [CrossRef]
  38. Tha, S.; Shakya, S.; Malla, M.; Aryal, P. Prospects of Indole derivatives as methyl transfer inhibitors: Antimicrobial resistance managers. BMC Pharm. Toxicol. 2020, 21, 33–44. [Google Scholar] [CrossRef]
  39. Kaur, H.; Singh, J.; Narasimhan, B. Indole hybridized diazenyl derivatives: Synthesis, antimicrobial activity, cytotoxicity evaluation and docking studies. BMC Chem. 2019, 13, 65–83. [Google Scholar] [CrossRef]
  40. Qin, H.-L.; Jing, L.; Fang, W.-Y.; Rakesh, K.P. Indole-based derivatives as potential antibacterial activity against methicillin-resistance Staphylococcus aureus (MRSA). Eur. J. Med. Chem. 2020, 194, 112245. [Google Scholar] [CrossRef]
  41. Horishny, V.; Geronikaki, A.; Kartsev, V.; Matiychuk, V.; Petrou, A.; Pogodin, P.; Poroikov, V.; Theodoroula, N.F.; Vizirianakis, I.S.; Kostic, M.; et al. Synthesis, biological evaluation and molecular docking studies of 5-indolylmethylen-4-oxo-2-thioxothiazolidine derivatives. Molecules 2020, 25, 1964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Michael, G.P.; Chennaiah, A.; Klara, H.; Sven, N.H.; Andrea, V.; Erik, C.B.; David, C. Importance of Co-operative Hydrogen Bonding in the Apramycin-Ribosomal Decoding A-Site Interaction. ChemMedChem 2022, e202200486. [Google Scholar]
  43. Verma, A.K.; Chennaiah, A.; Dubbu, S.; Vankar, Y.D. Palladium catalyzed synthesis of sugar-fused indolines via C(sp2)-H/N-H activation. Carbohydr. Res. 2019, 473, 57–65. [Google Scholar] [CrossRef] [PubMed]
  44. Konidala, S.K.; Kotra, V.; Danduga, R.C.S.R.; Kola, P.K.; Bhandare, R.R.; Shaik, A.B. Design, multistep synthesis and in-vitro antimicrobial and antioxidant screening of coumarin clubbed chalcone hybrids through molecular hybridization approach. Arab. J. Chem. 2021, 14, 103154. [Google Scholar] [CrossRef]
  45. Liu, H.; Sun, D.; Du, H.; Zheng, C.; Li, J.; Piao, H.; Li, J.; Sun, S. Synthesis and biological evaluation of tryptophan-derived rhodanine derivatives as PTP1B inhibitors and anti-bacterial agents. Eur. J. Med. Chem. 2019, 172, 163–173. [Google Scholar] [CrossRef]
  46. Konechnyi, Y.T.; Lozynskyi1, A.V.; Horishny, V.Y.; Konechna, R.T.; Vynnytska, R.B.; Korniychuk, O.P.; Lesyk, R.B. Synthesis of indoline-thiazolidinone hybrids with antibacterial and antifungal activities. Biopolym. Cell 2020, 36, 381–391. [Google Scholar] [CrossRef]
  47. Akunuri, R.; Unnissa, T.; Kaul, G.; Akhir, A.; Saxena, D.; Wajidali, M.; Veerareddy, V.; Yaddanapudi, V.M.; Sidharth Chopra, S.; Nanduri, S. Synthesis and Antibacterial Evaluation of Rhodanine and Its Related Heterocyclic Compounds against S. aureus and A. baumannii. Chem. Biodivers. 2022, 19, e202200213. [Google Scholar] [CrossRef]
  48. Pardasavio, P.T.; Pardasavi, P.; Sherry, D.; Chatarverdi, V. Synthetic and antibacterial studies of rhodanine derivatives with indol-2,3-diones. Ind. J. Chem. 2001, 40B, 1275–1278. [Google Scholar]
  49. Song, M.X.; Li, S.H.; Peng, J.-Y.; Guo, T.-T.; Xu, W.-H.; Xiong, S.-F.; Deng, X.-Q. Synthesis and Bioactivity Evaluation of N-Arylsulfonylindole Analogs Bearing a Rhodanine Moiety as Antibacterial Agents. Molecules 2017, 22, 970. [Google Scholar] [CrossRef] [Green Version]
  50. Benson, T.E.; Walsh, C.T.; Massey, V. Kinetic characterization of wild-type and S229A mutant MurB: Evidence for the role of Ser 229 as a general acid. Biochemistry 1997, 36, 796–805. [Google Scholar] [CrossRef]
  51. Sohlenius-Sternbeck, A.K.; Terelius, Y. Evaluation of ADMET Predictor in Early Discovery Drug Metabolism and Pharmacokinetics Project Work. Drug Metab. Dispos. 2022, 50, 95–104. [Google Scholar] [CrossRef] [PubMed]
  52. Naga, D.; Parrott, N.; Ecker, G.F.; Morale, A.O. Evaluation of the Success of High-Throughput Physiologically Based Pharmacokinetic (HT-PBPK) Modeling Predictions to Inform Early Drug Discovery. Mol. Pharm. 2022, 19, 2203–2216. [Google Scholar] [CrossRef] [PubMed]
  53. Kuriwaki, I.; Kameda, M.; Iikubo, K.; Hisamichi, H.; Kawamoto, Y.; Kikuchi, S.; Moritomo, H.; Terasaka, T.; Iwai, Y.; Noda, A.; et al. Discovery of ASP5878: Synthesis and structure–activity relationships of pyrimidine derivatives as pan-FGFRs inhibitors with improved metabolic stability and suppressed hERG channel inhibitory activity. Bioorg. Med. Chem. 2022, 59, 116657. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, J.; He, K.; Cai, L.; Chen, Y.C.; Yang, Y.; Shi, Q.; Woolf, T.F.; Ge, W.; Guo, L.; Borlak, J.; et al. Inhibition of bile salt transport by drugs associated with liver injury in primary hepatocytes from human, monkey, dog, rat, and mouse. Chem. Biol. Interact. 2016, 255, 45–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Banerjee, P.; Eckert, A.O.; Schrey, A.K.; Preissner, R. ProTox-II: A webserver for the prediction of toxicity of chemicals. Nucleic Acids Res. 2018, 46, W257–W263. [Google Scholar] [CrossRef] [Green Version]
  56. 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]
  57. 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]
  58. Fesatidou, M.; Zagaliotis, P.; Camoutsis, C.; Perou, A.; Eleftheriou, P.; Tratrtat, 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]
  59. Lipinski, C.A. Lead- and drug-like compounds: The rule-of-five revolution. Drug Discov. Today Technol. 2004, 1, 337–341. [Google Scholar] [CrossRef]
  60. Tseligka, E.D.; Rova, A.; Amanatiadou, E.P.; Calabrese, G.; Tsibouklis, J.; Fatouros, D.G.; Vizirianakis, I.S. Pharmacological Development of Target-Specific Delocalized Lipophilic Cation-Functionalized Carboranes for Cancer Therapy. Pharm. Res. 2016, 33, 1945–1958. [Google Scholar] [CrossRef]
Figure 1. Structures of approved 4-thiazolidinone-based drugs.
Figure 1. Structures of approved 4-thiazolidinone-based drugs.
Pharmaceuticals 16 00131 g001
Figure 2. Structures of approved indole-based drugs.
Figure 2. Structures of approved indole-based drugs.
Pharmaceuticals 16 00131 g002
Figure 3. Antimicrobial agents containing indole and rhodanine ring [6,41,45,47,48,49].
Figure 3. Antimicrobial agents containing indole and rhodanine ring [6,41,45,47,48,49].
Pharmaceuticals 16 00131 g003
Scheme 1. Preparation of compounds 117 Reagents and conditions: CH3COONH4, CH3COOH, reflux 2 h.
Scheme 1. Preparation of compounds 117 Reagents and conditions: CH3COONH4, CH3COOH, reflux 2 h.
Pharmaceuticals 16 00131 sch001
Figure 4. (A) Superposition of compound 8 (magenta) and FAD (blue) in E. coli MurB. (B) Docked conformation of the most active compound 8 in E. coli MurB. Red dotted arrows indicate H-bond and yellow spheres indicate hydrophobic interactions.
Figure 4. (A) Superposition of compound 8 (magenta) and FAD (blue) in E. coli MurB. (B) Docked conformation of the most active compound 8 in E. coli MurB. Red dotted arrows indicate H-bond and yellow spheres indicate hydrophobic interactions.
Pharmaceuticals 16 00131 g004
Figure 5. Docked conformation of the most active compound 15 in lanosterol 14α-demethylase of C. albicans (CYP51ca). (A) 3D representation of compound 15. (B) 2D representation. Red dotted arrows indicate H-bond, blue arrows indicate aromatic interactions, and yellow spheres indicate hydrophobic interactions.
Figure 5. Docked conformation of the most active compound 15 in lanosterol 14α-demethylase of C. albicans (CYP51ca). (A) 3D representation of compound 15. (B) 2D representation. Red dotted arrows indicate H-bond, blue arrows indicate aromatic interactions, and yellow spheres indicate hydrophobic interactions.
Pharmaceuticals 16 00131 g005
Figure 6. Docked conformation of ketoconazole in lanosterol 14α-demethylase of C. albicans (CYP51ca).
Figure 6. Docked conformation of ketoconazole in lanosterol 14α-demethylase of C. albicans (CYP51ca).
Pharmaceuticals 16 00131 g006
Figure 7. (A) Boiled egg, (B) bioavailability radar of compound 5, (C) drug−likeness model. In bioavailability radar, the pink area represents the optimal range for each property for oral bioavailability, (Lipophilicity (LIPO): XLOGP3 between −0.7 and +5.0, Molecular weight (SIZE): MW between 150 and 500 g/mol, Polarity (POLAR) TPSA between 20 and 130 Å2, Solubility (INSOLU): log S not higher than 6, Saturation (INSATU): fraction of carbons in the sp3 hybridization not less than 0.25, and Flexibility (FLEX): no more than nine rotatable bonds.
Figure 7. (A) Boiled egg, (B) bioavailability radar of compound 5, (C) drug−likeness model. In bioavailability radar, the pink area represents the optimal range for each property for oral bioavailability, (Lipophilicity (LIPO): XLOGP3 between −0.7 and +5.0, Molecular weight (SIZE): MW between 150 and 500 g/mol, Polarity (POLAR) TPSA between 20 and 130 Å2, Solubility (INSOLU): log S not higher than 6, Saturation (INSATU): fraction of carbons in the sp3 hybridization not less than 0.25, and Flexibility (FLEX): no more than nine rotatable bonds.
Pharmaceuticals 16 00131 g007
Figure 8. Metabolism of compound 1.
Figure 8. Metabolism of compound 1.
Pharmaceuticals 16 00131 g008
Figure 9. Metabolism of compound 11.
Figure 9. Metabolism of compound 11.
Pharmaceuticals 16 00131 g009
Figure 10. Cell viability levels (%) of MRC-5 cells exposed for 48h at various concentrations in culture to compounds 1, 2, 8, and 11. All values are presented as mean ± standard deviation (SD) of triplicate independent cellular experiments. With asterisk (*), the significant differences between each concentration and the control group are presented. The significance level was determined at p < 0.05.
Figure 10. Cell viability levels (%) of MRC-5 cells exposed for 48h at various concentrations in culture to compounds 1, 2, 8, and 11. All values are presented as mean ± standard deviation (SD) of triplicate independent cellular experiments. With asterisk (*), the significant differences between each concentration and the control group are presented. The significance level was determined at p < 0.05.
Pharmaceuticals 16 00131 g010
Table 1. The structures of evaluated compounds.
Table 1. The structures of evaluated compounds.
Pharmaceuticals 16 00131 i001
NR1R2R3NR1R2R3NR1R2R3
1HH-(CH2)5COOH7HH2-methylfuran13-OCH3H3-FC6H4
2HH4-COOHC6H48HF-C(CH3)2COOH14HF-(CH2)2COOH
3HH3-OHC6H49HF-(CH2)3COOH15HF-CH3
4HH4-OHC6H410HF4-OHC6H416HFmorpholine
5HH4-propylmorpholine11HH-CH2COOH17HH-(CH2)2COOH
6HHmorpholine12HH-C(CH3)2COOH
Table 2. Antibacterial activity of target compounds (MIC and MBC in mg/mL).
Table 2. Antibacterial activity of target compounds (MIC and MBC in mg/mL).
Compounds S.a.B.c.M.f.L.m.P.a.E. coliEn.clS.t.
1MIC0.022 ± 0.0050.015 ± 0.0090.022 ± 0.0050.022 ± 0.0040.022 ± 0.0050.015 ± 0.0090.022 ± 0.0050.015 ± 0.009
MBC0.03 ± 0.010.03 ± 0.010.03 ± 0.010.06 ± 0.0060.06 ± 0.0060.03 ± 0.010.03 ± 0.010.03 ± 0.01
2MIC0.015 ± 0.0090.015 ± 0.0090.022 ± 0.0050.03 ± 0.010.03 ± 0.010.022 ± 0.0050.015 ± 0.0090.022 ± 0.005
MBC0.03 ± 0.010.03 ± 0.010.03 ± 0.010.06 ± 0.0060.06 ± 0.0060.03 ± 0.010.03 ± 0.010.03 ± 0.01
3MIC0.015 ± 0.0090.015 ± 0.0090.045 ± 0.0030.03 ± 0.010.03 ± 0.010.022 ± 0.0050.015 ± 0.0090.015 ± 0.009
MBC0.03 ± 0.010.03 ± 0.010.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.03 ± 0.010.03 ± 0.010.03 ± 0.01
4MIC0.03 ± 0.010.03 ± 0.010.045 ± 0.0030.045 ± 0.0010.03 ± 0.010.045 ± 0.0030.015 ± 0.0090.045 ± 0.003
MBC0.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.03 ± 0.010.06 ± 0.006
5MIC0.015 ± 0.0090.03 ± 0.010.045 ± 0.0030.03 ± 0.010.03 ± 0.010.03 ± 0.010.015 ± 0.0090.022 ± 0.005
MBC0.03 ± 0.010.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.03 ± 0.010.03 ± 0.01
6MIC0.015 ± 0.0090.03 ± 0.010.045 ± 0.0030.03 ± 0.010.03 ± 0.010.06 ± 0.0060.015 ± 0.0090.03 ± 0.01
MBC0.03 ± 0.010.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.12 ± 0.010.03 ± 0.010.06 ± 0.006
7MIC0.015 ± 0.0090.03 ± 0.010.045 ± 0.0030.03 ± 0.010.03 ± 0.010.03 ± 0.010.015 ± 0.0090.015 ± 0.009
MBC0.03 ± 0.010.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.03 ± 0.010.03 ± 0.01
8MIC0.03 ± 0.010.03 ± 0.010.03 ± 0.010.015 ± 0.0010.004 ± 0.0050.008 ± 0.0010.004 ± 0.0050.008 ± 0.009
MBC0.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.03 ± 0.010.008 ± 0.00060.015 ± 0.0090.008 ± 0.00060.015 ± 0.009
9MIC0.03 ± 0.010.03 ± 0.010.03 ± 0.010.03 ± 0.010.03 ± 0.010.03 ± 0.010.03 ± 0.010.03 ± 0.01
MBC0.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.006
10MIC0.03 ± 0.010.03 ± 0.010.045 ± 0.0030.03 ± 0.010.03 ± 0.010.03 ± 0.010.03 ± 0.010.045 ± 0.003
MBC0.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.006
11MIC0.008 ± 0.00060.008 ± 0.00060.03 ± 0.010.03 ± 0.010.011 ± 0.010.015 ± 0.0090.011 ± 0.010.015 ± 0.009
MBC0.015 ± 0.0090.015 ± 0.0090.06 ± 0.0060.06 ± 0.0060.03 ± 0.010.03 ± 0.010.03 ± 0.010.03 ± 0.01
12MIC0.03 ± 0.010.03 ± 0.010.03 ± 0.010.015 ± 0.0010.015 ± 0.0090.015 ± 0.0090.03 ± 0.010.004 ± 0.005
MBC0.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.03 ± 0.010.03 ± 0.010.03 ± 0.010.06 ± 0.0060.008 ± 0.009
13MIC0.015 ± 0.0090.03 ± 0.010.045 ± 0.0030.03 ± 0.010.03 ± 0.010.045 ± 0.0030.015 ± 0.0090.03 ± 0.01
MBC0.03 ± 0.010.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.03 ± 0.010.06 ± 0.006
14MIC0.03 ± 0.010.03 ± 0.010.045 ± 0.0030.045 ± 0.0030.015 ± 0.0090.045 ± 0.0030.015 ± 0.0090.045 ± 0.003
MBC0.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.03 ± 0.010.06 ± 0.0060.03 ± 0.010.06 ± 0.006
15MIC0.022 ± 0.0050.03 ± 0.010.045 ± 0.0030.045 ± 0.0030.03 ± 0.010.045 ± 0.0030.03 ± 0.010.045 ± 0.003
MBC0.03 ± 0.010.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.006
16MIC0.022 ± 0.0050.015 ± 0.0090.045 ± 0.0030.045 ± 0.0030.03 ± 0.010.03 ± 0.010.045 ± 0.0030.045 ± 0.003
MBC0.03 ± 0.010.03 ± 0.010.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.06 ± 0.006
17MIC0.008 ± 0.00060.03 ± 0.010.03 ± 0.010.03 ± 0.010.008 ± 0.0000.045 ± 0.0030.008 ± 0.00060.03 ± 0.01
MBC0.015 ± 0.000.06 ± 0.0060.06 ± 0.0060.06 ± 0.0060.015 ± 0.0090.06 ± 0.0060.015 ± 0.0090.06 ± 0.006
AmpicillinMIC0.10 ± 0.050.006 ± 0.0030.25 ± 0.090.15 ± 0.030.30 ± 0.030.10 ± 0.050.25 ± 0.090.15 ± 0.03
MBC0.15 ± 0.050.025 ± 0.000.50 ± 0.10.30 ± 0.030.50 ± 0.10.20 ± 0.000.50 ± 0.10.20 ± 0.009
StreptomycinMIC0.10 ± 0.000.0015 ± 0.00020.20 ± 0.000.15 ± 0.030.10 ± 0.050.10 ± 0.050.20 ± 0.0050.10 ± 0.05
MBC0.20 ± 0.000.003 ± 0.00050.30 ± 0.030.30 ± 0.030.20 ± 0.000.20 ± 0.000.30 ± 0.030.20 ± 0.009
B.c.B. cereus, M.f.M. flavus, S.a.S. aurues, L.m.L. monocytogenes, En.c.En. cloacae, P.a.P. aeruginosa, S.t.S. typhimurium.
Table 3. Antifungal activity of target compounds (MIC and MFC in mg/mL).
Table 3. Antifungal activity of target compounds (MIC and MFC in mg/mL).
Compounds A.f.A.v.A.o.A.n.T.v.P.o.P.f.P.v.c.
1MIC0.06 ± 0.006 0.03 ± 0.010.008 ± 0.00060.022 ± 0.0050.015 ± 0.003 0.004 ± 0.001 0.015 ± 0.0030.03 ± 0.01
MFC0.12 ± 0.040.06 ± 0.0060.015 ± 0.0030.03 ± 0.010.03 ± 0.010.008 ± 0.0006 0.03 ± 0.01 0.06 ± 0.006
2MIC0.03 ± 0.01 0.015 ± 0.0030.11 ± 0.010.022 ± 0.005 0.008 ± 0.00060.015 ± 0.0030.015 ± 0.0030.03 ± 0.01
MFC0.06 ± 0.0060.03 ± 0.01 0.015 ± 0.0030.03 ± 0.010.015 ± 0.003 0.03 ± 0.010.03 ± 0.01 0.06 ± 0.006
3MIC0.03 ± 0.010.008 ± 0.00060.008 ± 0.00060.11 ± 0.010.004 ± 0.0010.008 ± 0.00060.008 ± 0.00060.015 ± 0.003
MFC0.06 ± 0.0060.015 ± 0.0030.015 ± 0.003 0.015 ± 0.003 0.008 ± 0.00060.015 ± 0.003 0.015 ± 0.0030.03 ± 0.01
4MIC0.06 ± 0.0060.008 ± 0.00060.008 ± 0.00060.015 ± 0.0030.008 ± 0.00060.015 ± 0.0030.015 ± 0.0030.03 ± 0.01
MFC0.12 ± 0.04 0.015 ± 0.003 0.015 ± 0.0030.03 ± 0.01 0.015 ± 0.0030.03 ± 0.01 0.03 ± 0.010.06 ± 0.006
5MIC0.06 ± 0.0060.008 ± 0.0006 0.008 ± 0.00060.015 ± 0.003 0.004 ± 0.0010.11 ± 0.01 0.015 ± 0.003 0.015 ± 0.003
MFC0.12 ± 0.040.015 ± 0.0030.015 ± 0.0030.03 ± 0.010.008 ± 0.00060.015 ± 0.0030.03 ± 0.01 0.03 ± 0.01
6MIC0.06 ± 0.0060.008 ± 0.00060.008 ± 0.0006 0.11 ± 0.01 0.008 ± 0.0006 0.008 ± 0.00060.015 ± 0.0030.015 ± 0.003
MFC0.12 ± 0.040.015 ± 0.0030.015 ± 0.0030.03 ± 0.010.015 ± 0.003 0.015 ± 0.0030.03 ± 0.010.03 ± 0.01
7MIC0.06 ± 0.0060.008 ± 0.00060.008 ± 0.0006 0.008 ± 0.00060.004 ± 0.001 0.008 ± 0.00060.015 ± 0.003 0.015 ± 0.003
MFC0.12 ± 0.040.015 ± 0.0030.015 ± 0.0030.015 ± 0.003 0.008 ± 0.0006 0.015 ± 0.0030.03 ± 0.01 0.03 ± 0.01
8MIC0.06 ± 0.006 0.015 ± 0.003 0.008 ± 0.00060.015 ± 0.0030.11 ± 0.01 0.015 ± 0.003 0.015 ± 0.0030.06 ± 0.006
MFC0.12 ± 0.040.03 ± 0.010.015 ± 0.0030.03 ± 0.010.015 ± 0.003 0.03 ± 0.01 0.03 ± 0.010.12 ± 0.04
9MIC0.09 ± 0.0030.015 ± 0.003 0.004 ± 0.0010.015 ± 0.0030.008 ± 0.0006 0.015 ± 0.003 0.015 ± 0.0030.022 ± 0.005
MFC0.12 ± 0.040.03 ± 0.010.008 ± 0.0006 0.03 ± 0.010.015 ± 0.003 0.03 ± 0.010.03 ± 0.01 0.015 ± 0.003
10MIC0.06 ± 0.0060.008 ± 0.0006 0.004 ± 0.0010.008 ± 0.00060.004 ± 0.0010.008 ± 0.0006 0.008 ± 0.0006 0.015 ± 0.003
MFC0.12 ± 0.04 0.015 ± 0.0030.008 ± 0.00060.015 ± 0.0030.008 ± 0.00060.015 ± 0.003 0.015 ± 0.0030.03 ± 0.01
11MIC0.06 ± 0.0060.015 ± 0.003 0.008 ± 0.0006 0.015 ± 0.0030.008 ± 0.0006 0.015 ± 0.003 0.015 ± 0.0030.015 ± 0.003
MFC0.12 ± 0.040.03 ± 0.010.015 ± 0.003 0.03 ± 0.010.015 ± 0.003 0.03 ± 0.01 0.03 ± 0.01 0.03 ± 0.01
12MIC0.06 ± 0.0060.008 ± 0.00060.008 ± 0.0006 0.022 ± 0.0050.008 ± 0.0006 0.015 ± 0.003 0.015 ± 0.003 0.03 ± 0.01
MFC0.12 ± 0.04 0.015 ± 0.003 0.015 ± 0.0030.03 ± 0.010.015 ± 0.0030.03 ± 0.01 0.03 ± 0.01 0.06 ± 0.006
13MIC0.06 ± 0.0060.015 ± 0.0030.008 ± 0.0006 0.015 ± 0.0030.11 ± 0.01 0.015 ± 0.0030.015 ± 0.003 0.015 ± 0.003
MFC0.12 ± 0.04 0.03 ± 0.010.015 ± 0.0030.03 ± 0.010.03 ± 0.01 0.03 ± 0.010.03 ± 0.01 0.03 ± 0.01
14MIC0.06 ± 0.0060.015 ± 0.003 0.015 ± 0.0030.015 ± 0.0030.11 ± 0.01 0.015 ± 0.003 0.015 ± 0.003 0.015 ± 0.003
MFC0.12 ± 0.04 0.03 ± 0.01 0.03 ± 0.01 0.03 ± 0.010.03 ± 0.010.03 ± 0.010.03 ± 0.01 0.03 ± 0.01
15MIC0.015 ± 0.0030.008 ± 0.0006 0.008 ± 0.00060.008 ± 0.00060.008 ± 0.0006 0.008 ± 0.00060.008 ± 0.0006 0.008 ± 0.0006
MFC0.03 ± 0.010.015 ± 0.0030.015 ± 0.0030.015 ± 0.0030.015 ± 0.0030.015 ± 0.0030.015 ± 0.003 0.015 ± 0.003
16MIC0.015 ± 0.0030.015 ± 0.0030.015 ± 0.0030.015 ± 0.0030.008 ± 0.00060.015 ± 0.003 0.015 ± 0.0030.015 ± 0.003
MFC0.03 ± 0.01 0.03 ± 0.010.03 ± 0.01 0.03 ± 0.010.015 ± 0.003 0.03 ± 0.010.03 ± 0.01 0.03 ± 0.01
17MIC0.06 ± 0.006 0.015 ± 0.003 0.008 ± 0.0006 0.022 ± 0.0050.11 ± 0.01 0.015 ± 0.003 0.015 ± 0.003 0.03 ± 0.01
MFC0.12 ± 0.04 0.03 ± 0.010.015 ± 0.0030.015 ± 0.0030.015 ± 0.0030.03 ± 0.010.03 ± 0.010.06 ± 0.006
KetoconazoleMIC0.20 ± 0.01 0.20 ± 0.010.25 ± 0.050.20 ± 0.011.00 ± 0.12.50 ± 0.30.20 ± 0.01 0.20 ± 0.01
MFC0.50 ± 0.0010.50 ± 0.0020.50 ± 0.0060.50 ± 0.0041.50 ± 0.093.50 ± 0.030.50 ± 0.003 0.30 ± 0.01
BifonazoleMIC0.15 ± 0.050.10 ± 0.0020.15 ± 0.050.15 ± 0.050.15 ± 0.050.20 ± 0.010.20 ± 0.010.10 ± 0.009
MFC0.20 ± 0.010.20 ± 0.010.20 ± 0.010.20 ± 0.010.20 ± 0.010.25 ± 0.050.25 ± 0.050.20 ± 0.01
A.f.A. fumigatus, A.v.A. versicolor, A.o.A. ochraceus, A.n.A. niger, T.v.T. viride, P.f.P. funiculosum, P.o.P. ochrochloron, P.v.c.P. cyclpoium var verucosum.
Table 4. Molecular docking free binding energies (kcal/mol) to antibacterial targets.
Table 4. Molecular docking free binding energies (kcal/mol) to antibacterial targets.
Comp.Est. Binding Energy (kcal/mol)Residues Involved in H-Bond Formation in
E. coli MurB
E. coli Gyrase
1KZN
Thymidylate Kinase
4QGG
E. coli Primase
1DDE
E. coli MurA JV4TE. coli MurB 2Q85
1−4.15−1.68-−5.63−9.82Arg213, Ser229
2−3.17--−5.23−10.04Arg158, Ser229
3−4.63−2.55−1.28−6.27−9.53Arg213, Ser229
4−5.10--−4.63−7.90Arg158, Arg213
5−3.51--−5.20−8.97Arg213, Ser229
6−4.15-−1.39−4.82−7.68Arg213
7−4.38--−6.12−9.14Arg158, Ser229
8−5.37−2.85-−5.92−10.88Ser50, Ser229
9−4.12--−5.37−8.11Ser50, Arg158
10−5.47--−5.42−7.54Ser50
11−5.21−3.47−2.61−6.20−10.46Ser116, Ser229
12−3.50−2.51−5.73−9.56Ser229
13−4.39-−5.14−8.55Ser229
14−4.26-−1.29−5.32−8.42Ser229
15−3.94-−2.57−5.18−7.60Arg213
16−5.28--−4.92−8.30Ser229
17−4.36--−5.16−9.46Arg158, Ser229
Table 5. Molecular docking free binding energies (kcal/mol) to antifungal targets.
Table 5. Molecular docking free binding energies (kcal/mol) to antifungal targets.
Est. Binding Energy (kcal/mol)Residues Involved in H-Bond FormationResidues Involved in Hydrophobic InteractionsResidues Involved in Aromatic InteractionsInteractions with HEM601
Comp.DNA TopoIV
1S16
CYP51 of C. albicans
5V5Z
1−3.69−7.54-Tyr118, Thr311, Phe380, Met508, Hem601-Hydrophobic
2−3.18−9.70Tyr132Tyr118, Leu300, Ile304, Thr311, Hem601Tyr118Hydrophobic
3−3.25−12.34Tyr132Tyr118,Ile131, Ile304, Hem601Hem601Hydrophobic, aromatic, Fe binding
4−2.66−8.70-Tyr118, Leu300, Thr311, Leu376, Phe380, Met508, Hem601Tyr118, Hem601Hydrophobic, aromatic
5−4.25−9.34Tyr118Tyr118, Leu376, Met508, Hem601Tyr118Hydrophobic
6−3.14−9.65Tyr64Tyr118, Tyr122, Thr311, Leu376, Phe380, Hem601Tyr118Hydrophobic
7−4.17−9.82Tyr132Tyr118, Leu121, Thr311, Phe380, Met508, Hem601Hem601Hydrophobic, aromatic
8−2.59−7.11-Tyr118, Leu376, Met508, Hem601-Hydrophobic
9−4.39−9.54Tyr132Tyr118, Phe380, Met508, Hem601-Hydrophobic
10−2.67−10.11Tyr64Tyr118, Leu300, Thr311, Met508, Hem601Tyr118, Hem601Hydrophobic, aromatic
11−2.73−9.31Tyr118Leu300, Met508, Hem601Hem601Hydrophobic, aromatic
12−4.37−8.62-Tyr118, Tyr122, Thr311, Met508, Hem601Tyr122, Hem601Hydrophobic, aromatic
13−3.56−8.81-Tyr118, Tyr122, Thr311, Leu376, Phe380, Hem601Tyr118Hydrophobic
14−3.28−7.64-Tyr118, Thr311, Leu376, Met508, Hem601-Hydrophobic
15−3.10−12.95Thr311Ile131, Leu300, Ile304, Hem601Hem601Hydrophobic, aromatic, Fe binding
16−2.67−10.26Tyr132Tyr118, Tyr122, Leu300, Ile304, Hem601Hem601Hydrophobic, aromatic
17−2.57−7.96-Tyr118, Tyr122, Thr311, Leu376, Met508, Hem601Tyr118, Hem601Hydrophobic, aromatic
Ketoconazole-−8.23Tyr64Tyr118, Ile131, Tyr132, Leu300, Ile304, Leu376, Met508, Hem601Hem601Hydrophobic, aromatic
Table 6. Drug-likeness predictions of tested compounds.
Table 6. Drug-likeness predictions of tested compounds.
Comp.MW a
(≤500)
HBAs b
(≤10)
HBDs c
(≤5)
Lipophilicity
(ClogP) d
(≤5)
RBs e
(≤10)
TPSA f
(≤140)
Lipinski ViolationsBioavailability
Score
Drug-Likeness Score
1432.51523.309157.0900.55−0.14
2438.48523.455157.0900.11−0.64
3410.47423.464140.0200.55−0.78
4410.47423.474140.0200.55−0.81
5445.56512.867132.2600.55+0.24
6403.48512.484132.2600.55−0.38
7398.46413.285132.9300.55−0.69
8436.48623.316157.0900.55−0.70
9422.45622.997157.0900.11−0.37
10428.46523.784140.0200.55−0.77
11376.41522.115157.0900.55−0.71
12418.49523.026157.0900.11−0.24
13442.48514.155129.0200.55−0.95
14408.42622.656157.0900.11−0.40
15350.39413.023119.7900.55−0.15
16421.47612.794132.2600.55−0.14
17390.43522.356157.0900.11−0.59
a Molecular weight; b number of hydrogen bond acceptors; c number of hydrogen bond donors; d consensus logarithm of partition coefficient (octanol/water) predicted as an average of five methods (iLOGP, XLOGP3, WLOGP, MLOGP, SILICOS-IT); e rotatable bonds; f topological polar surface area (Å).
Table 7. Absorption and distribution properties for the examined compounds 117.
Table 7. Absorption and distribution properties for the examined compounds 117.
NoAbsorption Distribution
Peff aMDCK bSw cSpH dFaSSGF eFaSSIF fFeSSIF gAbsorption
Risk
BBB hLogBBB ifu% jVd kRBP l
11.64218.1882310.0090.0090.0670.1600.000Low
(59%)
−1.2314.5880.3280.661
22.232112.63351.2950.0080.0400.1151.000Low
(66%)
−0.7934.4460.2440.692
32.491160.41520.0020.0260.0100.1551.000High (89%) −0.0714.5480.6910.738
42.439192.62420.0020.0260.0080.1111.000High (88%)−0.1274.4490.7420.737
51.979244.487300.0490.9140.0310.0890.000High
(99%)
−0.0069.9251.2320.748
62.608289.45680.0080.1240.0710.1970.329High (99%)−0.1309.6311.0970.767
72.846695.38220.0020.0130.0130.1591.000High (94%)−0.3935.2330.7800.744
81.71320.994286.9040.0090.0350.3810.000Low
(84%)
−0.9645.5250.3250.680
91.82411.5243714.5680.0140.0900.2070.000Low
(66%)
−1.1515.4910.3280.677
102.552238.50520.0020.0240.0050.1041.000High (77%)−0.0464.7220.7460.741
111.55718.324377.4290.0170.0920.3800.000Low
(74%)
−1.0475.9540.3130.684
121.62416.857297.1790.0100.0510.3080.000Low
(84%)
−1.0565.4020.3270.677
133.755512.70010.0010.0180.0060.0851.918High (96%)0.1294.7680.8610.731
141.86439.8383813.6990.0140.1780.3220.000Low
(90%)
−0.9975.9630.3130.683
153.536572.26240.0040.0990.0180.2111.000High (99%)0.1379.9100.7790.783
163.229352.37780.0080.1220.0610.1690.370High
(99%)
−0.03710.2361.1410.769
171.65229.5833813.7310.0150.2360.2600.000Low
(84%)
−1.0855.8380.3140.680
a Human jejunal effective permeability (104 cm/s); b MDCK Transwell permeability (107 cm/s); c water solubility (μg/mL); d water solubility in mg/mL at pH 7.4; e solubility expressed in mg/mL in fast state simulated gastric fluid; f solubility expressed in mg/mL in fast state simulated intestinal fluid; g solubility expressed in mg/mL in fed state simulated intestinal fluid; h possibility to penetrate blood–brain barrier; i logarithm of the brain/blood partition coefficient; j human %fraction unbound; k human volume of distribution in steady state (L/kg); l blood-to-plasma ratio in humans.
Table 8. Studies on the metabolism characteristics of compounds 117.
Table 8. Studies on the metabolism characteristics of compounds 117.
NoMetabolism
CYP
1A2
Inh.
CYP1A2
Substr.
Km
CYP
2C9
Inh
CYP2C9
Substr.
Km
CYP
2C19
Inh.
CYP2C19
Substr.
Km
CYP
2D6
Inh.
CYP2D6
Substr.
Km
CYP
3A4
Inh.
CYP3A4
Substr.
Km
CYP
CLint a
HEP
CLint b
UGTs
Subs. c
CYP
Risk
1NoNon
Subs.
No86.146No24.703NoNon
Subs.
NoNon
Subs.
6.56811.3621A3, 1A90.000
2No23.682No8.127No39.264NoNon
Subs.
NoNon
Subs.
100.39816.3591A8, 1A9, 1A102.000
3No30.916No14.403No29.495No127.051YesNon
Subs.
65.01371.1461A1, 1A8, 1A9, 1A10, 2B152.372
4No29.485No8.423No93.004No204.398YesNon
Subs.
80.36589.5571A1, 1A8, 1A9, 1A10, 2B152.985
5No18.129No83.216No20.465No13.006Yes22.76778.71023.7391A4, 1A91.147
6No7.486No87.569No44.972No28.012Yes31.235101.60318.418Non
Subs.
1.000
7No26.192No15.906No4.882No38.409Yes21.94389.26121.8571A90.981
8NoNon
Subs.
No20.918No203.738NoNon
Subs.
NoNon
Subs.
19.3587.0881A3, 1A8, 1A9, 1A100.935
9NoNon
Subs.
No123.319No16.370NoNon
Subs.
YesNon
Subs.
6.8688.3001A3, 1A90.000
10No34.151No7.744No94.454No255.204YesNon
Subs.
131.92193.6391A1, 1A8, 1A9, 1A10, 2B153.000
11NoNon
Subs.
No36.930No220.331NoNon
Subs.
NoNon
Subs.
9.9185.0801A3, 1A8, 1A90.000
12NoNon
Subs.
No25.953No199.456NoNon
Subs.
NoNon
Subs.
12.3486.6891A3, 1A8, 1A90.234
13No3.514No15.364No66.722No27.176Yes11.550259.95580.5541A1, 1A93.228
14NoNon
Subs.
No96.500No5.165NoNon
Subs.
YesNon
Subs.
13.2866.3701A3, 1A90.000
15No20.385No167.240No60.458No91.889Yes130.32457.43021.7821A1, 1A90.740
16No9.212No64.353No48.228No32.873Yes24.123102.34118.5061A8, 1A91.055
17NoNon
Subs.
No100.364No4.815NoNon
Subs.
YesNon
Subs.
10.2765.9661A3, 1A90.000
a A total intrinsic human cytochrome P-450 mediated clearance based on the sum of CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 individual clearances; b human hepatocytes intrinsic clearance (μL/min/106 cells); c potential isoenzyme substrates of UDP-glucuronosyltransferases (UGTs).
Table 9. Studies on selected transporters for the examined compounds 117.
Table 9. Studies on selected transporters for the examined compounds 117.
NoPgp
Sub.
Pgp
Inh.
BCRP
Sub.
BCRP
Inh.
BSEP
Inh.
1YesNo (49%)YesNoNo
2YesYes (62%)YesNoNo
3YesYes (97%)YesNoNo
4YesYes (97%)YesNoNo
5YesYes (88%)YesNoNo
6YesYes (60%)YesNoNo
7YesYes (97%)YesNoNo
8YesNo (93%)YesNoNo
9YesNo (49%)YesNoNo
10YesYes (97%)YesNoNo
11YesNo (93%)YesNoNo
12YesNo (93%)NoNoNo
13YesYes (97%)YesNoNo
14YesNo (68%)YesNoNo
15YesNo (46%)YesNoNo
16YesYes (57%)YesNoNo
17YesNo (78%)YesNoNo
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Petrou, A.; Geronikaki, A.; Kartsev, V.; Kousaxidis, A.; Papadimitriou-Tsantarliotou, A.; Kostic, M.; Ivanov, M.; Sokovic, M.; Nicolaou, I.; Vizirianakis, I.S. N-Derivatives of (Z)-Methyl 3-(4-Oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylates as Antimicrobial Agents—In Silico and In Vitro Evaluation. Pharmaceuticals 2023, 16, 131. https://doi.org/10.3390/ph16010131

AMA Style

Petrou A, Geronikaki A, Kartsev V, Kousaxidis A, Papadimitriou-Tsantarliotou A, Kostic M, Ivanov M, Sokovic M, Nicolaou I, Vizirianakis IS. N-Derivatives of (Z)-Methyl 3-(4-Oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylates as Antimicrobial Agents—In Silico and In Vitro Evaluation. Pharmaceuticals. 2023; 16(1):131. https://doi.org/10.3390/ph16010131

Chicago/Turabian Style

Petrou, Anthi, Athina Geronikaki, Victor Kartsev, Antonios Kousaxidis, Aliki Papadimitriou-Tsantarliotou, Marina Kostic, Marija Ivanov, Marina Sokovic, Ioannis Nicolaou, and Ioannis S. Vizirianakis. 2023. "N-Derivatives of (Z)-Methyl 3-(4-Oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylates as Antimicrobial Agents—In Silico and In Vitro Evaluation" Pharmaceuticals 16, no. 1: 131. https://doi.org/10.3390/ph16010131

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop