Next Article in Journal
Current Management and Future Perspectives in the Treatment of Lp(a) with a Focus on the Prevention of Cardiovascular Diseases
Previous Article in Journal
Curcumin Stereoisomer, Cis-Trans Curcumin, as a Novel Ligand to A1 and A3 Adenosine Receptors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Indole-Acrylonitrile Derivatives as Potential Antitumor and Antimicrobial Agents—Synthesis, In Vitro and In Silico Studies

1
Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdansk, Poland
2
Department of Oral Microbiology, Medical Faculty, Medical University of Gdansk, 80-204 Gdansk, Poland
3
Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznań, Poland
4
Department of Inorganic Chemistry, Faculty of Chemistry and Advanced Materials Centers, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(7), 918; https://doi.org/10.3390/ph16070918
Submission received: 10 May 2023 / Revised: 16 June 2023 / Accepted: 20 June 2023 / Published: 22 June 2023
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
A series of 2-(1H-indol-2-yl)-3-acrylonitrile derivatives, 2ax, 3, 4ab, 5ad, 6ab, and 7, were synthesized as potential antitumor and antimicrobial agents. The structures of the prepared compounds were evaluated based on elemental analysis, IR, 1H- and 13NMR, as well as MS spectra. X-ray crystal analysis of the representative 2-(1H-indol-2-yl)-3-acrylonitrile 2l showed that the acrylonitrile double bond was Z-configured. All compounds were screened at the National Cancer Institute (USA) for their activities against a panel of approximately 60 human tumor cell lines and the relationship between structure and in vitro antitumor activity is discussed. Compounds of interest 2l and 5ad showed significant growth inhibition potency against various tumor cell lines with the mean midpoint GI50 values of all tests in the range of 0.38–7.91 μM. The prominent compound with remarkable activity (GI50 = 0.0244–5.06 μM) and high potency (TGI = 0.0866–0.938 μM) against some cell lines of leukemia (HL-60(TB)), non-small cell lung cancer (NCI-H522), colon cancer (COLO 205), CNS cancer (SF-539, SNB-75), ovarian cancer ((OVCAR-3), renal cancer (A498, RXF 393), and breast cancer (MDA-MB-468) was 3-[4-(dimethylamino)phenyl]-2-(1-methyl-1H-indol-2-yl)acrylonitrile (5c). Moreover, the selected 2-(1H-indol-2-yl)-3-acrylonitriles 2ac and 2ex were evaluated for their antibacterial and antifungal activities against Gram-positive and Gram-negative pathogens as well as Candida albicans. Among them, 2-(1H-indol-2-yl)-3-(1H-pyrrol-2-yl)acrylonitrile (2x) showed the most potent antimicrobial activity and therefore it can be considered as a lead structure for further development of antimicrobial agents. Finally, molecular docking studies as well as drug-likeness and ADME profile prediction were carried out.

Graphical Abstract

1. Introduction

Since the indole motif is a key building block of pharmacologically active natural as well as synthetic molecules, there has been increasing interest in the synthesis and biological research of indole derivatives [1,2,3,4,5]. The importance of the indole skeleton to date has led to the development of diverse bioactive compounds that have been identified with anti-inflammatory [6,7], antioxidant and cytoprotective [8,9], antidepressant [10], anti-migraine [11], antihypertensive [12], antidiabetic [13,14] or antiviral [15] effects and as antitubercular agents [16]. Moreover, indole derivatives represent a significant source of novel antibacterial agents that may exhibit their biological activity through the inhibition of efflux pumps, biofilm or filamentous temperature-sensitive protein Z (FtsZ), and methicillin-resistant Staphylococcus aureus pyruvate kinase [17,18,19,20,21,22].
Particular attention has been paid to indole-containing compounds with anticancer properties, which exert their activity by affecting numerous biological targets [23,24,25]. For example, the tubulin inhibitors vincristine and vinblastine isolated from Catharanthus roseus are used in the treatment of various cancers [26], whereas dacomitinib is a well-known antitumor agent that blocks the activity of epidermal growth factor receptor (EGFR) [27]. Furthermore, indole derivatives have been identified as potent myeloid cell leukemia-1 (Mcl-1) [28] or Pim kinase [29] inhibitors. Antiproliferative effects of the indole-based compounds may also result from the selective inhibition of silent information regular sirtuin 1 (SIRT1) protein, a highly conserved NAD+-dependent deacetylase belonging to the sirtuin family [30], as well as σ2 receptors, the expression of which is increased in tumor cells with respect to quiescent cells. The σ2 receptor ligands can rapidly internalize into tumor cells and induce apoptosis through multiple pathways [31]. On the other hand, an indole-derived histone deacetylase (HDAC) inhibitor, panobinostat, has been approved for the treatment of multiple myeloma [32]. Recently, some indole derivatives of ursolic acid have been described as promising DNA topoisomerase II inhibitors with anticancer activity [33].
In addition, 2,3-disubstituted acrylonitriles containing a heteroaromatic core at position 2 of the acrylonitrile moiety have also gained much interest due to their versatile biological activities [34]. These compounds have been shown to possess anti-inflammatory [35], antioxidant [36], antihyperglycemic [37], antiviral [38,39], antimalarial [40] and antimycobacterial [41] properties as well as the ability to inhibit acetylcholinesterase (AChE) activity [42,43]. More recently, some benzazole acrylonitrile-based compounds I (Figure 1) were found to be active against both Escherichia coli and Pseudomonas aeruginosa. The antibacterial activity of these compounds is related to the inhibition of penicillin-binding protein (PBP) and/or β-lactamase enzyme [44].
Otherwise, several studies have shown the anticancer potential of heteroaryl-acrylonitriles [34]. For example, some acrylonitriles bearing N-substituted benzimidazole II (Figure 1) [45,46], benzotriazole III (Figure 1) [47], or triazolo [4,5-b]pyridine IV (Figure 1) [48] fragment are able to exert antiproliferative effects against tumor cell lines by interacting with tubulin in the colchicine-binding site. Other heteroaryl-acrylonitrile-based compounds such as 3-aryl-2-(benzimidazol-2-yl)acrylonitriles V (Figure 1) have shown significant interaction with ct-DNA, supporting the fact that their antitumor properties could be the consequence of DNA binding [49]. Meanwhile, some of the compounds obtained in our laboratory, such as 3-aryl-2-(1H-benzimidazol-2-yl)acrylonitriles, have been identified as potential caspase-9 activators, possessing cytotoxic activity against human cancer cell lines. The most active compound, 2-(benzimidazol-2-yl)-3-(5-nitrothiophen-2-yl)acrylonitrile (VI), was on average 10- and 3-fold more potent than cisplatin and etoposide, respectively, in inhibiting cancer cell growth [50]. In turn, the antiproliferative activity of N-alkylindole-substituted 2-(pyrid-3-yl)-acrylonitrile VII is probably due to the inhibition of EGFR and VEGFR-2 tyrosine kinases [51], while 2-(indol-3-yl)acrylonitrile (paprotrain) and its analogs have been reported as kinesin-like protein 2 (MKLP-2) inhibitors, which could be valuable tools to treat tumors overexpressing MKLP-2 [52,53].
Despite the importance of indole [23] and acrylonitrile [34] scaffolds in the design and discovery of new anticancer agents, indoles substituted at position 2 with the acrylonitrile group have remained unexplored for their biological activities.
In this context, and in connection with a research program on the chemistry and biological activities of 3-aryl-2-heteroaryl-acrylonitriles undertaken in our laboratory years ago [50,54], we considered that 2-(1H-indol-2-yl)acrylonitriles of type A (Figure 1) may act as potential anticancer agents. These compounds were evaluated for their antimicrobial activity against selected microbial species. In addition, to investigate the importance of the acrylonitrile moiety on biological activity, analogs lacking double bonds were prepared. Furthermore, molecular docking techniques were carried out to rationalize the possible mechanism of action of the most active compounds.

2. Results and Discussion

2.1. Chemistry

All 2-indolyl-3-acrylonitriles 2ax were synthesized by the Knoevenagel condensation of 2-(1H-indol-2-yl)acetonitrile (1) [55] with the appropriate aromatic and heteroaromatic aldehydes. It is worth noting that despite various procedures [56], the Knoevenagel reaction is one of the most useful approaches for the preparation of heteroaryl-acrylonitriles [34,57,58].
As described in Scheme 1, 2-(1H-indol-2-yl)acrylonitriles 2as were prepared upon treatment of a methanolic solution of 2-(1H-indol-2-yl)acetonitrile (1) and aldehydes with sodium methoxide solution at ambient temperature (method A). In turn, 2-(1H-indol-2-yl)-3-acrylonitriles 2tx were synthesized by reacting acetonitrile 1 with the corresponding aldehydes in ethanol at ambient temperature in the presence of triethylamine as a catalyst (method B).
It should be mentioned that 2-(1H-indol-2-yl)-3-acrylonitriles 2a [59,60,61,62,63] and 2b [61,62,63] have been previously reported. However, there have been no reports of their biological activities.
Compound 1 was then subjected to reaction with p-nitrosodimethylaniline in anhydrous methanol in the presence of sodium methoxide to yield the desired iminoacetonitrile 3 (Scheme 1).
The identities of the prepared compounds 2ax and 3 were confirmed by elemental analysis (C, H, N) as well as spectroscopic data (IR, NMR, and MS) presented in the experimental section (see Section 3).
It is worth noting that regular NMR methods could not establish the configuration of the acrylonitrile double bond. Therefore, X-ray crystallography was performed on representative 2-(1H-indol-2-yl)acrylonitrile 2l. As shown in Figure 2, in crystal form, compound 2l adopted a flattened conformation with a Z configuration at the C10–C11 double bond.
To determine whether readily available compounds lacking the acrylonitrile double bond would retain any activity of the parent compounds 2d, 2l, and 2p, the 2-(1H-indol-2-yl)-3-phenylpropanenitriles 4ac were synthesized by selective hydrogenation of the olefinic bond using NaBH4 in DMF at ambient temperature, according to Scheme 2.
The structures of the compounds 4ac were confirmed by elemental analysis, IR, NMR, as well as MS spectroscopic data presented in Section 3.
Recently, it has been reported that the introduction of an alkyl or aryl group at position N1 of benzimidazole-derived acrylonitriles can result in promising antiproliferative agents [45,46]. Therefore, to explore the structure–activity relationships of the synthesized 2-(1H-indol-2-yl)-3-acrylonitriles in more detail, we turned our attention to their N-substituted analogs.
As depicted in Scheme 3, the reaction procedure leading to the target 1-methyl derivatives 5ad consisted of the reaction of the corresponding indole-acrylonitrile sodium salts, generated by the treatment of 2-(1H-indol-2-yl)-3-acrylonitriles 2b, 2d, 2l, and 2p with sodium hydride in anhydrous dimethylformamide, with methyl iodide at ambient temperature.
In a similar manner, reaction of 2-(1H-indol-2-yl)-3-acrylonitriles 2d and 2l with acetyl chloride gave rise to the formation of the corresponding 1-acetyl products 6a and 6b, respectively (Scheme 3).
Finally, from reaction of the sodium salt of 2-(1H-indol-2-yl)-3-acrylonitrile 2d with methanesulfonyl chloride, 1-mesyl derivative 7 was isolated (Scheme 3).
The structures of the newly prepared N-substituted 2-(1H-indol-2-yl)-3-acrylonitriles 5ad, 6ab, and 7 were confirmed by elemental analysis and spectroscopic methods (Section 3).

2.2. Biological Evaluation

2.2.1. In Vitro Anticancer Activity

Evaluation of anticancer activity was performed at the National Cancer Institute (NCI, Bethesda, MD, USA), following the known in vitro disease-oriented antitumor screening program against a panel of approximately 60 human cancer cell lines derived from nine cancer types (leukemia, lung, colon, CNS, melanoma, ovarian, renal, prostate, and breast) [64,65,66,67].
Firstly, indole-acrylonitrile derivatives 2ax, 3, 4ac, 5ad, 6ab, and 7 were subjected to preliminary screening at a single concentration of 10 μM in approximately 60 cell lines within nine tumor type subpanels. Results for each compounds were reported as a mean graph of the percent growth (%GP) of the treated cells relative to the no-drug control. According to the data analysis of the one-dose mean graphs, it is clear that low mean growth values represented better inhibition activity (%GI = 100 − GP) (values between 0 and 100), while negative values corresponded to lethal activity (values less than 0) (Table 1).
The following can be noted with respect to the mean growth data presented in Table 1 for the tested compounds.
In the series of indole-acrylonitriles 2ap with aromatic substituent R at position 3 of the acrylonitrile moiety, the substituents that gave indisputable anticancer activity were 4-methoxyphenyl (compound 2d), 4-(dimethylamino)phenyl (compound 2l), and 2-naphthyl (compound 2p).
The incorporation of the heteroaromatic ring as substituent R at position 3 of the acrylonitrile moiety afforded compounds 2qx with weak activity.
Replacement of the acrylonitrile moiety by imino-acetonitrile led to a dramatic decrease in activity (comparing compounds 2l and 3). Additionally, hydrogenation of the acrylonitrile double bond was associated with very poor activity (compounds 4ac).
The introduction of a methyl substituent (5ad) into position 1 of the indole ring of the parent structure led to compounds with greater or equivalent activity compared with their acrylonitrile counterparts 2b, 2d, 2l, and 2p. Interestingly, the introduction of an acetyl group into position 1 of the indole ring of the acrylonitrile analogs had a more varied effect on activity than the introduction of a methyl group (comparing compounds 2b with 6a, and 2l with 6b). Moreover, a decrease in activity was observed when a methylsulfonyl group was attached to the nitrogen atom of the indole scaffold (comparing compounds 2l and 7).
From the pattern of the mean growth graph it was apparent that compounds 2l and 5ad exerted significant growth inhibition against various cancer cell lines representing different cancer types. Therefore, these compounds were selected by NCI for a secondary screening at five concentration levels (0.01–100 μM).
Data for the selected indole-acrylonitrile derivatives 2l and 5ad are recorded in Table 2 and Table 3 and Figure 3. The antitumor activity of the tested compounds is reported for each cell line by GI50 value (GI50 = molar concentration of the compound that inhibits 50% net cell growth) and TGI value (TGI = molar concentration of the compound leading to total inhibition). Furthermore, a mean graph midpoint (MG-MID) is depicted for the GI50 parameter, giving the averaged activity parameter over all cell lines.
As shown in Table 2, indole-acrylonitrile 2l bearing 4-(dimethylamino)phenyl as substituent R was characterized by relatively high inhibitory activity, with GI50 values in the range of 0.228–6.6 μM. In addition, derivative 2l was found to exert a significant cytostatic effect against some cell lines of leukemia (CCRF-CEM, RPMI-8226), lung cancer (HOP-62, HOP-92), colon cancer (COLO 205, HT29), CNS cancer (SF-295, SNB-75, U251), and melanoma (MDA-MB-435), with TGI values between 0.641 and 18.6 μM. In regard to the effect in the entire panel of tumor cell lines, compound 2l demonstrated pronounced antiproliferative activity with GI50 MG-MID values ranging from 0.66 to 2.28 μM, especially against leukemia cells (GI50 MG-MID = 0.66 μM) (Table 3, Figure 3).
As expected, N-methyl-substituted analogue 5c showed improved antitumor properties in comparison with its indole-acrylonitrile counterpart 2l (GI50 = 0.0193–5.06 μM vs. 0.228–6.6 μM) (Table 2). In addition, compound 5c exhibited remarkable cytostatic activity at low TGI level < 5.3 μM towards 12 cancer cell lines, being particularly effective against 9 various cell lines out of 7 subpanels with TGI values in the submicromolar range of 0.0866–0.938 μM. It was observed that non-small cell lung cancer cell line NCI-H522 was the most susceptible cell line with TGI = 0.0866 μM (Table 2). On the other hand, the highest overall sensitivity to this compound was found for the subpanels of leukemia and colon cancer cell lines, for which GI50 MG-MID values were 0.17 and 0.15 μM, respectively (Table 3, Figure 3). Notably, compared with its counterpart 2l, derivative 5c was 3–9-fold more potent in individual subpanels of cancer cell lines (GI50 MG-MID = 0.15–0.56 μM vs. GI50 MG-MID = 0.66–2.28 μM) (Table 3, Figure 3).
Moreover, N-methyl-substituted compound 5b with 4-methoxyphenyl as substituent R also demonstrated significant antitumor activity against all of the tested cell lines, with GI50 values ranging from 0.153 to 6.96 μM (Table 2). It is worth noting that derivative 5b acted as a potent inhibitor against 18 human tumor cell lines of 8 subpanels, with calculated TGI values in the range of 0.403–21.3 μM. Submicromolar TGI values were found for HL-60(TB) and SR leukemia (TGI = 0.582 and 0.94 μM, respectively), NCI-H522 non-small cell lung cancer (TGI = 0.554 μM), SNB-75 CNS cancer (TGI = 0.514 μM), MDM-MB-435 melanoma (TGI = 0.403 μM), OVCAR-3 ovarian cancer (TGI = 0.955 μM), A498 and RXF 393 renal cancer (TGI = 0.867 and 0.751 μM, respectively), and MDA-MB-468 breast cancer (TGI = 0.826 μM) cell lines (Table 2). Nevertheless, concerning overall activity, it should be noted that compound 5b proved to be slightly less potent than derivative 5c (GI50 MG-MID = 0.29–1.17 μM vs. 0.15–0.56 μM) (Table 3, Figure 3). A further decrease in potency was observed when 4-(dimethylamino)phenyl as substituent R in compound 5c was replaced with 4-methylphenyl moiety in analogue 5a (GI50 = 0.0193–5.06 μM vs.0.0726–100 μM and GI50 MG-MID = 0.15–0.56 μM vs. 0.35–16.98 μM) (Table 2 and Table 3, Figure 3). However, the latter derivative still retained pronounced growth inhibitory properties especially against certain cell lines, including HL-60(TB) leukemia, NCI-H522 non-small cell lung cancer, SF-295 and SF-539 CNS cancer, MDA-MB-435 melanoma, OVCAR-3 ovarian cancer, A498 renal cancer, and BT-549 breast cancer, with TGI values in the range of 0.254–7.12 μM (Table 2).
Another modification of 5c, consisting of replacement of 4-(dimethylamino)phenyl as substituent R at position 3 of the acrylonitrile moiety with a 2-naphthyl group, resulted in compound 5d, which generally exhibited higher activity than 5c against cell lines from the non-small cell lung cancer and CNS cancer subpanels (GI50 MG-MID = 0.24 and 0.11 μM vs. 0.89 and 0.47 μM, respectively) (Table 3, Figure 3). In addition, compound 5d acted selectively as a potent inhibitor against HL-60(TB) leukemia (GI50 = 0.179 μM, TGI = 0.703 μM), SF-539 CNS cancer (GI50 = 0.0613 μM, TGI = 0.552 μM), MDA-MB-435 melanoma (GI50 = 0.0318 μM, TGI = 0.103 μM), and A498 renal cancer (GI50 = 0.03 μM, TGI = 0.418 μM) cell lines (Table 2). On the other hand, taking into account overall potency, compound 5d proved to be the least active in this series (full panel GI50 MG-MID > 7.91 μM) (Table 3).
From these results it was concluded that the combination of the N-methyl group in the indole ring with the 4-dimethylaminophenyl group at position 3 of the acrylonitrile moiety resulted in derivative 5c with optimal properties (full panel GI50 MD-MIG = 0.38 μM vs. 0.60–7.91 μM) (Table 3).

2.2.2. Antimicrobial Activity against Reference Microbial Strains

The synthesized indole-acrylonitriles 2ac and 2ex were evaluated for their in vitro antimicrobial activity against Gram-positive bacteria (MSSA Staphylococcus aureus ATCC 6538, American Type Culture Collection, USA, Staphylococcus epidermis PMC 2118, Polish Collection of Microorganisms, Poland, Enterococcus faecalis ATCC 11420), Gram-negative bacteria (Escherichia coli ATCC 11229, Pseudomonas aeruginosa ATCC 15442), as well as a fungal species (Candida albicans ATCC 10231). The tests were performed using a serial dilution method, allowing the determination of the minimal concentration inhibiting bacterial growth (MIC), minimal bactericidal concentration (MBC), and minimal fungicidal concentration (MFC). The obtained results are presented in Table 4.
Most of the investigated compounds showed no or negligible antimicrobial activity; their MIC, MBC, and MFC values were between ≥128 and ≥256 μg/mL. On the other hand, in the series of indole-acrylonitriles 2ap with aromatic substituent R in position 3 of the acrylonitrile moiety, 3-chlorophenyl derivative 2i was characterized by relatively high antibacterial activity against Gram-positive bacteria S. aureus ATCC 6538 and S. epidermis PMC 2118, with MIC and MBC values ranging from 8 to 16 μg/mL (Table 4). Interestingly, replacement of the 3-chlorophenyl group in 2i for either 2-chlorophenyl (compound 2h) or 4-chlorophenyl (compound 2j) resulted in a severe reduction in activity (MIC = 128–256 μg/mL, MBC/MFC ≥ 256 μg/mL, Table 4). In turn, indole-acrylonitrile 2n bearing 4-nitrophenyl as substituent R exhibited moderate bacteriostatic activity against S. aureus ATCC 6538 (MIC = 64 μg/mL), while its bactericidal effect against this strain was weak (MBC = 128 μg/mL, Table 4).
In the series of heteroaromatic analogs 2qx, promising antimicrobial activity against some bacterial species was demonstrated by compounds 2q, 2s, and 2x (Table 4). Thus, derivative 2q containing pyridine as substituent R in position 3 of the acrylonitrile moiety was found to have pronounced potential against two bacterial Gram-positive strains: S. aureus ATCC 6538 and S. epidermis PMC 2118. The MIC and MBC values obtained for this compound against both strains were 8 and 16 μg/mL, respectively (Table 4). Changing the pyridine ring of 2q to a thiazole (compound 2s) led to a slight decrease in activity against the same bacterial strains (MIC = 16–32 μg/mL, MBC = 32 μg/mL) (Table 4). Otherwise, thiazole derivative 2s displayed significant antifungal activity against C. albicans with an MIC value of 16 μg/mL (Table 4). However, the highest activity among all the tested compounds was exhibited by 3-pyrole derivative 2x. Thus, compound 2x displayed relatively high antibacterial potency against all Gram-positive bacteria tested, with MIC values in the range of 8–32 μg/mL and MBC values of 32 μg/mL. Furthermore, it was the only compound that was found to be effective against Gram-negative bacteria, presenting antibacterial activity against E. coli with MIC and MBC values of 32 μg/mL. In addition, 3-pyrole derivative 2x was characterized by great antifungal activity against C. albicans, with MIC and MFC values of 4 and 8 μg/mL, respectively (Table 4).
It is notable that despite testing a large number of compounds, it was not possible to derive a relationship between structure and activity in the studied series of indole-acrylonitrile derivatives 2ac and 2ex. On the other hand, compounds 2i, 2q, 2s, and 2x with interesting antimicrobial activity did not exhibit antitumor effects against the tested cancer cell lines.

2.2.3. Antibacterial Activity against Clinical Staphylococcus Aureus Strains

The most pronounced compounds 2i, 2q, 2s, and 2x were further evaluated for their bacteriostatic and bactericidal activities against a panel of clinical isolates of Staphylococcus aureus (79, 124, 128, 143, 177, 220 and 244) derived from various human infections. The MIC and MBC values of the tested compounds are shown in Table 5.
As revealed by the data in Table 5, indole-acrylonitriles 2i, 2q, and 2s were inactive against the tested Staphylococcus aureus strains isolated from clinical specimens; their MIC and MBC values were between >128 and >256 μg/mL. On the other hand, satisfactory MIC and MBC values were obtained for compound 2x. It was shown that this compound had ability to inhibit the growth of the clinical isolates at a low concentration of 16 μg/mL. In addition, indole-acrylonitrile 2x was characterized by relatively strong or moderate bactericidal activity, with MBC values of 32 and 64 μg/mL.
Further studies also indicated relatively high and moderate bactericidal activities of compound 2x against both clinical methicillin-resistant and -sensitive Staphylococcus aureus strains (MRSA 1–5, MSSA 6–10), with MIC and MBC values of 16 and 64 μg/mL, respectively (Table 6). It should be noted that both MRSA and MSSA strains exhibited similar sensitivity to the tested compound 2x, while MRSA strains are generally more resistant to antibiotics and antimicrobial compounds than MSSA strains [68].

2.3. Docking Studies

2.3.1. Docking to Anticancer Targets

In order to rationalize the experimentally assessed antiproliferative properties of the synthesized compounds against cancer cell lines, computational analysis was undertaken. As mentioned above, heteroaryl-acrylonitriles can exhibit antiproliferative effects by inhibiting tubulin polymerization due to their ability to bind to the colchicine-binding site [45,46,47,48]. Although some synthetic compounds that bind to the colchicine site have been evaluated in clinical trials [69], none have been approved for cancer therapy to date. Therefore, this binding site still offers challenging opportunities for drug development [70].
The caspase signaling pathway has also generated considerable attention as a promising cancer therapeutic strategy [71]. Previously our research group identified a series of 3-aryl-2-(1H-benzimidazol-2-yl)acrylonitriles as potential caspase-3 and -9 activators with cancer cell growth inhibitory properties [50]. On the basis of these results, we considered the induction of the activity of apoptotic enzymes such as caspase-3 and -9.
With the above in mind, molecular docking studies of the obtained series of compounds were performed in the binding pockets of the following proteins: caspase-3, caspase-9, and tubulin (PDB codes: 2xyp [72], 2ar9 [73], and 5eyp [74], respectively).
From the obtained FRED Chemgauss4 scores (Table S1, Supplementary Materials), it was concluded that most of the proposed ligands exhibited significant affinity to caspase-3, including the active derivatives 2l, 5c, and 5d (Chemgauss4 scores ranging from −5.32 to −6.09 for the top ranked poses). Furthermore, the active compounds 2l and 5d were ranked relatively high for the caspase-9 binding pocket (Chemgauss4 scores of −3.57 and −3.05, respectively). On the other hand, the potent analogs 5ac as well as 5d were characterized by relatively high affinity for the tubulin binding pocket (Chemgauss4 scores ranging from −11.53 to −13.30).
The highest ranked poses of the most potent compound 5c docked in the target proteins are presented in Figure 4, while 2D diagrams of the interactions of the active derivatives 2l and 5ad can be found in the Supplementary Materials (Tables S2–S4).
As shown in Figure 4A, in the binding site of caspase-3, the aromatic rings of ligand 5c made hydrophobic contacts with the Met61 and Cys163 residues present in the p17 subunit as well as Arg207 in the p12 subunit. Side chains of His121 and Tyr204 from p17 and p12, respectively, formed π-π stacking interactions with the ligand core. Van der Waals forces were created with Thr64, Gly122, Glu123, and Phe128 in p17 as well as Tyr204, Ser205, Trp206, and Arg207 in p12.
In the case of caspase-9 (Figure 4B), van der Waals interactions were formed between ligand 5c and Thr181, Asp356, Trp362, Gly395, Ile396, and Tyr397. The NH group from the main chain of Arg355 formed a hydrogen bond with a length of 2.6 A with a nitrile nitrogen atom of 5c. Hydrophobic contacts between the ligand and Trp354 as well as Pro357 were also found.
According to Figure 4C, ligand 5c created van der Waals interactions with Gln11, Asn101, Ser178, Thr179, Tyr224, and Asn249 from α-tubulin as well as Leu248, Ala250, Lys254, Leu255, Asn258, Thr314, Val315, and Asn350 from β-tubulin in the colchicine-binding site of tubulin. Hydrophobic contacts were formed with Ala180 and Val181 from α-tubulin as well as Met259, Ala316, and Lys352 from β-tubulin. Glu183 from α-tubulin interacted via anion-π contact.
Based on the above results, it was concluded that the most important features of the pharmacophore were the two aromatic rings separated by two carbon atoms connected via double bound along with the nitrile moiety, which served as a hydrogen bond acceptor, as shown in Figure 4D. Bulky substituents were not allowed on the indole nitrogen atom, since only compounds without a substituent (2l) or bearing a methyl group (5ad) presented pronounced activity. For a more beneficial effect, the additional pendant aromatic ring should be 2-naphthyl or para-substituted phenyl. Especially advantageous was the introduction of a dimethylamine moiety, which was consistent with literature data [45,46].

2.3.2. Docking to Antibacterial Targets

The antibacterial activity of the acrylonitrile-based compounds could be potentially associated with their affinity to bacterial enzymes involved in the synthesis of peptidoglycan, which is the major component of bacterial cell walls, i.e., penicillin-binding protein 4 (PBP4) and/or β-lactamase [44]. Thus, the novel ligands were docked in the active sites of the aforementioned proteins from E. coli (PDB codes 2ex8 [75] and 1fqg [76], respectively).
As revealed by the FRED Chemgauss4 scores (Table S1, Supplementary Materials), some of the ligands were ranked higher than the original ligand in the crystal structures, penicillin G. For example, the active derivatives 2i and 2x were bound more strongly in the PBP4 active site than benzylpenicillin (−5.90 and −6.00 vs. −5.66, respectively). In the β-lactamase-binding pocket, the ligand 2i also achieved a greater score than the original ligand (−9.63 vs. −9.55), while the most potent ligand 2x was ranked lower than penicillin G with Chemgauss4 score of −7.75. The molecular structure of the most active derivative 2x docked in the active pockets of the analyzed proteins is presented in Figure 5, while the 2D diagrams of interactions are included in Supplementary Materials (Tables S5 and S6).
Within PBP4 (Figure 5A), van der Waals interactions were formed between ligand 2x and Ser62, Phe160, Ser306, Arg361, Ser398, Arg402, Thr418, Leu421, Gln422, and Arg459. The indole NH group of the ligand created a hydrogen bond with a length of 1.6 A with the oxygen atom of the carboxyl group from the main chain of Ser420. However, the hydroxyl group present in the side chain of this amino acid residue formed an unfavorable hydrogen donor–donor type interaction with the NH group of the pyrrole ring present within the ligand structure (length of 1.7 A).
In the binding site of β-lactamase (Figure 5B), Ala237 formed hydrophobic contacts with the indole ring of the derivative 2x. The main chain of this amino acid formed a hydrogen bond of 2.2 A with the NH group of the indole ring. Another hydrogen bond was created between the nitrile nitrogen atom and the Asp170 residue (3.0 A). Van der Waals forces were detected with Ser70, Tyr105, Met129, Ser130, Asn132, Pro167, Val216, Ser235, Gly236, Gly238, Glu239, and Arg243.

2.4. In Silico Physicochemical, Pharmacokinetic and Drug-Likeness Predictions

The free available SwissADME web tool (http://www.swissadme.ch (accessed on 9 May 2023)) accessed on 16 February 2023 was employed to evaluate the physicochemical characteristics and predict the pharmacokinetic and drug-likeness properties of the most potent 2-(1H-indol-2-yl)-3-acrylonitriles 2l, 2x, and 5ad [77]. The results are presented in Table 7 and Figure 6 and Figure 7 (see Table S7 in the Supplementary Materials for more details).
As can be seen from the data in Table 7, the tested molecules were characterized by reasonable polarity (TPSA values in the range of 28.78–55.37 Å2) and suitable lipophilicity (ClogP values ranging from 3.49 to 4.41), so they were expected to be soluble or moderately soluble in water.
Moreover, according to Table 7, the bioavailable radar charts in Figure 6, and the BOILED-Egg plot in Figure 7, the investigated compounds were predicted to possess high gastrointestinal tract (GI) absorption and blood–brain barrier (BBB) permeability. In this regard, all of the tested molecules showed the same bioavailability score of 0.55, which suggested desirable pharmacokinetic properties (Table 7). Additionally, as shown in Table 7, compounds 2l, 2x, and 5ad met all the criteria according to Lipinski’s “rule of five” as one of the key drug-likeness characteristics [78].
Other drug-likeness predictions, namely Caco-2 cell and MDCK cell permeabilities, were calculated using the PreADMET online server (http://preadmet.bmdrc.kr (accessed on 9 May 2023)) accessed on 1 June 2023. The in vitro Caco-2 cell permeability results classified indole-acrylonitriles 2l, 2x, and 5ad as moderate permeability compounds (12.20–57.65 nm/s), with 2x possessing the worst predicted permeability (12.20 nm/s). On the other hand, the in vitro MDCK permeability was more varied: moderate permeability was obtained for compounds 2x, 5a, and 5d (46.73–54.34 nm/s), compound 5b showed low permeability (14.80 nm/s), while analogs 2l and 5c bearing the dimethylamino moiety could be characterized as poorly permeable compounds (0.09 and 0.16 nm/s, respectively) (Table S8 in Supplementary Materials).

3. Materials and Methods

3.1. Chemistry

3.1.1. General Information

Melting points were measured using a Boetius apparatus (VEB Analytik Dresden, Germany) and are uncorrected. IR spectra were obtained in KBr pellets using a Nicolet 380 FTIR 1600 spectrometer. Magnetic resonance spectra (NMR) (Agilent, Karlsruhe, Germany) were recorded using a Varian Mercury-VX 300 or Bruker Avance III HD 400 spectrometer. 1H and 13C chemical shifts (δ) are reported in ppm relative to the residual solvent signals at 2.50 and 39.5 ppm (DMSO-d6). Coupling constants (J) are given in hertz (Hz). The mass spectra were recorded on a Shimadzu LCMS-2010 EV (Tokyo, Japan) spectrometer equipped with an electrospray source. The ESI-MS spectra were registered in positive- or negative-ion mode.
Diffraction data for 2l were collected at room temperature using an Oxford Diffraction SuperNova diffractometer (Agilent Technologies Inc., Santa Clara, CA, USA) with Cu Kα radiation and processed using CrysAlisPro software version 1.171.33.48 [79]. The structure was solved using the program SHELXT [80] and refined using the full-matrix least-squares method on F2 with SHELXL-2018/3 [81] with Olex2 software version 1.5 [82].
Preparative thin-layer chromatography was performed on silica gel 60 PF254 containing gypsum (Merck KGaA, Darmstadt, FRG) with the aid of a Chromatotron® using the reported solvent systems. 2-(1H-Indol-2-yl)acetonitrile (1) was obtained according to the published method [55].

3.1.2. General Procedure for the Preparation of 2-(1H-Indol-2-yl)-3-acrylonitriles 2as

To a solution of 2-(1H-indol-2-yl)acetonitrile (1) (312 mg, 2.0 mmol) in anhydrous methanol (10 mL) was added dropwise a solution of sodium methoxide (60 mg of sodium in 6 mL of anhydrous methanol). The reaction mixture was stirred for 30 min, and then the appropriate aldehyde was added (4.0 mmol). After stirring overnight at ambient temperature, the product that precipitated was collected by vacuum filtration, washed with methanol, and if necessary subjected to silica gel column chromatography with dichloromethane as the eluent. In this manner, the following compounds were obtained.
  • 2-(1H-Indol-2-yl)-3-phenylacrylonitrile (2a). Yield: 39% (yellow solid); m.p. 197–199 °C (m.p. 173–174 °C [63]); IR νmax (KBr, cm−1): 3360, 3085, 3058, 3023, 2224, 1433, 1341, 1302, 914, 782, 726, 676; 1H NMR (300 MHz, DMSO-d6) δ (ppm): 11.74 (s, 1H, NH), 7.92 (s, 1H, CH), 7.87 (d, J = 7.1 Hz, 2H, 2 × C-Harom), 7.62–7.45 (m, 4H, 4 × C-Harom), 7.41 (d, J = 8.9 Hz, 1H, C-Harom), 7.18 (t, J = 7.1 Hz, 1H, C-Harom), 7.04 (t, J = 7.5 Hz, 1H, C-Harom), 6.80 (s, 1H, C3-H, indole); 13C NMR (75 MHz, DMSO-d6) δ (ppm): 138.6, 138.2, 134.0, 133.7, 130.9, 129.6 (two overlapping signals), 129.2 (two overlapping signals), 128.2, 123.9, 121.3, 120.4, 117.5, 111.8, 104.2, 103.7; MS (ESI) m/z: 243 [M − H]. Anal. calcd for C17H12N2 (244.29) (%): C, 83.58; H, 4.95; N, 11.47. Found: C, 83.32; H, 5.01; N, 11.67.
  • 2-(1H-Indol-2-yl)-3-(p-tolyl)acrylonitrile (2b). Yield: 62% (yellow solid); m.p. 223–224 °C (192–194 °C [63]); IR νmax (KBr, cm−1): 3356, 3045, 2918, 2223, 1611, 1435, 1302, 898, 798, 782, 730, 607; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.72 (s, 1H, NH), 7.90 (s, 1H, CH), 7.80 (d, J = 8.2 Hz, 2H, 2 × C-Harom), 7.60 (d, J = 7.9 Hz, 1H, C-Harom), 7.43–7.37 (m, 1H, C-Harom), 7.38 (d, J = 8.0 Hz, 2H, 2 × C-Harom), 7.20 (t, J = 7.0 Hz, 1H, C-Harom), 7.05 (t, J = 7.0 Hz, 1H, C-Harom), 6.79 (s, 1H, C3-H, indole), 2.39 (s, 3H, CH3); MS (ESI) m/z: 257 [M − H]. Anal. calcd for C18H14N2 (258.32) (%): C, 83.69; H, 5.46; N, 10.84. Found: C, 83.57; H, 5.25; N, 11.18.
  • 2-(1H-Indol-2-yl)-3-(4-isopropylphenyl)acrylonitrile (2c). Yield: 21% (yellow solid); m.p. 176–178 °C; IR νmax (KBr, cm−1): 3327, 2956, 2869, 2233, 1606, 1417, 1302, 897, 827, 784, 747, 730, 613; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.74 (s, 1H, NH), 7.91 (s, 1H, CH), 7.83 (d, J = 8.3 Hz, 2H, 2 × C-Harom), 7.60 (d, J = 7.8 Hz, 1H, C-Harom), 7.45–7.42 (m, 3H, 3 × C-Harom), 7.20 (t, J = 8.1 Hz, 1H, C-Harom), 7.06 (t, J = 7.5 Hz, 1H, C-Harom), 6.80 (s, 1H, C3-H, indole), 3.02–2.91 (m, 1H, CH), 1.24 (d, J = 6.9 Hz, 6H, 2 × CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 151.8, 138.6, 138.2, 133.9, 131.7, 129.4 (two overlapping signals), 128.3, 127.7 (two overlapping signals), 123.8, 121.2, 120.4, 117.7, 111.8, 103.8, 102.7, 33.9, 24.0 (two overlapping signals); MS (ESI) m/z: 285 [M − H]. Anal. calcd for C20H18N2 (286.37) (%): C, 83.88; H, 6.34; N, 9.78. Found: C, 83.96; H, 6.29; N, 9.75.
  • 2-(1H-Indol-2-yl)-3-(4-methoxyphenyl)acrylonitrile (2d). Yield: 67% (yellow solid); m.p. 210–212 °C; IR νmax (KBr, cm−1): 3352, 3045, 3009, 2949, 2927, 2831, 2222,1609, 1589, 1506, 1261, 1181, 1035, 814, 779, 729, 606; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.67 (s, 1H, NH), 7.88 (d, J = 9.2 Hz, 3H, 2 × C-Harom + CH), 7.58 (d, J = 7.8 Hz, 1H, C-Harom), 7.41 (d, J = 8.1 Hz, 1H, C-Harom), 7.23–7.13 (m, 3H, 3 × C-Harom), 7.04 (t, J = 7.5 Hz, 1H, C-Harom), 6.75 (s, 1H, C3-H, indole), 3.86 (s, 3H, CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 161.5, 138.6, 138.1, 134.1, 131.1 (two overlapping signals), 128.3, 126.6, 123.6, 121.1, 120.4, 118.0, 115.2 (two overlapping signals), 111.7, 103.2, 100.7, 55.9; MS (ESI) m/z: 273 [M − H]. Anal. calcd for C18H14N2O (274.32): C, 78.81; H, 5.14; N, 10.21. Found: C, 78.75; H, 5.27; N, 10.19.
  • 3-(4-Ethoxyphenyl)-2-(1H-indol-2-yl)acrylonitrile (2e). Yield: 58% (yellow solid); m.p. 174–175 °C; IR νmax (KBr, cm−1): 3344, 3046, 2976, 2925, 2876. 2223, 1608, 1588, 1505, 1259, 1183, 1051, 779, 726; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.67 (s, 1H, NH), 7.86–7.88 (m, 3H, C-Harom + CH), 7.58 (d, J = 7.9 Hz, 1H, C-Harom), 7.41 (d, J = 8.9 Hz, 1H, C-Harom), 7.18 (t, J = 8.1 Hz, 1H, C-Harom), 7.11 (d, J = 8.8 Hz, 2H, 2 × C-Harom), 7.04 (t, J = 7.5 Hz, 1H, C-Harom), 6.74 (s, 1H, C3-H, indole), 4.12 (q, J = 7.0 Hz, 2H, CH2), 1.36 (t, J = 7.0 Hz, 3H, CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 160.8, 138.6, 138.1, 134.2, 131.2 (two overlapping signals), 128.4, 126.4, 123.5, 121.1, 120.3, 118.0, 115.6 (two overlapping signals), 111.7, 103.1, 100.6, 63.9, 15.0; MS (ESI) m/z: 287 [M − H]. Anal. calcd for C19H16N2O (288.34): C, 79.14; H, 5.59; N, 9.72. Found: C, 78.95; H, 5.49; N, 9.85.
  • 3-(Benzo[d][1,3]dioxol-5-yl)-2-(1H-indol-2-yl)acrylonitrile (2f). Yield: 15% (yellow solid); m.p. 233–234 °C; IR νmax (KBr, cm−1): 3354, 3082, 3044, 2897, 2222, 1501, 1453, 1256, 1049, 782, 733, 622; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.67 (s, 1H, NH), 7.83 (s, 1H, CH), 7.59 (d, J = 8.1 Hz, 1H, C-Harom), 7.55 (s, 1H, C-Harom), 7.41 (d, J = 8.1 Hz, 1H, C-Harom), 7.35 (d, J = 8.8 Hz, 1H, C-Harom), 7.19 (t, J = 7.0 Hz, 1H, C-Harom), 7.12 (d, J = 8.1 Hz, 1H, C-Harom), 7.05 (t, J = 7.0 Hz, 1H, C-Harom), 6.75 (s, 1H, C3-H, indole), 6.16 (s, 2H, CH2); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 149.8, 148.5, 138.5, 138.2, 134.0, 128.3, 128.2, 126.0, 123.7, 121.1, 120.4, 117.9, 111.7, 109.5, 107.4, 103.4, 102.5, 101.2; MS (ESI) m/z: 287 [M − H]. Anal. calcd for C18H12N2O2 (288.30) (%): C, 74.99; H, 4.20; N, 9.72. Found: C, 74.92; H, 4.32; N, 9.68.
  • 3-(4-Fluorophenyl)-2-(1H-indol-2-yl)acrylonitrile (2g). Yield: 50% (yellow solid); m.p. 203–205 °C; IR νmax (KBr, cm−1): 3349, 3062, 2224, 1603, 1593, 1504, 1242, 1164, 892, 818, 781, 729, 605; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.73 (s, 1H, NH), 7.96–7.93 (m, 3H, 2 × C-Harom + CH), 7.61 (d, J = 7.9 Hz, 1H, C-Harom), 7.44–7.40 (m, 3H, 3 × C-Harom), 7.21 (t, J = 7.1 Hz, 1H, C-Harom), 7.06 (t, J = 7.5 Hz, 1H, C-Harom), 6.82 (s, 1H, C3-H, indole); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 163.39 (d, J(C-F) = 250.0 Hz), 138.3, 137.5, 133.6, 131.59 (d, J(C-F) = 8.0 Hz, two overlapping signals), 130.66 (d, J(C-F) = 3.0 Hz), 128.2, 123.9, 121.3, 120.5, 117.5, 116.81 (d, J(C-F) = 22.0 Hz, two overlapping signals), 111.8, 104.1, 103.6; MS (ESI) m/z: 261 [M − H]; Anal. calcd for C17H11FN2 (262.28): C, 77.85; H, 4.23; N, 10.68. Found: C, 77.96; H, 4.25; N, 10.83.
  • 3-(2-Chlorophenyl)-2-(1H-indol-2-yl)acrylonitrile (2h). Yield: 49% (yellow solid); m.p. 201–203 °C; IR νmax (KBr, cm−1): 3355, 3059, 2225, 1593, 1433, 1340, 1304, 1258, 1045, 785, 747, 731, 611; 1H NMR (300 MHz, DMSO-d6) δ (ppm): 11.94 (s, 1H, NH), 8.05 (s, 1H, CH), 8.02–7.98 (m, 1H, C-Harom), 7.65–7.59 (m, 2H, 2 × C-Harom), 7.52–7.46 (m, 2H, 2 × C-Harom), 7.42 (d, J = 8.2 Hz, 1H, C-Harom), 7.21 (t, J = 7.1 Hz, 1H, C-Harom), 7.05 (t, J = 7.1 Hz, 1H, C-Harom), 6.86 (s, 1H, C3-H, indole); 13C NMR (75 MHz, DMSO-d6) δ (ppm): 138.5, 134.6, 133.8, 133.0, 132.6, 132.1, 130.3, 130.0, 128.1, 128.0, 124.4, 121.6, 120.6, 116.8, 111.9, 107.9, 105.4; MS (ESI) m/z: 277 [M − H]. Anal. calcd for C17H11ClN2 (278.74) (%): C, 73.25; H, 3.98; Cl, 12.72; N, 10.05. Found: C, 73.29; H, 3.82; N, 10.15.
  • 3-(3-Chlorophenyl)-2-(1H-indol-2-yl)acrylonitrile (2i). Yield: 22% (yellow solid); m.p. 188–189 °C; IR νmax (KBr, cm−1): 3351, 3056, 2223, 1562, 1481, 1416, 1339, 1303, 1208, 1082, 784, 732, 674; 1H NMR (300 MHz, DMSO-d6) δ (ppm): 11.74 (s, 1H, NH), 7.89 (m, 2H, C-Harom + CH), 7.82–7.77 (m, 1H, C-Harom), 7.60–7.53 (m, 3H, 3 × C-Harom), 7.42 (d, J = 8.1 Hz, 1H, C-Harom), 7.19 (t, J = 7.6 Hz, 1H, C-Harom), 7.04 (t, J = 8.0 Hz, 1H, C-Harom), 6.83 (s, 1H, C3-H, indole); 13C NMR (75 MHz, DMSO-d6) δ (ppm): 138.4, 136.7, 136.1, 134.2, 133.4, 131.5, 130.4, 128.4, 128.2, 127.9, 124.2, 121.5, 120.5, 117.1, 111.9, 105.3, 104.8; MS (ESI) m/z: 277 [M − H]. Anal. calcd for C17H11ClN2 (278.74) (%): C, 73.25; H, 12.72; N, 10.05. Found: C, 73.17; H, 3.86; N, 10.12.
  • 3-(4-Chlorophenyl)-2-(1H-indol-2-yl)acrylonitrile (2j). Yield: 67% (yellow solid); m.p. 241–242 °C; IR νmax (KBr, cm−1): 3357, 3082, 3050, 3015, 2225, 1597, 1585, 1488, 1409, 1101, 806, 787, 732; 1H NMR (300 MHz, DMSO-d6) δ (ppm): 11.73 (s, 1H, NH), 7.89–7.85 (m, 3H, 2 × C-Harom + CH), 7.62–7.57 (m, 3H, 3 × C-Harom), 7.40 (d, J = 8.0 Hz, 1H, C-Harom), 7.19 (t, J = 7.3 Hz, 1H, C-Harom), 7.04 (t, J = 7.2 Hz, 1H, C-Harom), 6.81 (s, 1H, C3-H, indole); 13C NMR (75 MHz, DMSO-d6) δ (ppm): 138.3, 137.1, 135.3, 133.5, 132.9, 130.8 (two overlapping signals), 129.7 (two overlapping signals), 128.2, 124.1, 121.4, 120.5, 117.3, 111.8, 104.5, 104.3; MS (ESI) m/z: 274 [M − H]. Anal. calcd for C17H11ClN2 (278.74) (%): C, 73.25; H, 3.98; N, 10.05. Found: C, 73.43; H, 3.75; N, 9.85.
  • 3-(4-Bromophenyl)-2-(1H-indol-2-yl)acrylonitrile (2k). Yield: 46% (yellow solid); m.p. 239–241 °C; IR νmax (KBr, cm−1): 3358, 3082, 3056, 3019, 2225, 1581, 1485, 1406, 1078, 1010, 894, 803, 782, 733, 608; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.76 (s, 1H, NH), 7.90 (s, 1H, CH), 7.83–7.76 (m, 4H, 4 × C-Harom), 7.61 (d, J = 7.9 Hz, 1H, C-Harom), 7.43 (d, J = 9.0 Hz, 1H, C-Harom), 7.22 (t, J = 7.6 Hz, 1H, C-Harom), 7.06 (t, J = 8.0 Hz, 1H, C-Harom), 6.84 (s, 1H, C3-H, indole); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 138.3, 137.2, 133.6, 133.2, 132.7 (two overlapping signals), 131.0 (two overlapping signals), 128.2, 124.2, 124.1, 121.4, 120.5, 117.3, 111.9, 104.6, 104.4; MS (ESI) m/z: 322 [M − H]; Anal. calcd for C17H11BrN2 (323.19): C, 63.18; H, 3.43; N, 8.67. Found: C, 62.97; H, 3.59; N, 8.53.
  • 3-[4-(Dimethylamino)phenyl]-2-(1H-indol-2-yl)acrylonitrile (2l). Yield: 37% (brown solid); m.p. 234–236 °C; IR νmax (KBr, cm−1): 3334, 3079, 3013, 2989, 2814, 2211, 1611, 1575, 1364, 1198, 810, 788, 746; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.55 (s, 1H, NH), 7.80 (d, J = 8.9 Hz, 2H, 2 × C-Harom), 7.75 (s, 1H, CH), 7.55 (d, J = 7.8 Hz, 1H, C-Harom), 7.39 (d, J = 8.1 Hz, 1H, C-Harom), 7.15 (t, J = 7.5 Hz, 1H, C-Harom), 7.02 (t, J = 7.4 Hz, 1H, C-Harom), 6.84 (d, J = 9.0 Hz, 2H, 2 × C-Harom), 6.64 (s, 1H, C3-H, indole), 3.03 (s, 6H, 2 × CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 152.1, 139.4, 138.0, 135.0, 131.1 (two overlapping signals), 128.6, 123.0, 121.2, 120.7, 120.2, 118.9, 112.3 (two overlapping signals), 111.5, 101.7, 96.3, 40.1 (two overlapping signals); MS (ESI) m/z: 286 [M − H]. Anal. calcd for C19H17N3 (287.36) (%): C, 79.41; H, 5.96; N, 14.62. Found: C, 79.31; H, 5.87; N, 14.82.
  • Crystal data for 2l: C19H17N3 (M = 287.35 g/mol), monoclinic, space group P21/n, a = 6.76580(10) Å, b = 21.9867(4) Å, c = 10.2650(2) Å, β = 93.006(2)°, V = 1524.90(5) Å3, Z = 4, μ(Cu Kα) = 0.587 mm−1, Dcalc = 1.252 g/cm3, 6428 reflections measured, 3114 unique (Rint = 0.0141, Rsigma = 0.0179), which were used in all calculations. The final R1 was 0.0412 (I > 2σ(I)) and wR2 was 0.1225 (all data).
  • 3-[4-(Diethylamino)phenyl]-2-(1H-indol-2-yl)acrylonitrile (2m). Yield: 7% (brown solid); m.p. 183–185 °C; IR νmax (KBr, cm−1): 3336, 3047, 2967, 2925, 2866, 2219, 1609, 1584, 1512, 1356, 1200, 812, 799, 731; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.54 (s, 1H, NH), 7.78 (d, J = 8.9 Hz, 2H, 2 × C-Harom), 7.72 (s, 1H, CH), 7.54 (d, J = 7.8 Hz, 1H, C-Harom), 7.38 (d, J = 8.1 Hz, 1H, C-Harom), 7.14 (t, J = 7.4 Hz, 1H, C-Harom), 7.02 (t, J = 7.4 Hz, 1H, C-Harom), 6.81 (d, J = 8.9 Hz, 2H, 2 × C-Harom), 6.62 (s, 1H, C3-H, indole), 3.44 (q, J = 6.9 Hz, 4H, 2 × CH2), 1.14 (t, J = 6.9 Hz, 6H, 2 × CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 149.6, 139.4, 137.9, 135.1, 131.5 (two overlapping signals), 128.6, 122.9, 120.7, 120.4, 120.2, 119.0, 111.8 (two overlapping signals), 111.5, 101.5, 95.5, 44.3 (two overlapping signals), 12.9 (two overlapping signals); MS (ESI) m/z: 314 [M − H]; MS (ESI) m/z: 316 [M + H]+. Anal. calcd for C21H21N3 (315.41) (%): C, 79.97; H, 6.71; N, 13.32. Found: C, 79.75; H, 6.87; N, 13.38.
  • 2-(1H-Indol-2-yl)-3-(4-nitrophenyl)acrylonitrile (2n). Yield: 27% (orange solid); m.p. 210–212 °C; IR νmax (KBr, cm−1): 3344, 2227, 1595, 1578, 1512, 1341, 1111, 787, 753, 684; 1H NMR (300 MHz, DMSO-d6) δ (ppm): 11.83 (s, 1H, NH), 8.36 (d, J = 8.9 Hz, 2H, C-Harom), 8.05 (d, J = 8.9 Hz, 2H, 2 × C-Harom), 8.00 (s, 1H, CH), 7.61 (d, J = 7.9 Hz, 1H, C-Harom), 7.42 (d, J = 7.7 Hz, 1H, C-Harom), 7.22 (t, J = 7.6 Hz, 1H, C-Harom), 7.05 (t, J = 7.9 Hz, 1H, C-Harom), 6.91 (s, 1H, C3-H, indole); 13C NMR (75 MHz, DMSO-d6) δ (ppm): 148.0, 140.2, 138.6, 135.6, 133.3, 130.2 (two overlapping signals), 128.1, 124.7, 124.6, 121.7, 120.7, 115.8, 114.9, 112.0, 107.2, 105.9; MS (ESI) m/z: 288 [M − H]. Anal. calcd for C17H11N3O2 (289.29) (%): C, 70.58; H, 3.83; N, 14.53. Found: C, 70.67; H, 3.65; N, 14.87.
  • 2-(1H-Indol-2-yl)-3-(naphthalen-1-yl)acrylonitrile (2o). Yield: 46% (yellow solid); m.p. 218–220 °C; IR νmax (KBr, cm−1): 3372, 3058, 3013, 2921, 2847, 2222, 1587, 1431, 1301, 882, 777, 728, 674; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.98 (s, 1H), 8.67 (s, 1H, CH), 8.31 (d, J = 8.2 Hz, 1H, C-Harom), 8.13–8.05 (m, 3H, 3 × C-Harom), 7.71–7.64 (m, 4H, 4 × C-Harom), 7.48 (d, J = 8.2 Hz, 1H, C-Harom), 7.25 (t, J = 7.6 Hz, 1H, C-Harom), 7.09 (t, J = 7.5 Hz, 1H, C-Harom), 6.89 (s, 1H, C3-H, indole); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 138.4, 136.4, 133.7, 133.7, 131.6, 131.2, 131.0, 129.3, 128.2, 127.6, 127.2, 126.9, 126.1, 124.3, 124.2, 121.5, 120.5, 117.4, 111.8, 107.1, 104.8; MS (ESI) m/z: 293 [M − H]. Anal. calcd for C21H14N2 (294.35) (%): C, 85.69; H, 4.79; N, 9.52. Found: C, 85.58; H, 4.82; N, 9.32.
  • 2-(1H-Indol-2-yl)-3-(naphthalen-2-yl)acrylonitrile (2p). Yield: 50% (yellow solid); m.p. 229–230 °C; IR νmax (KBr, cm−1): 3364, 3052, 2920, 2224, 1594, 1424, 1342, 915, 818, 769, 734, 673; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.83 (br. s, 1H, NH), 8.35 (s, 1H, CH), 8.11–8.09 (m, 3H, 3 × C-Harom), 8.06–8.03 (m, 1H, C-Harom), 8.01–7.98 (m, 1H, C-Harom), 7.66–7.60 (m, 3H, 3 × C-Harom), 7.47–7.44 (m, 1H, C-Harom), 7.22 (t, J = 7,1 Hz, 1H, C-Harom), 7.07 (t, J = 7.1 Hz, 1H, C-Harom), 6.87 (s, 1H, C3-H, indole); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 138.5, 138.4, 133.9, 133.2, 131.7, 130.2, 129.2, 129.1, 128.3, 128.24, 128.20, 127.6, 125.1, 124.0, 121.4, 120.5, 117.7, 111.9, 104.3, 103.9; MS (ESI) m/z: 293 [M − H]. Anal. calcd for C21H14N2 (294.35) (%): C, 85.69; H, 4.79; N, 9.52. Found: C, 85.75; H, 4,67; N, 9.58.
  • 2-(1H-Indol-2-yl)-3-(pyridin-2-yl)acrylonitrile (2q). Yield: 24% (yellow solid); m.p. 179–181 °C; IR νmax (KBr, cm−1): 3318, 3056, 3008, 2924, 2229, 1577, 1423, 1305, 1149, 895, 787, 723, 608; 1H NMR (300 MHz, DMSO-d6) δ (ppm): 11.81 (s, 1H, NH), 8.73 (m, 1H, C-Harom), 7.98–7.90 (m, 2H, C-Harom + CH), 7.67 (d, J = 7.9 Hz, 1H, C-Harom), 7.60 (d, J = 7.9 Hz, 1H, C-Harom), 7.45–7.40 (m, 2H, 2 × C-Harom), 7.20 (t, J = 7.6 Hz, 1H, C-Harom), 7.04 (t, J = 7.5 Hz, 1H, C-Harom), 6.90 (s, 1H, C3-H, indole); 13C NMR (75 MHz, DMSO-d6) δ (ppm): 152.0, 150.3, 138.5, 137.8, 136.4, 133.9, 128.2, 125.7, 125.0, 124.3, 121.5, 120.5, 117.0, 111.9, 106.3, 105.5; MS (ESI) m/z: 244 [M − H]; MS (ESI) m/z: 246 [M + H]+. Anal. calcd for C16H11N3 (245.28) (%): C, 78.35; H, 4.52; N, 17.13. Found: C, 78.27; H, 4.38; N, 17.35.
  • 2-(1H-Indol-2-yl)-3-(quinolin-2-yl)acrylonitrile (2r). Yield: 17% (orange solid); m.p. > 300 °C; IR νmax (KBr, cm−1): 3329, 3048, 2923, 2853, 2225, 1587, 1548, 1343, 886, 829, 796, 751, 728; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.93 (s, 1H, NH), 8.53 (d, J = 8.4 Hz, 1H, C-Harom), 8.11 (s, 1H, CH), 8.06 (t, J = 8.6 Hz, 2H, 2 × C-Harom), 7.89–7.84 (m, 2H, 2 × C-Harom), 7.71–7.65 (m, 2H, 2 × C-Harom), 7.46 (d, J = 8.2 Hz, 1H, C-Harom), 7.25 (t, J = 7.1 Hz, 1H, C-Harom), 7.09 (t, J = 7.0 Hz, 1H, C-Harom), 7.00 (s, 1H, C3-H, indole); MS (ESI) m/z: 294 [M − H]; MS (ESI) m/z: 296 [M + H]+. Anal. calcd for C20H13N3 (295.34) (%): C, 81.34; H, 4.44; N, 14.23. Found: C, 81.42; H, 4.32; N, 14.26.
  • 2-(1H-Indol-2-yl)-3-(thiophen-2-yl)acrylonitrile (2s). Yield: 21% (yellow solid); m.p. 178–180 °C; IR νmax (KBr, cm−1): 3351 3107, 3040, 2924, 2853, 2220, 1577, 1225, 1408, 1301, 885, 779, 691, 607; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.70 (s, 1H, NH), 8.13 (s, 1H, CH), 7.92 (d, J = 5.6 Hz, 1H, C-Harom), 7.68 (d, J = 4.6 Hz, 1H, C-Harom), 7.59 (d, J = 8.1 Hz, 1H, C-Harom), 7.41 (d, J = 9.0 Hz, 1H, C-Harom), 7.28–7.30 (m, 1H, C-Harom), 7.19 (t, J = 8.1 Hz, 1H, C-Harom), 7.05 (t, J = 7.1 Hz, 1H, C-Harom) 6.76 (s, 1H, C3-H, indole); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 138.3, 137.8, 133.6, 133.4, 131.8, 131.7, 128.8, 128.4, 123.8, 121.2, 120.5, 117.7, 111.7, 103.8, 100.1; MS (ESI) m/z: 249 [M − H]. Anal. calcd for C15H10N2S (250.32) (%): C, 71.97; H, 4.03; N, 11.19. Found: C, 71.85; H, 3.97; N, 11.27.

3.1.3. General Procedure for the Preparation of 2-(1H-Indol-2-yl)-3-acrylonitriles 2tx

To a stirred solution of 2-(1H-indol-2-yl)acetonitrile (1) (312 mg, 2.0 mmol) in anhydrous ethanol (2 mL) was added the appropriate carboxaldehyde (3.0 mmol) and ten drops of triethylamine. Stirring was continued under reflux for 15 min and then at ambient temperature for 48 h. The precipitate thus obtained was collected by vacuum filtration, washed with anhydrous ethanol, and dried. In this manner, the following compounds were obtained.
  • 3-(5-Chlorothiophen-2-yl)-2-(1H-indol-2-yl)acrylonitrile (2t). Yield: 33% (green solid); m.p. 259–261 °C; IR νmax (KBr, cm−1): 3348, 3054, 2218, 1501, 1433, 1414, 1308, 1228, 786, 741, 608; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.70 (s, 1H, NH), 8.03 (s, 1H, CH), 7.59 (d, J = 7.9 Hz, 1H, C-Harom), 7.51–7.49 (m, 1H, CHarom), 7.40 (d, J = 8.1 Hz, 1H, C-Harom), 7.34–7.32 (m, 1H, C-Harom), 7.19 (t, J = 7.6 Hz, 1H, C-Harom), 7.05 (t, J = 7.4 Hz, 1H), 6.77 (s, 1H, C3-H, indole); MS (ESI) m/z: 155 [M − H]. Anal. calcd for C15H9ClN2S (284.76) (%): C, 63.27; H, 3.19; N, 9.84. Found: C, 63.15; H, 3.21; N, 9.72.
  • 3-(Furan-2-yl)-2-(1H-indol-2-yl)acrylonitrile (2u). Yield: 53% (yellow solid); m.p. 185–186 °C; IR νmax (KBr, cm−1): 3321, 3056, 2924, 2231, 1607, 1469, 1346, 1303, 1019, 884, 781, 724; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.71 (s, 1H, NH), 8.02 (s, 1H, CH), 7.76 (s, 1H, C-Harom), 7.58 (d, J = 7.9 Hz, 1H, C-Harom), 7.39 (d, J = 8.0 Hz, 1H, C-Harom), 7.19 (t, J = 7.6 Hz, 1H, C-Harom), 7.15 (d, J = 3.5 Hz, 1H, C-Harom), 7.05 (t, J = 7.3 Hz, 1H, C-Harom), 6.82–6.74 (m, 2H, 2 × C-Harom); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 149.9, 146.7, 138.3, 133.5, 128.3, 124.7, 123.9, 121.2, 120.5, 117.3, 116.4, 113.8, 111.7, 104.0, 99.6; MS (ESI) m/z: 233 [M − H]. Anal. calcd for C15H10N2O (234.25) (%): C, 76.91; H, 4.30; N, 11.96. Found: C, 76.83; H, 4.38; N, 11.86.
  • 2-(1H-Indol-2-yl)-3-(5-methylfuran-2-yl)acrylonitrile (2v). Yield: 45% (yellow solid); m.p. 190–191 °C; IR νmax (KBr, cm−1): 3322, 3053, 2920, 2850, 2226, 1517, 1539, 1489, 1290, 1025, 781, 748; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.64 (s, 1H, NH), 7.67 (s, 1H, CH), 7.56 (d, J = 7.9 Hz, 1H, C-Harom), 7.38 (d, J = 8.2 Hz, 1H, C-Harom), 7.17 (t, J = 7.6 Hz, 1H, C-Harom), 7.08–7.01 (m, 2H, 2 × C-Harom), 6.73 (s, 1H, C3-H, indole), 6.43 (d, J = 3.2 Hz, 1H, C-Harom), 2.41 (s, 3H, CH3); MS (ESI) m/z: 247 [M − H]. Anal. calcd for C16H12N2O (248.28) (%): C, 77.40; H, 4.87; N, 11.28. Found: C, 77.46; H, 4.75; N, 11.32.
  • 3-(5-Chlorofuran-2-yl)-2-(1H-indol-2-yl)acrylonitrile (2w). Yield: 26% (yellow solid); m.p. 223–228 °C; IR νmax (KBr, cm−1): 3333, 3045, 2227, 1519, 1469, 1345, 1020, 876, 774, 729; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.71 (s, 1H, NH), 7.67 (s, 1H, CH), 7.59 (d, J = 7.9 Hz, 1H, C-Harom), 7.39 (d, J = 8.1 Hz, 1H, C-Harom), 7.23–7.16 (m, 2H, 2 × C-Harom), 7.05 (t, J = 7.8 Hz, 1H, C-Harom), 6.82 (d, J = 3.6 Hz, 1H, C-Harom), 6.80 (s, 1H, C3-H, indole); MS (ESI) m/z: 267 [M − H]. Anal. calcd. for C15H9ClN2O (268.70) (%): C, 67.05; H, 3.38; N, 10.43. Found: C, 67.17; H, 3.46; N, 10.37.
  • 2-(1H-Indol-2-yl)-3-(1H-pyrrol-2-yl)acrylonitrile (2x). Yield: 8% (brown, solid); m.p. 134–136 °C; IR νmax (KBr, cm−1): 3427, 3331, 3113, 3014, 2924, 2215, 1596, 1402, 1342, 1038, 786, 748, 663, 544; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.62 (s, 1H, NH), 11.54 (s, 1H, NH), 7.64 (s, 1H, CH), 7.54 (d, J = 7.7 Hz, 1H, C-Harom), 7.38 (d, J = 8.0 Hz, 1H, C-Harom), 7.16–7.12 (m, 3H, 3 × C-Harom), 7.02 (t, J = 7.3 Hz, 1H, C-Harom), 6.62 (s, 1H, C3-H, indole), 6.36 (s, 1H, C-Harom); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 138.0, 134.4, 129.4, 128.6, 127.6, 124.2, 122.9, 120.6, 120.2, 118.8, 112.9, 111.7, 111.6, 101.6, 95.2; MS (ESI) m/z: 232 [M − H]; MS (ESI) m/z: 234 [M + H]+. Anal. calcd for C15H11N3 (233.27) (%): C, 77.23; H, 4.75; N, 18.01. Found: C, 77.31; H, 4.83; N, 17.86.

3.1.4. Synthesis of N-[4-(Dimethylamino)phenyl]-1H-indole-2-carbimidoyl cyanide (3)

To a solution of 2-(1H-indol-2-yl)acetonitrile (1) (312 mg, 2.0 mmol) in anhydrous methanol (10 mL) was added dropwise a solution of sodium methoxide (60 mg of sodium in 6 mL of anhydrous methanol) and the reaction mixture was stirred at ambient temperature for 30 min. Then, 4-nitroso-N,N-dimethylaniline (600 mg, 4.0 mmol) was added and stirring was continued at ambient temperature for 5 h. The product 3 that precipitated was filtered, washed with methanol, and dried. Yield: 42% (brown solid); m.p. 181–183 °C; IR νmax (KBr, cm−1): 3399, 3049, 2888, 2805, 2220, 1608, 1510, 1361, 1339, 1220, 1177, 824, 785, 743, 688; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.91 (s, 1H, NH), 7.69 (d, J = 7.9 Hz, 1H, C-Harom), 7.48–7.46 (m, 3H, 3 × C-Harom), 7.27 (t, J = 7.6 Hz, 1H, C-Harom), 7.14 (s, 1H, C3-H, indole), 7.09 (t, J = 7.4 Hz, 1H, C-Harom), 6.85 (d, J = 9.1 Hz, 2H, 2 × C-Harom), 3.02 (s, 6H, 2 × CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 150.8, 138.9, 136.9, 135.4, 127.9, 125.4, 124.5 (two overlapping signals), 124.3, 122.3, 120.7, 113.2, 112.7, 112.5 (two overlapping signals), 108.6, 40.4 (two overlapping signals); MS (ESI) m/z: 287 [M − H]. Anal. calcd for C18H16N4 (288.35) (%): C, 74.98; H, 5.59; N, 19.43. Found: C, 75.02; H, 5.51; N, 19.47.

3.1.5. General Procedure for the Preparation of 2-(1H-Indol-2-yl)-3-phenylpropanenitriles 4ac

To a suspension of sodium borohydride (57 mg, 1.5 mmol) in a mixture of dimethylformamide (4 mL) and methanol (1 mL) was added the appropriate 2-(1H-indol-2-yl)-3-acrylonitrile 2d, 2l, or 2p (0.75 mmol). The mixture was stirred overnight and then diluted with water, neutralized with hydrochloric acid, and extracted with dichloromethane. The organic phase was dried (Na2SO4), concentrated under vacuum, and subjected to preparative thin-layer chromatography eluting with petroleum ether/ethyl acetate (9:2 v/v). In this manner, the following compounds were obtained.
  • 2-(1H-Indol-2-yl)-3-(4-methoxyphenyl)propanenitrile (4a). Yield: 73% (white solid); m.p. 151–153 °C; IR νmax (KBr, cm−1): 3397, 3044, 3009, 2935, 2839, 2242, 1612, 1512, 1242, 1177, 1032, 800, 739, 667; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.43 (s, 1H, NH), 7.50 (d, J = 7.9 Hz, 1H, C-Harom), 7.39 (d, J = 8.9 Hz, 1H, C-Harom), 7.21 (d, J = 8.7 Hz, 2H, 2 × C-Harom), 7.11 (t, J = 7.0 Hz, 1H, C-Harom), 7.00 (t, J = 7.5 Hz, 1H, C-Harom), 6.88 (d, J = 8.7 Hz, 2H, 2 × C-Harom), 6.39 (s, 1H, C3-H, indole), 4.72–4.65 (m, 1H, CH), 3.73 (s, 3H, CH3), 3.32–3.18 (m, 2H, CH2); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 158.7, 136.9, 133.2, 130.6 (two overlapping signals), 129.3, 127.8, 122.0, 120.5, 120.4, 119.7, 114.2 (two overlapping signals), 111.7, 100.8, 55.4, 37.7, 33.0; MS (ESI) m/z: 275 [M − H]. Anal. calcd for C18H16N2O (276.33): C, 78.24; H, 5.84; N, 10.14; O, 5.79. Found: C, 78.32; H, 5.92; N, 10.06.
  • 3-[4-(Dimethylamino)phenyl]-2-(1H-indol-2-yl)propanenitrile (4b). Yield: 75% (beige solid); m.p. 180–183 °C; IR νmax (KBr, cm−1): 3400, 3355, 3048, 2923, 2857, 2810, 2240, 1613, 1524, 1357, 1344, 1191, 796, 752, 743, 667; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 11.42 (s, 1H, NH), 7.50 (d, J = 7.9 Hz, 1H, C-Harom), 7.40 (d, J = 8.1 Hz, 1H, C-Harom), 7.14–7.09 (m, 3H, 3 × C-Harom), 7.00 (t, J = 7.0 Hz, 1H, C-Harom), 6.67 (d, J = 8.7 Hz, 2H, 2 × C-Harom), 6.40 (s, 1H, C3-H, indole), 4.60–4.64 (m, 1H, CH), 3.28–3.14 (m, 2H, CH2), 2.86 (s, 6H, 2 × CH3); MS (ESI) m/z: 288 [M − H]. Anal. calcd for C19H19N3 (289.37) (%): C, 78.86; H, 6.62; N, 14.52. Found: C, 78.76; H, 6.58; N, 14.66.
  • 2-(1H-Indol-2-yl)-3-(naphthalen-2-yl)propanenitrile (4c). Yield: 63% (beige solid); m.p. 164–166 °C; IR νmax (KBr, cm−1): 3392, 3046, 2924, 2242, 1455, 1426, 1290, 802, 752, 743, 667; 1H NMR (300 MHz, DMSO-d6) δ (ppm): 11.50 (s, 1H, NH), 7.88–7.83 (m, 4H, 4 × C-Harom), 7.51–7.38 (m, 5H, 5 × C-Harom), 7.10 (t, J = 7.7 Hz, 1H, C-Harom), 6.98 (t, J = 7.6 Hz, 1H, C-Harom), 6.42 (s, 1H, C3-H, indole), 4.90–4.85 (m, 1H, CH), 3.58–3.41 (m, 2H, CH2); 13C NMR (75 MHz, DMSO-d6) δ (ppm): 136.9, 135.1, 133.3, 133.1, 132.5, 128.4, 128.1, 128.0, 127.9, 127.8, 127.7, 126.7, 126.3, 122.0, 120.5, 120.3, 119.7, 111.7, 100.9, 38.6, 32.6; MS (ESI) m/z: 295 [M − H]. Anal. calcd for C21H16N2 (296.37) (%): C, 85.11; H, 5.44; N, 9.45. Found: C, 85.23; H, 5.38; N, 9.39.

3.1.6. General Procedure for the Preparation of 2-(1-Methyl-1H-indol-2-yl)-3-acrylonitriles 5ad

To a stirred solution of the appropriate 2-(1H-indol-2-yl)-3-acrylonitrile derivative 2b, 2d, 2l, or 2p (0.75 mmol) in anhydrous dimethylformamide (5 mL) was added sodium hydride (35 mg, 0.9 mmol, 60% oil dispersion) in one portion at 0 °C. The reaction mixture was stirred for an additional 30 min at ambient temperature. Then, the reaction mixture was cooled to 0 °C and treated with methyl iodide (124 mg, 54 µL, 0.9 mmol). After stirring overnight at ambient temperature, the mixture was diluted with water and the resulting precipitate was collected by filtration, washed with water, and dried. In this manner, the following compounds were obtained.
  • 2-(1-Methyl-1H-indol-2-yl)-3-(p-tolyl)acrylonitrile (5a). Yield: 77% (beige solid); m.p. 90–92 °C; IR νmax (KBr, cm−1): 3040, 2914, 2854, 2223, 1468, 1359, 1343, 1187, 815, 785, 750, 731; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 7.89 (d, J = 8.2 Hz, 2H, 2 × C-Harom), 7.73 (s, 1H, CH), 7.61 (d, J = 7.9 Hz, 1H, C-Harom), 7.54 (d, J = 8.3 Hz, 1H, C-Harom), 7.38 (d, J = 8.1 Hz, 2H, 2 × C-Harom), 7.26 (t, J = 7.3 Hz, 1H, C-Harom), 7.11 (t, J = 7.3 Hz, 1H, C-Harom), 6.80 (s, 1H, C3-H, indole), 3.87 (s, 3H, CH3), 2.40 (s, 3H, CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 146.9, 141.7, 139.1, 135.4, 131.2, 130.1 (two overlapping signals), 129.7 (two overlapping signals), 127.2, 123.3, 121.1, 120.6, 118.2, 110.8, 104.0, 101.0, 31.7, 21.6. Anal. calcd for C19H16N2 (272.34) (%): C, 83.79; H, 5.92; N, 10.29. Found: C, 83.73; H, 5.86; N, 10.41.
  • 3-(4-Methoxyphenyl)-2-(1-methyl-1H-indol-2-yl)acrylonitrile (5b). Yield: 96% (yellow solid); m.p. 113–114 °C; IR νmax (KBr, cm−1): 3050, 3000, 2924, 2853, 2210, 1601, 1506, 1461, 1253, 1180, 1031, 828, 798, 754, 744; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 7.98 (d, J = 8.8 Hz, 2H, 2 × C-Harom), 7.69 (s, 1H, CH), 7.60 (d, J = 7.8 Hz, 1H, C-Harom), 7.52 (d, J = 8.3 Hz, 1H, C-Harom), 7.25 (t, J = 7.1 Hz, 1H, C-Harom), 7.15–7.11 (m, 3H, 3 × C-Harom), 6.76 (s, 1H, C3-H, indole), 3.86 (s, 6H, NCH3 + OCH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 161.9, 146.8, 139.0, 135.6, 131.7 (two overlapping signals), 127.3, 126.5, 123.1, 121.0, 120.6, 118.5, 115.1 (two overlapping signals), 110.8, 103.6, 98.9, 56.0, 31.7. Anal. calcd for C19H16N2O (288.34) (%): C, 79.14; H, 5.59; N, 9.72. Found: C, 79.22; H, 5.51; N; 9.68.
  • 3-[4-(Dimethylamino)phenyl]-2-(1-methyl-1H-indol-2-yl)acrylonitrile (5c). Yield: 92% (brown solid); m.p. 131–132 °C; IR νmax (KBr, cm−1): 3106, 3035, 2887, 2804, 2218, 1608, 1588, 1361, 1197, 818, 792, 732; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 7.89 (d, J = 8.9 Hz, 2H, 2 × C-Harom), 7.57 (d, J = 7.8 Hz, 1H, C-Harom), 7.52–7.49 (m, 2H, C-Harom + CH), 7.22 (t, J = 7.3 Hz, 1H, C-Harom), 7.09 (t, J = 7.4 Hz, 1H, C-Harom), 6.83 (d, J = 9.0 Hz, 2H, 2 × C-Harom), 6.67 (s, 1H, C3-H, indole), 3.84 (s, 3H, CH3), 3.04 (s, 6H, 2 × CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 152.4, 147.6, 138.7, 136.5, 131.7 (two overlapping signals), 127.4, 122.6, 121.1, 120.7, 120.4, 119.4, 112.1 (two overlapping signals), 110.6, 102.7, 93.9, 40.1 (two overlapping signals), 31.6; MS (ESI) m/z: 302 [M + H]+. Anal. calcd. for C20H19N3 (301.38) (%): C, 79.70; H, 6.35; N, 13.94. Found: C, 79.76; H, 6.27; N, 13.97.
  • 2-(1-Methyl-1H-indol-2-yl)-3-(naphthalen-2-yl)acrylonitrile (5d). Yield: 84% (yellow solid); m.p. 160–163 °C; IR νmax (KBr, cm−1): 3054, 2925, 2853, 2207, 1599, 1461, 1356, 931, 803, 791, 744; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 8.46 (s, 1H, C-Harom), 8.17 (d, J = 8.7 Hz, 1H, C-Harom), 8.10 (d, J = 8.7 Hz, 1H, C-Harom), 8.02 (t, J = 6.8 Hz, 1H, C-Harom), 7.94 (s, 1H, CH), 7.69–7.60 (m, 3H, 3 × C-Harom), 7.56 (d, J = 8.3 Hz, 1H, C-Harom), 7.28 (t, J = 7.7 Hz, 1H, C-Harom), 7.13 (t, J = 7.2 Hz, 1H, C-Harom), 6.87 (s, 1H, C3-H, indole), 3.93 (s, 3H, CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 146.6, 139.2, 135.3, 134.2, 133.1, 131.6, 131.1, 129.2, 129.1, 128.5, 128.3, 127.6, 127.2, 125.3, 123.4, 121.2, 120.7, 118.2, 110.9, 104.3, 102.5, 31.9. Anal. calcd for C22H16N2 (308.38) (%): C, 85.69; H, 5.23; N, 9.08. Found: C, 85.75; H, 5.31; N, 8.94.

3.1.7. General Procedure for the Preparation of 2-(1-Acetyl-1H-indol-2-yl)-3-acrylonitriles 6a and 6b and 3-(4-Methoxyphenyl)-2-(1-(methylsulfonyl)-1H-indol-2-yl)acrylonitrile (7)

To a stirred solution of the appropriate 2-(1H-indol-2-yl)-3-acrylonitrile derivative (2d or 2l) (0.75 mmol) in anhydrous dimethylformamide (5 mL) was added sodium hydride (36 mg, 0.9 mmol, 60% oil dispersion) in one portion at 0 °C. The reaction mixture was stirred for an additional 30 min at ambient temperature. Then, the reaction mixture was cooled to 0 °C and treated with acetyl chloride (71 mg, 64 µL, 0.9 mmol) or methanesulfonyl chloride (103 mg, 70 µL, 0.9 mmol). After stirring overnight at ambient temperature, the mixture was diluted with water and the resulting precipitate was collected by filtration, washed with water, and dried. Thus, the obtained crude products 6a, 6b, and 7 were purified on silica gel by column chromatography with dichloromethane as the eluent. In this manner, the following compounds were obtained.
  • 2-(1-Acetyl-1H-indol-2-yl)-3-(4-methoxyphenyl)acrylonitrile (6a). Yield: 42% (beige solid); m.p. 119–122 °C; IR νmax (KBr, cm−1): 3112, 3085, 3022, 2962, 2932, 2836, 2207, 1702, 1604, 1515, 1450, 1366, 1306, 1263, 1189, 1027, 837, 827, 740; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 8.01 (d, J = 8.4 Hz, 1H, C-Harom), 7.94 (d, J = 8.9 Hz, 2H, 2 × C-Harom), 7.68 (d, J = 7.6 Hz, 1H, C-Harom), 7.60 (s, 1H, CH), 7.42 (t, J = 7.3 Hz, 1H, C-Harom), 7.33 (t, J = 7.5 Hz, 1H, C-Harom), 7.13 (d, J = 8.8 Hz, 2H, 2 × C-Harom), 7.05 (s, 1H, C3-H, indole), 3.86 (s, 3H, CH3), 2.80 (s, 3H, CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 170.4, 161.9, 144.8, 136.5, 135.4, 131.6 (two overlapping signals), 129.1, 126.3, 126.0, 124.0, 121.9, 118.0, 115.6, 115.1 (two overlapping signals), 113.3, 101.7, 56.0, 27.3. Anal. calcd for C20H16N2O2 (316.35) (%): C, 75.93; H, 5.10; N, 8.86. Found: C, 75.85; H, 4.88; N, 8.94.
  • 2-(1-Acetyl-1H-indol-2-yl)-3-[4-(dimethylamino)phenyl]acrylonitrile (6b). Yield: 31% (yellow solid); m.p. 164–166 °C; IR νmax (KBr, cm−1): 3115, 2899, 2817, 2204, 1692, 1612, 1579, 1524, 1450, 1367, 1303, 1169, 809, 761, 742; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 8.04 (d, J = 8.2 Hz, 1H, C-Harom), 7.85 (d, J = 9.0 Hz, 2H, 2 × C-Harom), 7.65 (d, J = 7.6 Hz, 1H, C-Harom), 7.47 (s, 1H, CH), 7.39 (t, J = 7.2 Hz, 1H, C-Harom), 7.31 (t, J = 7.4 Hz, 1H, C-Harom), 6.97 (s, 1H, C3-H, indole), 6.83 (d, J = 9.0 Hz, 2H, 2 × C-Harom), 3.05 (s, 6H, 2 × CH3), 2.76 (s, 3H, CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 170.6, 152.5, 146.0, 136.7, 136.1, 131.6 (two overlapping signals), 129.1, 125.7, 124.0, 121.5, 120.8, 115.6, 112.6, 112.1 (two overlapping signals), 112.0, 96.5, 49.1 (two overlapping signals), 27.3; MS (ESI) m/z: 330 [M + H]+. Anal. calcd for C21H19N3O (329.40) (%): C, 76.57; H, 5.81; N, 12.76. Found: C, 76.65; H, 5.75; N, 12.82.
  • 3-(4-Methoxyphenyl)-2-(1-(methylsulfonyl)-1H-indol-2-yl)acrylonitrile (7). Yield: 22% (white solid); m.p. 169–171 °C; IR νmax (KBr, cm−1): 3109, 3075, 3003, 2972, 2922, 2214, 1608. 1512, 1369, 1167, 1070, 833, 773, 747, 556; 1H NMR (400 MHz, DMSO-d6) δ (ppm): 7.95–7.93 (m, 3H, 3 × C-Harom), 7.74–7.71 (m, 2H, C-Harom + CH), 7.47 (t, J = 7.2 Hz, 1H, C-Harom), 7.39 (t, J = 7.5 Hz, 1H, C-Harom), 7.16–7.14 (m, 3H, 2 × C-Harom + C3-H indole), 3.86 (s, 3H, CH3), 3.31 (s, 3H, CH3); 13C NMR (100 MHz, DMSO-d6) δ (ppm): 162.1, 147.0, 137.3, 135.7, 131.8 (two overlapping signals), 129.5, 126.3, 126.1, 124.8, 122.2, 118.1, 115.1 (two overlapping signals), 115.0, 114.0, 99.7, 56.0, 40.9. Anal. calcd for C19H16N2O3S (352.41) (%): C, 64.76; H, 4.58; N, 7.95. Found: C, 64.68; H, 4.64; N, 7.87.

3.2. Biology

3.2.1. Evaluation of In Vitro Antiproliferative Activity

The in vitro anticancer assay was conducted at the National Cancer Institute (NCI) in Bethesda, USA against approximately 60 cancer cell lines [64,65,66,67].
The one-dose data were reported as a mean graph of the percent growth of treated cells. The number reported for the one-dose assay was the growth relative to the no-drug control and relative to the number of cells at time zero. This allowed detection of both growth inhibition (values between 0 and 100) and lethality (values less than 0). For example, a value of 100 meant no growth inhibition. A value of 40 would mean 60% growth inhibition. A value of 0 meant no net growth over the course of the experiment. A value of −40 would mean 40% lethality. A value of −100 meant all cells were dead.
The human tumor cell lines of the cancer screening panel were grown in RPMI 1640 medium containing 5% fetal bovine serum and 2 mM L-glutamine. For a typical screening experiment, 100 μL of cells was inoculated into 96-well microtiter plates at plating densities ranging from 5000 to 40,000 cells/well depending on the doubling time of the individual cell lines. After cell inoculation, the microtiter plates were incubated at 37 °C, under 5% CO2 and 95% air, and 100% relative humidity for 24 h prior to the addition of the tested compounds. After 24 h, two plates of each cell line were fixed in situ with TCA to represent a measurement of the cell population for each cell line at the time of sample addition (Tz). The tested compounds were solubilized in dimethyl sulfoxide at 400-fold the desired final maximum test concentration and stored frozen prior to use. At the time of compound addition, an aliquot of frozen concentrate was thawed and diluted to twice the desired final maximum test concentration with complete medium containing 50 μg/mL gentamicin. Additional 4-fold, 10-fold, or ½ log serial dilutions were made to provide a total of five compound concentrations plus the control. Aliquots of 100 μL of these different compound dilutions were added to the appropriate microtiter wells already containing 100 μL of medium, resulting in the required final compound concentrations. Following compound addition, the plates were incubated for an additional 48 h at 37 °C, under 5% CO2 and 95% air, and 100% relative humidity. For adherent cells, the assay was terminated by the addition of cold TCA. Cells were fixed in situ by the gentle addition of 50 μL of cold 50% (w/v) TCA (final concentration, 10% TCA) and incubated for 60 min at 4 °C. The supernatant was discarded, and the plates were washed five times with tap water and air dried. Sulforhodamine B (SRB) solution (100 μL) at 0.4% (w/v) in 1% acetic acid was added to each well, and plates were incubated for 10 min at room temperature. After staining, unbound dye was removed by washing five times with 1% acetic acid and the plates were air dried. Bound stain was subsequently solubilized with 10 mM Trizma Base, and the absorbance was measured using an automated plate reader at a wavelength of 515 nm. For cells in suspension, the methodology was the same except that the assay was terminated by fixing the settled cells at the bottom of the wells by gently adding 50 μL of 80% TCA (final concentration, 16% TCA). Using seven absorbance measurements [time zero, (Tz), control growth, (C), and growth in the presence of a test compound at five concentration levels (Ti)], the percentage growth was calculated at each of the drug concentration levels. Percentage growth was calculated as:
[(Ti − Tz)/(C − Tz)] × 100 for concentrations for which Ti >/= Tz
[(Ti − Tz)/Tz] × 100 for concentrations for which Ti < Tz
Three dose–response parameters were calculated for each tested compound. Growth inhibition of 50% (GI50) was calculated from [(Ti − Tz)/(C − Tz)] × 100 = 50, which refers to the compound concentration resulting in 50% reduction in the net protein increase (as measured by SRB staining) in control cells during the compound incubation. The compound concentration resulting in total growth inhibition (TGI) was calculated from Ti = Tz. The LC50 (concentration of compound resulting in 50% reduction in the measured protein at the end of the drug treatment as compared to that at the beginning) indicating a net loss of cells following treatment was calculated from [(Ti − Tz)/Tz] × 100 = −50. Values were calculated for each of these three parameters if the level of activity was reached; however, if the effect was not reached or was exceeded, the value for that parameter was expressed as greater or less than the maximum or minimum concentration tested. Furthermore, a mean graph midpoint (MG-MID) was calculated for each of the mentioned parameters, giving an averaged activity parameter over all cell lines. For calculation of the MG-MID, insensitive cell lines were included with the highest concentration tested.

3.2.2. In Vitro Antimicrobial Activity

Antimicrobial activity was tested using the following reference strains: Staphylococcus aureus ATCC 6538, Staphylococcus epidermidis PCM 2118, Escherichia coli ATCC 11229, Pseudomonas aeruginosa ATCC 15442, Enterococcus faecalis ATCC 11420, and Candida albicans ATCC 10231. Antibacterial activity of selected compounds (2i, 2q, 2s, and 2x) was evaluated against clinical S. aureus strains isolated from various clinical samples (nasal mucus, saliva, sputum, pus, and blood) derived from the Department of Oral Microbiology collection. Minimal inhibitory concentrations (MICs) for tested compounds were determined using a broth dilution method as recommended by the Clinical Laboratory Standards Institute (CLSI) guidelines [83]. Polypropylene 96-well plates with the investigated compounds serially diluted in Mueller Hinton Broth 2 (Sigma-Aldrich, St. Louis, MO, USA) (or in Sabouraud Dextrose Broth (BD Difco) for C. albicans) and initial inoculum 5 × 105 CFU/mL were incubated at 37 °C for 18 h (or 24–48 h for C. albicans). MIC was taken as the lowest compound concentration at which observable growth was inhibited. Minimal bactericidal concentration (MBC) was determined in a sample taken from each test tube in which no growth was observed in the MIC assay. A loopful (10 µL) of the tested sample was transferred to Tryptic Soy Agar (TSA, BD Difco) (or Sabouraud Dextrose Agar (Sigma-Aldrich) for C. albicans) and incubated at 37 °C for 48 h. MBC was taken as the lowest concentration of tested compound that resulted in more than 99.9% reduction of the initial inoculum. Solutions of compounds were made fresh on the day of the assay. All experiments were performed in triplicate. The reference strains were stored at −80 °C in Tryptic Soy Broth (TSB, Oxoid, England) supplemented with 15% glycerol.

3.3. Computational Studies

3.3.1. Preparation of Ligands and Proteins for Modeling

Crystal structures of the enzyme–DNA complexes were obtained from the Protein Data Bank [84]. In the study, the following proteins were used: caspase-3 from Homo sapiens (PDB code 2xyp) [72], caspase-9 from H. sapiens (PDB code 2ar9) [73], tubulin from Ovis aries (PDB code 5eyp) [74], penicillin-binding protein 4 from E. coli (PDB code 2ex8) [75], and β-lactamase from E. coli (PDB code 1fqg) [76]. The proteins were prepared using MAKE RECEPTOR software [85,86,87]. The pocket around the ligand bound in the crystal structure was generated automatically and was not adjusted, which resulted in grid boxes of various sizes (specifically 4946, 2022, 6206, 4292, and 4491 Ǻ for 2xyp, 2ar9, 5eyp, 2ex8, and 1fqg, respectively). A slow and effective “Molecular” method was used for “Cavity detection,” i.e., detection of binding sites. Outer and inner contours of the grid box were also calculated automatically using the “Balanced” settings for the “Site Shape Potential” calculation, which once more resulted in different outer contour sizes depending on the bound ligand, specifically 1645, 1219, 1339, 1562, and 1520Ǻ, respectively. The inner contours were disabled. No constraints for docking calculations were used.
The structures of compounds were prepared in SMILES notation. A library of conformers was generated with the OMEGA default settings, which resulted in a maximum of 200 conformers per ligand [87,88].

3.3.2. Molecular Docking

The compounds were docked using the FRED algorithm [85,86]. The docking resolution was set to high while the other settings were set as default. Ten docking solutions were inspected visually and the best-ranked HYBRID-calculated conformations were used for analysis and representation. The docking protocols were validated by re-docking the co-crystallized ligands with RMSD values below 2 Å for each binding pocket.

3.3.3. ADME/Drug-Likeness Calculation

The physicochemical, pharmacokinetic, and drug-likeness properties of compounds 2l, 2x, and 5ad were predicted using the SwissADME web tool and PreADMET server, which are available online [89,90].

4. Conclusions

A series of 2-(1H-indol-2-yl)-3-acrylonitriles 2ax and their derivatives 3, 4ac, 5ad, 6ab, and 7 were synthesized and evaluated in terms of their anticancer and antimicrobial properties.
Analysis of the structure–activity relationship for the antiproliferative activity of all of the prepared compounds against a panel of approximately 60 human cancer cell lines revealed that heterocyclic replacement of the aromatic ring at position 3 of the acrylonitrile moiety led to a strong decrease in activity. The acrylonitrile moiety was required for cell growth inhibition because either replacing this moiety with an imino-acetonitrile group or its saturation yielded compounds with poor or lacking activity. Otherwise, the introduction of a methyl group at position 1 of the 2-(1H-indol-2-yl)-3-acrylonitrile scaffold improved antitumor potency. The 2-(1H-indol-2-yl)-3-acrylonitriles 2l and 5ad were the most potent of all tested derivatives, exhibiting significant activity against the tumor cell lines investigated (full panel GI50 MD-MIG = 0.38–7.91 μM). Particularly, compound 5c bearing a methyl group at position 1 of the indole ring and a 4-(dimethylamino)phenyl group at position 3 of the acrylonitrile moiety demonstrated optimal properties (full panel GI50 MD-MIG = 0.38 μM and TGI = 0.0866–5.06 μM against 13 cell lines of different cancer subpanels) and therefore may serve as a useful scaffold for further development of more potent antitumor agents.
Moreover, 2-(1H-indol-2-yl)-3-acrylonitriles 2ac and 2ex were tested against Gram-positive and Gram-negative bacterial strains, namely Staphylococcus aureus, Staphylococcus epidermi, Enterococcus faecalis, Escherichia coli, and Pseudomonas aeruginosa, as well as fungal species Candida albicans. The majority of the tested 2-(1H-indol-2-yl)-3-acrylonitriles were found to be rather inactive; however, compounds 2i, 2q, 2s, and 2x showed promising potential against some bacterial species. An interesting compound is 2-(1H-indol-2-yl)-3-(1H-pyrrol-2-yl)acrylonitrile (2x), which exhibited relatively high antibacterial activity against all Gram-positive bacteria tested (MIC = 8–32 μg/mL, MBC = 32 μg/mL). Furthermore, it was the only compound among those tested that showed activity against Gram-negative bacteria, e.g., E. coli (MIC = MBC = 32 μg/mL). The analogue 2x also effectively inhibited the growth of various strains of S. aureus isolated from clinical specimens (MIC values of 16 μg/mL). In addition, derivative 2x was found to have pronounced antifungal activity against C. albicans (MIC = 4 μg/mL, MFC = 8 μg/mL). Based on these findings, compound 2x seems to be leading compound for further development as an antimicrobial agent.
The results of the docking studies suggested that, like other heteroaryl-acrylonitriles, the obtained compounds may exert their cancer cell growth inhibitory effects through interaction with tubulin in the colchicine-binding site and/or apoptotic caspase-3 as well as caspase-9, whereas their antibacterial activity may be related to interaction with PBP4 and/or β-lactamase. However, it is too early to speculate on the mode of action of these indole-acrylonitriles. For example, a number of mechanisms could be responsible for their antitumor effects, e.g., specific interaction with cellular receptors and enzymes or 1,4-nucleophilic addition of thiol to the acrylonitrile double bond [49,50]. As Michael acceptors, acrylonitriles react in vitro or in vivo with sulfur-containing nucleophiles such as glutathione (GSH) and proteins. It is worth noting that acrylonitrile-based Michael acceptors activated by aryl or heteroaryl electron-withdrawing groups have been reported as reversible, cysteine-targeted kinase inhibitors [91]. It is well known that such covalent, electrophilic “warheads” targeting cysteine residues constitute a promising approach in drug development [92]. Therefore, it remains a challenge to explore in depth the mode of action and the pharmacodynamic features of these compounds, which will be the next goal of this project.
According to the predicted ADME/drug-likeness properties, the most active compounds 2l, 2x, and 5ad were shown to be drug-likeness molecules.

Supplementary Materials

CCDC 2130049 contains the supplementary crystallographic data for this paper. These data can be obtained free of charge via http://www.ccdc.cam.ac.uk/conts/retrieving.html (or from the CCDC, 12 Union Road, Cambridge CB2 1EZ, UK; Fax: +44-1223-336033; E-mail: deposit@ccdc.cam.ac.uk) (accessed on 1 January 2023). The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph16070918/s1, Table S1: FRED Chemgauss4 scores of the docked ligands 1, 2ax, 3, 4ac, 5ad, 6ab, and 7; Table S2: Two-dimensional diagrams of interactions created by compounds 2a and 5ad in the active site of caspase-3 (2xyp) generated by BIOVA Discovery Studio Visualizer and Pose View; Table S3: Two-dimensional diagrams of interactions created by compounds 2l and 5ad in the active site of caspase-9 (2ar9) generated by BIOVA Discovery Studio Visualizer and Pose View; Table S4: Two-dimensional diagrams of interactions created by compounds 2l and 5ad in the active site of tubulin (5eyp) generated by BIOVA Discovery Studio Visualizer and Pose View; Table S5: Two-dimensional diagrams of interactions created by compound 2x in the active site of penicillin-binding protein 4 (2ex8) generated by BIOVA Discovery Studio Visualizer and Pose View; Table S6. Two-dimensional diagrams of interactions created by compound 2x in the active site of β-lactamase (1fqg) generated by BIOVA Discovery Studio Visualizer and Pose View; Table S7: Predicted physicochemical, pharmacokinetic, and drug-likeness properties of compounds 2l, 2x, and 5ad; Table S8: Predicted human intestinal absorption, Caco-2 cell and MDCK cell permeabilities for compounds 2l, 2x, and 5ad.

Author Contributions

Conceptualization, A.K.; methodology, A.K., K.G. (Katarzyna Garbacz) and M.G.; software, J.F. and J.K.; validation, A.K., K.G. (Katarzyna Garbacz) and K.G. (Karol Gzella); formal analysis, A.K., K.G. (Katarzyna Garbacz), M.G. and Ł.B.; investigation, A.K., K.G. (Karol Gzella), M.J., M.G., J.F., J.K. and A.O.; writing—original draft preparation, A.K., M.G., J.F. and Ł.B.; writing—review and editing, A.K. and K.G. (Katarzyna Garbacz); visualization, A.K., M.G., J.F., Ł.B. and J.K.; supervision, A.K.; project administration, A.K.; funding acquisition, A.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was founded by the Statutory Activity of the Medical University of Gdansk (ST 01-50023/0004931/01/513/513/0/2023).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Acknowledgments

NMR spectra were partially acquired at The Nuclear Magnetic Resonance Spectroscopy Laboratory, Collegium Medicum, Jagiellonian University, Kraków, Poland. X-ray structure analysis was carried out in The Structural Research Laboratory, Faculty of Chemistry, Adam Mickiewicz University, Poznań, Poland. The research in this work was supported by Gdańsk University of Technology (DEC-2/2021/IDUB/V.6/Si grant under the SILICIUM—‘Excellence Initiative—Research University’ program) (A.O.). The authors are grateful to Joel Morris, Drug Synthesis and Chemistry Branch (DSCB), US National Cancer Institute (Bethesda, MD, USA), for the in vitro screening. We thank OpenEye Scientific Software, Santa Fe, NM., for free academic licenses for the use of their software issued to Jarosław Sączewski.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kumar, S.; Ritika. A brief review of the biological potential of indole derivatives. Future J. Pharm. Sci. 2020, 6, 121. [Google Scholar] [CrossRef]
  2. Sravanthi, T.V.; Manju, S.L. Indoles—A promising scaffold for drug development. Eur. J. Pharm. Sci. 2016, 91, 1–10. [Google Scholar] [CrossRef] [PubMed]
  3. Chadha, N.; Silakari, O. Indoles as therapeutics of interest in medicinal chemistry: Bird’s eye view. Eur. J. Med. Chem. 2017, 134, 159–184. [Google Scholar] [CrossRef]
  4. Kumaria, A.; Singh, R.K. Medicinal chemistry of indole derivatives: Current to future therapeutic prospectives. Bioorg. Chem. 2019, 89, 103021. [Google Scholar] [CrossRef]
  5. Kumar, D.; Sharma, S.; Kalra, S.; Singh, G.; Monga, V.; Kumar, B. Medicinal perspective of indole derivatives: Recent developments and structure-activity relationship studies. Curr. Drug Targets 2020, 21, 864–891. [Google Scholar]
  6. Ferreira, S.H.; Moncada, S.; Vane, J.R. Indomethacin and aspirin abolish prostaglandin release from the spleen. Nature 1971, 231, 237–239. [Google Scholar] [CrossRef] [PubMed]
  7. Mazzotta, S.; Frattaruolo, L.; Brindisi, M.; Ulivieri, C.; Vanni, F.; Brizzi, A.; Carullo, G.; Cappello, A.R.; Aiello, F. 3-Amino-alkylated indoles: Unexplored green products acting as anti-inflammatory agents. Future Med. Chem. 2020, 12, 5–17. [Google Scholar] [CrossRef]
  8. Jasiewicz, B.; Kozanecka Okupnik, W.; Przygodzki, M.; Warżajtis, B.; Rychlewska, U.; Pospieszny, T.; Mrówczyńska, L. Synthesis, antioxidant and cytoprotective activity evaluation of C-3 substituted indole derivatives. Sci. Rep. 2021, 11, 15425. [Google Scholar] [CrossRef]
  9. Gurer-Orhan, H.; Karaaslan, C.; Ozcan, S.; Firuzi, O.; Tavakkoli, M.; Saso, L.; Suzen, S. Novel indole-based melatonin analogues: Evaluation of antioxidant activity and protective effect against amyloid β-induced damage. Bioorg. Med. Chem. 2016, 24, 1658–1664. [Google Scholar] [CrossRef]
  10. Kerzare, D.R.K.; Menghani, S.S.; Khedekar, P.B. Synthesis, characterization, antidepressant activity and docking studies of some novel indole bearing azetidinone derivatives. Drugs 2018, 52, 110–120. [Google Scholar] [CrossRef] [Green Version]
  11. Gras, J.; Llenas, J.; Jansat, J.M.; Jáuregui, J.; Cabarrocas, X.; Palacios, J.M. Almotriptan, a new anti-migraine agent: A review. CNS Drug Rev. 2002, 8, 217–234. [Google Scholar] [CrossRef]
  12. Tan, C.; Yang, S.-J.; Zhao, D.-H.; Li, J.; Yin, L.-Q. Antihypertensive activity of indole and indazole analogues: A review. Arab. J. Chem. 2022, 15, 103756. [Google Scholar] [CrossRef]
  13. Muhammad 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]
  14. Zhu, Y.; Zhao, J.; Luo, L.; Gao, Y.; Bao, H.; Li, P.; Zhang, H. Research progress of indole compounds with potential antidiabetic activity. Eur. J. Med. Chem. 2021, 223, 113665. [Google Scholar] [CrossRef] [PubMed]
  15. Dorababu, A. Indole—A promising pharmacophore in recent antiviral drug discovery. RSC Med. Chem. 2020, 11, 1335–1353. [Google Scholar] [CrossRef]
  16. Reddy, G.S.; Manojit Pal, M. Indole derivatives as anti-tubercular agents: An overview on their synthesis and biological activities. Curr. Med. Chem. 2021, 28, 4531–4568. [Google Scholar] [CrossRef]
  17. Nieto, M.J.; Lupton, H.K. Indole and indoline scaffolds in antimicrobials: Overview, synthesis and recent advances in antimicrobial research. Curr. Med. Chem. 2021, 28, 4828–4844. [Google Scholar] [CrossRef] [PubMed]
  18. Liu, Y.; Cui, Y.; Lu, L.; Gong, Y.; Han, W.; Piao, G. Natural indole-containing alkaloids and their antibacterial activities. Arch. Pharm. 2020, 353, e2000120. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, Y.; Rosado-Lugo, J.D.; Datta, P.; Sun, Y.; Cao, Y.; Banerjee, A.; Yuan, Y.; Parhi, A.K. Evaluation of a conformationally constrained indole carboxamide as a potential efflux pump inhibitor in Pseudomonas aeruginosa. Antibiotics 2022, 11, 716. [Google Scholar] [CrossRef]
  20. Xuecheng, C.; Liang, H.; Yanpeng, X.; Yurong, Z.; Yue, L.; Yalan, P.; Zhong, C.; Jie, Z.; Zhijian, Y.; Shiqing, H. Development of 2-alkyl-5-((phenylsulfonyl)oxy)-1H-indole-3-carboxylate derivatives as potential anti-biofilm agents. ChemistrySelect 2023, 8, e202204226. [Google Scholar] [CrossRef]
  21. 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] [PubMed] [Green Version]
  22. Qin, H.-L.; Liu, J.; Fang, W.-Y.; Ravindar, L.; 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]
  23. Wan, Y.; Li, Y.; Yan, Y.; Yan, M.; Tang, Z. Indole: A privileged scaffold for the design of anti-cancer agents. Eur. J. Med. Chem. 2019, 183, 111691. [Google Scholar] [CrossRef]
  24. Dadashpour, S.; Emami, S. Indole in the target-based design of anticancer agents: A versatile scaffold with diverse mechanisms. Eur. J. Med. Chem. 2018, 150, 9–29. [Google Scholar] [CrossRef]
  25. Mehra, A.; Sharma, V.; Verma, A.; Venugopal, S.; Mittal, A.; Singh, G. Indole derived anticancer agents. ChemistrySelect 2022, 7, e202202361. [Google Scholar] [CrossRef]
  26. Almagro, L.; Fernández-Pérez, F.; Pedreño, M.A. Indole alkaloids from Catharanthus roseus: Bioproductiom and their effect on human health. Molecules 2015, 20, 2973–3000. [Google Scholar] [CrossRef] [Green Version]
  27. Shirley, M. Dacomitinib: First global approval. Drugs 2018, 78, 1947–1953. [Google Scholar] [CrossRef]
  28. Amin, S.A.; Ghosh, K.; Mondal, D.; Jha, T.; Gayen, S. Exploring indole derivatives as myeloid cell leukaemia-1 (Mcl-1) inhibitors with multi-QSAR approach: A novel hope in anti-cancer drug discovery. New J. Chem. 2020, 44, 17494–17506. [Google Scholar] [CrossRef]
  29. Alnabulsi, S.; Al-Hurani, E.A. Pim kinase inhibitors in cancer: Medicinal chemistry insights into their activity and selectivity. Drug Discov. 2020, 25, 2062–2069. [Google Scholar] [CrossRef]
  30. Panathur, N.; Gokhale, N.; Dalimba, U.; Koushik, P.V.; Yogeeswari, P.; Dharmarajan, S. New indole–isoxazolone derivatives: Synthesis, characterisation and in vitro SIRT1 inhibition studies. Bioorg. Med. Chem. Lett. 2015, 25, 2768–2772. [Google Scholar] [CrossRef] [PubMed]
  31. Xie, F.; Kniess, T.; Neuber, C.; Deuther-Conrad, W.; Mamat, C.; Lieberman, B.P.; Liu, B.; Mach, R.H.; Brust, P.; Steinbach, J.; et al. Novel indole-based sigma-2 receptor ligands: Synthesis, structure-affinity relationship and antiproliferative activity. Med. Chem. Commun. 2015, 6, 1093–1103. [Google Scholar] [CrossRef] [Green Version]
  32. Atadja, P. Development of the pan-DAC inhibitor panobinostat (LBH589): Successes and challenges. Cancer Lett. 2009, 280, 233–241. [Google Scholar] [CrossRef]
  33. Li, A.-L.; Hao, Y.; Wang, W.-Y.; Liu, Q.-S.; Sun, Y.; Gu, W. Design, synthesis, and anticancer evaluation of novel indole derivatives of ursolic acid as potential topoisomerase II inhibitors. Int. J. Mol. Sci. 2020, 21, 2876. [Google Scholar] [CrossRef] [Green Version]
  34. Tan, O.U.; Zengin, M. Insights into the chemistry and therapeutic potential of acrylonitrile derivatives. Arch. Pharm. 2022, 355, e2100383. [Google Scholar]
  35. Sivaramakarthikeyan, R.; Iniyaval, S.; Saravanan, V.; Lim, W.-M.; Mai, C.-W.; Ramalingan, C. Molecular hybrids integrated with benzimidazole and pyrazole structural motifs: Design, synthesis, biological evaluation, and molecular docking studies. ACS Omega 2020, 209, 10089–10098. [Google Scholar] [CrossRef]
  36. Ahamed, J.I.; Priya, M.; Vinothkumar, P.; Sathyamoorthy, K.; MuraliManohar, P.; Liu, J.; Valan, M.F. A combined experimental and DFT computations study of novel (E)-3-(benzofuran-2-yl)-2-(thiophen-2-yl)acrylonitrile(TACNBNF): Insight into the synthesis, single crystal XRD, NMR, vibrational spectral analysis, in vitro antioxidant and in silico molecular docking investigation with human peroxiredoxin 5 protein. J. Mol. Struct. 2020, 1202, 127241. [Google Scholar]
  37. Solangi, M.; Kanwal; Khan, K.M.; Saleem, F.; Hameed, S.; Iqbal, J.; Shafique, Z.; Qureshi, U.; Ul-Haq, Z.; Taha, M.; et al. Indole acrylonitriles as potential anti-hyperglycemic agents: Synthesis, α-glucosidase inhibitory activity and molecular docking studies. Bioorg. Med. Chem. 2020, 28, 115605. [Google Scholar] [CrossRef]
  38. Han, Y.-S.; Penthala, N.R.; Oliveira, M.; Mespléde, T.; Xu, H.; Quan, Y.; Crooks, P.A.; Wainberg, M.A. Identification of resveratrol analogs as potent anti-dengue agents using a cell-based assay. J. Med. Virol. 2017, 89, 397–407. [Google Scholar] [CrossRef] [PubMed]
  39. Pan, T.; He, X.; Chen, B.; Chen, H.; Geng, G.; Luo, H.; Zhang, H.; Bai, C. Developmnent of benzimidazole derivatives to inhibit HIV-1 replication through protecting APOBEC3G protein. Eur. J. Med. Chem. 2015, 95, 500–513. [Google Scholar] [CrossRef]
  40. Sharma, K.; Shrivastava, A.; Mehra, R.N.; Deora, G.S.; Alam, M.M.; Zaman, M.S.; Akhter, M. Synthesis of novel benzimidazole acrylonitriles for inhibition of Plasmodium falciparum growth by dual target inhibition. Arch. Pharm. Chem. Life Sci. 2017, 351, e1700251. [Google Scholar] [CrossRef]
  41. Sirim, M.M.; Krishna, V.S.; Sriram, D.; Tan, O.U. Novel benzimidazole-acrylonitrile hybrids and their derivatives: Design, synthesis and antimycobacterial activity. Eur. J. Med. Chem. 2020, 188, 112010. [Google Scholar] [CrossRef]
  42. De-la-Torre, P.; Saavedra, L.A.; Caballero, J.; Quiroga, J.; Alzate-Morales, J.H.; Cabrera, M.G.; Trilleras, J. A novel class of selective acetylcholinesterase inhibitors: Synthesis and evaluation of (E)-2-(benzo[d]thiazol-2-yl)-3-heteroarylacrylonitriles. Molecules 2012, 17, 12072–12085. [Google Scholar] [CrossRef] [Green Version]
  43. De-la-Torre, P.; Treuer, A.V.; Gutierrez, M.; Poblete, H.; Alzate-Morales, J.H.; Trilleras, J.; Astudillo-Saavedra, L.; Caballero, J. Synthesis and in silico analysis of the quantitative structure-activity relationship of heteroaryl-acrylonitriles as AChE inhibitors. J. Taiwan Inst. Chem. Eng. 2016, 59, 45–60. [Google Scholar] [CrossRef]
  44. AlNeyadi, S.; Salem, A.A.; Ghattas, M.A.; Atatrech, N.; Abdou, I. Antibacterial activity and mechanism of action of the benzazole acrylonitrile-based compounds: In vitro, spectroscopic, and docking studies. Eur. J. Med. Chem. 2017, 136, 270–282. [Google Scholar] [CrossRef] [PubMed]
  45. Perin, N.; Hok, L.; Beč, A.; Vanstreels, E.; Daelemans, D.; Vianello, R.; Hranjec, M. N-substituted benzimidazole acrylonitryles as in vitro tubulin polymerization inhibitors: Synthesis, biological activity and computational analysis. Eur. J. Med. Chem. 2021, 211, 113003. [Google Scholar] [CrossRef] [PubMed]
  46. Beč, A.; Hok, L.; Persoons, L.; Vanstreels, E.; Daelemans, D.; Vianello, R.; Hranjec, M. Synthesis, computational analysis, and antiproliferative activity of novel benzimidazole acrylonitriles as tubulin polymerization inhibitors: Part 2. Pharmaceuticals 2021, 14, 1052. [Google Scholar] [CrossRef] [PubMed]
  47. Riu, F.; Sanna, L.; Ibba, R.; Piras, S.; Bordono, V.; Scorciapino, M.A.; Lai, M.; Sestito, S.; Bagella, L.; Carta, A. A comprehensive assessment of a new series of 5′,6′-difluorobenzotriazole-acrylonitrile derivatives as mictrotubule targeting agents (MTAs). Eur. J. Med. Chem. 2021, 222, 113590. [Google Scholar] [CrossRef]
  48. Briguglio, I.; Laurini, E.; Pirisi, M.A.; Piras, S.; Corona, P.; Fermeglia, M.; Pricl, S.; Carta, A. Triazolopyridinyl-acrylonitrile derivatives as antimicrotubule agents: Synthesis, in vitro and in silico characterization of antiproliferative activity, inhibition of tubulin polymerization and binding thermodynamics. Eur. J. Med. Chem. 2017, 141, 460–472. [Google Scholar] [CrossRef] [PubMed]
  49. Hranjec, M.; Pavlović, G.; Marjanović, M.; Kralj, M.; Kaminska-Zamola, G. Benzimidazole derivatives related to 2,3-acrylonitriles, benzimidazo[1,2-a]quinolines and fluorenes: Synthesis, antitumor evaluation in vitro and crystal structure determination. Eur. J. Med. Chem. 2010, 45, 2405–2417. [Google Scholar] [CrossRef]
  50. Sączewski, F.; Reszka, P.; Gdaniec, M.; Grünert, R.; Bednarski, P.J. Synthesis, X-ray crystal structures, stabilities, an in vitro cytotoxic activities of new heteroarylacrylonitriles. J. Med. Chem. 2004, 47, 3438–3449. [Google Scholar] [CrossRef]
  51. Oberhuber, N.; Ghosh, H.; Nitzsche, B.; Dandawate, P.; Höpfner, M.; Schobert, R.; Biersack, B. Synthesis and anticancer evaluation of new indole-based tyrphostin derivatives and their (p-cymene)dichloridoruthenium(II) complexes. Int. J. Mol. Sci. 2023, 24, 854. [Google Scholar] [CrossRef] [PubMed]
  52. Tcherniuk, S.; Skoufias, D.A.; Labriere, C.; Rath, O.; Gueritte, F.; Guillou, C.; Kozielski, F. Relocation of Aurora B and surviving from centromeres to the central spindle impaired by a kinesin-specific MKLP-2 inhibitor. Angew. Chem. Int. Ed. 2010, 49, 8228–8231. [Google Scholar] [CrossRef]
  53. Labriere, C.; Talapatra, S.K.; Thoret, S.; Bougeret, C.; Kozielski, F.; Guillou, C. New MKLP-2 inhibitors in the paprotrain series: Design, synthesis and biological evaluations. Bioorg. Med. Chem. 2016, 24, 721–734. [Google Scholar] [CrossRef] [PubMed]
  54. Sączewski, F.; Stencel, A.; Bieńczak, A.M.; Langowska, K.A.; Michaelis, M.; Werel, W.; Hałasa, R.; Reszka, P.; Bednarski, P.J. Structure-activity relationships of novel heteroaryl-acrylonitriles as cytotoxic and antibacterial agents. Eur. J. Med. Chem. 2008, 43, 1847–1857. [Google Scholar] [CrossRef]
  55. Baciocchi, E.; Muraglia, E.; Sleiter, G. Homolytic substitution reactions of electron-rich pentatomic heteroaromatics by electrophilic carbon-centered radicals. Synthesis of α-heteroarylacetic acids. J. Org. Chem. 1992, 57, 6817–6820. [Google Scholar] [CrossRef]
  56. Jiang, Y.; Wang, B.; Liu, D.; Xia, D.; Liu, Z.; Li, L.; Deng, G.; Yang, X. Aryl acrylonitriles synthesis enable by palladium-catalyzed α-alkenylation of arylacetonitriles with vinyl halides/triflates. Front. Chem. 2022, 10, 1091566. [Google Scholar] [CrossRef] [PubMed]
  57. Treuer, A.V.; De-La-Torre, P.; Gutiérrez, M.I. Synthesis of new (E)-2-(1H-indole-3-ylcarbonyl)-3-heteroaryl-acrylonitriles via microwave-assisted Knoevenagel condensation. J. Chem. 2017, 2017, 8418930. [Google Scholar] [CrossRef] [Green Version]
  58. Schaller, E.; Ma, A.; Gosch, C.L.; Klefenz, A.; Schaller, D.; Goehringer, N.; Kaps, L.; Schuppan, D.; Volkamer, A.; Schobert, R.; et al. New 3-aryl-2-(2-thienyl)acrylonitriles with high activity against hepatoma cells. Int. J. Mol. Sci. 2021, 22, 2243. [Google Scholar] [CrossRef] [PubMed]
  59. Ying, A.; Wang, L.; Qiu, F.; Hu, H.; Yang, J. Magnetic nanoparticle supported amine: An efficient and environmental benign catalyst for versatile Knoevenagel condensation under ultrasound irradiation. Comptes Rendus Chim. 2015, 18, 223–232. [Google Scholar] [CrossRef]
  60. Wilkens, J.; Kühling, A.; Blechert, S. Hetero-Cope rearrangements-VI. Short and stereoselective synthesis of 2-vinylindoles by a tandem process. Tetrahedron 1987, 43, 3237–3246. [Google Scholar] [CrossRef]
  61. Ying, A.; Qiu, F.; Wu, C.; Hu, H.; Yang, J. Ionic tagged amine supported on magnetic nanoparticles: Synthesis and application for versatile catalytic Knoevenagel condensation in water. RSC Adv. 2014, 4, 33175–33183. [Google Scholar] [CrossRef]
  62. Ying, A.; Ni, Y.; Xu, S.; Liu, S.; Yang, J.; Li, R. Novel DABCO based ionic liquids: Green and efficient catalysts with dual catalytic roles for aqueous Knoevenagel condensation. Int. Eng. Chem. Res. 2014, 53, 5678–5682. [Google Scholar] [CrossRef]
  63. Gao, X.; Gao, C.; Gao, R. Knoevenagel condensation reaction using ionic liquid [ADPQ][CF3SO3] as green and reusable catalyst. Asian J. Chem. 2015, 27, 2145–2148. [Google Scholar] [CrossRef]
  64. Grever, M.R.; Schepartz, S.A.; Chabner, B.A. The National Cancer Institute: Cancer drug discovery and development program. Semin. Oncol. 1992, 19, 622–638. [Google Scholar] [PubMed]
  65. Boyd, M.R.; Paull, K.D. Some practical considerations and applications of the National Cancer Institute in vitro anticancer drug discovery screen. Drug Dev. Res. 1995, 34, 91–109. [Google Scholar] [CrossRef]
  66. Shoemaker, R.H. The NCI60 human tumour cell line anticancer drug screen. Nat. Rev. 2006, 6, 813–823. [Google Scholar] [CrossRef]
  67. NCI-60 Screening Methodology. Available online: https://dtp.cancer.gov/discovery_development/nci-60/methodology.htm (accessed on 27 December 2022).
  68. Horváth, A.; Dobay, O.; Sahin-Tóth, J.; Juhász, E.; Pongrácz, J.; Iván, M.; Fazakas, E.; Kristóf, K. Characterisation of antibiotic resistance, virulence, clonality and mortality in MRSA and MSSA bloodstream infections at a tertiary-level hospital in Hungary: A 6 year retrospective study. Ann. Clin. Microbiol. Antimicrob. 2020, 19, 17. [Google Scholar] [CrossRef]
  69. McLoughlin, E.C.; O’Boyle, N.M. Colchicine-binding site inhibitors from chemistry to clinic: A review. Pharmaceuticals 2020, 13, 8. [Google Scholar] [CrossRef] [Green Version]
  70. Sun, K.; Sun, Z.; Zhao, F.; Shan, G.; Meng, Q. Recent advances in research of colchicine binding site inhibitors and their interaction modes with tubulin. Future Med. Chem. 2021, 13, 839–858. [Google Scholar] [CrossRef]
  71. Carneiro, B.A.; El-Deiry, W.S. Targeting apoptosis in cancer therapy. Nat. Rev. Clin. Oncol. 2020, 17, 395–417. [Google Scholar] [CrossRef]
  72. Ganesan, R.; Jelakovic, S.; Mittl, P.R.E.; Caflisch, A.; Grutter, M.G. In silico identification and crystal structure validation of caspase-3 inhibitors without a P1 aspartic acid moiety. Acta Cryst. 2011, F67, 842–850. [Google Scholar]
  73. Chao, Y.; Shiozaki, E.N.; Srinivasula, S.M.; Rigotti, D.J.; Fairman, R.; Shi, Y. Engineering a dimeric caspase-9: A re-evaluation of the induced proximity model for caspase activation. PLoS Biol. 2005, 3, e183. [Google Scholar] [CrossRef] [Green Version]
  74. Ahmad, S.; Pecqueur, L.; Dreie, B.; Hamdane, H.; Aumont-Nicaise, L.; Plückthun, A.; Knossow, M.; Gigant, B. Destabilizing an interacting motif strengthens the association of adesigned ankyrin repeat protein with tubulin. Sci. Rep. 2016, 6, 28922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Kishida, H.; Unzai, S.; Roper, D.I.I.; Lloyd, A.; Park, S.-Y.; Tame, J.R.H. Crystal Structure of Penicillin Binding Protein 4 (dacB) from Escherichia coli, both in the Native Form and Covalently Linked to Various Antibiotics. Biochemistry 2006, 45, 783–792. [Google Scholar] [CrossRef]
  76. Strynadka, N.C.J.; Adachi, H.; Jensen, S.E.; Johns, K.; Sielecki, A.; Betzel, C.; Sutoh, K.; James, M.N.G. Molecular structure of the acyl-enzyme intermediate in β-lactam hydrolysis at 1.7 Å resolution. Nature 1992, 359, 700–705. [Google Scholar] [CrossRef]
  77. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef] [Green Version]
  78. 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. 2012, 64, 4–17. [Google Scholar] [CrossRef]
  79. Rigaku Oxford Diffraction. CrysAlisPro Software System; Version 1.171.38.46; Rigaku Corporation: Oxford, UK, 2015. [Google Scholar]
  80. Sheldrick, G.M. SHELXT-Integrated space-group and crystal-structure determination. Acta Cryst. 2015, A71, 3–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Sheldrick, G.M. Crystal structure refinement with SHELXL. Acta Cryst. 2015, C71, 3–8. [Google Scholar]
  82. Dolomanov, O.V.; Bourhis, L.J.; Gildea, R.J.; Howard, J.A.K.; Puschmann, H. OLEX2: A complete structure solution, refinement and analysis program. J. Appl. Cryst. 2009, 42, 339–341. [Google Scholar] [CrossRef]
  83. Clinical and Laboratory Standards Institute. Performance Standards for Antimicrobial Susceptibility Testing; Twenty-Second Informational Supplement; Document M100-S22; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2017. [Google Scholar]
  84. RCBS Protein Data Bank. Available online: https://www.rcsb.org/ (accessed on 3 April 2023).
  85. Release 4.1.2.2; OpenEye Scientific Software, Inc.: Santa Fe, NM, USA; Available online: www.eyesopen.com (accessed on 3 April 2023).
  86. McGann, M.R.; Almond, H.R.; Nicholls, A.; Grant, J.A.; Brown, F.K. Gaussian docking functions. Biopolymers 2003, 68, 76–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. McGaughey, G.B.; Sheridan, R.P.; Bayly, C.I.; Culberson, J.C.; Kreatsoulas, C.; Lindsley, S.; Maiorov, V.; Truchon, J.-F.; Cornell, W.D. Comparisan of topological, shape, and docking methods in virtual screening. J. Chem. Inf. Model. 2007, 47, 1504–1519. [Google Scholar] [CrossRef] [PubMed]
  88. Release 4.2.0.1; OpenEye Scientific Software, Inc.: Santa Fe, NM, USA; Available online: www.eyesopen.com (accessed on 3 April 2023).
  89. SwissADME: A Free Web Tool to Compute Physicochemical Descriptors as Well as to Predict ADME Parameters, Pharmacokinetic Properties, Druglike Nature and Medicinal Chemistry Friendliness of One or Multiple Small Molecules to Support Drug Discovery. Available online: http://www.swissadme.ch (accessed on 16 February 2023).
  90. PreADMET: A Free Web Server. Available online: http://preadmet.bmdrc.kr (accessed on 1 June 2023).
  91. Krishnan, S.; Miller, R.M.; Tian, B.; Mullins, R.D.; Jacobson, M.P.; Taunton, J. Design of reversible, cysteine-targeted Michael acceptors guided by kinetic and computational analysis. J. Am. Chem. Soc. 2014, 136, 12624–12630. [Google Scholar] [CrossRef] [Green Version]
  92. Huang, F.; Han, X.; Xiao, X.; Zhou, J. Covalent warheads targeting cysteine residue: The promising approach in drug development. Molecules 2022, 27, 7728. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structures of heteroaryl-acrylonitriles reported and studied in this work.
Figure 1. Structures of heteroaryl-acrylonitriles reported and studied in this work.
Pharmaceuticals 16 00918 g001
Scheme 1. Synthesis of 2-(1H-indol-2-yl)-3-acrylonitriles 2ax and N-(4-(dimethylamino)phenyl)-1H-indole-2-carbimidoyl cyanide (3).
Scheme 1. Synthesis of 2-(1H-indol-2-yl)-3-acrylonitriles 2ax and N-(4-(dimethylamino)phenyl)-1H-indole-2-carbimidoyl cyanide (3).
Pharmaceuticals 16 00918 sch001
Figure 2. Molecular structure of 2l. Displacement ellipsoids are shown at the 50% probability level.
Figure 2. Molecular structure of 2l. Displacement ellipsoids are shown at the 50% probability level.
Pharmaceuticals 16 00918 g002
Scheme 2. Synthesis of 2-(1H-indol-2-yl)-3-phenylpropanenitriles 4ac.
Scheme 2. Synthesis of 2-(1H-indol-2-yl)-3-phenylpropanenitriles 4ac.
Pharmaceuticals 16 00918 sch002
Scheme 3. Synthesis of N-substituted 2-(1H-indol-2-yl)-3-acrylonitriles 5ad, 6ab, and 7.
Scheme 3. Synthesis of N-substituted 2-(1H-indol-2-yl)-3-acrylonitriles 5ad, 6ab, and 7.
Pharmaceuticals 16 00918 sch003
Figure 3. Comparison of the mean GI50 values per panel displayed by compounds 2l and 5ad.
Figure 3. Comparison of the mean GI50 values per panel displayed by compounds 2l and 5ad.
Pharmaceuticals 16 00918 g003
Figure 4. The highest-scored poses of compound 5c (cyan sticks) docked in the active site of: caspase-3 (p17 and p12 subunits in green and orange, respectively), (A), caspase-9 (B), and tubulin (α- and β-tubulin in green and orange, respectively), (C); graphic representation of compound 5c fitted into the ligand-based pharmacophore (D). Hydrogen bonds are indicated as black dotted lines. For clarity, only relevant amino acids are presented. (AC) were prepared using PyMOL 1.5.0.3., and (D) was generated by BIOVA Discovery Studio Visualizer (for interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article).
Figure 4. The highest-scored poses of compound 5c (cyan sticks) docked in the active site of: caspase-3 (p17 and p12 subunits in green and orange, respectively), (A), caspase-9 (B), and tubulin (α- and β-tubulin in green and orange, respectively), (C); graphic representation of compound 5c fitted into the ligand-based pharmacophore (D). Hydrogen bonds are indicated as black dotted lines. For clarity, only relevant amino acids are presented. (AC) were prepared using PyMOL 1.5.0.3., and (D) was generated by BIOVA Discovery Studio Visualizer (for interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article).
Pharmaceuticals 16 00918 g004
Figure 5. The highest-scored poses of compound 2x (cyan sticks) docked in the active site of E. coli enzyme (green), PBP4 (A), and β-lactamase (B). Hydrogen bonds are indicated as black dotted lines. For clarity, only relevant amino acids are presented. The figure was prepared using PyMOL 1.5.0.3. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article).
Figure 5. The highest-scored poses of compound 2x (cyan sticks) docked in the active site of E. coli enzyme (green), PBP4 (A), and β-lactamase (B). Hydrogen bonds are indicated as black dotted lines. For clarity, only relevant amino acids are presented. The figure was prepared using PyMOL 1.5.0.3. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article).
Pharmaceuticals 16 00918 g005
Figure 6. Oral bioavailability radar charts for the studied compounds 2l, 2x, and 5ad. In bioavailability radar, the pink area represents the optimal range for each physicochemical property of oral bioavailability (LIPO—lipophilicity, SIZE—size, POLAR—polarity, INSOLU—solubility, INSATU—saturation and FLEX—flexibility), while the red lines represent compounds: (A) 2l, (B) 2x, (C) 5a, (D) 5b, (E) 5c, and (F) 5d.
Figure 6. Oral bioavailability radar charts for the studied compounds 2l, 2x, and 5ad. In bioavailability radar, the pink area represents the optimal range for each physicochemical property of oral bioavailability (LIPO—lipophilicity, SIZE—size, POLAR—polarity, INSOLU—solubility, INSATU—saturation and FLEX—flexibility), while the red lines represent compounds: (A) 2l, (B) 2x, (C) 5a, (D) 5b, (E) 5c, and (F) 5d.
Pharmaceuticals 16 00918 g006
Figure 7. BOILED-Egg plot for the studied compounds 2l, 2x, and 5ad.
Figure 7. BOILED-Egg plot for the studied compounds 2l, 2x, and 5ad.
Pharmaceuticals 16 00918 g007
Table 1. Mean growth (%), growth inhibition percent (%GI), and lethality values for the most sensitive cell lines among approximately NCI-60 human cancer cell lines after treatment with the tested compounds at 10 μM a.
Table 1. Mean growth (%), growth inhibition percent (%GI), and lethality values for the most sensitive cell lines among approximately NCI-60 human cancer cell lines after treatment with the tested compounds at 10 μM a.
CompoundMean GrowthMost Sensitive Cell LineGrowth Inhibition Percent (%GI) b/Lethality c
2a85.56T-47D (breast cancer)59.79
2b49.80MDA-MB-435 (melanoma)−5.10 c
2c78.15K-562 (leukemia)72.20
2d30.17MDA-MB-435 (melanoma)−32.56 c
2e54.83MDA-MB-435 (melanoma)−28.75 c
2f66.94MDA-MB-435 (melanoma)87.84
2g93.62A549/ATCC (non-small cell lung cancer)39.97
2h84.56CCRF-CEM (leukemia)−4.12 c
2i84.18HOP-92 (non-small cell lung cancer)46.93
2j93.18MCF7 (breast cancer)36.48
2k91.86CCRF-CEM (leukemia)−42.43 c
2l26.05MDA-MB-435 (melanoma)−25.37 c
2m53.62MDA-MB-435 (melanoma)98.20
2n73.11K-562 (leukemia)71.26
2o85.25T-47D (breast cancer)43.76
2p36.99MDA-MB-435 (melanoma)−18.97 c
2q83.41MCF7 (breast cancer)79.89
2r88.94CCRF-CEM (leukemia)53.33
2s83.36T-47D (breast cancer)84.11
2t88.41SNB-75 (CNS cancer)56.11
2u92.81MCF7 (breast cancer)67.84
2v95.03SNB-75 (CNS cancer)46.36
2w91.64SNB-75 (CNS cancer)59.89
2x66.37MDA-MB-468 (breast cancer)−3.15 c
390.03UO-31 (renal cancer)45.43
4a87.87CCRF-CEM (leukemia)−27.23 c
4b94.94UO-31 (renal cancer)36.32
4c89.04CNB-75 (CNS cancer)60.83
5a32.33OVCAR-3 (ovarian cancer)−12.57 c
5b24.19MDA-MB-435 (melanoma)−49.06 c
5c25.80MDA-MB-435 (melanoma)−26.92 c
5d25.56SK-MEL-5 (melanoma)−26.68 c
6a81.55MDA-MB-435 (melanoma)90.31
6b33.80SNB-75 (CNS cancer)−6.19 c
782.53MDA-MB-435 (melanoma)93.95
a Data obtained from NCI-60 DTP human tumor cell line screen [67]; b %GI (growth inhibition percent) = 100 − GP (growth percent); c negative values mean lethality of the respective cancer cell line.
Table 2. Overview of the results of the in vitro antitumor screening for compounds 2l and 5ad a.
Table 2. Overview of the results of the in vitro antitumor screening for compounds 2l and 5ad a.
Compounds
2l5a5b5c5d
Panel NameCell NameGI50 bTGI cGI50 bTGI cGI50 bTGI cGI50 bTGI cGI50 bTGI c
LeukemiaCCRF-CEM0.81314.30.373>1000.333>1000.213>1000.218>100
HL-60(TB)0.455>1000.3056.640.2170.5820.1030.5550.1790.703
K-5620.244>1000.216>1000.0364>1000.0406>100
MOLT-40.906>1000.507>1000.41716.20.338>1000.446>100
RPMI-82260.634.910.39>1000.395>1000.2960.285>100
SR0.497>1000.269>1000.1790.940.03571.810.0578>100
Non-small cell lung cancerA549/ATCC1.55>1000.438>1000.525>1000.159>1000.174>100
EKVX2.94>1000.549>1000.821>1000.64>1000.442>100
HOP-621.164.540.4>1000.411>1000.206>1000.387>100
HOP-921.425.530.61>1000.633>1000.1895.290.0395>100
NCI-H2261.800.753>1004.26>1005.06>1000.325>100
NCI-H233.21>1000.536>1000.739>1000.734>1000.361>100
NCI-H322M2.88>1000.722>1000.57>1000.671>100>100
NCI-H4600.476>1000.366>1000.39721.30.3070.182>100
NCI-H5220.542>1000.2594.570.190.5540.02440.08660.0277
Colon cancerCOLO 2050.5667.390.405>1000.3211.040.1340.4980.29>100
HCC-29986.6>1000.522>1001.055.860.323>1000.308>100
HCT-1160.456>1000.367>1000.409>1000.0954>1000.042>100
HCT-150.509>1000.328>1000.359>1000.0587>1000.064>100
HT290.37515.90.351>1000.33611.60.04911.430.153
KM120.495>1000.431>1000.504>1000.285>1000.0922>100
SW-6200.3961.90.389>1000.366>1000.0825>1000.1>100
CNS cancerSF-2682.52>1004.02>1001.19>1001.9>100>100
SF-2950.617.330.2851.460.4037.940.08281.280.0599
SF-5391.9735.10.3131.090.2830.8910.170.5480.06130.552
SNB-192.23>1000.448>1000.487>1000.344>1000.207>100
SNB-751.314.590.2320.150.5140.01930.1960.055485.9
U2510.91318.60.389>1000.551>1000.299>1000.167>100
MelanomaLOX IMVI0.692>1000.532>1000.654>1000.471>1000.0705>100
MALME-3M2.32>100>1000.36>1000.0725>100>100
M140.551>1000.2030.338>1000.08550.0666>100
MDA-MB-
435
0.2280.6410.07260.2540.1530.4030.02850.03180.103
SK-MEL-20.454>100>1000.417>1000.328>10072.9>100
SK-MEL-282.63>100>1006.96>10016.6>100
SK-MEL-51.3632.80.717>1000.44614.30.3260.455>100
UACC-2574.86>100>100>1000.679>1002.98>100>100>100
UACC-620.679>1000.34>1000.54>1000.163>1000.0436>100
Ovarian cancerIGROV11.6>1000.555>1000.505>1000.165>1000.15>100
OVCAR-30.4094.060.3332.240.3020.9550.1420.6630.179
OVCAR-43.75>100>1000.67>1000.521>100>100>100
OVCAR-53.80>1000.607>1000.995>1000.428>1000.329>100
OVCAR-82.35>1000.584>1000.577>1000.338>1000.392>100
NCI/ADR-
RES
0.49>1000.379>1000.3178.240.0736>1000.0607>100
SK-OV-32.28>100>1000.517>1000.377>100>100
Renal cancer786-02.52>1000.676>1000.306>100
A4980.3916.610.2350.8160.2770.8670.07950.9380.030.418
ACHN2.42>1000.763>1000.629>1000.39>1000.0605>100
CAKI-11.6>1000.329>1000.444>1000.0736>1000.0545>100
RXF 3930.2430.2170.7510.1330.5490.0475
SN12C2.37>1000.69>1000.508>1000.392>1000.362>100
TK-102.66>100>100>1000.902>100>100>100>100
UO-312.01>1000.792>1000.514>1000.786>1000.0731>100
Prostate cancerPC-31.1531.50.431>1000.432>1000.166>1000.2>100
DU-1453.4135.40.469>1000.529>1000.488>1000.45>100
Breast cancerMCF70.416>1000.356>1000.317>1000.0309>1000.0692>100
MDA-MB-231/ATCC1.63>1000.713>1000.615>1000.428>1000.406>100
HS 578T1.639.490.484>1000.375>1000.427>1000.201>100
BT-5493.86>1000.657.120.7040.806>1000.251
T-47D0.585>1000.631>1000.31>1000.0764>100>100>100
MDA-MB-4680.3734.20.406>1000.2360.8260.03840.5010.55>100
a Data obtained from the NCI in vitro disease-oriented human tumor cell lines screen in μM [67]; b GI50—the molar concentration that inhibits 50% net cell growth; c TGI—the molar concentration leading to total growth inhibition;—not tested.
Table 3. GI50 mean-graph midpoint (MG-MID) a of subpanel cancer cell lines for compounds 2l and 5ad.
Table 3. GI50 mean-graph midpoint (MG-MID) a of subpanel cancer cell lines for compounds 2l and 5ad.
Subpanel Cancer Cell LineCompounds
2l5a5b5c5d
Leukemia0.660.350.290.170.20
Non-small cell lung cancer1.780.510.950.890.24
Colon cancer1.340.400.480.150.15
CNS cancer1.590.950.510.470.11
Melanoma1.53>16.981.170.5623.77
Ovarian cancer2.100.490.550.2916.85
Renal cancer2.00>12.970.500.3112.62
Prostate cancer2.280.450.480.330.33
Breast cancer1.420.540.430.3016.91
Full panel MG-MID b1.63>3.740.600.38>7.91
a Median value calculated based on the data from the NCI in vitro cytotoxicity screening for each cancer type cell line; b GI50 full panel mean-graph midpoint (MG-MID) is the average GI50 value against all cancer cell lines.
Table 4. Minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) of 2ax against Gram-positive and Gram-negative reference bacteria as well as minimal fungicidal concentration (MFC) against Candida albicans.
Table 4. Minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) of 2ax against Gram-positive and Gram-negative reference bacteria as well as minimal fungicidal concentration (MFC) against Candida albicans.
CompoundStrain
S. aureus
ATCC 6538
S. epidermidis PCM 2118E. faecalis ATCC 11420E. coli
ATCC 11229
P. aeruginosa ATCC 15442C. albicans ATCC 10231
MICMBCMICMBCMICMBCMICMBCMICMBCMICMFC
(μg/mL)
2a>256>256>256>256>256>256>256>256>256>256128256
2b>256>256>256>256>256>256>256>256>256>256128256
2c128>256128>256>256>256>256>256>256>256128128
2e>256>256>256>256>256>256>256>256>256>256256256
2f>256>256>256>256>256>256>256>256>256>256256256
2g128>256128>256>256>256>256>256>256>256128128
2h>256>256128256>256>256>256>256>256>256128256
2i1616816>256>256>256>256>256>256128256
2j>256>256>256>256>256>256>256>256>256>256256256
2k128>256128>256>256>256>256>256>256>256256256
2l>256>256>256>256>256>256>256>256>256>256128256
2n64128128256>256>256>256>256>256>256256256
2o>256>256128256>256>256>256>256>256>256256256
2p128>256128>256>256>256>256>256>256>256>256>256
2q881616>256>256>256>256>256>256128256
2r>256>256>256>256>256>256>256>256>256>256128256
2s16323232>256>256>256>256>256>25616128
2t>128>128>128>128>128>128>128>128>128>128>128>128
2u>128>128>128>128>128>128>128>128>128>128>128>128
2v>128>128>128>128>128>128>128>128>128>128>128>128
2w>128>128>128>128>128>128>128>128>128>128>128>128
2x83283232323232>256>25648
Table 5. Minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) of 2i, 2q, 2s, and 2x against clinical S. aureus strains.
Table 5. Minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) of 2i, 2q, 2s, and 2x against clinical S. aureus strains.
CompoundStrain
79124128143177220244
MICMBCMICMBCMICMBCMICMBCMICMBCMICMBCMICMBC
(μg/mL)
2i>128>128>128>128>128>128>128>128>128>128>128>128>128>128
2q>256>256>256>256>256>256>256>256>256>256>256>256>256>256
2s>128>128>128>128>128>128>128>128>128>128>128>128>128>128
2x1664163216321632163216321632
Table 6. Minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) of 2x against clinical methicillin-sensitive S. aureus (MSSA) and methicillin-resistant S. aureus (MRSA) strains.
Table 6. Minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) of 2x against clinical methicillin-sensitive S. aureus (MSSA) and methicillin-resistant S. aureus (MRSA) strains.
StrainMICMBC
(μg/mL)
Compound 2x
MSSA
S. aureus 11664
S. aureus 21664
S. aureus 31664
S. aureus 41664
S. aureus 51664
MRSA
S. aureus 61664
S. aureus 71664
S. aureus 81664
S. aureus 91664
S. aureus 101664
Table 7. Predicted physicochemical, pharmacokinetic, and drug-likeness properties of compounds 2l, 2x, and 5ad.
Table 7. Predicted physicochemical, pharmacokinetic, and drug-likeness properties of compounds 2l, 2x, and 5ad.
Physicochemical Properties LipophilicityWater SolubilityPharmacokineticsDrug–Likeness
mol. wt.
(g/mol)
ROTB
(n)
HBA
(n)
HBD
(n)
TPSACLogP
o/w
Solubility ClassGI
Absorption
BBB
Permeant
Lipinski
Filter
BS
Rule<500<10<10<5-<5-----
2l287.3631142.823.49Soluble (m)HighYesYes (0)0.55
2x233.2721255.372.61SolubleHighYesYes (0)0.55
5a272.3421028.723.83Soluble (m)HighYesYes (0)0.55
5b288.3432037.953.50Soluble (m)HighYesYes (0)0.55
5c301.3831031.963.51Soluble (m)HighYesYes (0)0.55
5d308.3821028.724.41Soluble (m)HighYesYes (0)0.55
mol. wt.—molecular weight; n—number, ROTB—rotable bonds; HBA—hydrogen bond acceptors; HBD—hydrogen bond donors; TPSA—topological polar surface area calculated in Å2; CLogPo/w—consensus logarithm of partition coefficient between n-octanol and water; m—moderate; Lipinski filter with number of violations in bracket; GI—gastrointestinal absorption; BBB—blood–brain barrier; BA—bioavailability score.
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

Kornicka, A.; Gzella, K.; Garbacz, K.; Jarosiewicz, M.; Gdaniec, M.; Fedorowicz, J.; Balewski, Ł.; Kokoszka, J.; Ordyszewska, A. Indole-Acrylonitrile Derivatives as Potential Antitumor and Antimicrobial Agents—Synthesis, In Vitro and In Silico Studies. Pharmaceuticals 2023, 16, 918. https://doi.org/10.3390/ph16070918

AMA Style

Kornicka A, Gzella K, Garbacz K, Jarosiewicz M, Gdaniec M, Fedorowicz J, Balewski Ł, Kokoszka J, Ordyszewska A. Indole-Acrylonitrile Derivatives as Potential Antitumor and Antimicrobial Agents—Synthesis, In Vitro and In Silico Studies. Pharmaceuticals. 2023; 16(7):918. https://doi.org/10.3390/ph16070918

Chicago/Turabian Style

Kornicka, Anita, Karol Gzella, Katarzyna Garbacz, Małgorzata Jarosiewicz, Maria Gdaniec, Joanna Fedorowicz, Łukasz Balewski, Jakub Kokoszka, and Anna Ordyszewska. 2023. "Indole-Acrylonitrile Derivatives as Potential Antitumor and Antimicrobial Agents—Synthesis, In Vitro and In Silico Studies" Pharmaceuticals 16, no. 7: 918. https://doi.org/10.3390/ph16070918

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