Next Article in Journal
Development of Fucoxanthin-Enriched Yogurt Using Nanoliposomal Carriers: A Strategy for Functional Dairy Products with Antioxidant and Erythroprotective Benefits
Previous Article in Journal
The Application of 2D Graphitic Carbon Nitride (g-C3N4) and Hexagonal Boron Nitride (h-BN) in Low-Temperature Fuel Cells: Catalyst Supports, ORR Catalysts, and Membrane Fillers
Previous Article in Special Issue
Anticancer Activity and Safety Profile of Novel 1-(4-Fluorophenoxyacetyl)-4-substituted Thio/Semicarbazide Derivatives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis, Evaluation of Biological Activity, and Structure–Activity Relationships of New Amidrazone Derivatives Containing Cyclohex-1-ene-1-Carboxylic Acid

by
Renata Paprocka
1,*,
Jolanta Kutkowska
2,
Ewelina Paczkowska
3,
Godwin Munroe Mwaura
4,
Andrzej Eljaszewicz
5 and
Anna Helmin-Basa
3
1
Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza Str. 2, 85-089 Bydgoszcz, Poland
2
Department of Genetics and Microbiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka Str. 19, 20-033 Lublin, Poland
3
Department of Immunology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Curie-Sklodowska Str. 9, 85-094 Bydgoszcz, Poland
4
Department of Pharmaceutical Chemistry, Pharmaceutics and Pharmacognosy, Faculty of Health Sciences, University of Nairobi, KNH, Nairobi P.O. Box 2149-00202, Kenya
5
Centre of Regenerative Medicine, Medical University of Bialystok, Waszyngtona 15 B, 15-269 Bialystok, Poland
*
Author to whom correspondence should be addressed.
Molecules 2025, 30(8), 1853; https://doi.org/10.3390/molecules30081853
Submission received: 21 March 2025 / Revised: 7 April 2025 / Accepted: 17 April 2025 / Published: 21 April 2025
(This article belongs to the Special Issue Design, Synthesis, and Analysis of Potential Drugs, 3rd Edition)

Abstract

:
In recent years, the incidence of acute and chronic inflammatory diseases has increased significantly worldwide, intensifying the search for new therapeutic agents, especially anti-inflammatory drugs. Therefore, the aim of this work was to synthesize, biologically assess, and explore the structure–activity relationships of new compounds containing the cyclohex-1-ene-1-carboxylic acid moiety. Six new derivatives, 2a2f, were synthesized through the reaction of amidrazones 1a1f with 3,4,5,6-tetrahydrophthalic anhydride. Their toxicity was evaluated in cultures of human peripheral blood mononuclear cells (PBMCs). Additionally, their antiproliferative properties and effects on the synthesis of TNF-α, IL-6, IL-10, and IL-1β were assessed in mitogen-stimulated PBMCs. The antimicrobial activity of derivatives 2a2f was determined by measuring the minimal inhibitory concentration (MIC) values against five bacterial strains—Staphylococcus aureus, Mycobacterium smegmatis, Escherichia coli, Yersinia enterocolitica, and Klebsiella pneumoniae—and the fungal strain Candida albicans. All compounds demonstrated antiproliferative activity, with derivatives 2a, 2d, and 2f at a concentration of 100 µg/mL being more effective than ibuprofen. Compound 2f strongly inhibited the secretion of TNF-α by approximately 66–81% at all studied doses (10, 50, and 100 µg/mL). Derivative 2b significantly reduced the release of cytokines, including TNF-α, IL-6, and IL-10, at a high dose (by approximately 92–99%). Compound 2c exhibited bacteriostatic activity against S. aureus and M. smegmatis, while derivative 2b selectively inhibited the growth of Y. enterocolitica (MIC = 64 µg/mL). Some structure–activity relationships were established for the studied compounds.

Graphical Abstract

1. Introduction

Inflammation refers to the body’s protective response to injury, infection, or harmful stimuli, aiming to eliminate the cause of damage, clear dead cells and debris, and trigger tissue repair. The inflammatory process is usually characterized by redness, swelling, heat, pain, and loss of tissue function, which results from vascular and cellular responses. These complex biological processes are orchestrated by immune cells and the associated inflammatory mediators. In fact, inflammation can be categorized into acute and chronic forms. Acute inflammation is an immediate and transient response caused by the direct disruption of epithelial barriers and involving the recruitment and activation of neutrophils and monocytes/macrophages. These processes are regulated by signals involving pathogen-associated molecular patterns (PAMPs) and danger/damage-associated molecular patterns (DAMPs), leading to the release of inflammatory cytokines, chemokines, and other inflammatory mediators (such as lipid mediators, including prostaglandins and leukotrienes) [1,2,3]. When acute inflammation fails to resolve, it can progress to a chronic process, persisting for months or even years. Chronic inflammation is characterized by the sustained activation of innate and adaptive immune responses involving the recruitment of T cells, B cells, and plasma cells to the affected site [4]. Both acute and chronic inflammation can also be modulated by neutrophil phagocytosis and the release of neutrophil extracellular traps (NETs) [5].
The global prevalence of inflammatory disorders has risen significantly in recent years, intensifying the search for novel therapeutic agents, particularly in the realm of anti-inflammatory drugs [6]. Chronic inflammation underlies a myriad of diseases, including autoimmune disorders, cardiovascular diseases, and cancer, necessitating ongoing research into more effective treatments. While current anti-inflammatory drugs demonstrate efficacy, they often come with severe side effects that limit their long-term use. This limitation has spurred the development of new agents [7]. Continuous research and development in this field aim to provide novel solutions to combat inflammatory diseases and improve the quality of life for affected individuals globally.
Amidrazone derivatives have emerged as promising candidates in drug discovery due to their diverse biological activities and structural versatility. These compounds have demonstrated notable anti-inflammatory properties, with studies showing their ability to modulate key cytokines such as IL-6, IL-10, and IL-1β, and tumor necrosis factor-alpha (TNF-α), which are critical mediators in inflammatory responses [8]. Specifically, triazole derivatives have exhibited strong anti-inflammatory effects through this mechanism [9]. Certain derivatives have also displayed antiproliferative activity in mitogen-activated peripheral blood mononuclear cell (PBMC) cultures, indicating their potential in controlling cell proliferation [10]. These effects highlight their broader therapeutic utility beyond inflammation management [10].
The significance of amidrazone derivatives in medicinal chemistry is underscored by their wide range of biological activities. They have demonstrated potential as tuberculostatic, antibacterial, antifungal, antiparasitic, antiviral, cytoprotective, and antitumor agents, as well as being inhibitors of furin and acetylcholinesterase [8,11].
Bioactive compounds containing the cyclohexene moiety can be isolated from raw materials of natural origin. For example, diisoprenylcyclohexene-type meroterpenoids isolated from Biscogniauxia sp. exhibited anti-inflammatory effects by inhibiting the production of pro-inflammatory cytokines TNF-α and IL-6 [12]. Among the polyoxygenated cyclohexene derivatives isolated from the stem of Uvaria rufa, 6-acetylzeylenol demonstrated activity against Mycobacterium tuberculosis [13]. The anti-inflammatory effect of (-)-zeylenone, isolated from Uvaria macclurei, has been demonstrated through the reduction in nitric oxide (NO) synthesis in mitogen-stimulated RAW 264.7 macrophages derived from mice and the inhibition of NF-κB signaling pathway activation. Additionally, it exhibited anti-sepsis activity in mice [14]. A synthetic analogue of zeylenone also showed strong antiproliferative activity in glioblastoma cells [15].
In our previous work, compounds derived from amidrazones containing the cyclohex-1-ane-1-carboxylic acid moiety demonstrated various biological activities, including antiviral, antibacterial, anti-inflammatory, and antinociceptive properties [16]. Encouraged by these results, we designed new amidrazone derivatives possessing an unsaturated cyclohex-1-ene-1-carboxylic acid system to evaluate their basic biological effects, such as toxicity and antiproliferative, anti-inflammatory, and antimicrobial activities. Additionally, we aim to investigate the influence of the double bond in the cyclohexene system and the type of substituents present in the synthesized compounds on their biological activity.

2. Results

2.1. The Synthesis of Compounds 2a2f

The reactions of amidrazones 1a1f with 3,4,5,6-tetrahydrophthalic anhydride yielded six new acyl derivatives, 2a2f (Scheme 1). All reactions proceeded with very high yields (above 90%), except for derivative 2d, which was synthesized from amidrazone containing a 4-nitrophenyl substituent at the R2 position. This substituent may have hindered the reaction.
In the 1H NMR and 13C NMR spectra of compounds 2a2f, a double number of signals is observed, consistent with the occurrence of E/Z isomerism around the C=N bond. This phenomenon has been previously described in acylamidrazones [17]. Elemental analysis of the synthesized compounds 2a2f, along with HRMS analysis, confirmed their elemental composition.

2.2. Toxicity of Compounds 2a2f

First, we aimed to assess the toxic effects of the new derivatives. The data from the toxicity assessments of compounds 2a2f and the reference anti-inflammatory agent, ibuprofen (IBU), on peripheral blood mononuclear cells (PBMCs) in 24 h in vitro cultures are presented in Figure 1. PBMC cultures incubated with a vehicle, namely dimethyl sulfoxide (DMSO) at concentrations of 0.05–0.50%, served as a control. As expected, approximately 86% of viable cells were observed in control cultures, with about 10% of cells in early apoptosis. In cultures with IBU at a concentration of 100 µg/mL, approximately 7.5% of early apoptotic and 7.5% of late apoptotic cells were observed. In PBMC cultures incubated with derivatives 2a, 2c, and 2e at the highest concentration, 100 µg/mL, approximately 76% of viable cells were present, and the level of late apoptotic cells was slightly elevated (approximately 11–15%).
Compound 2b showed strong dose-dependent toxicity, while derivatives 2d and 2f exhibited moderate dose-dependent toxicity. At a concentration of 100 µg/mL, compound 2b caused the highest frequencies of early apoptotic (23.66%), late apoptotic (35.51%), and necrotic (5.31%) cells compared to other PBMC cultures. In PBMC cultures with compounds 2d and 2f at a concentration of 100 µg/mL, approximately 6–9% of early apoptotic cells, 22.5% and 27% of late apoptotic cells, and 3–4% of necrotic cells were present. At the highest culture dose, the percentage of necrotic cells for individual compounds 2a2f was comparable to the percentage of necrotic cells in cultures containing IBU at the same dose.

2.3. Antiproliferative Activity of Compounds 2a2f

Having found no cytotoxic effects of the analyzed compounds, we next aimed to evaluate their impact on mitogen-induced PBMC proliferation. Again, PBMCs incubated in the presence of the vehicle (DMSO at the same concentrations as in cultures with the tested compounds) served as a control. No significant effect of the solvent on phytohemagglutinin (PHA)-stimulated T lymphocyte proliferation was observed (Figure 2). IBU at a dose of 10 µg/mL slightly increased the proliferation of T lymphocytes. However, at a concentration of 100 µg/mL, significant T cell inhibition (approximately 50%) was observed in the cultures. Compounds 2a, 2c, 2e, 2f, and IBU showed dose-dependent antiproliferative activity. The highest inhibition (approximately 95%) was observed for compound 2d at concentrations of 50 µg/mL and 100 µg/mL, as well as for derivatives 2a and 2f at a concentration of 100 µg/mL (approximately 90% inhibition). Among derivatives 2a2f, at a dose of 100 µg/mL, only compound 2b did not decrease the proliferation of T cells. However, compound 2b was the only one that inhibited T lymphocyte proliferation by approximately half at the lowest dose of 10 µg/mL.

2.4. Effect of Compounds 2a2f on TNF-α Production

Next, we assessed the effects of the analyzed compounds on the pro-inflammatory TNF-α production of endotoxin-exposed PBMCs. DMSO at the highest concentration of 100 µg/mL caused a significant reduction in TNF-α production in an LPS-stimulated PBMC culture (Figure 3). Both derivatives—2f at concentrations of 10 µg/mL and 50 µg/mL, and 2b at a concentration of 100 µg/mL—showed significant inhibition of TNF-α production.

2.5. Effect of Compounds 2a2f on IL-6 Production

Having found changes in TNF-α production, we next assessed the effects on IL-6 release. Derivative 2b at a dose of 100 µg/mL showed strong inhibition of the secretion of this pro-inflammatory cytokine (Figure 4). However, compound 2a at a concentration of 50 µg/mL significantly increased IL-6 production in an LPS-stimulated PBMC culture. No significant changes in IL-6 concentration were observed in the remaining cultures (p > 0.05).

2.6. Effect of Compounds 2a2fon IL-10 Production

Finally, we assessed the effect of the analyzed compounds on anti-inflammatory IL-10 production in the PBMC response to LPS. A significant reduction in the release of the IL-10 cytokine was observed in the case of the following compounds: IBU (at a concentration of 100 µg/mL), 2b (at concentrations of 50 and 100 µg/mL), and 2f (at a concentration of 100 µg/mL, Figure 5). The remaining compounds did not show significant differences in IL-10 levels compared to the LPS-stimulated culture.

2.7. The Influence of Compounds 2a2fon IL-1β Production

No significant changes in the level of IL-1β were observed for any of the studied compounds (p > 0.05, Figure S15 in the Supplementary Materials).

2.8. Antimicrobial Activity of Compounds 2a2f

The tested derivatives were found to possess moderate or weak activity against the tested strains; MIC values ranged from 64 to greater than 512 µg/mL (Table 1). The best anti-mycobacterial activity was shown by compounds 2a and 2c, which inhibited M. smegmatis growth at a concentration of 64 µg/mL. Moreover, they were effective against S. aureus at concentrations of 64 µg/mL (derivative 2c) and 256 µg/mL (derivative 2a).
Derivatives 2b and 2f showed moderate activity against Y. enterocolitica, with MIC values of 64 µg/mL and 128 µg/mL, respectively. Derivative 2b also inhibited the growth of E. coli and K. pneumoniae at a concentration of 256 µg/mL. Compound 2f revealed weak antifungal activity and inhibited the growth of C. albicans at a concentration of 256 µg/mL. The remaining compounds, 2d and 2e, did not exhibit antimicrobial activity, with MIC values of ≥512 µg/mL.
Four of the bacterial strains tested were sensitive to ampicillin, with MIC values of 0.5 and 8 µg/mL for S. aureus and E. coli and 16 µg/mL for Y. enterocolitica and M. smegmatis. K. pneumoniae was resistant to ampicillin (MIC > 256 µg/mL), whereas C. albicans was sensitive to fluconazole (MIC 0.25 µg/mL).

3. Discussion

Six new acyl derivatives, 2a2f, were easily synthesized in a reaction of N3-substituted amidrazones with 3,4,5,6-tetrahydrophthalic anhydride in anhydrous diethyl ether (Figure 6a).
Among closely related compounds, only a few amidrazone derivatives—3a, 3b, 3d, and 3e—containing the cyclohexanecarboxylic acid moiety (Figure 6b) [16] and only one derivative, 4b, containing the cyclohex-3-ene-1-carboxylic acid system (Figure 6c) [18], have been described. These compounds showed a wide range of biological activities and high potency, including antinociceptive and anti-inflammatory effects. This encouraged us to synthesize and investigate the biological activity of analogous compounds, 2a2f, containing an additional double bond in the aliphatic ring (cyclohex-1-ene-1-carboxylic acid moiety).
Six new acyl derivatives, 2a2f, were easily synthesized in a reaction of N3-substituted amidrazones with 3,4,5,6-tetrahydrophthalic anhydride in anhydrous diethyl ether. In previously described reactions, it was not possible to obtain the expected acyclic compounds, 3c and 3f, containing a 4-methylphenyl substituent in the R2 position, probably due to their tendency to undergo cyclisation to 1,2,4-triazole derivatives [16]. The successful synthesis of derivatives 2c and 2f in the current approach was facilitated, likely due to the shortening of the reaction time from 14 to 3 days or due to the more favorable properties of 3,4,5,6-tetrahydrophthalic anhydride compared to cis-1,2-cyclohexanedicarboxylic anhydride.
The next step in this work was the assessment of the toxicity of six new derivatives. Flow cytometric detection of apoptosis in PBMC culture showed toxicity for derivatives 2b, 2d, and 2f at a concentration of 100 µg/mL (about 35%, 64%, and 63% of viable cells, respectively). However, these compounds at concentrations of 10 and 50 µg/mL, as well as compounds 2a, 2c, and 2e, and IBU at concentrations of 10 µg/mL, 50 µg/mL, and 100 µg/mL, showed satisfactory cell survival in the range of 70–85%. For comparison, the previously studied compound 4b, having a double bond on the opposite side of the cyclohexene ring, was not significantly toxic to PBMCs in the concentration range of 1–100 µg/mL [18]. In turn, compound 3b, possessing a saturated cyclohexane ring, was studied in a human fibroblast CCD-18C0 cell culture and showed high toxicity at concentrations of 50 µg/mL and 100 µg/mL (about 35 and 24% of viable cells, respectively) [16].
Due to the small number of tested compounds and the use of different research models (fibroblast or PBMC), it is difficult to clearly indicate which structural element of these derivatives influenced cellular toxicity. It can be assumed that the presence of two 2-pyridyl substituents increases the toxicity of compounds 2b and 3b, while the presence of a cyclohex-3-ene-1-carboxylic acid moiety in derivative 4b reduces this effect.
The subsequent step of this work was to determine the antiproliferative effects of compounds 2a2f on PHA-stimulated lymphocytes. We showed that the inhibitory effect of compounds 2a2f depended on the type of their substituents, R1 and R2. Moreover, derivatives 2a, 2c, 2e, and 2f and IBU showed a dose-dependent inhibitory effect. In contrast, compound 2b caused about 50% inhibition of lymphocyte proliferation at the lowest dose, 10 µg/mL, but was ineffective at the other doses. For the most active compound, 2d, at a dose of 50 µg/mL, and for compounds 2a, 2d, and 2f at a dose of 100 µg/mL, showing about 90–95% inhibitory effect, the IBU activity (about 46%) was significantly exceeded. These results are consistent with the high toxicity of derivatives 2d and 2f. However, compounds 2a, 2c, 2e, and IBU were not toxic at a concentration of 100 µg/mL, so their observed dose-dependent antiproliferative effect was not associated with toxicity but rather with their anti-inflammatory effect.
Among the analogous compounds 3a, 3b, 3d, 3e, and 4d, also studied in PHA-stimulated PBMC cultures at a dose of 100 µg/mL, the strongest antiproliferative effect was observed for compound 3b, possessing two 2-pyridyl substituents (approximately 61% inhibition). Weaker activity was observed for compounds 3d and 3e (approximately 28% inhibition) and 3a (approximately 12% inhibition) [16]. No antiproliferative activity was detected for derivative 4b [18]. The presence of a double bond in the cyclohexene ring increased the antiproliferative activity of compounds 2a and 2e (which have a phenyl ring in the R2 position) and 2d (which has a 4-nitrophenyl substituent in the R2 position) in comparison to the cyclohexane derivatives 3a, 3d, and 3e. In the case of compounds 2b, 3b, and 4b, containing two 2-pyridyl rings in the R2 position, antiproliferative activity varied depending on their structures and the dose used (Figure 7).
To investigate the anti-inflammatory activity of compounds 2a2f, we also examined their effect on pro-inflammatory cytokine (IL-6, TNF-α, IL-1β) levels in LPS-stimulated PBMC cultures. Among them, only compound 2b showed a significant decrease in IL-6 secretion at the highest dose of 100 µg/mL (approximately 93% inhibition). In a previous study, we also observed a weak IL-6 inhibitory effect of compound 3b (approximately 10–12%) and a stronger effect for 3a (approximately 35%) at a concentration of 10 µg/mL [16].
A more complex situation occurred when examining the influence of compounds 2a2f on the secretion of TNF-α. Derivative 2f decreased the TNF-α level at all studied doses (by approximately 66–81%). Compound 2b was significantly inhibitory at a concentration of 100 µg/mL (approximately 92%). Only derivative 2d significantly increased the level of TNF-α at a concentration of 100 µg/mL. Analogous compounds 3a, 3b, 3d, 3e, and 4b, studied at a concentration of 10 µg/mL, caused TNF-α inhibition of approximately 40%.
The presence of a double bond in the cyclohexene ring significantly reduced the inhibitory effect of compounds 2a, 2b, 2d, and 2e at a concentration of 10 µg/mL on TNF-α production compared to derivatives 3a, 3b, 3d, and 3e, although it did not change the activity of compound 4b (having a double bond on the other side of the cyclohexane ring) compared to 3b.
We also studied the effect of compounds 2a2f on the secretion of the anti-inflammatory cytokine IL-10 in LPS-stimulated PBMC cultures. An inhibitory effect on IL-10 release was observed at the highest dose for compounds 2f and IBU, as well as for compound 2b at medium and the highest doses (Figure 5). All compounds, 2a2f, showed a tendency to inhibit IL-10 at a dose of 100 µg/mL, but only for compounds 2b and 2f was this effect significant. A summary of the effects of the studied compounds on cytokine levels is presented in Figure 8.
In the second part of the biological tests, the antimicrobial activity of compounds 2a2f was examined. The most active antibacterial compounds possessed a 2-pyridyl substituent in the R1 position (compounds 2a2c). The exception was compound 2d, containing a 4-nitrophenyl substituent in the R2 position, which was devoid of any antimicrobial activity. Compound 2b, containing two 2-pyridyl rings, showed selective activity against the Gram-negative strain of Y. enterocolitica. The presence of a 2-pyridyl in the R1 position and a phenyl (or 4-methylphenyl) substituent in the R2 position appears to have a beneficial effect on the antituberculosic activity of compounds 2a and 2c against M. smegmatis.
In a previous study, we also observed an enhancement of the antibacterial activity for compounds possessing the 4-methylphenyl group in the R2 position. For example, derivative 2c showed stronger activity than 2a against S. aureus and K. pneumoniae [16]. Similarly, the enhanced activity of compound 2f compared to 2e against Y. enterocolitica and C. albicans was also observed [16].
Compared to the previously described analogous compounds 3a, 3b, and 3e, adding a double bond to the cyclohexane ring resulted in the following changes in biological activity:
(1)
Increased activity of compound 2b against Y. enterocolitica;
(2)
Reduced bacteriostatic effect of compound 2a against S. aureus;
(3)
Reduced bacteriostatic effect of compound 2e against M. smegmatis.
A summary of these dependencies is presented in Figure 9.
The structure–activity relationships in Figure 7, Figure 8 and Figure 9 can be summarized in the following conclusions:
The 2-pyridyl substituent in the R1 position is crucial for the antiproliferative activity of the studied amidrazone derivatives. Moreover, 4-nitrophenyl or 4-methylphenyl substituents in the R2 position can enhance this activity. The presence of a double bond in the cyclohex-1-ene ring further enhances the antiproliferative activity.
The presence of two pyridyl substituents in the R1 and R2 positions is the most effective for the inhibition of cytokines (TNF-α, IL-6, IL-10). In general, saturation of the cyclohexane ring enhances the inhibitory effect of the compounds on the TNF-α level (except for compound 3b).
The 2-pyridyl substituent in the R1 position is crucial for the antibacterial activity of the studied compounds. The 4-CH3-phenyl substituent in the R2 position is beneficial for enhancing both antituberculosic and antibacterial activity, while the 4-nitrophenyl substituent is detrimental to antimicrobial activity. Saturation of the cyclohexene bond may alter activity.
The presented relationships have certain limitations related to the small number of substituents considered. Our future research aims to expand this scope to include halogen atoms, methoxy groups, and other functional groups, enabling a broader understanding of the relationship between structure and biological activity in acylamidrazone derivatives.

4. Materials and Methods

4.1. General

Reagents and solvents were acquired from Sigma-Aldrich or Avantor Performance Materials Poland. Melting points were measured using a Mel-Temp apparatus (Electrothermal). 1H NMR and 13C NMR analyses were performed in deuterated dimethyl sulfoxide (DMSO-d6) on a Bruker Avance III Spectrometer (700 MHz and 400 MHz) and on a Bruker Avance (300 MHz) Spectrometer (Bruker Corporation, Billerica, MA, USA). Elemental analysis was carried out using a Vario MACRO CHN ELEMENTAR (Analysensysteme GmbH, Langenselbold, Germany) elemental analyzer. HRMS (high-resolution mass spectrometry) was recorded on a Synapt G2-Si mass spectrometer (Waters Corporation, Milford, MA, USA) equipped with an ESI source and a quadrupole–time–of–flight mass analyzer. The results were analyzed using MassLynx 4.1 software (Waters Corporation, Milford, MA, USA). The course of the reactions was monitored using TLC chromatography (silica gel on TLC-PET foils, Sigma-Aldrich, St. Louis, MO, USA).

4.2. General Method for the Synthesis of Compounds 2a2f

The amidrazones 1a1f required for the synthesis were obtained by reacting thioamides with 64% hydrazine hydrate [19]. In general, 0.3 g of amidrazones 1a1f (about 0.9 mmol) and 0.2 g of 3,4,5,6-tetrahydrophthalic anhydride (approximately 0.9 mmol) were dissolved in 30 mL of diethyl ether. After 3 days, the obtained solid compounds 2a2f were filtered and washed with 10 mL of diethyl ether to remove unreacted substrates, and dried.
2-[(2Z)-2-{phenyl[(pyridin-2-yl)amino]methylidene}hydrazinecarbonyl]cyclohex-1-ene-1-carboxylic acid (2a)
m.p. 119–120 °C, yield 92.5%. 1H NMR (700 MHz, DMSO-d6) δ 12.40 (s, 1 H, COOH). 10.06 (s, 0.48H), 9.77 (s, 0.52H), 8.81–6.79 (m, 9H + 1NH), 6.61 (d, 2H, CH=), 2.36–2.08 (m, 4H), 1.69–1.50 (m, 4H) ppm. 13C NMR (75 MHz, DMSO-d6): 172.58, 172.02, 168.49, 168.10, 167.68, 166.47, 152.63, 149.29, 148.93, 148.79, 144.77, 144.76, 140.64, 138.74, 137.50, 137.30, 130.88, 130.11, 129.47, 129.37, 129.10, 126.30, 124.63, 123.78, 122.54, 120.50, 119.22, 118.93, 28.57, 28.40, 25.59, 25.04, 22.02, 21.77, 21.64, 21.54, 20.84, 20.68, 15.63 ppm. Elem. Anal. for C20H20N4O3: calculated, C, 65.92, H, 5.53, N, 15.38%; found, C, 66.01%, H, 5.70%, N, 15.21%. HR-MS m/z 363.1463 [M+ − 1] (calculated for C20H19N4O3: 363.1457).
2-[(2Z)-2-{(pyridin-2-yl)[(pyridin-2-yl)amino]methylidene}hydrazinecarbonyl]cyclohex-1-ene-1-carboxylic acid (2b)
m.p. 133–135 °C, yield 94.16%. 1H NMR (400 MHz, DMSO-d6): 12.41 (sb, 1H), 11.93 (s, 0.58H), 11.18 (s, 0.42H), 9.27 (d, 1H NH), 8.49 (d, 1H), 8.15–7.33 (m, 6 H), 7.07–6.81 (m, 2H, CH=), 2.30 (s, 4 H), 1.66 (d, 42), 1.60 (s, 2H) ppm. 13C NMR (100 MHz, DMSO-d6): 172.86, 168.63, 167.95, 166.50, 154.25, 153.94, 152.56, 152.39, 148.38, 147.25, 146.93, 144.71, 142.13, 140.93, 139.13, 138.78, 137.88, 137.56, 129.92, 126.44, 124.78, 124.46, 122.60, 121.84, 116.94, 116.60, 113.29, 112.72, 65.33, 28.45, 28.30, 25.74, 25.07, 22.07, 21.79, 21.61, 20.81, 20.61 ppm. Elem. Anal. for C19H19N5O3: calculated, C, 62.46, H, 5.24, N, 19.17%; found, C, 62.45%, H, 5.37%, N, 19.18%. HR-MS m/z 364.1419 [M+ − 1] (calculated for C19H18N5O3: 364.1410).
2-{(2Z)-2-[(4-methylanilino)(pyridin-2-yl)methylidene]hydrazinecarbonyl}cyclohex-1-ene-1-carboxylic acid (2c)
m.p. 114–118 °C, yield 91%. 1H NMR (300 MHz, DMSO-d6): 12.30 (sb, COOH), 9.98 (s, 0.48H), 9.62 (s, 0.52H), 8.60–6.87 (m, 8H + 1NH), 6.53 (d, CH=), 2.40–2.05 (m, 9H), 1.73–1.48 (m, 4H) ppm. 13C NMR (75 MHz, DMSO-d6): 172.57, 172.02, 168.49, 168.10, 167.69, 166.48, 152.88, 152.62, 149.30, 148.92, 148.81, 144.78, 143.11, 140.64, 138.72, 138.66, 137.46, 137.29, 129.44.129.38, 129.12, 126.30, 124.73, 124.63, 124.57, 123.80, 122.53, 120.50, 119.22, 118.93, 65.38, 28.57, 28.40, 25.60, 25.02, 22.03, 21.74, 21.63, 21.54, 20.84, 20.68, 15.6389 ppm. Elem. Anal. for C21H22N4O3: calculated, C, 65.66, H, 5.86, N, 14.81%; found, C, 66.22, H, 5.89, N, 14.77%. HR-MS m/z 377.1619 [M+ − 1] (calculated for C21H21N4O3: 377.1614).
2-[(2Z)-2-{[(5-nitropyridin-2-yl)amino](pyridin-2-yl)methylidene}hydrazinecarbonyl]-cyclohex-1-ene-1-carboxylic acid (2d)
m.p. 144–145 °C, yield 57.41%. 1H NMR (400 MHz, DMSO-d6) 12.45 (s, 1H, COOH), 10.58 (s, 0.37H, CONH), 10.46 (s, 0.63H, CONH), 9.28 (d, 1H, NH), 8.20–7.58 (m, 6H), 7.50–7.38 (m, 1H), 6.76–6.63 (m, 2H, CH=), 2.41–1.15 (m, 4H), 1.68 (s, 2H), 1.56 (s, 2H) ppm. 13C NMR (100 MHz, DMSO-d6): 173.19, 172.50, 168.11, 167.66, 167.45, 152.04, 149.49, 149.20, 149.05, 145.37, 144.91, 140.60, 140.25, 139.72, 138.88, 138.28, 137.74, 126.32, 125.43, 125.14, 124.96, 124.84, 123.08, 122.02, 117.76, 117.15, 65.43, 116.58, 28.74, 28.47, 25.42, 25.00, 22.02, 21.92, 21.68, 21.46 ppm. Elem. Anal. for C20H19N5O5: calculated, C, 58.68, H, 4.68, N, 17.11%; found, C, 58.64, H, 4.60, N, 16.86%. HR-MS m/z 408.1313 [M+ − 1] (calculated for C20H18N5O5: 408.1308).
2-{(2Z)-2-[anilino(pyridin-4-yl)methylidene]hydrazinecarbonyl}cyclohex-1-ene-1-carboxylic acid (2e)
m.p. 118–120 °C, yield 94.20%. 1H NMR (700 MHz, DMSO-d6) 12.40 (s, 1 H, COOH), 10.05 (s, 0.48H), 9.77 (s, 0.52H), 8.82–6.80 (9H arom + 1HN), 6.61 (d, CH=), 3.35–2.08 (m, 4H), 1.69–1.50 (m, 4H) ppm. 13C NMR (176 MHz, DMSO-d6): 173.13, 168.44, 168.35, 168.03, 167.59, 150.35, 150.26, 150.18, 144.54, 142.29, 141.55, 141.29, 140.41, 139.13, 129.36, 129.34, 129.05, 126.56, 124.74, 124.00, 123.71, 123.08, 122.35, 122.30, 121.46, 120.98, 120.45, 118.90, 28.69, 28.41, 25.53, 25.02, 22.00, 21.76, 21.64, 21.51, 21.33, 21.12, 19.98, 19.87 ppm. Elem. Anal. for C20H20N4O3: calculated, C, 65.92, H, 5.53, N, 15.38%; found, C, 66.00%, H, 5.61%, N, 15.02%. HR-MS m/z 363.1464 [M+ − 1] (calculated for C20H19N4O3: 363.1457).
2-{(2Z)-2-[(4-methylanilino)(pyridin-4-yl)methylidene]hydrazinecarbonyl}cyclohex-1-ene-1-carboxylic acid (2f)
m.p. 132–134 °C, yield 90.00%. 1H NMR (400 MHz, DMSO-d6): 12.30 (sb, 1 H, COOH), 10.30 (s, 0.5 H, NHCO), 10.24 (s, 0.5 H, NHCO), 8.80–6.92 (m, 8 H + 1NH), 6.57 (d, 1H CH=), 6.50 (d, 1H CH=), 2.34–2.21 (m, 4 H), 2.18 (s, 3 H), 1.64 (s, 2H), 1.61 (s, 2H) ppm. 13C NMR (100 MHz, DMSO-d6): 173.05, 168.48, 168.38, 168.03, 166.84, 150.18, 150.09, 144.56, 143.28, 142.43, 142.36, 140.35, 139.56, 131.63, 130.65, 129.82, 129.44, 128.70, 126.47, 124.32, 123.69, 123.18, 122.43, 122.30, 121.07, 119.55, 28.71, 28.38, 25.54, 25.03, 21.99, 21.76, 21.63, 21.53, 20.92, 20.80, 20.67, 19.99, 19.84 ppm. Elem. Anal. for C21H22N4O3: calculated, C, 66.65%; H, 5.86%; N, 14.75%; found, C, 66.82%; H, 5.89%; N, 14.81%. HR-MS m/z 377.1620 [M+ − 1] (calculated for C21H21N4O3: 377.1614).

4.3. Peripheral Blood Mononuclear Cell Cultures

Experiments using peripheral blood mononuclear cells (PBMCs) were conducted according to the guidelines of the Declaration of Helsinki and approved by the Collegium Medicum of Nicolaus Copernicus University Bioethical Commission (KB 39/2019). Informed consent for participation was obtained from all subjects involved in the study. After obtaining informed consent, 18 mL of fresh blood was collected into two heparin sodium blood tubes (Medlab Products, Raszyn, Poland) from four healthy donors (aged 22–56 years) at the Occupational Medicine Clinic located in Dr. Antoni Jurasz University Hospital in Bydgoszcz, Poland. PBMCs were isolated by density gradient centrifugation (Lymphosep, BioWest, Nuaille, France). After PBMC isolation, the cell count and viability were determined using 0.4% trypan blue stain (Logos Biosystems, Gyeonggi-do, Republic of Korea) and an automated cell counter (LUNA II, Logos Biosystems, Gyeonggi-do, Republic of Korea). The cell viability was greater than 90%. For all experiments, freshly isolated PBMCs (1.0–1.5 × 106 cells/mL) were cultured with compounds 2a2f and racemic ibuprofen (IBU) (Sigma Aldrich, Burlington, MA, USA) in RPMI 1640 medium (Biomed Lublin, Lublin, Poland) in the presence of 5% heat-inactivated fetal bovine serum (FBS) (Euroclone, Pero, Milan, Italy). All studied compounds and IBU were initially dissolved in DMSO (Sigma Aldrich, Burlington, MA, USA), and then in the culture medium to achieve concentrations of 10, 50, and 100 µg/mL.

4.4. Cell Toxicity Analysis Protocol

The toxic effects of the studied compounds were assessed in PBMC culture using flow cytometry. PBMCs were incubated with compounds 2a2f at concentrations of 10, 50, and 100 µg/mL in polypropylene, round-bottom tubes (FALCON, Corning Science Mexico, Tamaulipas, Mexico) for 24 h at 37 °C and in a 5% CO2 condition. Control cultures included PBMCs only or PBMCs with DMSO (0.05–0.50%, corresponding to the solvent concentrations in the cultures with the compounds), or PBMCs with DMSO and IBU (at concentrations of 10, 50, and 100 µg/mL). After incubation, apoptosis was assessed using annexin V-FITC and propidium iodide (PI) double staining (Annexin V Apoptosis Detection Kit I, BD Pharmingen, San Diego, CA, USA). The cells were then analyzed using a CytoFLEX flow cytometer (Beckman Coulter, Suzhou, China). Flow cytometry acquisition and analysis were performed on at least 10,000 acquired events using CytExpert 2.3 software (Beckman Coulter, Suzhou, China). The flow cytometric gating strategy is shown in Figure 10.

4.5. Lymphocyte Proliferation Assay Protocol

The antiproliferative effects of the studied compounds were assessed in BD Horizon Violet Proliferation Dye 450 (VPD450, BD Pharmingen)-stained PBMC cultures using flow cytometry. Freshly isolated PBMCs (10–20 × 106 cells/mL of PBS) were labelled with 1uM VPD450 for 11 min at 37 °C. The labelling reaction was stopped by medium containing 10% FBS, and the cells were then re-suspended to a concentration of 1 × 106 cells/mL in RPMI 1640 supplemented with 5% FBS.
500 µL of VPD450-stained PBMCs were cultured in conical polypropylene tubes (Googlab Scientific, Rokocin, Poland) for 72 h at 37 °C in a 5% CO2 atmosphere. The cultures were stimulated with phytohemagglutinin (PHA) (Sigma Aldrich, Burlington, MA, USA) (2 µL) and increasing concentration of compounds 2a2f (10, 50, and 100 μg/mL) dissolved in DMSO. Control samples included PHA alone (positive control), PHA and DMSO, or PHA and IBU. After incubation, the culture tubes were centrifuged at 400× g at room temperature (RT) for 5 min, washed once with PBS, and 10,000 cells per sample were acquired on a CytoFLEX flow cytometer (Beckman Coulter Life Sciences, Brea, CA, USA). Data analysis was performed using CytExpert 2.3 software. The flow cytometric gating strategy is shown in Figure 11.

4.6. Cytokine Assay Protocol

PBMCs were stimulated with lipopolysaccharide (LPS) from E. coli serotype O55:B5 (Sigma-Aldrich, Burlington, MA, USA) at a concentration of 1 μg/mL, along with increasing concentrations of compounds 2a2f, for 24 h in 24-well polypropylene, non-adherent plates (CytoGen, Zgierz, Poland). Control PBMC cultures contained either medium alone, LPS and DMSO, or LPS and IBU. After incubation, samples were centrifuged at room temperature for 5 min at a speed of 400× g. The culture supernatants were collected into 1.5 mL tubes (Nest Biotechnology, Wuxi, Jiangsu, China) and frozen at −30 °C. After thawing, the supernatants were used to assess the concentrations of IL-1β, IL-6, TNF-α, and IL-10 using the enzyme-linked immunosorbent method (ELISA) and BD OptEIA Set Human ELISA kits (Becton Dickinson, Franklin Lakes, NJ, USA). The standard concentration ranges were as follows: IL-1β: 3,9–250 pg/mL, IL-6: 4,7–300 pg/mL, IL-10 and TNF-α: 7,8–500 pg/mL. The samples were analyzed with iEMS Reader MF (Labsystems, Helsinki, Finland), and cytokine concentrations were calculated using Ascent 2.4 software (Coimbatore, India).

4.7. Antimicrobial Activity Assay

The antimicrobial activity of derivatives 2a2f was tested against bacterial strains derived from the American Type Culture Collection (ATCC), including Gram-negative bacteria Escherichia coli ATCC 25922, Yersinia enterocolitica O3, and Klebsiella pneumoniae ATCC 700603; Gram-positive bacterium Staphylococcus aureus ATCC 25923; pathogenic yeast Candida albicans ATCC 90,028; and Mycobacterium smegmatis, an isolate deposited in the collection of the Department of Genetics and Microbiology, Maria Curie-Skłodowska University.
The minimum inhibitory concentration (MIC) value was determined in vitro using the microdilution method in sterile 96-well microplates, following the guidelines of the Clinical and Laboratory Standards Institute (CLSI) [20]. The tested samples were dissolved in DMSO at a concentration of 10.24 mg/mL, then diluted ten-fold, followed by two-fold serial dilutions (ranging from 512 to 0.5 μg/mL) in Mueller–Hinton (MH) broth.

4.8. Statistical Analysis

Data were analyzed and visualized using GraphPad software (ver. 10.2.3, Dotmatics, Boston, MA, USA). All p-values were calculated using the nonparametric Mann–Whitney U test.

5. Conclusions

Six new compounds, 2a2f, containing a cyclohex-1-ene-1-carboxylic acid moiety, were designed, synthesized, and studied for biological activity. Derivatives 2a and 2c2f showed dose-dependent antiproliferative activity against PBMCs, comparable to or stronger than IBU. Compounds 2a, 2d, and 2f almost completely inhibited lymphocyte proliferation at the highest concentration. Moreover, derivative 2d, at the highest dose, elevated TNF-α levels, suggesting potential anticancer activity. Compound 2f consistently reduced TNF-α production across all doses, indicating its potential as an anti-inflammatory drug. Compound 2b at a concentration of 100 µg/mL strongly inhibited IL-6, TNF-α, and IL-10 release and exhibited antibacterial activity against Y. eneterocolitica, but its toxicity may limit its therapeutic use. Derivative 2c demonstrated the strongest antibacterial activity against M. smegmatis and S. aureus. It is worth noting that the antibacterial activity of compound 2c was observed at a concentration of 64 µg/mL, where it was non-toxic to PBMCs.
Some structure–activity relationships were identified for the studied compounds. For example, the 2-pyridyl substituent at the R1 position enhances antiproliferative and antibacterial potency, while the presence of two 2-pyridyl substituents is essential for cytokine release inhibition. Additionally, the 4-NO2-phenyl substituent may increase antiproliferative activity, while the 4-CH3-phenyl substituent may enhance both antiproliferative and antibacterial effects. The presence of an unsaturated double bond in the cyclohexene ring may further boost antiproliferative activity and antibacterial efficacy against Gram-negative bacterial strains, such as Y. enterocolitica. These findings could guide the development of new drug candidates among amidrazone derivatives.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/molecules30081853/s1: Figure S1. 1H NMR spectrum of compound 2a. Figure S2. 1H NMR spectrum of compound 2b. Figure S3. 1H NMR spectrum of compound 2c. Figure S4. 1H NMR spectrum of compound 2d. Figure S5. 1H NMR spectrum of compound 2e. Figure S6. 1H NMR spectrum of compound 2f. Figure S7. 13C NMR spectrum of compound 2a. Figure S8. 13C NMR spectrum of compound 2b. Figure S9. 13C NMR spectrum of compound 2c. Figure S10. 13C NMR spectrum of compound 2d. Figure S11. 13C NMR spectrum of compound 2e. Figure S12. 13C NMR spectrum of compound 2f. Figure S13. Representative flow cytometric analysis of alive, early apoptotic, late apoptotic, and necrotic cells in PBMC cultures stimulated with different doses of DMSO (top row), IBU (middle row), and compound 2a (bottom row). Figure S14. Representative flow cytometry analysis of proliferating lymphocytes in PHA-stimulated VPD-450-labelled PBMC cultures exposed to different doses of DMSO (top row), IBU (middle row), and compound 2a (bottom row). Table S1. Toxicity of compounds 2a2f toward PBMCs (24h). Figure S15. The effect of compounds 2a2f on IL-1β production in 72 h PBMC cultures stimulated with lipopolysaccharide (LPS).

Author Contributions

Conceptualization, R.P.; methodology, R.P., A.H.-B. and J.K.; formal analysis, R.P.; investigation, R.P., E.P. A.H.-B. and J.K.; writing—original draft preparation, R.P.; writing—review and editing, A.H.-B., G.M.M., J.K. and A.E.; visualization, R.P.; supervision, R.P.; project administration, R.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been supported by Nicolaus Copernicus University (IDUB DEBIUTY4 grant for Renata Paprocka).

Institutional Review Board Statement

Experiments using peripheral blood mononuclear cells (PBMCs) were conducted according to the guidelines of the Declaration of Helsinki and approved by the Collegium Medicum of Nicolaus Copernicus University Bioethical Commission (KB39/2019).

Informed Consent Statement

Informed consent for participation was obtained from all subjects involved in the study.

Data Availability Statement

Data are available from the authors.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

ATCCAmerican Type Culture Collection
DAMPdanger/damage-associated molecular pattern
DMSOdimethyl sulfoxide
IBUibuprofen
ILinterleukin
LPSlipopolysaccharide
MICminimal inhibitory concentration
NETneutrophil extracellular traps
PAMPpathogen-associated molecular pattern
PBMCsperipheral blood mononuclear cells
PHAphytohemagglutinin
TNF-αtumor necrosis factor-alpha

References

  1. Actor, J.K. Chapter 2—The inflammatory response. In Introductory Immunology, 3rd ed.; Academic Press: Cambridge, MA, USA, 2023; pp. 21–38. [Google Scholar] [CrossRef]
  2. Kolaczkowska, E.; Kubes, P. Neutrophil recruitment and function in health and inflammation. Nat. Rev. Immunol. 2013, 13, 159–175. [Google Scholar] [CrossRef] [PubMed]
  3. Pittman, K.; Kubes, P. Damage-associated molecular patterns control neutrophil recruitment. J. Innate Immun. 2013, 5, 315–323. [Google Scholar] [CrossRef] [PubMed]
  4. Zeliger, H.I. Chapter 9—Inflammation. In Oxidative Stress; Academic Press: Cambridge, MA, USA, 2023; pp. 101–109. [Google Scholar] [CrossRef]
  5. Castanheira, F.V.S.; Kubes, P. Neutrophils and NETs in modulating acute and chronic inflammation. Blood 2019, 133, 2178–2185. [Google Scholar] [CrossRef] [PubMed]
  6. Muzammil, M.A.; Fariha, F.; Patel, T.; Sohail, R.; Kumar, M.; Khan, E.; Khanam, B.; Kumar, S.; Khatri, M.; Varrassi, G.; et al. Advancements in Inflammatory Bowel Disease: A Narrative Review of Diagnostics, Management, Epidemiology, Prevalence, Patient Outcomes, Quality of Life, and Clinical Presentation. Cureus 2023, 15, e41120. [Google Scholar] [CrossRef] [PubMed]
  7. Kaur, P.; Bernela, M.; Kaushal, P.; Verma, N.; Thakur, R.; Ahuja, M. Anti-Inflammatory Therapeutics: Conventional Concepts and Future with Nanotechnology. Recent. Adv. Inflamm. Allergy Drug Discov. 2023, 17, 7–19. [Google Scholar] [CrossRef] [PubMed]
  8. Paprocka, R.; Wiese-Szadkowska, M.; Kosmalski, T.; Frisch, D.; Ratajczak, M.; Modzelewska-Banachiewicz, B.; Studzińska, R. A Review of the Biological Activity of Amidrazone Derivatives. Pharmaceuticals 2022, 15, 1219. [Google Scholar] [CrossRef] [PubMed]
  9. Tullis, J.S.; VanRens, J.C.; Natchus, M.G.; Clark, M.P.; De, B.; Hsieh, L.C.; Janusz, M.J. The development of new triazole based inhibitors of tumor necrosis factor-α (TNF-α) production. Bioorganic Med. Chem. Lett. 2003, 13, 1665–1668. [Google Scholar] [CrossRef] [PubMed]
  10. Fernandes, S.P.S.; Gonçalves, J.M.; Silva, B.F.; Pereira, E.Q.; Coutinho, P.J.G.; Pereira-Wilson, C.; Dias, A.M. Novel Imidazole-based Hydrazonoyl Cyanides and Amidrazones Containing N(N,O)-Heterocycles: Selective Synthesis, Reaction Mechanisms and Preliminary Anticancer Evaluation. Eur. J. Org. Chem. 2024, 27, e202400253. [Google Scholar] [CrossRef]
  11. Habashneh, A.Y.; El-Abadelah, M.M.; Bardaweel, S.K.; Taha, M.O. Synthesis and Structure-Activity Relationship; Exploration of some Potent Anti-Cancer Phenyl Amidrazone Derivatives. Med. Chem. 2018, 14, 468–477. [Google Scholar] [CrossRef] [PubMed]
  12. Zhao, H.; Zou, J.; Xu, W.; Hu, D.; Guo, L.-D.; Chen, J.-X.; Chen, G.-D.; So, K.-F.; Yao, X.-S.; Gao, H. Diisoprenyl-cyclohexene/ane-Type Meroterpenoids from Biscogniauxia sp. and Their Anti-inflammatory Activities. J. Org. Chem. 2021, 86, 11177–11188. [Google Scholar] [CrossRef] [PubMed]
  13. Gurgul, A.; Wu, Z.; Han, K.-Y.; Shetye, G.; Sydara, K.; Souliya, O.; Johnson, J.J.; Che, C.-T. Polyoxygenated cyclohexene derivatives and other constituents of Uvaria rufa stem. Phytochemistry 2023, 216, 113884. [Google Scholar] [CrossRef] [PubMed]
  14. Cheng, Y.; Li, H.; Wu, D.; Hu, Y.; Li, J.; Yang, Y.; Li, J.; Zhou, H.; Zhang, H.; Xie, C.; et al. Anti-inflammatory polyoxygenated cyclohexene derivatives from Uvaria macclurei. Phytochemistry 2023, 214, 113797. [Google Scholar] [CrossRef] [PubMed]
  15. Su, R.; Cao, W.; Ma, G.; Li, W.; Li, Z.; Liu, Y.; Chen, L.; Chen, Z.; Li, X.; Cui, P.; et al. Cyclohexene oxide CA, a derivative of zeylenone, exhibits anti-cancer activity in glioblastoma by inducing G0/G1 phase arrest through interference with EZH2. Front. Pharmacol. 2024, 14, 1326245. [Google Scholar] [CrossRef] [PubMed]
  16. Modzelewska-Banachiewicz, B.; Ucherek, M.; Zimecki, M.; Kutkowska, J.; Kaminska, T.; Morak-Młodawska, B.; Paprocka, R.; Szulc, M.; Lewandowski, G.; Marciniak, J.; et al. Reactions of N3-substituted amidrazones with cis-1,2-cyclohexanedicarboxylic anhydride and biological activities of the products. Arch. Pharm. 2012, 345, 486–494. [Google Scholar] [CrossRef] [PubMed]
  17. Mazur, L.; Sączewski, J.; Jarzembska, K.N.; Szwarc-Karabyka, K.; Paprocka, R.; Modzelewska-Banachiewicz, B. Synthesis, structural characterization and reactivity of new trisubstituted N1-acylamidrazones: Solid state and solution studies. CrystEngComm 2018, 20, 4179–4193. [Google Scholar] [CrossRef]
  18. Mazur, L.; Modzelewska-Banachiewicz, B.; Paprocka, R.; Zimecki, M.; Wawrzyniak, U.E.; Kutkowska, J.; Ziółkowska, G. Synthesis, crystal structure and biological activities of a novel amidrazone derivative and its copper(II) complex—A potential antitumor drug. J. Inorg. Biochem. 2012, 114, 55–64. [Google Scholar] [CrossRef] [PubMed]
  19. Modzelewska, B.; Pyra, E. Synthesis of N3-substituted amidrazones. Ann. UMCS Sec. AA 1995, 50/51, 111. [Google Scholar]
  20. Clinical and Laboratory Standards Institute. Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard, 9th ed.; CLSI Document M07-A9; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2012. [Google Scholar]
Scheme 1. The synthesis of compounds 2a2f from the reaction of amidrazones 1a1f with 3,4,5,6-tetrahydrophthalic anhydride.
Scheme 1. The synthesis of compounds 2a2f from the reaction of amidrazones 1a1f with 3,4,5,6-tetrahydrophthalic anhydride.
Molecules 30 01853 sch001
Figure 1. Frequencies of viable, early apoptotic, late apoptotic, and necrotic cells in PBMC cultures (24 h) with compounds 2a2f and IBU at concentrations of 10, 50, and 100 µg/mL.
Figure 1. Frequencies of viable, early apoptotic, late apoptotic, and necrotic cells in PBMC cultures (24 h) with compounds 2a2f and IBU at concentrations of 10, 50, and 100 µg/mL.
Molecules 30 01853 g001
Figure 2. The antiproliferative effect of compounds 2a2f on a 24 h PBMC culture stimulated with phytohemagglutinin (PHA). Results (from 5 independent experiments, expressed as median and Q1–Q3 range) are presented as a percentage of the positive control (cultures stimulated with PHA alone). Dots in different colors indicate the results of independent experiments. The symbols indicate the statistical significance (Mann–Whitney U test): * vs. DMSO, p < 0.05; # vs. IBU, p < 0.05; ** vs. DMSO p < 0.01; ## vs. IBU, p < 0.01.
Figure 2. The antiproliferative effect of compounds 2a2f on a 24 h PBMC culture stimulated with phytohemagglutinin (PHA). Results (from 5 independent experiments, expressed as median and Q1–Q3 range) are presented as a percentage of the positive control (cultures stimulated with PHA alone). Dots in different colors indicate the results of independent experiments. The symbols indicate the statistical significance (Mann–Whitney U test): * vs. DMSO, p < 0.05; # vs. IBU, p < 0.05; ** vs. DMSO p < 0.01; ## vs. IBU, p < 0.01.
Molecules 30 01853 g002
Figure 3. The effect of compounds 2a2f on TNF-α production in a 72 h PBMC culture stimulated with lipopolysaccharide (LPS). Results (from 4 independent experiments, expressed as median and Q1–Q3 range) are presented as a percentage of the positive control (cultures stimulated with LPS alone). Dots in different colors indicate the results of independent experiments. The symbols indicate the statistical significance (Mann–Whitney U test): * vs. DMSO, p < 0.05; # vs. IBU, p < 0.05; $ vs. LPS alone, p < 0.05.
Figure 3. The effect of compounds 2a2f on TNF-α production in a 72 h PBMC culture stimulated with lipopolysaccharide (LPS). Results (from 4 independent experiments, expressed as median and Q1–Q3 range) are presented as a percentage of the positive control (cultures stimulated with LPS alone). Dots in different colors indicate the results of independent experiments. The symbols indicate the statistical significance (Mann–Whitney U test): * vs. DMSO, p < 0.05; # vs. IBU, p < 0.05; $ vs. LPS alone, p < 0.05.
Molecules 30 01853 g003
Figure 4. The effect of compounds 2a2f on IL-6 production in a 72 h PBMC culture stimulated with lipopolysaccharide (LPS). Results (from 4 independent experiments, expressed as median and Q1–Q3 range) are presented as a percentage of the positive control (cultures stimulated with LPS alone). Dots in different colors indicate the results of independent experiments. The symbols indicate the statistical significance (Mann–Whitney U test): * vs. DMSO, p < 0.05; # vs. IBU, p < 0.05.
Figure 4. The effect of compounds 2a2f on IL-6 production in a 72 h PBMC culture stimulated with lipopolysaccharide (LPS). Results (from 4 independent experiments, expressed as median and Q1–Q3 range) are presented as a percentage of the positive control (cultures stimulated with LPS alone). Dots in different colors indicate the results of independent experiments. The symbols indicate the statistical significance (Mann–Whitney U test): * vs. DMSO, p < 0.05; # vs. IBU, p < 0.05.
Molecules 30 01853 g004
Figure 5. The effect of compounds 2a2f on IL-10 production in a 72 h PBMC culture stimulated with lipopolysaccharide (LPS). Results (from 4 independent experiments, expressed as median and Q1–Q3 range) are presented as a percentage of the positive control (cultures stimulated with LPS alone). Dots in different colors indicate the results of independent experiments. The symbols indicate the statistical significance (Mann–Whitney U test): * vs. DMSO, p < 0.05; # vs. IBU, p < 0.05.
Figure 5. The effect of compounds 2a2f on IL-10 production in a 72 h PBMC culture stimulated with lipopolysaccharide (LPS). Results (from 4 independent experiments, expressed as median and Q1–Q3 range) are presented as a percentage of the positive control (cultures stimulated with LPS alone). Dots in different colors indicate the results of independent experiments. The symbols indicate the statistical significance (Mann–Whitney U test): * vs. DMSO, p < 0.05; # vs. IBU, p < 0.05.
Molecules 30 01853 g005
Figure 6. Amidrazone derivatives 2a2f containing (a) cyclohex-1-ene-1-carboxylic acid moiety, (b) derivatives 3a, 3b, 3d, and 3e with cyclohexanecarboxylic acid moiety, or (c) 4b with cyclohex-3-ene-1-carboxylic acid moiety, as described in previous works [16,18].
Figure 6. Amidrazone derivatives 2a2f containing (a) cyclohex-1-ene-1-carboxylic acid moiety, (b) derivatives 3a, 3b, 3d, and 3e with cyclohexanecarboxylic acid moiety, or (c) 4b with cyclohex-3-ene-1-carboxylic acid moiety, as described in previous works [16,18].
Molecules 30 01853 g006
Figure 7. Structure–antiproliferative activity relationships observed for amidrazone derivatives 2a2f, 3a, 3b, 3d, 3e, and 4b.
Figure 7. Structure–antiproliferative activity relationships observed for amidrazone derivatives 2a2f, 3a, 3b, 3d, 3e, and 4b.
Molecules 30 01853 g007
Figure 8. Structural elements of amidrazone derivatives 2a2f, 3a, 3b, 3d, 3e, and 4b and their impact on the levels of IL-6, TNF-α, and IL-10 cytokines.
Figure 8. Structural elements of amidrazone derivatives 2a2f, 3a, 3b, 3d, 3e, and 4b and their impact on the levels of IL-6, TNF-α, and IL-10 cytokines.
Molecules 30 01853 g008
Figure 9. Structure–antimicrobial activity relationships observed for amidrazone derivatives 2a2f, 3a, 3b, 3d, and 3e.
Figure 9. Structure–antimicrobial activity relationships observed for amidrazone derivatives 2a2f, 3a, 3b, 3d, and 3e.
Molecules 30 01853 g009
Figure 10. Flow cytometry gating strategy for identifying live, early apoptotic, late apoptotic, and necrotic cells in PBMC culture: (a) PBMCs were gated using a Forward Scatter Area (FSC-A)/Side Scatter Area (SSC-A) dot plot. (b) Singlet PBMCs were gated using FSC-A and Forward Scatter Height (FSC-H). (c) Cells were analyzed using an annexin V/propidium iodide (PI) dot plot. The identified regions are as follows: necrotic cells (annexin V−/PI+), late apoptotic cells (annexin V+/PI+), early apoptotic cells (annexin V+/PI−), live cells (annexin V−/PI−). The gating strategy shown represents PBMCs stimulated with 10 µL of DMSO. The same gating strategy applied to all samples.
Figure 10. Flow cytometry gating strategy for identifying live, early apoptotic, late apoptotic, and necrotic cells in PBMC culture: (a) PBMCs were gated using a Forward Scatter Area (FSC-A)/Side Scatter Area (SSC-A) dot plot. (b) Singlet PBMCs were gated using FSC-A and Forward Scatter Height (FSC-H). (c) Cells were analyzed using an annexin V/propidium iodide (PI) dot plot. The identified regions are as follows: necrotic cells (annexin V−/PI+), late apoptotic cells (annexin V+/PI+), early apoptotic cells (annexin V+/PI−), live cells (annexin V−/PI−). The gating strategy shown represents PBMCs stimulated with 10 µL of DMSO. The same gating strategy applied to all samples.
Molecules 30 01853 g010
Figure 11. Representative flow cytometry gating strategy identifying proliferating lymphocytes in PBMCs: (a) PBMCs were gated on a Forward Scatter Area (FSC-A)/Side Scatter Area (SSC-A) dot plot. (b) Singlet PBMCs were gated on FSA-A and FSC Height (FSA-H). (c) Proliferating cells in non-VPD450-labelled PBMCs were identified on a VPD450 histogram. (d) Proliferating cells in VPD450-labelled PBMCs were identified on a VPD450 histogram. The represented gating strategy refers to unstimulated non-VPD450- and VPD450-labelled PBMCs. The same gating strategy was applied to all culture samples.
Figure 11. Representative flow cytometry gating strategy identifying proliferating lymphocytes in PBMCs: (a) PBMCs were gated on a Forward Scatter Area (FSC-A)/Side Scatter Area (SSC-A) dot plot. (b) Singlet PBMCs were gated on FSA-A and FSC Height (FSA-H). (c) Proliferating cells in non-VPD450-labelled PBMCs were identified on a VPD450 histogram. (d) Proliferating cells in VPD450-labelled PBMCs were identified on a VPD450 histogram. The represented gating strategy refers to unstimulated non-VPD450- and VPD450-labelled PBMCs. The same gating strategy was applied to all culture samples.
Molecules 30 01853 g011
Table 1. Minimum inhibitory concentration (MIC) values (µg/mL) of compounds 2a2f against tested bacterial and fungal strains compared to ampicillin (Amp) and fluconazole (Flu).
Table 1. Minimum inhibitory concentration (MIC) values (µg/mL) of compounds 2a2f against tested bacterial and fungal strains compared to ampicillin (Amp) and fluconazole (Flu).
StrainMIC Values (µg/mL)
2a2b2c2d2e2fAmpFlu
Mycobacterium smegmatis6451264512512>51216-
Staphylococcus aureus ATCC 2592325625664512512>5120.25-
Escherichia coli ATCC 25922512256512512>5125128-
Yersinia enterocolitica O35126451251251212816-
Klebsiella pneumoniae ATCC 700603512256256512512>512>256-
Candida albicans ATCC 90028512>512512512512256-0.25
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

Paprocka, R.; Kutkowska, J.; Paczkowska, E.; Mwaura, G.M.; Eljaszewicz, A.; Helmin-Basa, A. Synthesis, Evaluation of Biological Activity, and Structure–Activity Relationships of New Amidrazone Derivatives Containing Cyclohex-1-ene-1-Carboxylic Acid. Molecules 2025, 30, 1853. https://doi.org/10.3390/molecules30081853

AMA Style

Paprocka R, Kutkowska J, Paczkowska E, Mwaura GM, Eljaszewicz A, Helmin-Basa A. Synthesis, Evaluation of Biological Activity, and Structure–Activity Relationships of New Amidrazone Derivatives Containing Cyclohex-1-ene-1-Carboxylic Acid. Molecules. 2025; 30(8):1853. https://doi.org/10.3390/molecules30081853

Chicago/Turabian Style

Paprocka, Renata, Jolanta Kutkowska, Ewelina Paczkowska, Godwin Munroe Mwaura, Andrzej Eljaszewicz, and Anna Helmin-Basa. 2025. "Synthesis, Evaluation of Biological Activity, and Structure–Activity Relationships of New Amidrazone Derivatives Containing Cyclohex-1-ene-1-Carboxylic Acid" Molecules 30, no. 8: 1853. https://doi.org/10.3390/molecules30081853

APA Style

Paprocka, R., Kutkowska, J., Paczkowska, E., Mwaura, G. M., Eljaszewicz, A., & Helmin-Basa, A. (2025). Synthesis, Evaluation of Biological Activity, and Structure–Activity Relationships of New Amidrazone Derivatives Containing Cyclohex-1-ene-1-Carboxylic Acid. Molecules, 30(8), 1853. https://doi.org/10.3390/molecules30081853

Article Metrics

Back to TopTop