Next Article in Journal
Nanostructured Lipid Carrier-Based Codelivery of Raloxifene and Naringin: Formulation, Optimization, In Vitro, Ex Vivo, In Vivo Assessment, and Acute Toxicity Studies
Next Article in Special Issue
Employ of Anthocyanins in Nanocarriers for Nano Delivery: In Vitro and In Vivo Experimental Approaches for Chronic Diseases
Previous Article in Journal
Computation-Aided Design of Albumin Affibody-Inserted Antibody Fragment for the Prolonged Serum Half-Life
Previous Article in Special Issue
Investigations on Cellular Uptake Mechanisms and Immunogenicity Profile of Novel Bio-Hybrid Nanovesicles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Microbiological Screening of 5-Functionalized Pyrazoles for the Future Development of Optimized Pyrazole-Based Delivery Systems

1
Department of Pharmacy (DIFAR), Section of Medicinal Chemistry, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy
2
Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy
3
Department of Pharmacy (DIFAR), Section of Organic Chemistry, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy
4
Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL 60612, USA
*
Author to whom correspondence should be addressed.
Pharmaceutics 2022, 14(9), 1770; https://doi.org/10.3390/pharmaceutics14091770
Submission received: 26 July 2022 / Revised: 16 August 2022 / Accepted: 22 August 2022 / Published: 24 August 2022

Abstract

:
The pyrazole ring represents a widely applied chemical scaffold in medicinal chemistry research and we have observed that the physicochemical and biological features of highly substituted pyrazoles can be successfully improved by their encapsulation in dendrimer nanoparticles (NPs). For the future development of new optimized antibacterial delivery systems, we report the synthesis and biological evaluation of 5-amino functionalized pyrazole library (compounds 27). In detail, new derivatives 27 were differently decorated in C3, C4 and C5 positions. An in silico study predicted pyrazoles 27 to exert good drug-like and pharmacokinetic properties. Compounds 3c and 4b were endowed with moderate, but nanotechnologically improvable activity against multidrug-resistant (MDR) clinical isolates of Gram-positive species, especially of the Staphylococcus genus (MICs = 32–64 µg/mL). In addition, derivatives 3c and 4a showed moderate activities against Mycobacterium tuberculosis and 4a evidenced activity also against MDR strains. Overall, the collected evidence supported that, upon nano-formulation with proper polymer matrices, the new synthesized compounds could provide new pyrazole-based drug delivery systems with an enhanced and enlarged-spectrum of antibacterial activity.

Graphical Abstract

1. Introduction

The five-membered heterocycle pyrazole ring represents a widely applied chemical scaffold in medicinal chemistry research. Several pyrazole derivatives show a broad spectrum of biological properties, such as antimicrobial [1], analgesic [2], antifungal [3], anti-inflammatory [4], and antioxidant activities [5]. Collectively, the pyrazole nucleus possesses almost all types of pharmacological activities, and throughout the years, many researchers have studied its skeleton both chemically and biologically. Currently, the pyrazole nucleus is present in numerous pharmacological agents belonging to different therapeutic categories (Figure 1), as anti-inflammatory/analgesic compounds (Celecoxib, Tepoxalin, Betazole), anticancer (Crizotimib), antiobesity (Surinabant, Difenamizole), antidepressant and tranquillizer (Fezolamine, Mepiprazole) and many others, confirming the pharmacological potential of this heterocyclic ring [6]. Particularly, 5-pyrazolyl-ureas (5-PUs) (Figure 1) are reported and studied for the treatment of different microbial infections caused by bacteria or protozoa of the genera Plasmodium, Toxoplasma, and others [7] or to treat cancer (e.g., antiangiogenic agents and Src, p38-MAPK and TrKa inhibitors) [7,8,9,10]. Notably, 5-PUs bearing a 2-hydroxy-2-phenylethyl chain at the N1 position were identified as effective anticancer, anti-inflammatory, and anti-Mycobacterium agents [8,9,10,11,12,13].
Additionally, we have observed that the water solubility and antibacterial effects of two differently functionalized pyrazoles synthetized by us (BBB4 and CB1H) (Figure 1) were remarkably improved by their encapsulation in polyester-based dendrimer nanoparticles (NPs) and in styrene-based copolymer NPs [6,14]. In particular, the obtained BBB4-G4K NPs and CB1H-P7 NPs were able to contrast multidrug-resistant (MDR) pathogenic species, also including clinical isolates particularly difficult to treat, due to the onset of resistance versus the most potent antibiotics, such as carbapenems and colistin [15,16]. Additionally, the developed encapsulated formulations showed lower cytotoxicity in human cells and higher selectivity indices than the free pyrazole compounds [15,16]. On the other hand, other 5-pyrrolyl-pyrazoles I (Figure 1), characterized as our previous 5-PUs by an N1-hydroxy-2-phenylethyl chain, evidenced some anti-Mycobacterium activity [13].
In the current scenario of bacterial infections, those caused by bacteria of the ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumonia, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter) family, which are recognized as dangerous superbugs by the Infectious Diseases Society of America (IDSA), are becoming a global concern because they are almost untreatable [6]. Unfortunately, ESKAPE bacteria are endowed with an increasing tendency to develop MDR to the bactericidal effect of conventional antibiotics, which are no longer effective and need substitution with more efficient new antibacterial agents suitable for clinical application [6]. To meet the global urgency of new antimicrobial compounds, the aim of this work is to investigate the antibacterial/anti-Mycobacterium activity of a small library of 5-amino functionalized pyrazoles (compounds 27, see the workflow of the whole study in Figure 2), structurally related to previously reported BBB4, CB1H, 5-PUs and I. To reduce the lipophilicity (and therefore increase water solubility) and preserve antimicrobial activity, the new pyrazole derivatives do not bear the 3,5-diphenyl groups but share with the parent compounds the N1 2-hydroxy-2-phenylethyl chain identified in previous studies as a relevant pharmacophoric portion [11,12,13]. Particularly, compounds 2 and 3 (Table 1) are characterized by an ureido moiety in the 5 position and a carboxyethyl (derivatives 2ad) or a tert-butyl (derivatives 3ad) substituent at C4 and C3, respectively. Compounds 46 (Table 1) share with derivatives 2 the 4-carboxyethyl substituent but bear at C5 a benzoyl-thiourea (derivatives 4ad), an amide (derivatives 5ad) or a sulfonamide function (derivative 6). Finally, compound 7 is a positional isomer of derivative 4c characterized by a carboxyethyl substituent moved from position 4 to position 3. Within each series of compounds, the substituent at position 5 was varied to define structure–activity relationships (SAR) on the effect of this functionalization on antibacterial/anti-Mycobacterium activity. In fact, except for compounds 5a, all compounds bear at different positions (ortho, meta, para) of the phenyl ring in the functional group at position 5 a fluorine atom or a trifluoromethyl group to increase lipophilicity.

2. Results and Discussion

2.1. Chemistry

Compounds 27 were synthesized by a divergent synthetic protocol starting from the pyrazole key intermediates 1ac (Scheme 1) [12,17] by functionalization of the 5-amino group with different electrophilic reagents as isocyanates, benzoylisocyanates, benzoyl chlorides and 4-fluorobenzenesulfonyl chloride. In general, reaction yields are good for compounds 24 (from 42% to 51%), and lower for compounds 5 and 6 (from 23% to 61%). In general, compounds derived from pyrazole 1a (substituted in C4 position) were obtained with yields higher than compounds obtained from pyrazoles 1b and 1c (substituted at the C3 position), as demonstrated comparing the reaction yields of compound 4c with its isomer 7 (51% versus 37%), evidencing a different reactivity versus electrophilic reagents of amino group at the 5 position of pyrazole 1a withrespect to pyrazoles 1b,c. As previously reported [11,12], compounds 2ad and 3ad, characterized by a urea moiety at the 5 position of the pyrazole nucleus, were synthesized by refluxing in anhydrous toluene 1a or 1c with a small excess (1.1. equivalents) of suitable phenyl isocyanate for 6 h. Compounds 4ac and 7, in which the urea moiety was transformed into thiourea, were obtained by reacting pyrazoles 1a,b with the suitable benzoyl isothiocyanate, previously prepared modifying a method from the literature [18], in anhydrous THF at reflux for 12 h. Finally, amides 5a-e and the sulphonamide 6 were obtained by condensing 1 with benzoyl chlorides (for derivatives 5) or 4-fluorobenzenesulfonyl chloride (for derivative 6) in anhydrous THF in the presence of triethylamine (TEA) at reflux for 18 h (compounds 5) or 3 days (compound 6).

2.2. Pharmacokinetic Properties and Druglikeness Prediction

To evaluate the pharmaceutical relevance of this 5-amino functionalized library, the pharmacokinetics properties, as well as the druglikeness of compounds 27 were calculated by SwissADME [19]. Collectively, an in silico study predicted these compounds to exert good drug-like and pharmacokinetic properties, particularly regarding physicochemical properties, lipophilicity and water solubility. Interestingly, no violations of the Lipinski rules were detected, as well as any pan assay interference compound (PAINS) alerts were reported. Exclusively for compounds 4 and 7, the presence of the thiocarbonyl functionality at the C5 position was spotted as problematic fragment(s) according to the Brenk filters [20]. Overall, the collected data support the pharmaceutical potentials of derivatives 3 and 4.
In detail, as reported in Table 2 and Table 3, the calculated physicochemical parameters (i.e., LogP range: 2.74–4.5; MW range: 396.46–462.42 g/mol; TPSA range: 79–137 Å2; Fraction Csp3 range: 0.18–0.3; number of rotatable bonds: 8–11) indicated a good oral bioavailability. In addition, compounds 2ac and 5a were predicted as water-soluble, whereas the other derivatives moderately water-soluble. Furthermore, the predicted gastrointestinal (GI) absorption would be high for derivatives 2,3,5,6 whereas thioureido compounds 4 and 7 would be poorly adsorbed in the GI tract. None of the analyzed compounds were predicted to penetrate the blood–brain barrier (BBB). According to the calculations, tested derivatives can inhibit some of the cytochrome (CYP) isoforms (1A2, 2C19, 2C9, 2D6, 3A4), but only 2ac and 3c are substrates of Pgp.

2.3. Antimicrobial Activity

Supported by the promising pharmacokinetic and druglikeness properties predicted for derivatives 27 and based on the structural similarities with the previous compounds BBB4, CB1H and I, the antibacterial activity of all compounds was preliminarily evaluated. Out of this initial screening, which established the inactivity of all compounds against bacteria of the Gram-negative species, compounds 3ac and 4ac, characterized by a urea moiety (3) or thiourea function (4) at the C5 position, evidenced in some cases promising antibacterial profiles. Based on these results, they have been selected for further evaluation against twenty-one clinical and MDR isolates of the genera Staphylococcus and Enterococcus (Table 4).
Compounds were considered inactive against a specific strain when MIC values higher than 128 µg/mL were observed. Thus, compound 3c showed widespread activity (MICs = 32–64 µg/mL) on selected isolates, emerging ineffective only against E. faecalis 365, and less clinically relevant enterococci and staphylococci, including E. gallinarum 150, E. casseliflavus 184, S. simulans 163 and S. haemolyticus 193 strains. Interestingly, the substitution pattern of the urea phenyl ring deeply affects the antibacterial properties of the compounds. Specifically, 5-PUs 3a (ortho-F substituted) and 3b (meta-F substituted) showed reduced activity in comparison with their 4-fluorophenyl analogue 3c, still capable of affecting the duplication of strains not influenced by 3c (namely, E. gallinarum 150 and E. casseliflavus 184). Collectively, compound 3c demonstrated significant antibacterial activity against practically all the isolates considered, regardless of their resistance patterns. Particularly, as reported in Table 4, compound 3c displayed MICs = 32–64 µg/mL against all staphylococci considered in this study, except for one isolate, and MICs = 64 µg/mL against all E. faecium isolates tested, two out of three isolates of E. faecalis and one out of three isolates of enterococci. Derivatives 4 emerged to be inactive against all strains of the Enterococcus genus, being moderately active (MICs = 32–64 µg/mL) on some isolates of the Staphylococcus genus (namely, S. aureus 18 and 187, S. epidermidis 180 and 181 and S. lugdunensis 129 (4b), S. aureus 187 and S. lugdunensis 129 (4c) and only S. lugdunensis 129 (4a)). Overall, compound 3c and, although with a very narrow spectrum on staphylococci, compound 4b resulted as the most promising molecules between compounds 3ac and 4ac for future nano-formulation studies. To further define their potency, the antibacterial effects of 3c and 4b were compared with those of commonly used antibiotics (Table 5). Both compounds showed similar or improved activities in comparison to the reference antibiotics (i.e., ampicillin, ciprofloxacin and oxacillin) against sensitive isolates, thus supporting the pharmaceutical potentials of these pyrazole derivatives as novel antibacterial agents.
Compounds 3c (para-F substituted) and 4b (meta-F substituted), characterized by a urea or thiourea function at C5 of the pyrazole nucleus, respectively, were found as the most active derivatives. On the contrary, the presence in this position of an amide (compounds 5) or sulfonamide (compounds 6) moiety negatively affected the antibacterial properties. Furthermore, relevant structural features for antibacterial activity include: (i) the carboxyethyl group at C4 and not at C3 (compare 4a with 7, Table 4); (ii) a tert-butyl substituent at C3 (as in compounds 3). However, the simultaneous presence of the carboxyethyl substituent at C4 and of a urea function at C5 (as in compounds 2) caused a reduction in biological activity. Finally, compounds in which the aromatic ring at the 5 position is not substituted (as in 5a) or presented a trifluoromethyl group (as in 3d and 5d) showed decreased activity with respect to fluoro-substituted analogues.
Data reported in Table 5 established that, both compounds 3c and 4b show promise in the development of new drug delivery systems to specifically counteract infections sustained by staphylococci, where both ciprofloxacin and most beta-lactam drugs are no longer active. Additionally, compound 3c also proved to be promising against enterococci isolates that are difficult to inhibit with ampicillin.

2.4. Anti-Mycobacterium Activity

All compounds were preliminary screened at 50 μg/mL for growth inhibition of replicating cultures of Mycobacterium tuberculosis using the Microplate Alamar Blue Assay (MABA). This concentration was selected according to literature data [13] and is normally used in screening protocols to test compounds against Mycobacterium tubercolosis. Additionally, pyrazoles 27 were tested against low oxygen-adapted M. tuberculosis containing a luxABCDE reporter under hypoxia using the low oxygen recovery assay (LORA) to assess compound activity against non-replicating cultures [22]. Most compounds 27 proved to be inactive, showing a percentage of inhibition concentration lower than 50% in the MABA test. Conversely, selected derivatives 3 and 4 evidenced a moderate activity (Table 6). Particularly, compounds 3c and 4a, characterized by a fluorophenyl substituent in para (3c) or ortho (4a) position, evidenced the most promising results, being able to block the duplication of M. tuberculosis at 50 μg/mL concentration. The derived MIC values for 3c and 4a were 35.8 and 39.3 μg/mL, respectively (Table 6). Unfortunately, none of the tested derivatives showed significant inhibition in the LORA test.
Finally, compounds 2-7 underwent extensive preliminary screening to assess their potential to inhibit the growth of MDR M. tuberculosis strains, responsible for a form of tuberculosis resistant to at least two first-line drugs for tuberculosis (Lilly TB Drug Discovery Initiative) [23]. From this large screening, only compound 4a evidenced weak activity at 20 μM concentration (31% of inhibition), representing a good starting point to develop new agents able to counteract MDR M. tuberculosis.

3. Materials and Methods

3.1. General Information

All chemicals were purchased by Chiminord srl and Merk (Aldrich Chemical) (Milan, Italy). Solvents were reagent grade. All commercial reagents were used without further purification. Aluminium-backed silica gel plates (Merck DC-Alufolien Kie-selgel 60 F254, Darmstad, Germany), were used in thin-layer chromatography (TLC) for routine monitoring of the reaction course. Merck silica gel, 230–400 mesh, was used for chromatography. Flash chromatography was performed using Isolera one instrument (Biotage, Uppsala, Sweden) using Silicagel column. Melting points are not “corrected” and were obtained with a Buchi M-560 instrument (Buchi instruments, Flawil, Switzerland). IR spectra were recorded with a Perkin-Elmer 398 spectrophotometer (Perkin-Elmer, Milan, Italy) for compounds 4, 5ac and 6 or Spectrum Two FT-IR Spectrometer (PerkinElmer, Inc., Waltham, MA, USA) for compounds 5d and 7. The 1H NMR spectra were recorded on a Varian Gemini 200 (200 MHz, Varian Gemini, Palo Alto, CA, USA); chemical shifts are reported as δ (ppm) relative to tetramethylsilane (TMS) as internal standard; signals were characterized as s (singlet), d (doublet), t (triplet), n t (near triplet), q (quartet), m (multiplet), br s (broad signal); J were reported in Hz (see Supporting Material). The 13C NMR spectra were recorded on a JEOL JNM ECZ-400S/L1 (101 MHz, Tokyo, Japan).
Elemental analysis was determined with an elemental analyzer EA 1110 (Fison-Instruments, Milan, Italy); products were considered pure when the difference between the calculated and found values is ± than 0.4 (see Table 7).

3.2. Synthesis of Compounds 2a-d and 3a-d

A mixture of 5-amino-pyrazoles 1a [17] or 1c [12] (10 mmol) and the suitable phenyl isocyanates (11 mmol) in anhydrous toluene (70 mL) was heated at reflux for 12 h. After cooling to room temperature, the organic solution was washed with 3 M HCl (2 × 20 mL), water (10 mL), dried (MgSO4) and evaporated under reduced pressure. The obtained crude oil was crystallized by adding a solution of diethyl ether/petroleum ether (b.p. 50–60 °C) (1:1) or purified by flash chromatography using a mixture of diethyl ether/petroleum ether (b.p. 50–60 °C) 1/1 as the eluent. Analytical data of compounds 2ad and 3ad are previously reported [11,12].

3.2.1. Synthesis of Compounds 4ac

A mixture of 5-amino-pyrazole 1a (275 mg, 1 mmol) [17] and the proper benzoyl isothiocyanate (1 mmol), previously prepared modifying a method from the literature [18], in anhydrous THF (10 mL) was refluxed for 12 h. After cooling to room temperature, the solution was concentrated under reduced pressure. The crude crystallized by adding a solution of diethyl ether/petroleum ether (b.p. 50–60 °C) (1:1) or purified by flash chromatography using a mixture of diethyl ether/petroleum ether (b.p. 50–60 °C) 1/1 as the eluent.

Synthesis of 2-Fluorobenzoyl isothiocyanate, 3-Fluorobenzoyl isothiocyanate and 4-Fluorobenzoyl isothiocyanate

To a suspension of anhydrous potassium thiocyanate (1.62 g, 16.7 mmoles) in an. toluene (15 mL), a solution of the suitable fluorobenzoyl chloride (13.9 mmoles) in an. toluene (5 mL) was added dropwise. The reaction was heated to 70 °C for 7 h. After the reaction was complete, the mixture was cooled to room temperature and filtered through a Celite pad. The solvent was evaporated to give an oil that crystallized upon standing in a refrigerator. The product was used without further purification (yield: 79%, 81% and 82% for 2-Fluorobenzoyl isothiocyanate, 3-Fluorobenzoyl isothiocyanate and 4-Fluorobenzoyl isothiocyanate, respectively).
Ethyl 5-(3-(2-fluorobenzoyl)-thioureido)-1-(2-hydroxy-2-phenylethyl)-1H-pyrazole-4-carboxylate4a. White solid. Yield 42%; mp 150–151 °C. 1H-NMR (400 MHz, CDCl3): δ 1.20 (t, J = 7.0, 3H, CH3), 3.92–4.10 (m, 4H, CH2O + CH2N), 4.87–4.90 (m, 1H, CHOH), 5.92 (br s, 1H, exchangeable), 7.19–7.43 (m, 9H Ar), 7.86 (s, 1H, H-3), 8.05 (br s, 1H, exchangeable), 12.00 (br s, 1H, exchangeable). 13C NMR (101 MHz, CDCl3): δ 177.0, 164.6, 161.4, 159.2, 142.3, 140.2, 132.2, 130.0, 128.4, 127.2, 125.1, 116.3, 103.4, 72.5, 60.1, 58.0, 14.3. FTIR (KBr): 3200–3032 (OH + NH), 1719 (COOEt), 1692 (CONH) cm−1. Anal. (C22H21N4O4SF) calcd for C, H, N, S (see Table 7).
Ethyl 5-(3-(3-fluorobenzoyl)-thioureido)-1-(2-hydroxy-2-phenylethyl)-1H-pyrazole-4-carboxylate4b. White solid. Yield 48%; mp 155–156 °C. 1H-NMR (400 MHz, DMSO-d6): δ 1.13 (t, J = 7.0, 3H, CH3), 4.06–4.17 (m, 4H, CH2O + CH2N), 4.91–4.94 (m, 1H, CHOH), 5.78 (br s, 1H, OH, exchangeable), 7.21–7.85 (m, 9H, Ar), 7.89 (s, 1H, H-3). 13C NMR (101 MHz, DMSO-d6): δ 177.0, 164.4, 161.3, 159.2, 142.3, 140.2, 132.3, 130.0, 128.4, 127.2, 125.1, 116.4, 103.5, 72.6, 60.2, 57.9, 14.3. FTIR (KBr): 3350–3050 (OH + NH), 1708 (COOEt), 1677 (CONH) cm−1. Anal. (C22H21N4O4SF) calcd for C, H, N, S (see Table 7).
Ethyl 5-(3-(4-fluorobenzoyl)-thioureido)-1-(2-hydroxy-2-phenylethyl)-1H-pyrazole-4-carboxylate4c. White solid. Yield 51%; mp 158–160 °C. 1H-NMR (400 MHz, DMSO-d6): δ 1.22 (t, J = 7.0, 3H, CH3), 3.95–4.15 (m, 4H, CH2O + CH2N), 4.89–4.94 (m, 1H, CHOH), 5.94 (br s, 1H, exchangeable), 7.21–7.45 (m, 9H Ar), 7.88 (s, 1H, H-3), 8.07 (br s, 1H, exchangeable), 12.03 (br s, 1H, exchangeable). 13C NMR (101 MHz, CDCl3): δ 177.0, 164.6, 161.6, 159.3, 142.4, 140.3, 132.4, 130.1, 128.6, 127.2, 125.1, 116.5, 103.7, 72.7, 60.1, 57.2, 14.4. FTIR (KBr): 3281–2900 (OH + NH), 1711 (COOEt), 1676 (CONH) cm−1. Anal. (C22H21N4O4SF) calcd for C, H, N, S (see Table 7).

3.2.2. Synthesis of Compounds 5a-d

To a mixture of 5-amino-pyrazole 1a [17] (1.37 g, 5 mmol) in anhydrous THF (10 mL), TEA (1 mL) and the suitable benzoyl chloride (6 mmol) were slowly added at 0 °C; then, the mixture was refluxed for 18 h. After cooling to room temperature, the solvent was removed under reduced pressure and the crude was solved in DCM (15 mL). The organic phase was washed with 4M NaOH (150 mL), 3M HCl (15 mL), water (15 mL), dried (MgSO4) and concentrated under reduced pressure. The crude crystallized by adding a solution of diethyl ether/petroleum ether (b.p. 50–60 °C) (1:1).
Ethyl 5-benzamido-1-(2-hydroxy-2-phenylethyl)-1H-pyrazole-4-carboxylate 5a. White solid. Yield 61%; mp 117–118 °C. 1H-NMR (200 MHz, CDCl3): δ 1.25 (t, J = 7.0, 3H, CH3), 4.30 (q, J = 7.0, 2H, CH2O), 4.35–4.90 (2m, 2H, CH2N), 6.29–6.47 (m, 1H, CHOH), 6.70 (br s, 1H, exchangeable), 7.53–8.25 (m, 11H, 10Ar + H-3). 13C NMR (101 MHz, CDCl3): δ 177.0, 164.6, 161.4, 159.2, 142.3, 140.2, 132.2, 130.0, 128.4, 127.2, 125.1, 116.3, 103.4, 72.5, 60.1, 58.0, 14.3. FTIR (KBr): 3461, 3360 (OH + NH), 1681 (COOEt), 1697 (CONH) cm−1. Anal. (C21H21N3O4) calcd for C, H, N (see Table 7).
Ethyl 5-(3-fluorobenzamido)-1-(2-hydroxy-2-phenylethyl)-1H-pyrazole-4-carboxylate 5b. White solid. Yield 35%; mp 149–151 °C. 1H-NMR (200 MHz, CDCl3): δ 1.49 (t, J = 7.0, 3H, CH3), 4.43 (q, J = 7.0, 2H, CH2O), 4.90–5.30 (m, 2H, CH2N), 6.42 (t, 1H, OH exchangeable), 6.58–6.67 (m, 1H, CHOH), 7.35–8.15 (m, 10H, 9Ar + H-3), 9.43 (br s, 1H, NH exchangeable). 13C NMR (101 MHz,CDCl3): δ 166.2, 163.9, 162.0, 142.3, 141.6, 140.2, 135.6, 130.6, 128.4, 128.0, 127.2, 123.5, 119.0, 114.3, 102.4, 72.5, 60.1, 58.0, 14.3. FTIR (KBr): 3200–2974 (OH + NH), 1732 (COOEt), 1710 (CONH) cm−1. Anal. (C21H20N3O4F) calcd for C, H, N (see Table 7).
Ethyl 5-(4-fluorobenzamido)-1-(2-hydroxy-2-phenylethyl)-1H-pyrazole-4-carboxylate 5c. White solid. Yield 30%; mp 98–100 °C. 1H-NMR (400 MHz, DMSO-d6): δ 1.16 (t, J = 7.0, 3H, CH3), 4.10 (q, J = 7.0, 2H, CH2O), 4.21–4.64 (2m, 2H, CH2N), 4.94 (br s, 1H, OH exchangeable), 6.09–6.20 (m, 1H, CHOH), 6.50 (br s, 1H, OH exchangeable), 7.20–7.42 (m, 7H, Ar), 7.51 (s, 1H, H-3), 8.21–8.28 (m, 2H, Ar). 13C NMR (101 MHz, CDCl3): δ 165.9, 163.8, 161.4, 142.3, 141.6, 140.2, 130.2, 130.2, 129.8, 129.8, 128.4, 128.0, 127.2, 116.0, 115.9, 102.4, 72.5, 60.1, 58.0, 14.3. FTIR (KBr): 3408, 3292, 3226 (OH + NH), 1708 (COOEt), 1684 (CONH) cm−1. Anal. (C21H20N3O4F) calcd for C, H, N (see Table 7).
Ethyl 1-(2-hydroxy-2-phenylethyl)-5-(3-(trifluoromethyl)-benzamido)-1H-pyrazole-4-carboxylate 5d. Yellow oil. Yield 23%. 1H-NMR (400 MHz, CDCl3): δ 1.15 (t, J = 7.0, 3H, CH3), 4.07 (q, J = 7.0, 2H, CH2O), 4.25–4.69 (2m, 2H, CH2N), 6.19–6.52 (m, 1H, CHOH), 6.52 (br s, 1H, exchangeable), 7.23–8.30 (m, 10H, 9Ar + H-3). 13C NMR (101 MHz, CDCl3): 166.3, 161.4, 142.3, 141.6, 140.2, 133.7, 131.6, 130.8, 129.0, 129.0, 127.2, 126.9, 124.9, 124.6, 122.7, 120.5, 102.4, 72.5, 60.1, 58.0, 14.3. FTIR (KBr): FTIR (KBr): 3450, 3346 (OH + NH), 1727 (COOEt), 1683 (CONH) cm−1. Anal. (C21H20N3O4F) calcd for C, H, N (see Table 7).

3.2.3. Synthesis of Ethyl 5-((4-fluorophenyl)-sulfonamido)-1-(2-hydroxy-2-phenylethyl)-1H-pyrazole-4-carboxylate 6

To a mixture of 5-amino-pyrazole 1a [17] (1.37 g, 5 mmol) in anhydrous THF (10 mL), TEA (1 mL) and the 4-fluorobenzenesulfonyl chloride (1.17 g, 6 mmol) were slowly added at 0 °C; then, the mixture was refluxed for 3 days. After cooling to room temperature, the solvent was removed under reduced pressure and the crude was solved in DCM (15 mL). The organic phase was washed with 4M NaOH (15 mL), 3 M HCl (15 mL), water (15 mL), dried (MgSO4) and concentrated under reduced pressure. The crude crystallized by adding a solution of diethyl ether/petroleum ether (b.p. 50–60 °C) (1:1). The white solid obtained was recrystallized from absolute ethanol. Yield 34%; mp 180–181 °C. 1H-NMR (400 MHz, DMSO-d6): δ 1.19 (t, J = 7.0, 3H, CH3), 3.94–4.16 (m, 4H, CH2O + CH2N), 4.90–4.93 (m, 1H, CHOH), 6.90 (br s, 1H, exchangeable), 7.07–7.30 (m, 8H, Ar), 7.55 (s, 1H, H-3), 7.58–7.61 (m, 1H, Ar). 13C NMR (101 MHz, CDCl3): δ 161.2, 158.1, 156.2, 143.7, 142.3, 140.0, 134.7, 131.3, 131.2, 128.4, 128.0, 127.1, 126.3, 126.1, 104.7, 72.5, 60.1, 57.8, 14.2. FTIR (KBr): 3399, 3303, 3177 (OH, NH), 1698 (COOEt) cm−1. Anal. (C20H20N3O5SF) calcd for C, H, N, S (see Table 7).

3.2.4. Synthesis of Ethyl 5-(3-(4-fluorobenzoyl)-thioureido)-1-(2-hydroxy-2-phenylethyl)-1H-pyrazole-3-carboxylate 7

A mixture of 5-amino-pyrazole 1b [11] (0.275 g, 1 mmol) and the 4-fluorobenzoyl isothiocyanate (0.181 g, 1 mmol), previously prepared following a method from the literature [18], in anhydrous THF (10 mL) was refluxed for 12 h. After cooling to room temperature, the white solid obtained was filtered and purified by flash chromatography using diethyl ether as the eluent. White solid. Yield 37%; mp 169–172 °C. 1H-NMR (400 MHz, DMSO-d6): δ 1.27 (t, J = 7.0, 3H, CH3), 4.13–4.30 (m, 4H, CH2O + CH2N), 4.86–4.91 (m, 1H, CHOH), 5.92 (br s, 1H, exchangeable), 7.07 (br s, 1H, exchangeable), 7.23–7.57 (m, 8H 7Ar + H-3), 8.03–8.09 (m, 2H Ar), 12.00 (br s, 1H, exchangeable). 13C NMR (101 MHz, CDCl3): δ 175.6, 165.8, 165.8, 163.8, 162.1, 143.3, 142.3, 141.2, 131.5, 131.5, 130.2, 130.1, 128.4, 128.0, 127.2, 116.0, 115.8, 100.6, 72.5, 60.9, 57.7, 14.3. FTIR (KBr): 3450, 3350, 3069 (OH + NH), 1712 (COOEt) cm−1. Anal. (C22H21N4O4SF) calcd for C, H, N, S (see Table 7).

3.3. Biological Evaluation

3.3.1. Microbiology

Bacterial Species Considered in This Study

Since in preliminary experiments none of the compounds reported in this study were active on bacteria of the Gram-negative species, in the following experiments several clinical isolates of Gram-positive species were tested for a total of 21 strains of Enterococcus and Staphylococcus genus. All bacteria belonged to a collection obtained from the School of Medicine and Pharmacy of University of Genoa (Italy). Their identification was carried out as described in our previous works [24,25]. Particularly, 9 strains were of the Enterococcus genus, including 3 vancomycin-resistant (VRE) E. faecalis strains, 3 VRE E. faecium isolates, 1 VRE E. durans, 1 VRE E. gallinarum and 1 VRE E. casseliflavus. The other clinical isolates were staphylococci. Particularly, 3 isolates were methicillin-resistant S. aureus (MRSA), 3 were methicillin-resistant S. epidermidis (MRSE), while 6 were strains belongonging to other species of the genus Stafilococcus, of which 3 resistant to methicillin (1 S. hominis, 1 S. simulans and 1 S. haemolyticus) and 3 susceptible to methicillin (including 1 S. saprophyticus, 1 S. warneri and 1 S. lugdunensis).

Determination of the Minimal Inhibitory Concentrations (MICs)

To assess the antimicrobial activity of compounds 3ac and 4ac their MICs were determined following the microdilution procedures detailed by the European Committee on Antimicrobial Susceptibility Testing EUCAST [26], as also reported in our previous studies [24,25]. Here, serial 2-fold dilutions of solutions of all the six samples (DMSO), ranging from 1 to 256 µg/mL, were used, using DMSO as control to verify the absence of antibacterial activity of the solvent used for the experiments. All MICs were obtained at least in triplicate, and results were expressed reporting the modal value, that is the value that has been observed most frequently. In the case of equivocal or unclear results, more than three determinations of MICs were carried out.

3.3.2. MABA and LORA Tests

MICs against replicating and non-replicating cultures of M. tuberculosis were determined using the Microplate Alamar Blue Assay (MABA) and the Low Oxygen Recovery Assay (LORA) [22].

4. Conclusions

To further extend the SARs of BBB4 and CB1H, 5-PUs and I derivatives, the synthesis, characterization, and microbiological evaluation of a small library of trisubstituted pyrazoles have been here reported. In addition, the compounds demonstrating more promise for future development of new drug delivery systems by nanoencapsulation have been found. Structurally, compounds 27 shared the N1 2-hydroxy-2-phenylethyl chain, previously identified as a relevant structural determinant for biological activity [8,9,10,11,12,13]. The compounds were prepared by a divergent synthetic protocol starting from the key intermediates 1ac and were tested on a panel of twenty-one MDR clinical isolates of the Staphylococcus and Enterococcus genus. Compounds 3c and 4b, characterized by a urea or thiourea function at C5 of the pyrazole nucleus, respectively, were found to be the most active derivatives as antibacterial agents, whereas derivatives 3c and 4a show significant, albeit moderate, activity against M. tuberculosis, with 4a being weakly active against an MDR TB strain. Collectively, the presence of an amide (compounds 5) or sulfonamide (compounds 6) moiety at the C5 position negatively affected the biological properties. In silico study predicted these compounds to exert good drug-like and pharmacokinetic properties.
Overall, the collected data support the pharmaceutical potentials of derivatives 3 and 4 as novel antibacterial agents. In fact, the biological results, particularly against clinical isolates of enterococci and staphylococci, confirm that the N1 2-hydroxy-2-phenylethyl chain induces an improvement in antimicrobial activity with respect to previously reported BBB4 and CB1H, which were completely inactive when not encapsulated in proper nanosized macromolecules. Based on the excellent antibacterial effects obtained encapsulating the inactive BBB4 and CB1H in NPs, pyrazoles 3c and 4b here reported represent even more promising candidates for future development of pyrazole-based nano-delivery systems with further enhanced antibacterial properties in terms of potency and spectrum of activity.
In conclusion, the pyrazole-based library here developed contains at least two compounds, namely 3c and 4b which, upon nano-formulation with proper polymer matrices, could provide new pyrazole-based drug delivery systems with enhanced and broader-spectrum antibacterial activity.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/pharmaceutics14091770/s1. Figure S1. 1H NMR of 4c. Figure S2. 13C NMR of 4c. Figure S3. IR of 4c. Figure S4. 1H NMR of 5c. Figure S5. 13C NMR of 5c. Figure S6. IR of 5c. Figure S7. 1H NMR of 5d. Figure S8. 13C NMR of 5d. Figure S9. IR of 5d. Figure S10. 1H NMR of 6. Figure S11. 13C NMR of 6. Figure S12. IR of 6. Figure S13. 1H NMR of 7. Figure S14. 13C NMR of 7. Figure S15. IR of 7.

Author Contributions

Conceptualization, C.B.; synthesis and characterization C.B. and B.T.; pharmacokinetic properties and druglikeness prediction A.S.; Writing—original draft preparation, C.B., A.S. and S.A.; D.C. and A.M.S., antimicrobial activity, S.G.F. anti-Mycobacterium activity. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank M. Anzaldi and R. Raggio for spectral recording and elemental analysis.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Patel, B.; Zunk, M.; Grant, G.; Rudrawar, S. Design, synthesis and bioactivity evaluation of novel pyrazole linked phenylthiazole derivatives in context of antibacterial activity. Bioorg. Med. Chem. Lett. 2021, 39, 127853. [Google Scholar] [CrossRef] [PubMed]
  2. Taher, A.T.; Sarg, M.T.M.; Ali, N.R.E.; Elnagdi, N.H. Design, synthesis, modeling studies and biological screening of novel pyrazole derivatives as potential analgesic and anti-inflammatory agents. Bioorg. Chem. 2019, 89, 103023. [Google Scholar] [CrossRef] [PubMed]
  3. Yu, B.; Zhou, S.; Cao, L.; Hao, Z.; Yang, D.; Guo, X.; Zhang, N.; Bakulev, V.A.; Fan, Z. Design, Synthesis, and Evaluation of the Antifungal Activity of Novel Pyrazole–Thiazole Carboxamides as Succinate Dehydrogenase Inhibitors. J. Agric. Food Chem. 2020, 68, 7093–7102. [Google Scholar] [CrossRef] [PubMed]
  4. Abdellatif, K.R.A.; El-Saadi, M.T.; Elzayat, S.G.; Amin, N.H. New substituted pyrazole derivatives targeting COXs as potential safe anti-inflammatory agents. Future Med. Chem. 2019, 11, 1871–1887. [Google Scholar] [CrossRef]
  5. Ebenezer, O.; Shapi, M.; Tuszynski, J.A. A Review of the Recent Development in the Synthesis and Biological Evaluations of Pyrazole Derivatives. Biomedicines 2022, 10, 1124. [Google Scholar] [CrossRef]
  6. Alfei, S.; Brullo, C.; Caviglia, D.; Zuccari, G. Preparation and Physicochemical Characterization of Water-Soluble Pyrazole-Based Nanoparticles by Dendrimer Encapsulation of an Insoluble Bioactive Pyrazole Derivative. Nanomaterials 2021, 11, 2662. [Google Scholar] [CrossRef]
  7. Brullo, C.; Rapetti, F.; Bruno, O. Pyrazolyl-Ureas as Interesting Scaffold in Medicinal Chemistry. Molecules 2020, 25, 3457. [Google Scholar] [CrossRef]
  8. Morretta, E.; Sidibè, A.; Spallarossa, A.; Petrella, A.; Meta, E.; Bruno, O.; Monti, M.C.; Brullo, C. Synthesis, functional proteomics and biological evaluation of new 5-pyrazolyl ureas as potential anti-angiogenic compounds. Eur. J. Med. Chem. 2021, 226, 113872. [Google Scholar] [CrossRef]
  9. Belvedere, R.; Morretta, E.; Novizio, N.; Morello, S.; Bruno, O.; Brullo, C.; Petrella, A. The pyrazolyl-urea GeGe3 inhibits the activity of ANXA1 in the angiogenesis induced by the pancreatic cancer derived EV. Biomolecules 2021, 11, 1758. [Google Scholar] [CrossRef]
  10. Signorello, M.G.; Rapetti, F.; Meta, E.; Sidibè, A.; Bruno, O.; Brullo, C. New Series of Pyrazoles and Imidazo-pyrazoles Targeting Different Cancer and Inflammation Pathways. Molecules 2021, 26, 5735. [Google Scholar] [CrossRef]
  11. Bruno, O.; Brullo, C.; Bondavalli, F.; Schenone, S.; Ranise, A.; Arduino, N.; Bertolotto, M.B.; Montecucco, F.; Ottonello, L.; Dallegri, F.; et al. Synthesis and biological evaluation of N-pyrazolyl-N′-alkyl/benzyl/phenylureas: A new class of potent inhibitors of interleukin 8-induced neutrophil chemotaxis. J. Med. Chem. 2007, 50, 3618–3626. [Google Scholar] [CrossRef] [PubMed]
  12. Meta, E.; Brullo, C.; Sidibè, A.; Imhof, B.A.; Bruno, O. Design, synthesis, and biological evaluation of new pyrazolyl-ureas and imidazopyrazolecarboxamides able to interfere with MAPK and PI3K upstream signalling involved in the angiogenesis. Eur. J. Med. Chem. 2017, 133, 24–35. [Google Scholar] [CrossRef] [Green Version]
  13. Meta, E.; Brullo, C.; Tonelli, M.; Franzblau, S.G.; Wang, Y.; Ma, R.; Baojie, W.; Orena, B.S.; Pasca, M.R.; Bruno, O. Pyrazole and imidazo[1,2-b]pyrazole derivatives as new potential anti-tuberculosis agents. Med. Chem. 2019, 15, 17–27. [Google Scholar] [CrossRef] [PubMed]
  14. Alfei, S.; Zuccari, G.; Caviglia, D.; Brullo, C. Synthesis and Characterization of Pyrazole-Enriched Cationic Nanoparticles as New Promising Antibacterial Agent by Mutual Cooperation. Nanomaterials 2022, 12, 1215. [Google Scholar] [CrossRef] [PubMed]
  15. Alfei, S.; Brullo, C.; Caviglia, D.; Piatti, G.; Zorzoli, A.; Marimpietri, D.; Zuccari, G.; Schito, A.M. Pyrazole-Based Water-Soluble Dendrimer Nanoparticles as a Potential New Agent against Staphylococci. Biomedicines 2022, 10, 17. [Google Scholar] [CrossRef]
  16. Schito, A.M.; Caviglia, D.; Brullo, C.; Zorzoli, A.; Marimpietri, D.; Alfei, S. Enhanced Antibacterial Activity of a Cationic Macromolecule by its Complexation with a Weakly Active Pyrazole Derivative. Biomedicines 2022, 10, 1607. [Google Scholar] [CrossRef]
  17. Bondavalli, F.; Botta, M.; Bruno, O.; Ciacci, A.; Corelli, F.; Fossa, P.; Lucacchini, A.; Manetti, F.; Martini, C.; Menozzi, G.; et al. Synthesis, Molecular Modeling Studies, and Pharmacological Activity of Selective A1 Receptor Antagonists. J. Med. Chem. 2002, 45, 4875–4887. [Google Scholar] [CrossRef]
  18. Lewis, R.T.; Bode, C.M.; Choquette, D.M.; Potashman, M.; Romero, K.; Stellwagen, J.C.; Teffera, Y.; Moore, E.; Whittington, D.A.; Chen, H.; et al. The discovery and optimization of a novel class of potent, selective, and orally bioavailable anaplastic lymphoma kinase (ALK) inhibitors with potential utility for the treatment of cancer. J. Med. Chem. 2012, 55, 6523–6540. [Google Scholar] [CrossRef]
  19. 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]
  20. Brenk, R.; Schipani, A.; James, D.; Krasowski, A.; Gilbert, I.H.; Frearson, J.; Wyatt, P.G. Lessons learnt from assembling screening libraries for drug discovery for neglected diseases. ChemMedChem 2008, 3, 435–444. [Google Scholar] [CrossRef]
  21. Delaney, J.S. ESOL: Estimating Aqueous Solubility Directly from Molecular Structure. J. Chem. Inf. Model. 2004, 44, 1000–1005. [Google Scholar] [CrossRef] [PubMed]
  22. Cho, S.; Lee, H.S.; Franzblau, S. Microplate Alamar Blue Assay (MABA) and Low Oxygen Recovery Assay (LORA) for mycobacterium tuberculosis. Methods Mol. Biol. 2015, 1285, 281–292. [Google Scholar] [CrossRef] [PubMed]
  23. Lilly Launches Open Innovation Drug Discovery Platform to Help Find Potential New Medicines Where Medical Need is Great. Available online: https://investor.lilly.com/news-releases/news-release-details/lilly-launches-open-innovation-drug-discovery-platform-help-find (accessed on 23 May 2022).
  24. Schito, A.M.; Piatti, G.; Caviglia, D.; Zuccari, G.; Alfei, S. Broad-Spectrum Bactericidal Activity of a Synthetic Random Copolymer Based on 2-Methoxy-6-(4-Vinylbenzyloxy)-Benzylammonium Hydrochloride. Int. J. Mol. Sci. 2021, 22, 5021. [Google Scholar] [CrossRef] [PubMed]
  25. Schito, A.M.; Piatti, G.; Caviglia, D.; Zuccari, G.; Zorzoli, A.; Marimpietri, D.; Alfei, S. Bactericidal Activity of Non-Cytotoxic Cationic Nanoparticles against Clinically and Environmentally Relevant Pseudomonas spp. Isolates. Pharmaceutics 2021, 13, 1411. [Google Scholar] [CrossRef]
  26. EUCAST. European Committee on Antimicrobial Susceptibility Testing. Available online: https://www.eucast.org/ast_of_bacteria (accessed on 23 May 2022).
Figure 1. Chemical structure of some drugs endowed with the pyrazole structure (Celecoxib, Crizotinib, Surinabant, Fezolamine) and 5-PUs, BBB4, CB1H and I previously reported as anticancer/anti-inflammatory, antibacterial and anti-Mycobacterium compounds, respectively.
Figure 1. Chemical structure of some drugs endowed with the pyrazole structure (Celecoxib, Crizotinib, Surinabant, Fezolamine) and 5-PUs, BBB4, CB1H and I previously reported as anticancer/anti-inflammatory, antibacterial and anti-Mycobacterium compounds, respectively.
Pharmaceutics 14 01770 g001
Figure 2. Workflow of the applied strategies leading to the design, biological evaluation and future development of novel pyrazole compounds 27.
Figure 2. Workflow of the applied strategies leading to the design, biological evaluation and future development of novel pyrazole compounds 27.
Pharmaceutics 14 01770 g002
Scheme 1. Synthesis of compounds 2–7. Reagents and conditions: (a) suitable benzoyl isothiocyanates [18], anh. THF, reflux, 12 h; (b) suitable phenyl isocyanates, anh. toluene, reflux, 6 h; (c) suitable benzoyl chlorides, TEA, anh. THF, reflux, 18 h; (d) 4-fluorobenzenesulfonyl chloride, TEA, anh. THF, reflux, 3 days.
Scheme 1. Synthesis of compounds 2–7. Reagents and conditions: (a) suitable benzoyl isothiocyanates [18], anh. THF, reflux, 12 h; (b) suitable phenyl isocyanates, anh. toluene, reflux, 6 h; (c) suitable benzoyl chlorides, TEA, anh. THF, reflux, 18 h; (d) 4-fluorobenzenesulfonyl chloride, TEA, anh. THF, reflux, 3 days.
Pharmaceutics 14 01770 sch001
Table 1. Chemical structure, melting point and reaction yield of new pyrazole derivatives 27.
Table 1. Chemical structure, melting point and reaction yield of new pyrazole derivatives 27.
Pharmaceutics 14 01770 i001
Compd.RR’R’’M.p (°C)Yield (%)
2aCOOEtH Pharmaceutics 14 01770 i002[11][11]
2bCOOEtH Pharmaceutics 14 01770 i003[11][11]
2cCOOEtH Pharmaceutics 14 01770 i004[11][11]
2dCOOEtH Pharmaceutics 14 01770 i005[11][11]
3aHC(CH3)3 Pharmaceutics 14 01770 i006[12][12]
3bHC(CH3)3 Pharmaceutics 14 01770 i007[12][12]
3cHC(CH3)3 Pharmaceutics 14 01770 i008[12][12]
3dHC(CH3)3 Pharmaceutics 14 01770 i009[12][12]
4aCOOEtH Pharmaceutics 14 01770 i010150–15142
4bCOOEtH Pharmaceutics 14 01770 i011155–15648
4cCOOEtH Pharmaceutics 14 01770 i012158–16051
5aCOOEtH Pharmaceutics 14 01770 i013117–11861
5bCOOEtH Pharmaceutics 14 01770 i014149–15135
5cCOOEtH Pharmaceutics 14 01770 i01598–10030
5dCOOEtH Pharmaceutics 14 01770 i016Oil23
6COOEtH Pharmaceutics 14 01770 i017180–18134
7HCOOEt Pharmaceutics 14 01770 i018169–17237
Table 2. Predicted pharmacokinetics and drug-like properties of compounds 2ad and 3ad.
Table 2. Predicted pharmacokinetics and drug-like properties of compounds 2ad and 3ad.
2a2b2c2d3a3b3c3d
Physicochemical Prop.
MW (g/mol)412.41412.41412.41462.42396.46396.46396.46446.47
Fraction Csp30.190.190.190.230.270.270.270.3
Rotatable bonds101010118889
H-bond acceptors66684446
H-bond donors33333333
TPSA a2)105.48105.48105.48105.4879.1879.1879.1879.18
Lipophilicity
LogP b2.782.782.783.563.723.723.724.5
Water solubility
Solubility (mg/mL) c0.0510.0510.0510.01150.01120.01120.01120.00256
Solubility classSolubleSolubleSolubleModerately solubleModerately solubleModerately solubleModerately solubleModerately soluble
Pharmacokinetics
GI absorptionHighHighHighHighHighHighHighHigh
BBB permeantNoNoNoNoNoNoNoNo
Pgp substrateYesYesYesNoNoNoYesNo
CYP1A2 inhibitorNoNoNoNoNoNoNoNo
CYP2C19 inhibitorNoYesYesYesYesYesYesYes
CYP2C9 inhibitorYesYesYesYesYesYesYesNo
CYP2D6 inhibitorNoNoNoNoNoNoNoNo
CYP3A4 inhibitorNoNoNoYesYesYesYesYes
Druglikeness
Lipinski violations00000000
Medicinal chemistry
PAINS alerts00000000
Brenk alerts00000000
a Topological Polar Surface Area. b Predicted according to XLOGP3 program. c Values predicted by ESOL method [21].
Table 3. Predicted pharmacokinetics and drug-like properties of compounds 47.
Table 3. Predicted pharmacokinetics and drug-like properties of compounds 47.
4a4b4c5a5b5c5d67
Physicochemical Properties
MW (g/mol)456.49456.49456.49379.41397.40397.40447.41433.45456.49
Fraction Csp30.180.180.180.190.190.190.230.200.18
Rotatable bonds11111199910911
H-bond acceptors666566876
H-bond donors333222223
TPSA a2)137.57137.57137.5793.4593.4593.4593.45118.9137.57
Lipophilicity
LogP b3.263.263.262.953.053.053.832.743.59
Water solubility
Solubility (mg/mL) c0.01850.01850.01850.04710.03420.03420.007780.03610.0115
Solubility classModerately solubleModerately solubleModerately solubleSolubleModerately solubleModerately solubleModerately solubleModerately solubleModerately soluble
Pharmacokinetics
GI absorptionLowLowLowHighHighHighHighHighLow
BBB permeantNoNoNoNoNoNoNoNoNo
Pgp substrateNoNoNoNoNoNoNoNoNo
CYP1A2 inhibitorNoNoNoNoYesYesNoNoNo
CYP2C19 inhibitorYesYesYesYesYesYesYesYesYes
CYP2C9 inhibitorYesYesYesYesYesYesYesYesYes
CYP2D6 inhibitorYesNoYesYesYesYesYesNoNo
CYP3A4 inhibitorYesYesYesNoNoNoYesYesYes
Druglikeness
Lipinski violations000000000
Medicinal chemistry
PAINS alerts000000000
Brenk alerts111000001
a Topological Polar Surface Area. b Predicted according to XLOGP3 program. c Values predicted by ESOL method [21].
Table 4. MIC values of pyrazoles 3ac and 4ac against bacteria of the Gram-positive species, obtained from experiments carried out at least in triplicate, expressed as µg/mL.
Table 4. MIC values of pyrazoles 3ac and 4ac against bacteria of the Gram-positive species, obtained from experiments carried out at least in triplicate, expressed as µg/mL.
StrainsMIC (µg/mL)
3a3b3c4a4b4c
Enterococcus genus
E. faecalis 365 *>128>128128>128>128>128
E. faecalis 450 *12812864>128>128>128
E. faecalis 451 *>12812864>128>128>128
E. faecium 182 *1286464>128>128>128
E. faecium 300 *>128>12864>128>128>128
E. faecium 364 *>128>12864>128>128>128
E. durans 10364>12864>128>128>128
E. gallinarum 150 *32>128128>128>128>128
E. casseliflavus 1846464128128>128>128
Staphylococcus genus
S. aureus 18 **>128>12864>12864128
S. aureus 187 **>128128321286464
S. aureus 195 **12812832>128128128
S. epidermidis 180 ***128643212864128
S. epidermidis 181 ***>1281286412864128
S. epidermidis 363 **12812832128128128
S. saprophyticus 41>128>12864>128>128>128
S. warneri 746412864128>128>128
S. hominis 125 #323264128128128
S. lugdunensis 129323232643232
S. simulans 163 #>128>128128>128>128>128
S. haemolyticus 193 #>128>128128>128>128>128
* Denotes vancomycin resistant (VRE), ** denotes methicillin resistant; *** denotes resistance toward methicillin and linezolid; # denotes methicillin resistant; bold data evidence promising antibacterial activity.
Table 5. MIC values of compounds 3c and 4b against the most clinically relevant bacteria of the Gram-positive species, obtained from experiments carried out at least in triplicate, and those of reference antibiotics expressed as µg/mL.
Table 5. MIC values of compounds 3c and 4b against the most clinically relevant bacteria of the Gram-positive species, obtained from experiments carried out at least in triplicate, and those of reference antibiotics expressed as µg/mL.
MIC (µg/mL)
Strains3c4bAmpicillinCiprofloxacinOxacillin
E. faecalis 365 *128>128128--
E. faecalis 450 *64>128128--
E. faecalis 451 *64>128128--
E. faecium 182 *64>128128--
E. faecium 300 *64>128128--
E. faecium 364 *64>128128--
S. aureus 18 **6464-128512
S. aureus 187 **3264-128512
S. aureus 195 **32128-128512
S. epidermidis 180 ***3264-64256
S. epidermidis 181 ***6464-64256
S. epidermidis 363 **3264-64256
* Denotes vancomycin resistant (VRE); ** denotes methicillin resistant; *** denotes resistance toward methicillin and linezolid; bold data evidence promising antibacterial activity.
Table 6. Percentage of inhibition at 50 μg/mL concentration and MIC values (µg/mL) in the MABA test for compounds 3ad and 4ac. These values are the result of the average of two independent experiments; bold data evidence promising antibacterial activity.
Table 6. Percentage of inhibition at 50 μg/mL concentration and MIC values (µg/mL) in the MABA test for compounds 3ad and 4ac. These values are the result of the average of two independent experiments; bold data evidence promising antibacterial activity.
Entry% inhib. at 50 µg/mLMIC (µg/mL)
3a30>50
3b55>50
3c9935.8
3d59>50
4a10039.3
4b75>50
4c88>50
Table 7. Elemental analysis of compounds 4-7.
Table 7. Elemental analysis of compounds 4-7.
Compd.Molecular FormulaElemental Analysis%S
Values%C%H%N
4aC22H21N4O4SFCalcd.57.884.6412.277.02
Found57.774.5012.296.76
4bC22H21N4O4SFCalcd.57.884.6412.277.02
Found57.874.5012.366.83
4cC22H21N4O4SFCalcd.57.884.6412.277.02
Found57.574.9112.407.24
5aC21H21N3O4Calcd.66.485.5811.08//
Found66.165.8011.09//
5bC21H20N3O4FCalcd.63.475.0710.57//
Found63.354.8710.46//
5cC21H20N3O4FCalcd.63.475.0710.57//
Found63.475.3210.57//
5dC22H20N3O4F3Calcd.59.064.519.39//
Found59.004.609.20//
6C20H20N3O5SFCalcd.55.424.659.697.40
Found55.884.779.417.14
7C22H21N4O4SFCalcd.57.884.6412.277.02
Found57.924.3612.277.15
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Brullo, C.; Caviglia, D.; Spallarossa, A.; Alfei, S.; Franzblau, S.G.; Tasso, B.; Schito, A.M. Microbiological Screening of 5-Functionalized Pyrazoles for the Future Development of Optimized Pyrazole-Based Delivery Systems. Pharmaceutics 2022, 14, 1770. https://doi.org/10.3390/pharmaceutics14091770

AMA Style

Brullo C, Caviglia D, Spallarossa A, Alfei S, Franzblau SG, Tasso B, Schito AM. Microbiological Screening of 5-Functionalized Pyrazoles for the Future Development of Optimized Pyrazole-Based Delivery Systems. Pharmaceutics. 2022; 14(9):1770. https://doi.org/10.3390/pharmaceutics14091770

Chicago/Turabian Style

Brullo, Chiara, Debora Caviglia, Andrea Spallarossa, Silvana Alfei, Scott G. Franzblau, Bruno Tasso, and Anna Maria Schito. 2022. "Microbiological Screening of 5-Functionalized Pyrazoles for the Future Development of Optimized Pyrazole-Based Delivery Systems" Pharmaceutics 14, no. 9: 1770. https://doi.org/10.3390/pharmaceutics14091770

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