Next Article in Journal
The Anti-Virulence Effect of Vismia guianensis against Candida albicans and Candida glabrata
Previous Article in Journal
Staphylococcus aureus-Cure-Associated Antigens Elicit Type 3 Immune Memory T Cells
Previous Article in Special Issue
Next-Generation Polymyxin Class of Antibiotics: A Ray of Hope Illuminating a Dark Road
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Optimization of Pyrazole Compounds as Antibiotic Adjuvants Active against Colistin- and Carbapenem-Resistant Acinetobacter baumannii

1
Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, Viale Bracci 16, 53100 Siena, Italy
2
Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
3
Lead Discovery Siena S.r.l., Via Aldo Moro 2, 53100 Siena, Italy
4
Centre d’Ingénierie des Protéines—InBioS, Université de Liège, Allée du six Août 11, 4000 Liège, Belgium
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Antibiotics 2022, 11(12), 1832; https://doi.org/10.3390/antibiotics11121832
Submission received: 4 November 2022 / Revised: 1 December 2022 / Accepted: 12 December 2022 / Published: 16 December 2022
(This article belongs to the Special Issue Latest Progress in the Polymyxin Class of Antibiotics)

Abstract

:
The diffusion of antibiotic-resistant, Gram-negative, opportunistic pathogens, an increasingly important global public health issue, causes a significant socioeconomic burden. Acinetobacter baumannii isolates, despite causing a lower number of infections than Enterobacterales, often show multidrug-resistant phenotypes. Carbapenem resistance is also rather common, prompting the WHO to include carbapenem-resistant A. baumannii as a “critical priority” for the discovery and development of new antibacterial agents. In a previous work, we identified several series of compounds showing either direct-acting or synergistic activity against relevant Gram-negative species, including A. baumannii. Among these, two pyrazole compounds, despite being devoid of any direct-acting activity, showed remarkable synergistic activity in the presence of a subinhibitory concentration of colistin on K. pneumoniae and A. baumannii and served as a starting point for the synthesis of new analogues. In this work, a new series of 47 pyrazole compounds was synthesized. Some compounds showed significant direct-acting antibacterial activity on Gram-positive organisms. Furthermore, an evaluation of their activity as potential antibiotic adjuvants allowed for the identification of two highly active compounds on MDR Acinetobacter baumannii, including colistin-resistant isolates. This work confirms the interest in pyrazole amides as a starting point for the optimization of synergistic antibacterial compounds active on antibiotic-resistant, Gram-negative pathogens.

Graphical Abstract

1. Introduction

Antibacterial agents of both natural and synthetic origins represent invaluable therapeutic resources that not only have allowed for the effective treatment of once deadly bacterial infections, but also have paved the way for (and still is a pillar of) modern medicine (including organ transplantations, invasive surgery, etc.). However, the effectiveness of antibacterial drugs is undermined by the emergence and spread of resistant strains, which have developed resistance mechanisms to evade the activity of virtually all classes of antibacterial drugs [1,2]. Several other factors (the low financial attractiveness of antibiotics and costs of discovery and development, the evolution of clinically relevant species with multidrug resistance, and more stringent requirements for the approval of new drugs) have led to a significant shortage of new antibacterial drugs on the market in the last two decades and, consequently, to the increase in resistance to antibiotics, especially carbapenems, which are increasingly used to treat infections caused by multidrug-resistant (MDR) bacteria [3]. Inevitably, the current crisis due to antibiotic resistance is characterized by a significant increase in the prevalence of carbapenem-resistant, Gram-negative, opportunistic pathogens (Enterobacterales, Pseudomonas spp., and Acinetobacter spp.), which have caused a large number of deaths and comorbidities all around the world and have been identified by the WHO as critical priorities for the development of new antibiotics [4]. Furthermore, the recent increase in the prevalence of colistin resistance (due to a variety of mechanisms depending on the host) has allowed relevant Gram-negative bacteria to evolve towards ultra-resistant phenotypes, and consequently, has further narrowed our therapeutic options.
Acinetobacter baumannii (Aba) is an emblematic example of this as it has rapidly evolved into having ultra-resistant phenotypes, especially in relation to the significant increase, over the last decade, in the use of last-resort drugs. Commonly resistant to colistin [5] and carbapenems (due to the production of a variety of carbapenemases), Aba isolates are also refractory to the more recent combinations of β-lactams with diazabicyclooctanone-based (DBO; ceftazidime/avibactam and imipenem/cilastatin/relebactam) or boronate-based (meropenem/vaborbactam) β-lactamase inhibitors [6,7]. Furthermore, only a few compounds show a spectrum of activity that includes this organism (eravacycline, approved in September 2018; and, to some extent, cefiderocol, approved in April 2020), and therapeutic solutions targeting extensively drug-resistant (XDR) Aba that is resistant to carbapenems and colistin (sulbactam/durlobactam and BV100, phase 1; meropenem/ANT3310, preclinical phase) are in the early stages of development [8]. There is still an urgent need to explore alternative strategies for treating infections caused by ultra-resistant Aba isolates.
One of the fastest ways to obtain valuable antibacterial drugs active against resistant bacteria and susceptible to clinical development is based on: (1) combinations of compounds that exhibit “antibiotic enhancer” [9] or “resistance breaker” [10] activity with available antibiotics and (2) the use of new technological platforms for the formulation of last-resort antibiotics (e.g., conjugated with antibodies or nanoparticles [11]). Other innovative approaches include the design of antivirulence compounds, e.g., by targeting important bacterial regulators of biofilm formation or virulence, or by directly inhibiting virulence factors.
Following an extensive screening campaign for the identification of synthetic compounds with direct or synergistic antibacterial activity, we recently identified a number of compounds showing synergistic activity with colistin on various Gram-negative pathogens, including K. pneumoniae, P. aeruginosa, and Aba [12]. Most of the compounds identified belong to two structural classes (substituted phenols and pyrazoles) that could serve as a basis for the optimization of colistin resistance breakers and could be used in combination for the treatment of multidrug-resistant Aba infections.
In view of the potentially problematic nature of phenol derivatives from the pharmacokinetic point of view, we have given priority to the pyrazole scaffold and selected compounds 1 and 2 (Figure 1) as a starting point for optimization, with the aim of identifying more active compounds as colistin resistance breakers acting on MDR Aba isolates.

2. Results

2.1. Chemistry

Considering the paucity of information available regarding the structural determinants for synergistic antibacterial activity with the colistin of hit compounds 1 and 2, we kept the pyrazole ring as the critical structural element and undertook a wide exploration of the chemical space around this ring to obtain, in the end, a library of 47 pyrazole derivatives. Based on the hit compound 1, a first group of 20 pyrazole 4-carboxamide derivatives (Figure 2) was synthesized by varying substituents R1, R2, and R3, whereas a wider investigation of chemical substitution at every position of the pyrazole ring, starting from the hit compound 2, led to a second family of derivatives characterized by the presence of a carboxamide substituent at position 3 (Figure 3). No particular attention was paid to the design of compounds with optimal physicochemical properties, favoring, in the first instance, the exploration of chemical space, and postponing the structural optimization of the most promising compounds to a later stage. Compounds 322, featuring a carboxamide group at position 4, were prepared as outlined in Scheme 1, Scheme 2 and Scheme 3, whereas the preparation of compounds 2349, featuring a carboxamide substituent at position 3 of the pyrazole nucleus, as shown by the hit compound 2, is described in Scheme 4 and Scheme 5. The reaction between the appropriate hydrazines and diethyl ethoxymethylenemalonate (Scheme 1) yielded the 5-aminopyrazole derivatives 5053, of which 5052 were converted into the pyrrol-1-yl derivatives 5456 by using the Clauson-Kaas procedure [13]. Compound 56 was subjected to a Suzuki–Miyaura reaction with p-tolylboronic acid to produce compound 57. Esters 54, 55, and 57 were, in turn, hydrolyzed [14] to the corresponding acids 5860, which were subjected to an amidation reaction, which was conducted by following previously published procedures [15], to give the final amides 314.
Alkyl esters 1113 (Scheme 2) were further elaborated by using hydrolysis on the corresponding acids 1517, and acid 15 was reacted with L-alanine ethyl ester to yield 18, which, in turn, was converted into acid 19. Ethyl 5-amino-1H-pyrazole-4-carboxylates 51 and 53 (Scheme 3) were acylated with di-tert-butyl dicarbonate to give the N-Boc derivatives 61 and 62, which were, subsequently, hydrolyzed to carboxylic acids 63 and 64, respectively. Amidation of these carboxylic acids using commercially available 4-bromobenzylamine or (5-(4-(trifluoromethoxy)phenyl)pyridine-2-yl)methanamine [16] led to the final compounds 2022.
Compounds 2339 (Scheme 4) were synthesized through a condensation reaction with the appropriate amines under usual conditions starting from the carboxylic acids 6572, which were prepared as previously reported [15]. Ester 32 was then hydrolyzed to obtain the corresponding acid 40.
To investigate whether the particular antibacterial properties of compound 40 (vide infra) might depend on the acidity of its carboxyl group or on the position of the same on the benzene ring, a small family of compounds was synthesized (Scheme 5) in which the carboxyl group was shifted to the meta- or ortho-position (compounds 42 and 44), or replaced by other functional groups with different pKa values, namely, a phenolic hydroxyl (45) or a sulfonamide (46), acylsulfonamide (47), or sulfoximine group (49). Acid 68 was amidated with methyl 3-aminobenzoate using EDC/HOBt as coupling reagents to give the intermediate 41, which was then hydrolyzed (NaOH/MeOH) to obtain the free carboxylic acid 42. Similarly, 68 was also converted to 43 using 2-aminobenzoate methyl ester, which was then hydrolyzed to 44, whereas for the preparation of 45, 4-aminophenol PyBOP/HOBt in dry DMF was used. The activation of 68 to the corresponding acyl chloride followed by a reaction with sulfanilamide led to 46, which was, subsequently, transformed into the acylsulfonamide 47 via a treatment with methyl chloroformate. The reaction between acid 68 and 4-methylthioaniline, as described for the synthesis of 41 and 43, gave the amide 48 which, when treated with (diacetoxyiodo)benzene in the presence of ammonium hydroxide as a nitrogen source, gave the sulfoximine derivative 49.

2.2. Biological Evaluation of Pyrazole-4- and Pyrazole-3-Carboxamide Derivatives

Compounds 340, 42, 4447, and 49 (Scheme 1, Scheme 2, Scheme 3, Scheme 4 and Scheme 5) were subjected to a detailed investigation of their direct and synergistic antibacterial activity and potential cytotoxicity by using several methods. The direct antibacterial activity of these compounds was determined using both agar diffusion and broth microdilution methods. The former was initially used to identify potentially active compounds and was tested on eight reference strains, with representatives of both Gram-positive and Gram-negative clinically relevant bacterial species or genera (Figure 4).
As somewhat expected, considering that the parent compounds were selected for their synergistic rather than direct-acting antibacterial activity [12], limited growth inhibition was observed for these compounds as the best compounds showed a growth inhibition zone with a diameter of a max. of 7 mm. Interestingly, some compounds, specifically 15, 16, 17, 19, and 42, showed a broad-spectrum, though moderate, direct-acting antibacterial activity. All these compounds, which are analogues of parent compound 1, but where no direct antibacterial activity was observed [12], are characterized by the presence of free carboxylic acid on the carboxamide substituent at position 4 of the pyrazole heterocycle. However, none of these compounds exhibited MIC values lower than 512 µg/mL when tested using a broth microdilution method.
Notable exceptions were represented by 40, 42, 44, and 47, which showed a remarkable antibacterial activity on Gram-positive organisms (although slightly lower on Enterococcus faecalis), with growth inhibition zones having diameters of up to 18 mm. This activity was confirmed in broth microdilution assays and translated in MIC values ranging from 32 µg/mL for Bacillus subtilis, Enterococcus faecalis, and Staphylococcus aureus, to 16 µg/mL for Streptococcus pyogenes. From a structural standpoint, these four appear to be related to parent compound 2, as they feature a 3-carboxamide (rather than a 4-carboxamide) substituent. Interestingly, the 3-carboxamide substituent also contains a free carboxylic group in three of these compounds, although the nature of the linker varies from that of the analogs of 1, being made up of a phenyl rather than an aliphatic chain. Overall, these data suggest that the introduction of a free carboxyl group into parent compounds 1 and 2 leads to an improvement, albeit modest overall, in their direct-acting antibacterial activity against Gram-positive organisms, which is maintained when the acid group is moved to other positions on the benzene ring or replaced by a functional group, such an acylsulfonamide, with a similar pKa value. Although potentially interesting, none of these compounds exhibited sufficient activity on Gram-negative bacteria, which nowadays, represent a far more relevant target for the discovery of novel antibacterial scaffolds, and were not further studied in this work.
Second, we established a rapid screening method to identify compounds showing synergistic activity with antibiotics on representative Gram-negative species. This method was based on the detection of growth inhibition in liquid medium (Mueller–Hinton II culture medium) in the presence of a single concentration of the compound (64 µg/mL) and subinhibitory concentrations (0.5 × MIC) of colistin (see the Materials and Methods section for details), as this antibiotic appears to be the best potentiator of parent compounds [12]. From this analysis, 11 compounds (8, 9, 13, 20, 22, 27, 28, 31, 39, 45, and 49) were identified that showed synergistic activity on at least one organism (out of the four representative Gram-negative bacteria) (Table 1).
Interestingly, these compounds were structurally different from those showing direct-acting activity, and were analogues of both parent compounds 1 and 2. Compound 39 showed synergistic activity with colistin on Aba only. A broader spectrum of activity was observed with all other compounds, most notably, 9, 28, 31, 45, and 49, which inhibited the growth of all tested bacteria except P. aeruginosa. None of the compounds actually proved active on that organism. This synergistic activity was further investigated by determining the MIC values of these compounds in the presence of a subinhibitory concentration of colistin and provided additional data regarding their antibacterial spectrum and potency (Table 2).
Compounds 9, 45, and 49 emerged as the most promising pyrazolo-carboxamide derivatives, with MAC values ranging from 4 to 8 µg/mL on both Aba and Enterobacterales, interestingly showing that both scaffolds (pyrazole-4- and pyrazole-3-carboxamides) allowed for the identification of potent compounds showing synergistic activity with colistin. A similarly potent synergistic activity was also observed with compound 8 on Aba. Compounds 8 and 9 are analogues of parent compound 1, with modifications of the nature of the 4-carboxamide substituent (e.g., adamantanyl). Other modifications of compound 1, including the introduction of butanoate ester on the 4-carboxamide substituent (13) or the substitution of the 5-pyrrole ring with a tert-butoxycarbonylamino moiety (20 and 22), did not translate into a similar increase in antibacterial synergistic activity. Regarding pyrazole-3-carboxamide derivatives, some new analogues of compound 2 (27, 28, 31, and 39) did not prove to be significantly better than the parent compound. However, compounds 45 and 49 did provide some form of significant improvement in their synergistic activity, when compared to that of parent compound 2. Compounds 8, 9, 45, and 49 showed the best overall activity, including on Aba, which we consider a more relevant bacterial target in our drug discovery programs than Enterobacterales, in relation to the paucity of antibiotics currently in clinical development targeting this specific pathogen (see Introduction), and were selected for further investigation. A chequerboard analysis, to further assess whether they would exhibit true synergistic activity or rather a simple additive effect with colistin, was carried out (Table 3, Figure 5). All new tested compounds (8, 9, 45, and 49) were confirmed to be synergistic with colistin on antibiotic-susceptible reference strains of E. coli, K. pneumoniae, and Aba. Compound 8 was slightly inferior to 9, as expected from the data reported in Table 2. A different picture emerged when these compounds were tested on clinical isolates showing a multidrug- or pandrug-resistant phenotype, including to polymyxin antibiotics. On one hand, synergistic activity could not be observed with the pan-resistant K. pneumoniae isolate, whose resistance to colistin was acquired through the insertional inactivation of mgrB (a negative regulator of PhoP/PhoQ) [17]. On the other hand, the potentiation effect was stronger on a colistin-resistant Aba strain when compared to that of the antibiotic-susceptible reference strains, as the resulting average FIC index values were lower when measured on the colistin-resistant Aba strain (Figure 5).
These data confirmed that the new analogues 8 and 9 were promising potentiators of polymyxin antibiotics, especially on colistin-resistant Aba. Encouraged by these results, we wanted to understand whether any of these two active compounds would show sufficient selectivity, and thus, validate the scaffold as a potentially useful new series of antibiotic adjuvants. The potential cytotoxic activity of both compounds was assessed using several methods. First, a simple membrane integrity assay carried out on HeLa cells showed that none of the compounds induced an LDH release at concentrations of up to 256 µg/mL after 24 h of exposure (Table 4). This is in agreement with the fact that no hemolytic activity could be observed, not only for compounds 8 and 9, but also for compounds 1, 2, 45, and 49. Considering that these compounds would show potent antibacterial activity in the presence of colistin (a membrane-interacting antibiotic known for its suboptimal safety profile [19,20]), we also wanted to investigate whether the cytotoxicity of our compounds would be affected by the presence of this antibiotic. Colistin, when tested alone in a similar assay, showed an IC50 value of 1240 µg/mL, confirming its membrane-damaging activity in eukaryotic cells at higher concentrations. Based on this result, the cytotoxicity of our compounds was measured in the presence of a subtoxic concentration of colistin (512 µg/mL). In this case, compounds 8 and 9 showed a significantly different behavior, as compound 8 appeared to enhance the membrane-damaging activity of colistin at rather low concentrations (IC50, 16.4 µg/mL). Interestingly, this was not observed with compound 9, which was devoid of any membrane-damaging activity both in the absence and in the presence of 512 µg/mL of colistin. Subsequently, the cytotoxicity of compound 9 was further assessed using cell proliferation/viability assays (see the Materials and \methods section for details). In these assays, the proliferation of HeLa cells was not significantly altered in the presence of 16 µg/mL of compound for up to 72 h. Moreover, similar results were obtained when this experiment was performed in the presence of 512 µg/mL of colistin in the medium.
Finally, and considering the promising selectivity showed by compound 9, its antibacterial activity with colistin on the colistin-resistant Aba N50 strain was further characterized using time-kill curves (Figure 6). The presence of colistin alone (tested at 2 µg/mL, i.e., the susceptibility breakpoint), although showing an initial moderate impact on bacterial survival, was not expectedly able to show any bactericidal effect (defined as the capacity of a compound to decrease the initial bacterial population by at least 3 log10 [21]). However, a combination of 2 µg/mL of colistin and 8 µg/mL of 9 did exhibit a significantly faster and better bacterial killing, with a 4 log10 reduction achieved within 5 h. Unfortunately, complete eradication could not achieved, leading to the recovery of the bacterial growth. This was confirmed by the determination of the MBC, which yielded values >64 µg/mL for compounds 8, 9, 45, and 49 when tested in the presence of 2 µg/mL of colistin.

3. Discussion and Conclusions

In this study, we investigated the direct-acting and synergistic antibacterial activity of 44 new analogues of pyrazole-3-carboxamides and pyrazole-4-carboxamides identified in a previous work [12]. A single compound showed significant direct-acting activity primarily on Gram-positive organisms, a result that was somewhat expected considering that the parent compounds were previously identified and characterized for their synergistic activity with colistin. Interestingly, nine new analogues of both parent compounds 1 and 2 maintained a synergistic activity on Gram-negative bacteria, with the exception of P. aeruginosa. Two compounds, 8 and 9, were significantly more active than their parent compound 1, and were characterized by a different substituent on the 4-carboxamide moiety. Apparently, this structural difference largely affects the properties of compound 8, which not only exhibited a narrower spectrum of synergistic activity, but also a more apparent enhancement of the membrane-damaging activity of colistin on eukaryotic cells. More strikingly, compound 9 showed synergistic activity on both colistin-susceptible Enterobacterales and Aba, but also proved, in combination with a subinhibitory concentration of colistin, to significantly enhance the rate of killing of a colistin-resistant Aba clinical isolate at concentrations as low as 8 µg/mL while showing a promising selectivity. Furthermore, compounds 45 and 49, which are pyrazole-3-carboxamide derivatives, also proved to have a better synergistic activity than their parent 2, although the improvement on Aba was less remarkable. These data, overall, further highlight the potential of pyrazole-4- and pyrazolo-3-carboxamide analogues to obtain selective and synergistic compounds to be further optimized, and hopefully represent a promising source of new therapeutic solutions that are able to restore the activity of the last-resort polymyxins for the treatment of infections caused by extensively drug- or pandrug-resistant clinical isolates of Acinetobacter baumannii. Furthermore, the synergistic activity exhibited by these compounds in the presence of colistin may rely on the permeabilizing effect of the latter, and provides hope that the further optimization of these compounds, particularly regarding their rate of diffusion through bacterial membranes, may allow for the identification of potent and direct-acting compounds.

4. Materials and Methods

4.1. Chemistry

Reagents were purchased from commercial suppliers and used without further purification. Anhydrous reactions were run under a positive pressure of dry N2. Merck silica gel 60 was used for flash chromatography (23–400 mesh). 1H NMR and 13C NMR were recorded at 400 and 100 MHz, respectively, on a Bruker Advance DPX400. Chemical shifts are reported relative to tetramethylsilane at 0.00 ppm. Mass spectral (MS) data were obtained using the Agilent 1100 LC/MSD VL system (G1946C) with a 0.4 mL/min flow rate and using a binary solvent system of 95:5 methanol/water. UV detection was monitored at 254 nm. Mass spectra were acquired either in positive or in negative mode by scanning over the mass range of 105–1500. Melting points were determined on a Gallenkamp apparatus and are uncorrected. Elemental analyses were performed on a PerkinElmer PE 2004 elemental analyzer and the data for C, H, and N are within 0.4% of the theoretical values. The chemical purity of most of the target compounds was determined using an ACQUITY Waters UPLC-MS system with a Waters BEH C18 reversed-phase column (2.1 mm × 50 mm, 1.7 µm); the method was carried out under the following conditions: gradient elution, solvent A (0.1% formic acid in water), solvent B (0.1% formic acid in acetonitrile) 90:10 to 0:100 over 2.9 min, flow rate of 0.5 mL/min, UV detector, and 254 nm. The chemical purity of compounds 10, 14, and 19 was determined using an Agilent 1260 Infinity instrument constituted of a binary pump, an autosampler, an UV-DAD, and an ESI-MS detector. The chromatographic separation was realized with a Symmetry® C18 column (4.6 × 75 mm, 3.5 µm). Analysis was carried out with methanol as the mobile phase at a flow rate of 0.5 mL/min. UV detection was monitored at 254 nm. The purity of each compound was ≥ 95% in either analysis, with the exception of compounds 3, 4, 6, 7, and 20, whose purity was in the range of 90–94%; accordingly, elemental analyses were not performed on these compounds.

4.1.1. Synthesis of Aminopyrazole Precursors 5053

Well-established procedures from the literature were applied for the synthesis of 50 [22], 51 [15], 52 [23], and 53 [24].

4.1.2. General Procedure for the Synthesis of Ethyl 5-Pyrrol-1-yl-1H-pyrazole-4-carboxylates 5456

Compounds 54 and 55 were prepared according to known procedures [15,25]. As an example, the preparation of the new compound 56 is reported below.

Ethyl 5-Amino-1-(6-bromopyridin-2-yl)-1H-pyrazole-4-carboxylate (56)

A solution of AcONa (1.6 g, 11.7 mmol) in water (2.5 mL) was added to a solution of ethyl (ethoxymethylene)cyanoacetate (988 mg, 5.85 mmol) in glacial AcOH (15 mL). After stirring for a few minutes, 6-bromo-2-pyridinylhydrazine (1.0 g, 5.32 mmol) was added and the mixture was stirred at 100 °C overnight. After cooling, the solution was poured into ice water and the precipitate which formed was suction-filtered and washed thoroughly with PE. Flash chromatographic purification on a silica gel with DCM/PE (3:1) as the eluent produced the title compound 56 (76% yield) as a white solid. Mp 163–166 °C. 1H NMR (400 MHz, CDCl3): δ 7.85 (d, J = 8.2 Hz, 1H), 7.71 (s, 1H), 7.61 (t, J = 8.0 Hz, 1H), 7.27 (d, J = 7.7 Hz, 1H), 4.24 (q, J = 14.2, 7.1 Hz, 2H), 1.30 (t, J = 7.1 Hz, 3H). MS (ESI): m/z 311 [M + Na]+.

4.1.3. Ethyl 5-(1H-Pyrrol-1-yl)-1-[6-(p-tolyl)pyridin-2-yl]-1H-pyrazole-4-carboxylate (57)

To a solution of 56 (100 mg, 0.27 mmol) in 1,4-dioxane (4 mL), Pd(OAc)2 (6.04 mg, 0.027 mmol), PPh3 (21.2 mg, 0.08 mmol), and p-tolylboronic acid (110 mg, 0.81 mmol) were added successively. After flushing with N2, a 1 M Na2CO3 solution (0.5 mL, 0.5 mmol) was added and the reaction mixture was refluxed for 4 h under N2. The solvent was removed under reduced pressure and the residue was taken up in EtOAc and filtered on celite. The solution was dried over anhydrous sodium sulfate and concentrated to leave a residue, which was purified through flash chromatography on a silica gel (PE/EtOAc, 3:1) to produce 57 (77% yield) as a white solid. Mp 124–126 °C. 1H NMR (400 MHz, CDCl3): δ 8.11 (s, 1H), 7.77 (t, J = 7.9 Hz, 1H), 7.61 (d, J = 7.8 Hz, 1H), 7.54 (d, J = 8.2 Hz, 2H), 7.37 (d, J = 7.9 Hz, 1H), 7.13 (d, J = 8.0 Hz, 2H), 6.80–6.72 (m, 2H), 6.26–6.24 (m, 2H), 4.19 (q, J = 14.2, 7.1 Hz, 2H), 2.31 (s, 3H), 1.20 (t, J = 7.1 Hz, 3H). MS (ESI): m/z 372 [M + H]+.

4.1.4. General Procedure for the Synthesis of 5-(1H-Pyrrol-1-yl)-1H-pyrazole-4-carboxylic Acids 5860

Compounds 58 and 59 were prepared according to known procedures [15,25]. As an example, the preparation of the new compound 60 (5-(1H-Pyrrol-1-yl)-1-[6-(p-tolyl)pyridin-2-yl]-1H-pyrazole-4-carboxylic acid) is reported below.
A mixture of 57 (200 mg, 0.54 mmol) in EtOH (4 mL) and NaOH (216 mg, 5.4 mmol) in water (4 mL) was heated at reflux for 1 h, then cooled in an ice bath and acidified with conc. HCl. The precipitate was extracted with EtOAc and the combined organic layer was dried over sodium sulfate and evaporated under reduced pressure. The solid residue was triturated with PE/Et2O to give the title compound 60 (81% yield) as a white solid. Mp 240–243 °C. 1H NMR (400 MHz, CDCl3): δ 8.17 (s, 1H), 7.78 (t, J = 8.0 Hz, 1H), 7.63 (d, J = 7.9 Hz, 1H), 7.54 (d, J = 8.1 Hz, 2H), 7.35 (d, J = 7.9 Hz, 1H), 7.13 (d, J = 7.9 Hz, 2H), 6.77–6.75 (m, 2H), 6.26 (d, J = 2.0 Hz, 2H), 2.32 (s, 3H). MS (ESI): m/z 344 [M − H].

4.1.5. General Procedure for the Synthesis of Amide Derivatives 314

To a solution of the carboxylic acids 5860 (1 mmol) in DCM (50 mL), HOBt (135 mg, 1 mmol), EDC (383 mg, 1.2 mmol), and the appropriate amine (1.5 mmol) were added successively. After stirring at room temperature for 12 h, the solution was washed with 1 N HCl, 10% NaHCO3 solution, and brine. After drying on sodium sulfate, the solvent was removed under reduced pressure and the residue was purified by recrystallization from MeOH or flash chromatography on a silica gel using the reported eluent system.

(4-Methylpiperazin-1-yl)[1-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]methanone (3)

Prepared from the acid 58 and 1-methylpiperazine. Eluent: DCM/MeOH (97:3). Yield: 68%. Mp 109–111 °C. 1H NMR (400 MHz, CDCl3): δ 7.64 (s, 1H), 6.79–6.77 (m, 2H), 6.37–6.35 (m, 2H), 3.72 (s, 3H), 3.65–3.57 (br m, 2H), 3.20–3.16 (br m, 2H), 2.33–2.28 (br m, 2H), 2.30 (s, 3H), 2.10–1.98 (br m, 2H). 13C NMR (100 MHz, CDCl3): δ 158.3, 140.6, 136.5, 122.3, 112.6, 112.4, 56.9, 35.5, 24.8, 23.1. MS (ESI): m/z 274 [M + H]+.

(4-Methylpiperazin-1-yl)[1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]methanone (4)

Prepared from the acid 59 and 1-methylpiperazine. Eluent: DCM/MeOH (97:3). Yield: 51%. Mp 156–158 °C. 1H NMR (400 MHz, CDCl3): δ 7.85 (s, 1H), 7.33 (s, 3H), 7.16–7.11 (m, 2H), 6.64 (s, 2H), 6.27 (s, 2H), 3.70 (br s, 2H), 3.23 (br s, 2H), 2.36 (br s, 2H), 2.22 (s, 3H), 2.04 (br s, 2H). 13C NMR (100 MHz, CDCl3): δ 162.1, 140.0, 137.8, 136.2, 136.2, 129.2, 128.2, 123.2, 121.8, 112.0, 111.4, 54.5, 45.9, 41.8. MS (ESI): m/z 336 [M + H]+.

1-Phenyl-N-(piperidin-1-yl)-5-(1H-pyrrol-1-yl)-1H-pyrazole-4-carboxamide (5)

Obtained from the acid 59 and 1-aminopiperidine. Eluent: DCM/MeOH (95:5). Yield: 33%. Mp 165–167 °C. 1H NMR (400 MHz, CDCl3): δ 8.24 (s, 1H), 7.34–7.30 (m, 3H), 7.19–7.17 (m, 2H), 6.76 (s, 2H), 6.45 (s, 2H), 2.56–2.52 (m, 4H), 1.67–1.63 (m, 4H), 1.36–1.33 (m, 2H). 13C NMR (100 MHz, CDCl3): δ 158.2, 141.7, 137.6, 135.8, 129.2, 128.2, 122.5, 122.4, 113.9, 112.4, 56.9, 24.8, 23.1. MS (ESI): m/z 336 [M + H]+.

N-(4-Chlorophenyl)-1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-4-carboxamide (6)

Prepared from the acid 59 and 4-chloroaniline. Yield: 38%. Mp 197–198 °C. 1H NMR (400 MHz, CDCl3): δ 8.31 (s, 1H), 7.35 (s, 3H), 7.26–7.22 (m, 4H), 6.87 (s, 2H), 6.79 (s, 2H), 6.55 (s, 2H). 13C NMR (100 MHz, CDCl3): δ 158.6, 141.8, 137.4, 136.3, 135.9, 129.4, 129.1, 128.9, 128.5, 122.8, 122.6, 120.8, 114.7, 113.0. MS (ESI): m/z 385 [M + Na]+.

[1-Phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl][4-(pyrimidin-2-yl)piperazin-1-yl]methanone (7)

Prepared from the acid 59 and 1-(2-pyrimidyl)piperazine. Yield: 73%. Eluent DCM/MeOH (95:5). Mp 218–219 °C. 1H NMR (400 MHz,CDCl3): δ 8.28 (m, 2H), 7.91 (s, 1H), 7.34 (m, 3H), 7.14–7.13 (m, 2H), 6.66 (m, 2H), 6.51 (m,1H), 6.27 (s, 2H), 3.81–3.62 (m, 4H), 3.44–3.22 (m, 4H). 13C NMR (100 MHz, CDCl3): δ 157.8, 140.2, 136.4, 129.3, 128.3, 127.5, 123.2, 122.6, 122.4, 121.7, 116.2, 111.7, 110.4, 56.4, 43.3. MS (ESI): m/z 422 [M + Na]+.

N-(Adamantan-1-yl)-1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-4-carboxamide (8)

Obtained through a reaction between acid 59 and 1-aminoadamantane. Yield: 73%. Mp 167–169 °C. 1H NMR (400 MHz, CD3COCD3): δ 7.29 (s, 1H), 6.63 (m, 3H), 6.50 (m, 2H), 6.25 (s, 2H), 6.65 (s, 2H), 4.54 (br s, 1H), 1.26 (s, 3H), 1.15 (s, 6H), 0.91 (s, 6H). 13C NMR (100 MHz, CDCl3): δ 159.8, 141.5, 137.7, 135.6, 129.2, 128.1, 122.4, 122.3, 115.8, 112.4, 51.9, 41.5, 36.3, 29.4. MS (ESI): m/z 409 [M + Na]+.

tert-Butyl (S)-3-Phenyl-2-[[[1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]carbonyl]amino]propanoate (9)

Prepared from the acid 59 and L-phenylalanine tert-butyl ester hydrochloride. Eluent: PE/EtOAc (3:1). Yield: 90%. Oil. 1H NMR (400 MHz,CDCl3): δ 8.19 (s, 1H), 7.33–7.16 (m, 6H), 7.09–7.00 (m, 4H), 6.64 (d, J = 1.9 Hz, 2H), 6.36 (d, J = 2.0 Hz, 2H), 5.82 (br s, 1H), 4.88–4.80 (m, 1H), 3.08–3.03 (m, 1H), 2.95–2.88 (m, 1H), 1.31 (s, 9H). 13C NMR (100 MHz, CDCl3): δ 170.3, 160.4, 141.5, 137.6, 136.6, 136.2, 129.4, 129.2, 128.4, 128.2, 126.9, 122.5, 122.3, 113.4, 112.4, 82.2, 53.6, 37.9, 27.9.. MS (ESI): m/z 479 [M + Na]+.

Ethyl 3-[[N-Benzil-N-[1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]carbonyl]amino]propanoate (10)

Obtained through a reaction between acid 59 and ethyl 3-(benzylamino)propionate. Eluent: PE/EtOAc (3:1). Yield: 65%. White solid. Mp 93–96 °C. 1H NMR (400 MHz, CDCl3): δ 7.87 and 7.79 (s, 1H overall, rotamers), 7.38–7.22 (m, 6H), 7.18–6.98 (m, 4H), 6.66 (s, 2H), 6.27 (s, 2H), 4.68 and 4.45 (br m, 2H overall, rotamers), 4.11 (q, J = 7.1 Hz, 2H), 3.61 and 3.43 (br m, 2H overall, rotamers), 2.60 and 2.22 (br m, 2H overall, rotamers), 1.24 (t, J = 7.1 Hz, 3H). 13C NMR (100 MHz, CDCl3): 171.7, 170.8 (1 C), 164.1, 139.0, 137.9, 136.9, 136.5, 129.2, 128.9, 128.1, 127.7, 126.9, 123.1, 122.1, 112.7, 112.1 (1 C), 111.3, 60.6, 53.1, 47.7 (1 C), 43.7, 41.6 (1 C), 32.8, 32.3 (1 C), 14.2. MS (ESI): m/z 442 [M + H]+.

Methyl [[[1-Phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]carbonyl]amino]acetate (11)

Prepared from acid 59 and glycine methyl ester hydrochloride. Eluent: PE/EtOAc (3:1 to 1:1). Yield: 70%. White needles. Mp 139–141 °C. 1H NMR (400 MHz, CDCl3): δ 8.15 (s, 1H), 7.25 (m, 3H), 7.05 (m, 2H), 6.71 (s, 2H), 6.36 (s, 2H), 5.79 (br s, 1H), 4.00 (d, 2H), 3.65 (s, 3H). 13C NMR (100 MHz, CDCl3): δ 169.9, 160.9, 141.5, 137.6, 136.7, 129.2, 128.3, 122.5, 122.4, 113.2, 112.5, 52.3, 41.1. MS (ESI): m/z 347 [M + Na]+.

Ethyl (S)-2-[[[1-Phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]carbonyl]amino]propanoate (12)

Obtained from acid 59 and L-alanine ethyl ester hydrochloride. Eluent PE/EtOAc (3:1). Yield: 60%. Amorphous white solid. Mp 90–92 °C. 1H NMR (400 MHz, CDCl3): δ 8.22 (s, 1H), 7.34–7.32 (m, 3H), 7.15 (dd, J = 7.6, 2.1 Hz, 2H), 6.79 (s, 2H), 6.45 (s, 2H), 5.77 (br d, 1H), 4.66–4.59 (m, 1H), 4.17 (qd, J = 7.2, 1.8 Hz, 2H), 1.31–1.23 (m, 6H). 13C NMR (100 MHz, CDCl3): δ 172.5, 160.3, 141.4, 137.6, 136.6, 129.2, 128.2, 122.5, 122.4, 113.7, 112.4, 61.4, 47.9, 18.2, 14.1. MS (ESI): m/z 352 [M + Na]+.

Ethyl (S)-3-Methyl-2-[[[1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]carbonyl]amino]butanoate (13)

Prepared from acid 59 and L-valine ethyl ester hydrochloride. Eluent PE/EtOAc (3:1). Yield: 53%. Yellow solid. Mp 75–78 °C. 1H NMR (400 MHz, CDCl3): δ 8.23 (s, 1H), 7.33–7.31 (m, 3H), 7.14 (dd, J = 7.6, 2.0, 2H), 6.83–6.82 (m, 2H), 6.45–6.44 (m, 1H), 5.78 (d, J = 8.4 Hz, 1H), 4.60 (dd, J = 8.6, 4.6 Hz, 1H), 4.16 (q, J = 7.1 Hz, 2H), 2.13–2.04 (m, 1H), 1.26 (t, J = 7.1 Hz, 3H), 0.85 (d, J = 6.9 Hz, 3H), 0.68 (d, J = 6.9 Hz, 3H). 13C NMR (100 MHz, CDCl3): δ 171.5, 160.8, 141.7, 137.8, 136.3, 129.2, 128.2, 122.5, 113.9, 112.6, 61.1, 56.8, 30.8, 19.0, 17.2, 14.2. MS (ESI): m/z 380 [2M + Na]+.

tert-Butyl (S)-3-Phenyl-2-[[[5-(1H-pyrrol-1-yl)-1-[6-(p-tolyl)pyridin-2-yl]-1H-pyrazol-4-yl]carbonyl]amino]propanoate (14)

Prepared from acid 60 and L-phenylalanine tert-butyl ester hydrochloride. Eluent: PE/EtOAc (2:1). Yield: 94%. Amorphous white solid. 1H NMR (400 MHz, CDCl3): δ 8.14 (s, 1H), 7.76 (t, J = 7.9 Hz, 1H), 7.58 (d, J = 7.8 Hz, 1H), 7.50–7.48 (m, 3H), 7.21–7.19 (m, 5H), 7.11 (d, J = 8.0 Hz, 2H), 7.01 (d, J = 7.7 Hz, 2H), 6.74 (d, J = 4.0 Hz, 2H), 6.33 (d, J = 3.9 Hz, 1H), 5.60 (d, J = 7.6 Hz, 1H), 4.74 (dd, J = 13.8, 7.1 Hz, 1H), 2.96 (dd, J = 14.0, 6.2 Hz, 1H), 2.80 (dd, J = 14.0, 7.1 Hz, 1H), 2.32 (s, 3H), 1.32 (s, 9H). 13C NMR (100 MHz, CDCl3): 170.3, 160.3, 156.3, 150.9, 141.9, 139.6, 139.2, 137.0, 136.3, 134.8, 129.4, 129.2, 128.4, 127.1, 126.8, 122.7, 118.8, 114.9, 114.8, 111.8. MS (ESI): m/z 547 [M + Na]+.

4.1.6. General Procedure for the Synthesis of Acids 1517

A 2 M solution of LiOH (0.7 mL, 1.4 mmol) was added to a solution of the appropriate esters 1113 (0.6 mmol) in THF (2 mL) and MeOH (1 mL). After stirring at room temperature for 2–4 h, the mixture was concentrated, diluted with water, and brought to a pH of 1–2 with conc. HCl. The solid was extracted with EtOAc and the combined organic layer was washed with brine and dried over anhydrous sodium sulfate. Evaporation of the solvent left a solid, which was purified by trituration with PE/Et2O.

[[[1-Phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]carbonyl]amino]acetic Acid (15)

Prepared through the hydrolysis of 11. Yield: 62%. White solid. Mp 191–194 °C. 1H NMR (400 MHz, CD3COCD3): δ 10.94 (br s, 1H), 8.08 (s, 1H), 7.31–7.30 (m, 3H), 7.12–7.10 (m, 2H), 6.83 (s, 2H), 6.77 (br s, 1H), 6.19 (s, 2H), 3.95 (d, J = 5.5 Hz, 2H). 13C NMR (100 MHz, CD3OD): δ 171.4, 162.3, 139.8, 138.6, 137.6, 128.9, 128.3, 123.2, 122.6, 112.1, 110.8, 40.4. MS (ESI): m/z 309 [M − H].

(S)-2-[[[1-Phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]carbonyl]amino]propanoic Acid (16)

Obtained via the hydrolysis of 12. Yield: 60%. Pale-pink solid. Mp 142–145 °C. 1H NMR (400 MHz, CDCl3): δ 9.43 (s, 1H), 8.23 (s, 1H), 7.33–7.32 (m, 3H), 7.17–7.15 (m, 2H), 6.77 (s, 2H), 6.43 (s, 1H), 5.78 (d, J = 6.7 Hz, 1H), 4.64–4.57 (m, 1H), 1.32 (d, J = 7.1 Hz, 3H). 13C NMR (100 MHz, CDCl3): δ 176.1, 161.0, 141.5, 137.4, 136.9, 129.3, 128.4, 122.6, 122.4, 113.4, 112.6, 48.0, 17.7. MS (ESI): m/z 324 [M − H].

(S)-3-Methyl-2-[[[1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]carbonyl]amino]butanoic Acid (17)

Prepared through the hydrolysis of 13. Yield: 77%. Light-grey solid. Mp 150–152 °C. 1H NMR (400 MHz, CDCl3): δ 8.25 (s, 1H), 7.34–7.32 (m, 3H), 7.16–7.13 (m, 2H), 6.82 (s,2H), 6.45 (s,2H), 5.75 (d, J = 8.3 Hz, 1H), 4.59 (dd, J = 8.3, 4.5 Hz, 1H), 2.15 (dq, J = 13.6, 6.9 Hz, 1H), 0.88 (d, J = 6.8 Hz, 3H), 0.71 (d, J = 6.9 Hz, 3H). 13C NMR (100 MHz, CDCl3): δ 175.6, 161.3, 141.8, 137.5, 136.5, 129.3, 128.3, 122.5, 113.6, 112.8, 56.9, 30.2, 19.1, 17.0. MS (ESI): m/z 352 [M − H].

4.1.7. N-[N-[[1-Phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]carbonyl]glycyl]-L-alanine Ethyl Ester (18)

Prepared from the acid 15 and L-alanine ethyl ester hydrochloride according to the general procedure used for the synthesis of amides 314. Eluent: PE/EtOAc (1:1). Yield: 80%. White solid. Mp 116–117 °C. 1H NMR (400 MHz, CDCl3): δ 8.22 (s, 1H), 7.33–7.31 (m, 3H), 7.14–7.11 (m, 2H), 6.79–6.78 (m, 3H), 6.42 (s, 2H), 6.10 (br t, 1H), 4.58–4.51 (m, 1H), 4.19 (q, J = 7.1, 2H), 4.0 (d, J = 3.6, 2H), 2.33 (br s, 1H), 1.40 (d, J = 7.2, 3H), 1.27 (t, J = 7.1 3H). 13C NMR (100 MHz, CDCl3): δ 172.7, 168.2, 161.4, 141.2, 137.6, 137.1, 129.2, 128.3, 122.6, 122.5, 113.0, 112.4, 61.5, 48.2, 43.0, 18.3, 14.1. MS (ESI): m/z 432 [M + Na]+.

4.1.8. N-[N-[[1-Phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-4-yl]carbonyl]glycyl]-L-alanine (19)

The hydrolysis of 18, according to the general procedure described for the synthesis of acids 1517, produced the title compound 19, which was purified via recrystallization from MeOH/H2O. Yield: 81%. Light-yellow solid. Mp 80–84 °C. 1H NMR (400 MHz, CD3OD): δ 8.10 (s, 1H), 7.28–7.26 (m, 3H), 7.08–7.05 (m, 2H), 6.71–6.70 (m, 2H), 6.19–6.18 (m, 2H), 4.82 (br s, 2H), 4.35 (q, J = 7.3, 1H), 3.90 (q, J = 16.7, 2H), 1.32 (d, J = 7.3, 3H). 13C NMR (100 MHz, CD3OD): δ 174.4, 169.6, 162.3, 139.9, 138.7, 137.6, 128.9, 128.3, 123.2, 122.6, 112.2, 110.8, 48.0, 41.8, 16.4. MS (ESI): m/z 380 [M − H].

4.1.9. General Procedure for the Synthesis of 61 and 62

A solution of the amino derivatives 51 and 53 (3.6 mmol), DMAP (44 mg, 0.36 mmol), and di-tert-butyl dicarbonate (1.57 g, 7.2 mmol) in dry DCM (40 mL) was stirred at room temperature for 2–4 h. Afterward, it was washed with 1 N HCl, then brine, and finally, dried over anhydrous sodium sulfate. The removal of the solvent left an oily residue, which was purified by using flash chromatography on a silica gel eluted with PE/EtOAc (4:1) to produce the pure compounds 61 and 62.

Ethyl 5-(tert-Butoxycarbonylamino)-1-phenyl-1H-pyrazole-4-carboxylate (61)

Prepared from 51. Yield: 90%. White solid. Mp 91–93 °C. 1H NMR (400 MHz, CDCl3): δ 8.04 (s, 1H), 7.41–7.35 (m, 5H), 4.24 (q, 2H), 1.29–1.24 (superimposed signals, 12H). MS (ESI): m/z 332 [M + H]+.

Ethyl 5-(tert-Butoxycarbonylamino)-1-(4-nitrophenyl)-1H-pyrazole-4-carboxylate (62)

Prepared from 52. Yield:79%. Pale-yellow solid. Mp 125–129 °C. 1H NMR (400 MHz, CDCl3): δ 8.25 (d, 2H), 8.02 (s, 1H), 7.61 (d, 2H), 4.20 (q, 2H), 1.20–1.30 (superimposed signals, 12H). MS (ESI): m/z 377 [M + H]+.

4.1.10. General Procedure for the Synthesis of Acids 63 and 64

To a solution of the esters 61 and 62 (4 mmol) in 50 mL of EtOH, 6 N KOH (27 mL, 0.16 mol) was added and the mixture was refluxed for 1 h. After cooling, the dark-orange solution was acidified with conc. HCl and the precipitate was extracted with DCM. The organic layer was washed with brine, dried (Na2SO4), and evaporated to leave a solid, which was sufficiently pure to be used in the next step.

5-(tert-Butoxycarbonylamino)-1-phenyl-1H-pyrazole-4-carboxylic Acid (63)

Prepared from ester 61. Yield: 73%. Mp 159–161 °C. 1H NMR (400 MHz, CDCl3): δ 7.99 (s, 1H), 7.53 (d, 2H), 7.43–7.20 (m, 3H), 1.19 (s, 9H). MS (ESI): m/z 302 [M − H].

5-(tert-Butoxycarbonylamino)-1-(4-nitrophenyl)-1H-pyrazole-4-carboxylic Acid (64)

Obtained from ester 62. Yield: 20%. Mp 212–213 °C. 1H NMR (400 MHz, CDCl3): δ 8.28 (d, 2H), 8.03 (s, 1H), 7.76 (d, 2H), 5.23 (s, 1H), 1.26 (s, 9H). MS (ESI): m/z 346 [M − H].

4.1.11. General Procedure for the Synthesis of Amide Derivatives 2022

To a solution of the carboxylic acids 63 and 64 (1 mmol) in DCM (50 mL), HOBt (135 mg, 1 mmol), EDC (230 mg, 1.2 mmol), and the appropriate amine (1.5 mmol) were added successively. After stirring at room temperature for 2–4 h, the solution was washed with 1 N HCl, 10% NaHCO3 solution, and brine. After drying on sodium sulfate, the solvent was removed under reduced pressure and the residue was purified as described below.

N-(4-Bromobenzyl)-5-(tert-butoxycarbonylamino)-1-phenyl-1H-pyrazole-4-carboxamide (20)

Prepared from acid 63 and 4-bromobenzylamine. Purified via flash chromatography on a silica gel eluted with PE/EtOAc (1:1). Yield: 39%. White solid. Mp 175–176 °C. 1H NMR (400 MHz, CD3OD): δ 7.97 (s, 1H), 7.45–7.37 (m, 7H), 7.22 (d, J = 8.3 Hz, 2H), 4.43 (s, 2H), 1.20 (s, 9H). 13C NMR (100 MHz, CD3OD): δ 160.0, 153.6, 139.0, 138.4, 138.1, 137.0, 134.7, 131.2, 129.1, 128.9, 128.3, 124.2, 120.5, 81.2, 41.9, 26.9. MS (ESI): m/z 493 [M + Na]+.

N-(4-Bromobenzyl)-5-(tert-butoxycarbonylamino)-1-(4-nitrophenyl)-1H-pyrazole-4-carboxamide (21)

Obtained through a reaction between acid 64 and 4-bromobenzylamine. Purified via recrystallization from MeOH. Yield: 51%. White solid. Mp 169–171 °C. 1H NMR (400 MHz, DMSO-d6): δ 9.17 (br s, 1H), 8.62 (br t, 1H), 8.33 (d, J = 8.7 Hz, 2H), 8.16 (d, J = 2.7, 1H), 7.76 (d, J = 8.7 Hz, 2H), 7.44 (d, J = 8.2 Hz, 2H), 7.21 (d, J = 8.1 Hz, 2H), 4.34 (d, J = 4.2 Hz, 2H), 1.17 (s, 9H). 13C NMR (100 MHz, DMSO-d6): δ 161.6, 157.3, 146.5, 144.1, 140.7, 139.5, 138.4, 131.6, 130.0, 125.3, 124.2, 120.3, 112.9, 80.9, 41.9, 28.1. MS (ESI): m/z 538 [M + Na]+.

5-(tert-Butoxycarbonylamino)-1-(4-nitrophenyl)-N-[[5-(4-trifluoromethoxyphenyl)pyridin-2-yl]methyl]-1H-pyrazole-4-carboxamide (22)

Prepared from acid 64 and [5-[4-(trifluoromethoxy)phenyl]pyridin-2-yl]methanamine [16]. The yellowish compound was purified through repeated triturations with EtOAc until a white solid was obtained. Yield: 59%. Mp 197–199 °C. 1H NMR (400 MHz, DMSO-d6): δ 9.22 (s, 1H), 8.76 (d, J = 7.0 Hz, 2H), 8.34 (d, J = 7.2 Hz, 2H), 8.22 (s, 1H), 8.00 (d, J = 8.2 Hz, 1H), 7.77 (d, J = 8.8 Hz, 3H), 7.46–7.36 (m, 2H), 4.51 (d, J = 5.6 Hz, 2H), 1.16 (s, 9H). 13C NMR (100 MHz, CDCl3): δ 163.1, 154.7, 151.6, 149.6, 146.3, 145.3, 141.0, 140.0, 138.6, 136.5, 135.4, 135.0, 128.6, 124.6, 122.9, 122.8, 121.7, 107.4, 82.6, 43.3, 27.9. MS (ESI): m/z 597 [M − H].

4.1.12. General Procedure for the Synthesis of Amides 2339

These compounds were prepared according to the general procedure reported for the synthesis of amides 314 and 2022, and purified as reported below.

N-(Adamantan-3-ol-1-yl)-1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (23)

Prepared from 65 and 3-aminoadamantan-1-ol. Purified through recrystallization from MeOH. Yield: 96%. Colorless crystals. Mp 217–221 °C. 1H NMR (400 MHz, CDCl3): δ 7.37–7.36 (m, 3H), 7.18–7.16 (m, 2H), 6.89 (s, 1H), 6.83 (s, 1H), 6.63–6.62 (m, 2H), 6.26–6.25 (m, 2H), 2.32 (s, 2H), 2.07 (q, J = 11.7, 4H), 1.91 (s, 2H), 1.75 (q, J = 11.6, 4H), 1.65–1.55 (m, 2H). 13C NMR (100 MHz, CDCl3): δ 160.4, 147.4, 140.6, 137.8, 129.3, 128.5, 123.5, 122.1, 110.9, 102.7, 69.1, 54.4, 49.1, 44.1, 40.4, 35.0, 30.7. MS (ESI): m/z 425 [M + Na] +.

N-(Adamantan-1-yl)-4-cyano-1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (24)

Prepared from 66 and 1-aminoadamantane. Purified through recrystallization from EtOH/DCM. Yield: 90%. White solid. Mp 227–229 °C. 1H NMR (400 MHz, CDCl3): δ 7.46–7.44 (m, 3H), 7.21–7.18 (m, 2H), 6.72–6.70 (m, 2H), 6.59 (br s, 1H), 6.34–6.32 (m, 2H), 2.16 (s, 9H), 1.73 (s, 6H). 13C NMR (100 MHz, CDCl3): δ 157.6, 148.2, 144.5, 136.7, 129.7, 123.9, 121.3, 112.4, 111.3, 88.8, 52.6, 41.6, 36.3, 29.5. MS (ESI): m/z 434 [M + Na]+.

tert-Butyl (S)-3-Phenyl-2-[[[4-cyano-1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-3-yl]carbonyl]amino]propanoate (25)

Prepared from 66 and L-phenylalanine tert-butyl ester hydrochloride. Purified via flash chromatography on a silica gel, eluent PE/EtOAc (3:1), followed by trituration with PE/Et2O. Yield: 56%. White solid. Mp 93–94 °C. 1H NMR (400 MHz, CDCl3): δ 7.46–7.37 (m, 5H), 7.30–7.17 (m, 6H), 6.74–6.73 (m, 2H), 6.35–6.34 (m, 2H), 4.99 (dd, J = 14.1, 6.2 Hz, 1H), 3.24 (d, J = 6.1 Hz, 2H), 1.42 (s, 9H). 13C NMR (100 MHz, CDCl3): δ 170.3, 158.3, 146.7, 144.5, 136.7, 136.1, 129.7, 129.6, 129.5, 128.5, 127.0, 123.8, 121.4, 112.5, 111.0, 89.1, 82.7, 53.6,38.3, 28.0. MS (ESI): m/z 504 [M + Na]+.

(4-Ethoxycarbonylpiperazin-1-yl)[4-methyl-1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-3-yl]methanone (26)

Prepared from 67 and ethyl 1-piperazinecarboxylate. Purified via flash chromatography on a silica gel, eluent PE/EtOAc (3:1). Yield: 81%. White solid. Mp 130–131 °C. 1H NMR (400 MHz, CDCl3): δ 7.41–7.21 (m, 3H), 7.06 (d, J = 8.2 Hz, 2H), 6.64 (s, 2H), 6.32 (s, 2H), 4.18 (q, J = 7.1 Hz, 2H), 3.97 (br s, 2H), 3.92 (br s, 2H), 3.61 (d, J = 4.2 Hz, 2H), 2.14 (s, 3H), 1.29 (t, J = 7.1 Hz, 3H). 13C NMR (100 MHz, CDCl3): δ 163.1, 155.5, 144.6, 138.0, 137.4, 129.2, 127.7, 122.2, 122.1, 115.1, 110.9, 61.4, 47.1, 44.3, 43.6, 42.2, 14.7, 7.9. MS (ESI): m/z 407 [M + Na]+.

N-(Adamantan-3-ol-1-yl)-4-methyl-1-phenyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (27)

Prepared from 67 and 3-aminoadamantan-1-ol. Purified via flash chromatography on a silica gel, eluent PE/EtOAc (2:1 to 1:1), followed by trituration with PE/Et2O. Yield: 34%. White solid. Mp 137–138 °C. 1H NMR (400 MHz, CDCl3): δ 7.32–7.31 (m, 3H), 7.11–7.09 (d, J = 8.0, 2H), 6.90 (s, 1H), 6.61 (s, 2H), 6.30 (s, 2H), 2.32 (s, 2H), 2.27 (s, 2H), 2.16 (s, 2H), 2.08 (s, 3H), 1.79–1.71 (m, 4H), 1.66–1.55 (m, 4H). 13C NMR (100 MHz, CDCl3): δ 161.8, 144.0, 138.0, 137.9, 129.2, 127.9, 122.5, 122.3, 115.1, 110.8, 69.2, 54.3, 49.3, 44.2, 40.5, 35.0, 30.7, 8.2. MS (ESI): m/z 439 [M + Na]+, 417 [M + H]+.

[1-(4-Chlorophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-3-yl](4-methylpiperazin-1-yl)methanone (28)

Prepared from 68 and 1-methylpiperazine. Purified via flash chromatography on a silica gel eluted with EtOAc, followed by recrystallization from MeOH. Yield: 91%. White needles. Mp 178–182 °C. 1H NMR (400 MHz, CDCl3): δ 7.26 (d, J = 8.8 Hz, 2H), 6.97 (d, J = 8.8 Hz, 2H), 6.63 (s, 2H), 6.34 (s, 2H), 3.92–3.90 (m, 2H), 3.88–3.85 (m, 2H), 2.56–2.48 (m, 4H), 2.36 (s, 3H), 2.11 (s, 3H). 13C NMR (100 MHz, CDCl3): δ 162.7, 145.4, 137.1, 136.6, 133.2, 129.3, 123.1, 122.1, 114.9, 111.2, 55.6, 54.8, 47.1, 46.0, 42.1, 7.8. MS (ESI): m/z 384 [M + H]+.

N-(Benzo[d][1,3]dioxol-5-ylmethyl)-1-(4-chlorophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (29)

Prepared from 68 and piperonylamine. Purified via flash chromatography on a silica gel eluted with PE/EtOAc 4:1, followed by recrystallization from MeOH. Yield: 74%. White crystals. Mp 144–148 °C. 1H NMR (400 MHz, CDCl3): δ 7.30–7.26 (m, 3H), 6.99 (d, J = 7.8 Hz, 2H), 6.89 (d, J = 1.4 Hz, 1H), 6.84 (dd, J = 8.0, 1.4 Hz, 1H), 6.78 (d, J = 7.9 Hz, 1H), 6.62 (s, 2H), 6.34 (s, 2H), 5.95 (s, 2H), 4.56 (d, J = 6.0 Hz, 2H), 2.30 (s, 3H). 13C NMR (100 MHz, CDCl3): δ 162.1, 148.0, 147.0, 143.6, 137.9, 136.4, 133.6, 132.3, 129.3, 123.3, 122.2, 121.2, 115.8, 111.2, 108.5, 108.3, 101.1, 42.8, 8.1. MS (ESI): m/z 457 [M + Na]+.

[1-(4-Chlorophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-3-yl](morpholin-4-yl)methanone (30)

Prepared from 68 and morpholine. Purified via flash chromatography on a silica gel (eluent: EtOAc), followed by recrystallization from MeOH. Yield: 89%. White needles. Mp 174–177 °C. 1H NMR (400 MHz, CDCl3): δ 7.20–7.18 (m, 2H), 6.90–6.87 (m, 2H), 6.57–6.55 (m, 2H), 6.27–6.24 (m, 2H), 3.88–3.86 (m, 2H), 3.76–3.73 (m, 4H), 3.68–3.66 (m, 2H), 2.04 (s, 3H). 13C NMR (100 MHz, CDCl3): δ 162.7, 144.9, 137.2, 136.5, 133.3, 129.3, 123.1, 115.3, 111.2, 67.2, 66.9, 47.8, 42.6, 7.8. MS (ESI): m/z 393 [M + Na]+.

N-Benzyl-1-(4-chlorophenyl)-N-(2-hydroxyethyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (31)

Obtained through a reaction between 68 and N-benzylethanolamine. Purified via flash chromatography on a silica gel eluted with PE/EtOAc (1:1). Yield: 82%. Oil. Two rotamers: 1H NMR (400 MHz, CDCl3): δ 7.37–7.34 (m), 7.28 (d, J = 10.7 Hz), 7.19 (d, J = 7.5 Hz), 6.96 (d, J = 7.9 Hz), 6.82 (d, J = 7.5 Hz), 6.63 (d, J = 7.7 Hz, 2H), 6.35 (s), 5.04 (s), 4.88 (s), 4.42 (br s), 3.86 (s), 3.77–3.70 (m, 2H), 2.20 (s), 2.13 (s). 13C NMR (100 MHz, CDCl3): δ 166.2 and 164.3 (1 C), 145.2 and 145.1 (1 C), 137.7 and 137.3 (1 C), 137.2 and 137.0 (1 C), 136.5 and 136.1 (1 C), 133.7 and 133.2 (1 C), 129.6, 129.3, 128.7, 128.1, 127.6, 123.2, 122.8, 122.1, 115.9 and 115.7 (1 C), 111.4, 61.5, 59.6, 54.1, 49.5, 48.4, 7.9. MS (ESI): m/z 435 [M + H]+.

Ethyl 4-[[[1-(4-Chlorophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-3-yl]carbonyl]amino]benzoate (32)

Prepared through a reaction between 68 and ethyl 4-aminobenzoate. Purified via flash chromatography on a silica gel eluted with DCM, followed by recrystallization from EtOH. Yield: 70%. White needles. Mp 168–170 °C. 1H NMR (400 MHz, CDCl3): δ 8.98 (br s, 1H), 8.07 (d, J = 8.7 Hz, 2H), 7.80 (d, J = 8.7 Hz, 2H), 7.33 (d, J = 8.8 Hz, 2H), 7.06 (d, J = 8.8 Hz, 2H), 6.65–6.64 (m, 2H), 6.37–6.36 (m, 2H), 4.38 (q, J = 7.2 Hz, 2H), 2.34 (s, 3H), 1.41 (t, J = 7.1 Hz, 3H). 13C NMR (100 MHz, CDCl3): δ 166.2, 160.2, 143.3, 141.8, 138.4, 136.2, 134.0, 130.9, 129.5, 125.9, 123.5, 122.2, 118.8, 116.3, 111.4, 60.9, 14.4, 8.2. MS (ESI): m/z 449 [M + H]+.

1-(4-Chlorophenyl)-4-methyl-N-(pyridin-4-ylmethyl)-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (33)

Prepared through a reaction between 68 and 4-aminomethylpyridine. Purified via flash chromatography on a silica gel eluted with EtOAc, followed by recrystallization from PE/Et2O/DCM. Yield: 75%. White crystals. Mp 189–193 °C. 1H NMR (400 MHz, CDCl3): δ 8.60 (d, J = 5.0 Hz, 2H), 7.41 (t, J = 5.9 Hz, 1H), 7.37–7.31 (m, 4H), 7.01 (d, J = 8.9 Hz, 2H), 6.63–6.62 (m, 2H), 6.36–6.35 (m, 2H), 4.67 (d, J = 6.3 Hz, 2H) 2.30 (s, 3H). 13C NMR (100 MHz, CDCl3): δ 162.5, 149.1, 148.8, 143.1, 138.1, 136.3, 133.8, 129.4, 123.3, 122.6, 122.2, 115.9, 111.3, 41.8, 8.06. MS (ESI): m/z 392 [M + H]+.

N-Benzyl-1-(4-chlorophenyl)-N,4-dimethyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (34)

Obtained through a reaction between 68 and N-benzylmethylamine. Purified via trituration with PE. Yield: 53%. White solid. Mp 104–108 °C. Two rotamers: 1H NMR (400 MHz, CDCl3): δ 7.46–7.13 (m), 6.99 (d, J = 7.4 Hz), 6.86 (d, J = 7.9 Hz), 6.63 (s), 6.35 (s), 4.95 (s), 4.81 (s), 3.25 (s), 3.08 (s), 2.15 (s). 13C NMR (100 MHz, CDCl3): δ 164.7 and 164.3 (1 C), 137.3 and 136.9 (1 C), 137.1 and 136.6 (1C), 133.2 and 133.1 (1 C), 129.3, 128.7, 128.2, 127.6, 127.4, 123.1, 122.8, 122.1, 115.2 and 115.1 (1 C), 111.2, 54.9 and 51.2 (1 C), 36.6 and 33.4 (1 C), 7.8. MS (ESI): m/z 405 [M + H]+.

1-(4-Chlorophenyl)-4-cyano-N-(pyridin-4-ylmethyl)-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (35)

Prepared through a reaction between 69 and 4-aminomethylpyridine. Purified via flash chromatography on a silica gel eluted with DCM/MeOH (99:1). Yield: 54%. Glassy solid. Mp 85–88 °C. 1H NMR (400 MHz, CDCl3): δ 8.59 (d, J = 5.4 Hz, 2H), 7.40 (d, J = 8.7 Hz, 2H), 7.34 (d, J = 5.3 Hz, 2H), 7.12 (d, J = 8.7 Hz, 2H), 6.73 (s, 2H), 6.38 (s, 2H), 4.70 (d, J = 6.3 Hz, 2H). 13C NMR (100 MHz, CDCl3): δ 158.9, 149.3, 147.8, 146.8, 144.6, 135.9, 135.0, 129.9, 124.9, 122.7, 121.3, 113.0, 110.6, 89.5, 42.1. MS (ESI): m/z 403 [M + H]+.

1-(3,4-Dichlorophenyl)-N-(4-fluorobenzyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (36)

Prepared through a reaction between 70 and 4-fluorobenzylamine. Purified via flash chromatography on a silica gel eluted with PE/EtOAc (2:1). Yield: 77%. White solid. Mp 148–151 °C. 1H NMR (400 MHz, CDCl3): δ 7.51–7.29 (m, 4H), 7.25 (d, J = 2.3 Hz, 1H), 7.05 (t, J = 8.6, 2H), 6.73 (dd, J = 8.7, 2.3, 1H), 6.63 (s, 2H), 6.39 (s, 2H), 4.63 (d, J = 6.1, 2H), 2.30 (s, 3H)). 13C NMR (100 MHz, CDCl3): δ 163.4, 162.0, 161.0, 143.9, 137.9, 137.0, 134.3, 133.2, 131.7, 130.7, 129.6, 129.5, 123.8, 122.1, 120.4, 116.3, 115.6, 115.4, 111.7, 42.3, 8.1. MS (ESI): m/z 465 [M + Na]+.

1-(3,4-Dichlorophenyl)-N-[4-(4-fluorophenyl)thiazol-2-yl]-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (37)

Prepared through a reaction between 70 and 2-amino-4-(4-fluorophenyl)thiazole. Purified via flash chromatography on a silica gel eluted with PE/EtOAc (5:1). Yield: 20%. White solid. Mp 207–210 °C. 1H NMR (400 MHz, CDCl3): δ 10.54 (br s, 1H), 7.86–7.82 (m, 2H), 7.40 (d, J = 8.8 Hz, 1H), 7.27 (s, 1H), 7.15–7.08 (m, 3H), 6.83 (d, J = 8.7 Hz, 1H), 6.67 (s, 2H), 6.43 (s, 2H), 2.35 (s, 3H). 13C NMR (100 MHz, CDCl3): δ 163.9, 161.4, 159.4, 157.5, 149.0, 141.9, 138.3, 136.6, 133.4, 132.1, 130.8, 130.4, 127.8, 127.7, 123.6, 122.1, 120.4, 117.2, 115.7, 115.5, 111.9, 107.3, 7.9. MS (ESI): m/z 511 [M − H].

N-(Benzo[d][1,3]dioxol-5-ylmethyl)-1-(3,4-dichlorophenyl)-4-methyl-5-(2,5-dimethyl-1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (38)

Prepared from 71 and piperonylamine. Purified via flash chromatography on a silica gel eluted with PE/EtOAc (2:1). Yield: 97%. Light-yellow solid. Mp 141–145 °C. 1H NMR (400 MHz, CDCl3): δ 7.46–7.30 (m, 4H), 7.22 (d J = 2.5 Hz, 1H), 6.92 (s, 1H), 6.88 (d, J = 7.9 Hz, 1H), 6.80 (d, J = 7.9 Hz, 1H), 6.82 (dd, J = 8.8, 2.5 Hz, 1H), 5.97 (d, J = 9.6 Hz, 2H), 4.58 (d, J = 6.0 Hz, 2H), 2.25 (s, 3H), 1.87 (s, 6H). 13C NMR (100 MHz, CDCl3): δ 162.0, 148.0, 147.0, 144.0, 137.4, 135.7, 133.4, 132.2, 131.2, 131.0, 128.9, 122.9, 121.2, 119.4, 118.5, 108.6, 108.3, 101.1, 42.8, 12.2, 8.3. MS (ESI): m/z 498 [M + H]+.

tert-Butyl (S)-3-Phenyl-2-[[[4-methyl-1-(2,4,6-trichlorophenyl)-5-(2,5-dimethyl-1H-pyrrol-1-yl)-1H-pyrazol-3-yl]carbonyl]amino]propanoate (39)

Prepared from 72 and L-phenylalanine tert-butyl ester hydrochloride. Purified via flash chromatography on a silica gel eluted with DCM. Yield: 58%. Glassy solid. Mixture of rotamers. Mp 72–81 °C. 1H NMR (400 MHz, CDCl3): δ 7.41–7.31 (m, 3H), 7.22–7.17 (m, 5H), 5.79–5.76 (m, 2H), 4.93–4.86 (m, 1H), 3.14 (t, J = 6.4 Hz, 2H), 2.09 and 2.07 (s, 3H overall), 1.94 and 1.92 (s, 3H overall), 1.89 and 1.87 (s, 3H overall), 1.36 and 1.34 (s, 9H overall). 13C NMR (100 MHz, CDCl3): δ 170.4, 161.6, 144.4, 138.5, 136.4, 135.4, 134.9, 132.8, 130.5, 130.3, 129.7, 129.6, 129.2, 129.1, 128.4, 126.9, 117.3, 108.1, 82.2, 53.3, 38.5, 28.0, 12.8, 12.7, 8.7. MS (ESI): m/z 639 [M + H]+.

4.1.13. 4-[[[1-(4-Chlorophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-3-yl]carbonyl]amino]benzoic Acid (40)

A mixture of 32 (176 mg, 0.4 mmol) in MeOH (20 mL) and NaOH (208 mg, 0.52 mmol) in water (20 mL) was refluxed for 3 h. After cooling to room temperature, the solution was brought to a pH of 1–2 with conc. HCl. The precipitate was filtered under suction and washed thoroughly with water and then PE. The solid product was recrystallized with DMF to give the title compound (69 mg, 41% yield) as a white solid. Mp > 250 °C. 1H NMR (400 MHz, DMSO-d6): δ 12.67 (s, 1H), 10.42 (s, 1H), 7.90 (d, J = 8.7 Hz, 2H), 7.86 (d, J = 8.7 Hz, 2H), 7.42 (d, J = 8.6 Hz, 2H), 7.17 (d, J = 8.6 Hz, 2H), 6.86 (s, 2H), 6.23 (s, 2H), 2.09 (s, 3H).13C NMR (100 MHz, DMSO-d6): δ 167.4, 161.2, 143.9, 143.1, 138.5, 136.9, 133.2, 130.7, 129.6, 126.1, 125.0, 123.3, 120.1, 115.6, 111.3, 8.3. MS (ESI): m/z 421 [M + H]+.

4.1.14. General Procedure for the Synthesis of Amides 41, 43, and 48

These compounds were prepared according to the general procedure reported for the synthesis of amides 2339 and purified as reported below.

Methyl 3-[[[1-(4-Chlorophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-3-yl]carbonyl]amino]benzoate (41)

Prepared through a reaction between 68 and methyl 3-aminobenzoate. Purified via flash chromatography on a silica gel eluted with DCM/PE (1:1), followed by recrystallization from MeOH. Yield: 55%. White crystals. Mp 158–160 °C. 1H NMR (400 MHz, CDCl3): δ 8.93 (s, 1H), 8.23 (s, 1H), 8.10 (d, J = 7.6 Hz, 1H), 7.83 (d, J = 7.0 Hz, 1H), 7.47 (t, J = 6.6 Hz, 1H), 7.33 (d, J = 6.6 Hz, 2H), 7.07 (d, J = 6.7 Hz, 2H), 6.65 (s, 2H), 6.36 (s, 2H), 3.94 (s, 3H), 2.34 (s, 3H). MS (ESI): m/z 457 [M + Na]+.

Methyl 2-[[[1-(4-Chlorophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-3-yl]carbonyl]amino]benzoate (43)

Prepared through a reaction between 68 and methyl 2-aminobenzoate. Purified via trituration with Et2O/PE. Yield: 53%. White solid. Mp 217–219 °C. 1H NMR (400 MHz, CDCl3): δ 12.36 (s, 1H), 8.87 (dd, J = 8.5, 1.0 Hz, 1H), 8.06 (dd, J = 8.0, 3.7 Hz, 1H), 7.57 (ddd, J = 8.6, 7.4, 1.5 Hz, 1H), 7.31–7.29 (m, 2H), 7.11–7.09 (m, 3H), 6.64 (dd, J = 5.0, 2.9 Hz, 2H), 6.35 (dd, J = 5.0, 2.8 Hz, 2H), 3.94 (s, 3H), 2.30 (s, 3H). MS (ESI): m/z 457 [M + Na]+.

1-(4-Chlorophenyl)-N-(4-methylthiophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (48)

Prepared through a reaction between 68 and 4-(methylthio)aniline. Purified via trituration with Et2O/PE. Yield: 94%. Beige solid. Mp 102–105 °C. 1H NMR (400 MHz, CDCl3): δ 8.76 (s, 1H), 7.63 (d, J = 8.7 Hz, 2H), 7.33–7.28 (m, 4H), 7.10–7.07 (m, 2H), 6.63–6.62 (m, 2H), 6.34–6.32 (m, 2H), 2.47 (s, 3H), 2.30 (s, 3H). MS (ESI): m/z 445 [M + Na]+.

4.1.15. General Procedure for the Synthesis of Acids 42 and 44

These acids were prepared according to the same procedure used for the synthesis of 40.

3-[[[1-(4-Chlorophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-3-yl]carbonyl]amino]benzoic Acid (42)

Obtained from 41. Purified via recrystallization from DMF. Yield: 82%. White crystals. Mp 248–250 °C. 1H NMR (400 MHz, CDCl3): δ 10.39 (s, 1H), 8.49–8.47 (m, 1H), 8.02 (ddd, J = 8.1, 2.1, 1.0 Hz, 1H), 7.66–7.64 (m, 1H), 7.47–7.43 (m, 3H), 7.23–7.20 (m, 2H), 6.91–6.89 (m, 2H), 6.28–6.27 (m, 2H), 2.13 (s, 3H). 13C NMR (100 MHz, DMSO-d6): δ 167.66, 161.09, 143.88, 139.25, 138.38, 136.88, 133.03, 131.74, 129.59, 129.18, 124.92, 124.72 (x2), 123.28, 121.62, 115.31, 111.53. MS (ESI): m/z 443 [M + Na]+.

2-[[[1-(4-Chlorophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazol-3-yl]carbonyl]amino]benzoic Acid (44)

Prepared from 43. No further purification was required. Yield: 61%. White solid. Mp 270–272 °C. 1H NMR (400 MHz, DMSO-d6): δ 12.58 (s, 1H), 8.76 (d, J = 8.2 Hz, 1H), 8.02 (dd, J = 7.9, 1.4 Hz, 1H), 7.62–7.60 (m, 1H) 7.45 (d, J = 8.8 Hz, 2H), 7.16–7.14 (m, 1H), 7.11 (d, J = 8.8, 2H), 6.92–6.91 (m, 2H), 6.28–6.27 (m, 2H), 2.15 (s, 3H). 13C NMR (100 MHz, DMSO-d6): δ 169.49, 160.72, 143.63, 141.30, 138.64, 136.56, 134.68, 133.03, 131.60, 129.66 (x2), 124.12, 123.24, 120.03, 116.68, 115.86, 111.33, 8.33. MS (ESI): m/z 443 [M + Na]+.

4.1.16. 1-(4-Chlorophenyl)-N-(4-hydroxyphenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (45)

To a solution of the carboxylic acid 68 (0.10 g, 0.33 mmol) in dry DMF (5 mL), PyBOP (0.26 g, 0.50 mmol), HOBt (40 mg, 0.33 mmol), and 4-aminophenol (40 mg, 0.36 mmol) were added successively. After stirring at room temperature for 4 h, the solution was washed with a 5% LiCl solution and brine. After drying on sodium sulfate, the solvent was removed under reduced pressure and the residue was purified through flash chromatography on a silica gel eluted with PE/AcOEt (2:1) to give 58 mg (45%) of the title compound as a beige solid. Mp 226–228 °C. 1H NMR (400 MHz, DMSO-d6): δ 9.94 (s, 1H), 9.21 (s, 1H), 7.53 (d, 2H), 7.46–7.44 (m, 2H), 7.19–7.17 (m, 2H), 6.89 (t, 2H), 6.70 (d, 2H), 6.27–6.26 (m, 2H), 2.10 (s, 3H). 13C NMR (100 MHz, DMSO-d6): δ 160.41, 154.19, 144.42, 138.23 136.93 132.92, 130.49, 129.47 (x2), 124.76, 123.27, 122.57, 115.23, 111.21, 8.01. MS (ESI): m/z 415 [M + Na]+.

4.1.17. 1-(4-Chlorophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-N-(4-sulfamoylphenyl)-1H-pyrazole-3-carboxamide (46)

Oxalyl chloride (60 µL, 0.668 mmol) and a drop of dry DMF were added to a solution of 68 (0.18 g, 0.608 mmol) in dry DCM (15 mL), which was cooled to 0 °C. After stirring for 1 h at room temperature, volatiles were evaporated and the solid residue was taken up in dry THF (10 mL). When the solution was cooled to 0 °C, sulfanilamide (0.10 g, 0.608 mmol) and TEA (0.13 mL, 0.91 mmol) were added and the mixture was stirred at room temperature for 2 h. The solvent was evaporated, and the residue was redissolved in DCM and washed with saturated NaHCO3 solution and brine. After drying on sodium sulfate, the solution was evaporated to leave a solid residue, which was triturated with PE/AcOEt to give 0.211 g (76%) of 46 as a white solid. Mp 238–239 °C. 1H NMR (400 MHz, DMSO-d6): δ 10.50 (s, 1H), 7.98 (d, J = 8.8, 2H), 7.77 (d, J = 8.8, 2H), 7.50–7.46 (m, 2H), 7.22 (dd, J = 11.0, 4.0 Hz, 4H), 6.90 (t, J = 2.1, 2H), 6.28–6.27 (m, 2H), 2.13 (s, 3H). 13C NMR (100 MHz, DMSO-d6): δ 161.23, 143.51, 141.79, 139.23, 138.40, 136.81, 132.96, 129.43 (x2), 126.93, 125.00, 123.30, 120.38, 115.54, 110.94. MS (ESI): m/z 478 [M + Na]+.

4.1.18. Ethyl [[4-[1-(4-Chlorophenyl)-4-methyl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamido]phenyl]sulfonyl]carbamate (47)

To a suspension of 46 (50 mg, 0.11 mmol) in dry DCM (10 mL), a solution of ethyl chloroformate (0.02 mL, 0.22 mmol) and TEA (0.03 mL, 0.22 mmol) in 5 mL of DCM was added dropwise. After stirring for 1 h at room temperature, the mixture was diluted with further DCM and washed with an aqueous 10% citric acid solution, then brine, and dried over sodium sulfate. The removal of the solvent produced a residue, which was purified through flash chromatography on a silica gel eluted with PE/AcOEt (2:1 to 1:1) to provide 38 mg (65%) of 47, which was recrystallized from AcOEt (white solid). Mp 188–190 °C. 1H NMR (400 MHz, acetone-d6): δ 10.00 (s, 1H), 8.11 (d, J= 8.8, 2H), 7.98 (d, J= 8.8, 2H), 7.42 (d, J= 8.8, 2H), 7.19 (d, J= 8.8, 2H), 6.31 (d, J = 1.8, 2H), 2.22 (s, 3H). 13C NMR (100 MHz, acetone-d6): δ 205.25, 160.64, 150.97, 143.95, 143.08, 138.19, 136.80, 133.89, 133.07, 129.18, 124.09, 122.50, 116.26, 115.74, 110.90, 61.82, 13.49, 7.01. MS (ESI): m/z 550 [M + Na]+.

4.1.19. 1-(4-Chlorophenyl)-4-methyl-N-[4-(S-methylsulfonimidoyl)phenyl]-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamide (49)

To a solution of 48 (70 mg, 0.16 mmol) in MeOH (15 mL), an aqueous 25% ammonia solution (9 µL, 0.24 mmol) and (diacetoxyiodo)benzene (0.12 g, 0.38 mmol) were added, and after 2 h, the volatiles were removed under reduced pressure. The residue was dried in vacuo and triturated with PE/AcOEt to give 66 mg (88%) of compound 48 as a brown solid. Mp 207–210 °C. 1H NMR (400 MHz, CDCl3): δ 9.13 (s, 1H), 8.01 (d, 2H), 7.94 (d, 2H), 7.30 (d, 2H), 7.05 (d, 2H), 6.61 (s, 2H), 6.33 (s, 2H), 3.27 (s, 3H), 2.29 (s, 3H). 13C NMR (100 MHz (CDCl3): δ 159.91, 142.73, 138.48, 136.10, 135.11, 134.01, 129.30 (x2), 123.50, 122.15 (x2), 119.75, 116.39, 111.30, 46.16, 7.58. MS (ESI): m/z 454 [M + Na]+.

4.2. Antibacterial Susceptibility Testing

4.2.1. Agar Diffusion-Based Methods

The compounds were provided as powders and dissolved in 100% DMSO at a final concentration of 50 mg/mL. When a compound was not soluble, DMSO was further added to lower the concentration to 25 mg/mL or, when necessary, 12.5 or 6.25 mg/mL, until the compound was completely solubilized. The primary screen used in the evaluation of the direct antibacterial activity through an agar diffusion test was implemented on a 8-channel, automated liquid-handling platform (PerkinElmer Janus Integrator, Waltham, Mass.), which was located under a custom-made class 1000 laminar flow cabinet (Faster, Cornaredo, Italy). Briefly, 1-well plates (Thermo Fisher Scientific, Waltham, Mass.), previously filled with Mueller–Hinton Agar medium, were inoculated with a suspension of the bacterium in a sterile 0.9% NaCl solution containing 1.5 × 108 CFU/mL, using a sterile swab. Indicator organisms included 4 Gram-positive (Bacillus subtilis ATCC 6633, Enterococcus faecalis ATCC 29212, Staphylococcus epidermidis ATCC 14990, and Streptococcus pyogenes ATCC 12344) and 4 Gram-negative (Escherichia coli CCUGT, Klebsiella pneumoniae ATCC 13833, Pseudomonas aeruginosa ATCC 27853, and Acinetobacter baumannii ATCC 17978) strains, which were routinely propagated using Mueller–Hinton Agar plates. In total, 2 μL of each solution of compounds was dispensed on the plate surface (corresponding to 100, 50, 25, or 12.5 μg of compound according to its solubility). The controls included the solvent (100% DMSO), kanamycin, piperacillin, ceftazidime, vancomycin, and teicoplanin. The results were recorded as the diameter of the growth inhibition zone. The results were recorded as the diameter of the growth inhibition zone. The inhibition zones of the controls (max variation from expected diameter, ± 1 mm) were used as the validation criteria. The assays were performed at least in duplicate, and the position in which a single compound was spotted on the plate was randomly modified in any successive experiment.

4.2.2. Broth Microdilution Methods and MIC Determinations

A rapid test to identify compounds showing potential synergistic activity with specific antibiotics was implemented in a microplate format (final volume of 100 μL). The compounds were tested at a fixed concentration of 64 µg/mL in Mueller–Hinton broth (residual DMSO concentration ≤0.07%, except for compounds 12, 28, and 36, for which the residual DMSO concentration was 1%) in the absence and presence of a subinhibitory concentration (0.5 × MIC) of colistin (fixed concentration of 0.12 μg/mL for Escherichia coli CCUGT and Klebsiella pneumoniae ATCC 13833, and of 0.25 μg/mL for Acinetobacter baumannii ATCC 17978 and Pseudomonas aeruginosa ATCC 27853). Briefly, 10-fold concentrated (640 µg/mL) solutions of each compound were prepared in Mueller–Hinton broth and diluted in 80 μL of either unsupplemented or colistin-supplemented Mueller–Hinton II broth (Becton Dickinson, Eysins, Switzerland) in the microplate well, prior to the addition of the bacterial inoculum (5 × 105 CFU/mL, by adding 10 μL of a 5 × 106 CFU/mL bacterial suspension prepared extemporaneously in Mueller–Hinton broth). After incubation for 18–24 h at 35 ± 2 °C, plates were inspected for growth inhibition.
The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of the compounds were determined in triplicate using Mueller–Hinton broth and a bacterial inoculum of 5 × 105 CFU/mL, as recommended by the CLSI [26]. The minimum antibacterial concentration (MAC) of the compounds was determined in the presence of a fixed and subinhibitory concentration of colistin (as mentioned above) [21].
Carbapenem-resistant, colistin-resistant, and XDR clinical isolates were kindly provided by Prof. Gian Maria Rossolini (University of Florence, Italy). Klebsiella pneumoniae SI-004Bo is a KPC-producing, pandrug-resistant isolate (KKBO-4 [17]) selected in vivo during colistin therapy (colistin resistance mechanism, IS-mediated insertional inactivation of mgrB). Acinetobacter baumannii N50 is an OXA-24-producing clinical isolate resistant to all β-lactams, aminoglycosides, and fluoroquinolones [18].

4.2.3. Chequerboard Analysis and FIC Determinations

The chequerboard analysis is a two-dimensional array in which individual microplate wells contain a unique combination of the concentrations of the tested compound and colistin. Tested concentrations of the compound ranged from 64 to 0.06 μg/mL along the x-axis, whereas polymyxin E1 concentrations ranged from 2 to 0.03 μg/mL (or 32–0.5 μg/mL when polymyxin E1-resistant clinical isolates were used) and varied along the y-axis. Determining the fractional inhibitory concentration (FIC) of both compounds and computing the average FIC index allowed us to determine whether a synergistic, additive, or antagonistic activity between the tested compounds could be observed. A synergistic activity occurred when the average FIC index was equal to or less than 0.5 [21,27].

4.2.4. Kill-Curve Analysis

The time-dependent bacterial killing of colistin, compound 9, or a combination thereof was investigated using established methods [25]. Briefly, a culture medium containing a starting inoculum of 5.0 × 106 CFU/mL of Acinetobacter baumannii N50 clinical isolate was incubated aerobically at 35 °C under orbital agitation (200 rpm) in Mueller–Hinton broth, in the absence (growth control) or presence of either 2 μg/mL of colistin (a concentration equivalent to the susceptibility breakpoint), 8 µg/mL of compound 9, or both. The bacterial count (expressed in CFU/mL) in the control and antibiotic-containing media was determined every hour for up to 6 h of incubation via serial 10-fold dilution and direct plating of 0.1 mL of the sample on the Mueller–Hinton Agar medium (without antibiotic), which was incubated for 24 h at 35 °C.

4.3. Cytotoxicity Assays

4.3.1. Membrane Integrity Assays

The potential cytotoxic activity of compounds was evaluated using the commercially available integrity assay (CytoTox 96® non-radioactive cytotoxicity assay, Promega). The compounds were tested for their ability to induce the lysis of HeLa cells by measuring the release of lactate dehydrogenase (LDH) after incubating the HeLa cell cultures (20,000 cells/well) for 24 h (37 °C, 5% CO2) in DMEM (Dulbecco’s Modified Eagle Medium) supplemented with 10% fetal bovine serum, 4.5 mg/mL glucose, and 2 mM L-glutamine in the absence and presence of varying concentrations of the compounds (up to 256 µg/mL). Further controls included samples containing the medium only or in which cell lysis was induced by the addition of 9% Triton X-100 (maximum LDH release control). The percentage of cytotoxicity was computed as 100 × (sample LDH release)/(maximum LDH release). The variation of the percentage of cytotoxicity allowed us to compute an IC50 value (compound concentration inducing 50% cytotoxicity).

4.3.2. Cell Viability and Proliferation Assays

The cytotoxicity of the compounds was also evaluated by measuring the number of viable cells in the culture with respect to the control culture (cells treated with DMSO only) using the RealTime-Glo™ MT Cell Viability Assay in the presence of varying concentrations of the compound. The assay is a nonlytic homogeneous, bioluminescent method that can be used to measure cell viability/proliferation in real time using NanoLuc® luciferase and a cell-permeant prosubstrate (MT Cell Viability Substrate). The potential cytotoxicity was tested by incubating Hela cells (1500 cells/well) in DMEM and in the presence of up to 128 µg/mL of the compound for up to 48 h.

4.3.3. Hemolytic Activity

The hemolytic activity of compounds 1, 2, 8, 9, 45, and 49 (0.025–10 μg) was investigated using the method described by Bechlars et al. [28]. Triton X-100 was used as the hemolysis control and DMSO as the negative control.

Author Contributions

Conceptualization, F.C., C.M. and J.-D.D.; methodology, F.S., A.B., R.D.P., M.A., T.S., C.P. and C.M.; validation, F.S., A.B., T.F., F.D.L., M.P., F.C., C.M. and J.-D.D.; writing—original draft preparation, F.S., F.C., C.M. and J.-D.D.; writing—review and editing, F.S., F.C., C.M. and J.-D.D.; funding acquisition, F.C. and J.-D.D. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge the partial financial support of the Italian MIUR “Progetto Dipartimenti di Eccellenza 2018–2022”, grant No. L. 232/2016.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Antibiotic Resistance. Available online: https://www.who.int/news-room/fact-sheets/detail/antibiotic-resistance (accessed on 24 October 2022).
  2. Ferri, M.; Ranucci, E.; Romagnoli, P.; Giaccone, V. Antimicrobial Resistance: A Global Emerging Threat to Public Health Systems. Crit. Rev. Food Sci. Nutr. 2017, 57, 2857–2876. [Google Scholar] [CrossRef] [PubMed]
  3. Cassini, A.; Högberg, L.D.; Plachouras, D.; Quattrocchi, A.; Hoxha, A.; Simonsen, G.S.; Colomb-Cotinat, M.; Kretzschmar, M.E.; Devleesschauwer, B.; Cecchini, M.; et al. Attributable Deaths and Disability-Adjusted Life-Years Caused by Infections with Antibiotic-Resistant Bacteria in the EU and the European Economic Area in 2015: A Population-Level Modelling Analysis. Lancet Infect. Dis. 2019, 19, 56–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Tacconelli, E.; Carrara, E.; Savoldi, A.; Harbarth, S.; Mendelson, M.; Monnet, D.L.; Pulcini, C.; Kahlmeter, G.; Kluytmans, J.; Carmeli, Y.; et al. Discovery, Research, and Development of New Antibiotics: The WHO Priority List of Antibiotic-Resistant Bacteria and Tu-berculosis. Lancet Infect. Dis. 2018, 18, 318–327. [Google Scholar] [CrossRef]
  5. Trebosc, V.; Gartenmann, S.; Tötzl, M.; Lucchini, V.; Schellhorn, B.; Pieren, M.; Lociuro, S.; Gitzinger, M.; Tigges, M.; Bumann, D.; et al. Dissecting Colistin Resistance Mechanisms in Extensively Drug-Resistant Acinetobacter baumannii Clinical Isolates. mBio 2019, 10, e01083-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Papp-Wallace, K.M. The Latest Advances in β-Lactam/β-Lactamase Inhibitor Combinations for the Treatment of Gram-Negative Bacterial Infections. Expert Opin. Pharmacother. 2019, 20, 2169–2184. [Google Scholar] [CrossRef] [PubMed]
  7. Docquier, J.-D.; Mangani, S. An Update on β-Lactamase Inhibitor Discovery and Development. Drug Resist. Updat. 2018, 36, 13–29. [Google Scholar] [CrossRef]
  8. Theuretzbacher, U.; Outterson, K.; Engel, A.; Karlén, A. The Global Preclinical Antibacterial Pipeline. Nat. Rev. Microbiol. 2020, 18, 275–285. [Google Scholar] [CrossRef] [Green Version]
  9. Melander, R.J.; Melander, C. The Challenge of Overcoming Antibiotic Resistance: An Adjuvant Approach? ACS Infect. Dis. 2017, 3, 559–563. [Google Scholar] [CrossRef]
  10. Laws, M.; Shaaban, A.; Rahman, K.M. Antibiotic Resistance Breakers: Current Approaches and Future Directions. FEMS Microbiol. Rev. 2019, 43, 490–516. [Google Scholar] [CrossRef] [Green Version]
  11. Van Giau, V.; An, S.S.A.; Hulme, J. Recent Advances in the Treatment of Pathogenic Infections Using Antibiotics and Nano-Drug Delivery Vehicles. Drug Des. Devel. Ther. 2019, 13, 327–343. [Google Scholar] [CrossRef]
  12. Mugnaini, C.; Sannio, F.; Brizzi, A.; Del Prete, R.; Simone, T.; Ferraro, T.; De Luca, F.; Corelli, F.; Docquier, J.-D. Screen of Unfocused Libraries Identified Compounds with Direct or Synergistic Antibacterial Activity. ACS Med. Chem. Lett. 2020, 11, 899–905. [Google Scholar] [CrossRef] [PubMed]
  13. Artico, M.; Massa, S.; Stefancich, G.; Corelli, F.; Ortenzi, G. Heterocyclic Systems. V. Synthesis of 1H,4H,6H-Pyrazolo[3,4-e]pyrrolo[2,1-c][1,4]oxazepine Derivatives. Heterocycles 1985, 23, 1417. [Google Scholar] [CrossRef]
  14. Massa, S.; Mai, A.; Artico, M.; Corelli, F. Heterocyclic System. XI. Synthesis of 1H,4H-Pyrazolo[4,3-b]pyrrolizine and 2H,4H-Pyrazolo[4,3-b]pyrrolizine Derivatives. J. Heterocycl. Chem. 1990, 27, 1805–1808. [Google Scholar] [CrossRef]
  15. Silvestri, R.; Cascio, M.G.; La Regina, G.; Piscitelli, F.; Lavecchia, A.; Brizzi, A.; Pasquini, S.; Botta, M.; Novellino, E.; Di Marzo, V.; et al. Synthesis, Cannabinoid Receptor Affinity, and Molecular Modeling Studies of Substituted 1-Aryl-5-(1H-pyrrol-1-yl)-1H-pyrazole-3-carboxamides. J. Med. Chem. 2008, 51, 1560–1576. [Google Scholar] [CrossRef] [PubMed]
  16. Tang, J.; Wang, B.; Wu, T.; Wan, J.; Tu, Z.; Njire, M.; Wan, B.; Franzblauc, S.G.; Zhang, T.; Lu, X.; et al. Design, Synthesis, and Biological Evaluation of Pyrazolo[1,5-a]pyridine-3-carboxamides as Novel Antitubercular Agents. ACS Med. Chem. Lett. 2015, 6, 814–818. [Google Scholar] [CrossRef] [Green Version]
  17. Cannatelli, A.; D’Andrea, M.M.; Giani, T.; Di Pilato, V.; Arena, F.; Ambretti, S.; Gaibani, P.; Rossolini, G.M. In vivo emergence of colistin resistance in Klebsiella pneumoniae producing KPC-type carbapenemases mediated by insertional inactivation of the PhoQ/PhoP mgrB regulator. Antimicrob. Agents Chemother. 2013, 57, 5521–5526. [Google Scholar] [CrossRef] [Green Version]
  18. D’Andrea, M.M.; Giani, T.; D’Arezzo, S.; Capone, A.; Petrosillo, N.; Visca, P.; Luzzaro, F.; Rossolini, G.M. Characterization of PABVA01, a Plasmid Encoding the OXA-24 Carbapenemase from Italian Isolates of Acinetobacter baumannii. Antimicrob. Agents Chemother. 2009, 53, 3528–3533. [Google Scholar] [CrossRef] [Green Version]
  19. Kelesidis, T.; Falagas, M.E. The Safety of Polymyxin Antibiotics. Expert Opin. Drug Saf. 2015, 14, 1687–1701. [Google Scholar] [CrossRef]
  20. Amaya-Villar, R.; Garnacho-Montero, J. How Should We Treat Acinetobacter Pneumonia? Curr. Opin. Crit. Care 2019, 25, 465–472. [Google Scholar] [CrossRef]
  21. Terminology Relating to Methods for the Determination of Susceptibility of Bacteria to Antimicrobial Agents. Clin. Microbiol. Infect. 2000, 6, 503–508. [CrossRef]
  22. Wahab Khan, M.; Uddin, M.K.; Ali, M.; Rahman, M.S.; Rashid, M.A.; Chowdhury, R. A Convenient Synthesis of New Annelated Pyrimidines and Their Biological Importance: Synthesis of Annelated Pyrimidines. J. Heterocycl. Chem. 2014, 51, E216–E221. [Google Scholar] [CrossRef]
  23. Christopher, A.; Takeru, E. Indane Derivatives and the Use Thereof as Soluble Guanylate Cyclase Activators. WIPO (PCT) Patent WO2017103888 (A1), 18 December 2018. [Google Scholar]
  24. Chen, H.; Wang, B.; Li, P.; Yan, H.; Li, G.; Huang, H.; Lu, Y. The Optimization and Characterization of Functionalized Sulfonamides Derived from Sulfaphenazole against Mycobacterium Tuberculosis with Reduced CYP 2C9 Inhibition. Bioorg. Med. Chem. Lett. 2021, 40, 127924. [Google Scholar] [CrossRef] [PubMed]
  25. Kopp, M.; Lancelot, J.-C.; Dallemagne, P.; Rault, S. Synthesis of Novel Pyrazolopyrrolopyrazines, Potential Analogs of Sildenafil. J. Heterocycl. Chem. 2001, 38, 1045–1050. [Google Scholar] [CrossRef]
  26. Approved Standard; Clinical and Laboratory Standards Institute (Eds.) Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically: M07-A10, 10th ed.; Documents/Clinical and Laboratory Standards Institute; Committee for Clinical Laboratory Standards: Wayne, PA, USA, 2015; ISBN 978-1-56238-987-1. [Google Scholar]
  27. Hall, M.J.; Middleton, R.F.; Westmacott, D. The Fractional Inhibitory Concentration (FIC) Index as a Measure of Synergy. J. Antimicrob. Chemother. 1983, 11, 427–433. [Google Scholar] [CrossRef] [PubMed]
  28. Bechlars, S.; Wüstenhagen, D.A.; Drägert, K.; Dieckmann, R.; Strauch, E.; Kubick, S. Cell-free synthesis of functional thermostable direct hemolysins of Vibrio parahaemolyticus. Toxicon 2013, 76, 132–142. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structure of hit compounds identified during a previous antibacterial screening campaign of an unfocused library [12].
Figure 1. Chemical structure of hit compounds identified during a previous antibacterial screening campaign of an unfocused library [12].
Antibiotics 11 01832 g001
Figure 2. Exploration of the chemical space around pyrazole-4-carboxamides.
Figure 2. Exploration of the chemical space around pyrazole-4-carboxamides.
Antibiotics 11 01832 g002
Figure 3. Exploration of the chemical space around pyrazole-3-carboxamides.
Figure 3. Exploration of the chemical space around pyrazole-3-carboxamides.
Antibiotics 11 01832 g003
Scheme 1. Synthesis of pyrazole-4-carboxamide derivatives 314. Reagents and conditions: (a) diethyl ethoxymethylenemalonate, EtOH, reflux; (b) 2,5-dimethoxytetrahydrofuran, glacial acetic acid, 120 °C; (c) p-tolylboronic acid, Pd(OAc)2, PPh3, 1 M Na2CO3, dioxane, reflux, N2 atmosphere, 4 h; (d) NaOH, H2O, reflux; (e) appropriate amine, HOBt, EDC, DCM, RT.
Scheme 1. Synthesis of pyrazole-4-carboxamide derivatives 314. Reagents and conditions: (a) diethyl ethoxymethylenemalonate, EtOH, reflux; (b) 2,5-dimethoxytetrahydrofuran, glacial acetic acid, 120 °C; (c) p-tolylboronic acid, Pd(OAc)2, PPh3, 1 M Na2CO3, dioxane, reflux, N2 atmosphere, 4 h; (d) NaOH, H2O, reflux; (e) appropriate amine, HOBt, EDC, DCM, RT.
Antibiotics 11 01832 sch001
Scheme 2. Synthesis of pyrazole-4-carboxamide derivatives 1519. Reagents and conditions: (a) LiOH, H2O/THF/MeOH, RT, 4 h; (b) (R)-alanine ethyl ester hydrochloride or (R)-valine ethyl ester hydrochloride, HOBt, EDC, DCM, RT.
Scheme 2. Synthesis of pyrazole-4-carboxamide derivatives 1519. Reagents and conditions: (a) LiOH, H2O/THF/MeOH, RT, 4 h; (b) (R)-alanine ethyl ester hydrochloride or (R)-valine ethyl ester hydrochloride, HOBt, EDC, DCM, RT.
Antibiotics 11 01832 sch002
Scheme 3. Synthesis of pyrazole-4-carboxamide derivatives 2022. Reagents and conditions: (a) Boc2O, DCM, RT, 2–4 h; (b) KOH, EtOH/THF, 55 °C, 1 h; (c) appropriate amine, HOBt, EDC, DCM, RT.
Scheme 3. Synthesis of pyrazole-4-carboxamide derivatives 2022. Reagents and conditions: (a) Boc2O, DCM, RT, 2–4 h; (b) KOH, EtOH/THF, 55 °C, 1 h; (c) appropriate amine, HOBt, EDC, DCM, RT.
Antibiotics 11 01832 sch003
Scheme 4. Synthesis of pyrazole-3-carboxamide derivatives 2340. Reagents and conditions: (a) appropriate amine, HOBt, EDC, DCM, RT; (b) NaOH, MeOH, H2O, reflux, 3 h.
Scheme 4. Synthesis of pyrazole-3-carboxamide derivatives 2340. Reagents and conditions: (a) appropriate amine, HOBt, EDC, DCM, RT; (b) NaOH, MeOH, H2O, reflux, 3 h.
Antibiotics 11 01832 sch004
Scheme 5. Synthesis of pyrazole-3-carboxamide derivatives 4149. Reagents and conditions: (a) appropriate aniline, HOBt, EDC, DCM, RT; (b) NaOH, MeOH, H2O, reflux, 3 h; (c) 4-aminophenol, PyBOP, HOBt, dry DMF, RT, 4 h; (d) (COCl)2, DCM, RT, 1 h, and then sulfanilamide, TEA, dry THF, RT, 2 h; (e) methyl chloroformate, TEA, DCM, RT, 1 h; (f) PhI(OAc)2, NH4OH, MeOH, RT, 2 h.
Scheme 5. Synthesis of pyrazole-3-carboxamide derivatives 4149. Reagents and conditions: (a) appropriate aniline, HOBt, EDC, DCM, RT; (b) NaOH, MeOH, H2O, reflux, 3 h; (c) 4-aminophenol, PyBOP, HOBt, dry DMF, RT, 4 h; (d) (COCl)2, DCM, RT, 1 h, and then sulfanilamide, TEA, dry THF, RT, 2 h; (e) methyl chloroformate, TEA, DCM, RT, 1 h; (f) PhI(OAc)2, NH4OH, MeOH, RT, 2 h.
Antibiotics 11 01832 sch005
Figure 4. Direct antibacterial activity of compounds 340, 42, 4447, and 49 (agar diffusion method) on a panel of eight representative bacterial species (type strains), including four Gram-negative and four Gram-positive organisms. (a) Pyrazole-4-carboxamide analogues (data for parent compound 1 are shown for comparison); (b) pyrazole-3-carboxamide analogues (data for parent compound 2 are shown for comparison) [12].
Figure 4. Direct antibacterial activity of compounds 340, 42, 4447, and 49 (agar diffusion method) on a panel of eight representative bacterial species (type strains), including four Gram-negative and four Gram-positive organisms. (a) Pyrazole-4-carboxamide analogues (data for parent compound 1 are shown for comparison); (b) pyrazole-3-carboxamide analogues (data for parent compound 2 are shown for comparison) [12].
Antibiotics 11 01832 g004
Figure 5. Isobolograms showing the strong synergistic activity of compounds 8 (top) and 9 (bottom) with colistin on A. baumannii reference strain (ATCC 17978) (left) and an MDR, colistin-resistant, and carbapenem-resistant A. baumannii clinical isolate (N50) (right).
Figure 5. Isobolograms showing the strong synergistic activity of compounds 8 (top) and 9 (bottom) with colistin on A. baumannii reference strain (ATCC 17978) (left) and an MDR, colistin-resistant, and carbapenem-resistant A. baumannii clinical isolate (N50) (right).
Antibiotics 11 01832 g005
Figure 6. Time-dependent kill-curve analysis of the MDR, colistin-resistant A. baumannii clinical isolate (N50) in the presence 2 µg/mL of colistin (squares), 2 µg/mL of colistin, and 8 µg/mL of 9 (circles). The growth control is shown as triangles (similar results were obtained in the presence of 8 µg/mL of 9, as this compound alone does not exhibit antibacterial activity).
Figure 6. Time-dependent kill-curve analysis of the MDR, colistin-resistant A. baumannii clinical isolate (N50) in the presence 2 µg/mL of colistin (squares), 2 µg/mL of colistin, and 8 µg/mL of 9 (circles). The growth control is shown as triangles (similar results were obtained in the presence of 8 µg/mL of 9, as this compound alone does not exhibit antibacterial activity).
Antibiotics 11 01832 g006
Table 1. Identification of compounds exhibiting synergistic activity on Gram-negative bacteria (Eco, Escherichia coli CCUGT; Kpn, Klebsiella pneumoniae ATCC 13833; Pae, Pseudomonas aeruginosa ATCC 27853; Aba, Acinetobacter baumannii ATCC 17978) with colistin (0.5 × MIC). A green square indicates the absence of growth under these conditions.
Table 1. Identification of compounds exhibiting synergistic activity on Gram-negative bacteria (Eco, Escherichia coli CCUGT; Kpn, Klebsiella pneumoniae ATCC 13833; Pae, Pseudomonas aeruginosa ATCC 27853; Aba, Acinetobacter baumannii ATCC 17978) with colistin (0.5 × MIC). A green square indicates the absence of growth under these conditions.
Cpd aSynergistic Activity (Growth Inhibition) b
EcoKpnPaeAba
3
4
5
6
7
8
9
10
11 - c
12
13
14
15 -
16
17
18- -
19- -
20
21---
22
23
24
25
26
27
28
29
30
31
32---
33---
34---
35
36
37
38
39
40
42
44
45
46
47
49
aCompounds tested at 64 µg/mL, 1% DMSO was used as the negative control (no growth inhibition observed); bcolistin concentration was 0.12 µg/mL (Eco and Kpn) or 0.25 µg/mL (Pae and Aba), corresponding to 0.5 × MIC; c-, not determined due to limited solubility of the compound in these conditions.
Table 2. MAC (minimum antibacterial concentration) values of selected compounds on representative Gram-negative bacteria (Eco, Escherichia coli CCUGT; Kpn, Klebsiella pneumoniae ATCC 13833; Aba, Acinetobacter baumannii ATCC 17978), when tested in the presence of 0.5 × MIC colistin. The minimal ratio between the MIC of the compound alone and the MAC in the presence of colistin is given between parentheses and used to quantify the improvement of the antibacterial activity of the tested compounds when tested in the presence of colistin. Data for parent compounds 1 and 2 are provided for comparison [12].
Table 2. MAC (minimum antibacterial concentration) values of selected compounds on representative Gram-negative bacteria (Eco, Escherichia coli CCUGT; Kpn, Klebsiella pneumoniae ATCC 13833; Aba, Acinetobacter baumannii ATCC 17978), when tested in the presence of 0.5 × MIC colistin. The minimal ratio between the MIC of the compound alone and the MAC in the presence of colistin is given between parentheses and used to quantify the improvement of the antibacterial activity of the tested compounds when tested in the presence of colistin. Data for parent compounds 1 and 2 are provided for comparison [12].
CompoundMAC (µg/mL) (MIC/MAC Ratio)
EcoKpnAba
164 (8)64 (8)32 (16)
832 (16)- a4 (128)
98 (64)8 (64)4 (128)
1364 (8)32 (16)-
20-64 (8)64 (8)
22-64 (8)64 (8)
264 (8)32 (16)16 (32)
2732 (16)32 (16)-
2864 (8)64 (8)64 (8)
3132 (16)16 (32)32 (16)
39--32 (16)
454 (128)4 (128)8 (64)
498 (64)4 (128)8 (64)
aNo synergistic activity.
Table 3. Chequerboard analysis and average FIC index (FICI) values determined via the combination of colistin and pyrazolo-4-carboxamides 1, 8, and 9 and pyrazolo-3-carboxamides 2, 45, and 49 on E. coli, K. pneumoniae, and A. baumannii strains, including colistin-resistant clinical isolates.
Table 3. Chequerboard analysis and average FIC index (FICI) values determined via the combination of colistin and pyrazolo-4-carboxamides 1, 8, and 9 and pyrazolo-3-carboxamides 2, 45, and 49 on E. coli, K. pneumoniae, and A. baumannii strains, including colistin-resistant clinical isolates.
Bacterial StrainAverage FIC Index
18924549
E. coli CCUGT0.50.380.330.310.310.32
K. pneumoniae ATCC 138330.50.50.320.50.310.39
K. pneumoniae SI-004Bo a111111
A. baumannii ATCC 179780.380.310.340.390.310.38
A. baumannii N50 b0.50.240.230.40.260.31
a Klebsiella pneumoniae exhibiting a pandrug-resistant phenotype (colistin MIC, 64 μg/mL) [17]. bA multidrug-resistant (MDR) Acinetobacter baumannii clinical isolate (colistin MIC, 32 μg/mL) [18].
Table 4. Cytotoxicity (membrane-damaging activity) of compounds 8 and 9, tested alone or in the presence of 512 µg/mL of colistin.
Table 4. Cytotoxicity (membrane-damaging activity) of compounds 8 and 9, tested alone or in the presence of 512 µg/mL of colistin.
CompoundIC50 (μg/mL)
Colistin>256
8>256
8 + colistin16.4
9>256
9 + colistin>256
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sannio, F.; Brizzi, A.; Del Prete, R.; Avigliano, M.; Simone, T.; Pagli, C.; Ferraro, T.; De Luca, F.; Paolino, M.; Corelli, F.; et al. Optimization of Pyrazole Compounds as Antibiotic Adjuvants Active against Colistin- and Carbapenem-Resistant Acinetobacter baumannii. Antibiotics 2022, 11, 1832. https://doi.org/10.3390/antibiotics11121832

AMA Style

Sannio F, Brizzi A, Del Prete R, Avigliano M, Simone T, Pagli C, Ferraro T, De Luca F, Paolino M, Corelli F, et al. Optimization of Pyrazole Compounds as Antibiotic Adjuvants Active against Colistin- and Carbapenem-Resistant Acinetobacter baumannii. Antibiotics. 2022; 11(12):1832. https://doi.org/10.3390/antibiotics11121832

Chicago/Turabian Style

Sannio, Filomena, Antonella Brizzi, Rosita Del Prete, Marialuce Avigliano, Tiziana Simone, Carlotta Pagli, Teresa Ferraro, Filomena De Luca, Marco Paolino, Federico Corelli, and et al. 2022. "Optimization of Pyrazole Compounds as Antibiotic Adjuvants Active against Colistin- and Carbapenem-Resistant Acinetobacter baumannii" Antibiotics 11, no. 12: 1832. https://doi.org/10.3390/antibiotics11121832

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