Next Article in Journal
Terminal Residue and Dietary Risk Assessment of Atrazine and Isoxaflutole in Corn Using High-Performance Liquid Chromatography–Tandem Mass Spectrometry
Next Article in Special Issue
Dimethyl Bisphenolate Ameliorates Carbon Tetrachloride-Induced Liver Injury by Regulating Oxidative Stress-Related Genes
Previous Article in Journal
Analysis of Volatile Compounds in Processed Cream Cheese Models for the Prediction of “Fresh Cream” Aroma Perception
Previous Article in Special Issue
3-Ferrocenyl-estra-1,3,5 (10)-triene-17-one: Synthesis, Crystal Structure, Hirshfeld Surface Analysis, DFT Studies, and Its Binding to Human Serum Albumin Studied through Fluorescence Quenching and In Silico Docking Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Pyrrole-Based Enaminones as Building Blocks for the Synthesis of Indolizines and Pyrrolo[1,2-a]pyrazines Showing Potent Antifungal Activity

by
Diter Miranda-Sánchez
1,
Carlos H. Escalante
1,
Dulce Andrade-Pavón
2,3,
Omar Gómez-García
1,
Edson Barrera
1,
Lourdes Villa-Tanaca
3,
Francisco Delgado
1 and
Joaquín Tamariz
1,*
1
Departamento de Química Orgánica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Mexico City 11340, Mexico
2
Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu S/N, Mexico City 07738, Mexico
3
Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Mexico City 11340, Mexico
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(20), 7223; https://doi.org/10.3390/molecules28207223
Submission received: 8 September 2023 / Revised: 9 October 2023 / Accepted: 10 October 2023 / Published: 23 October 2023
(This article belongs to the Special Issue Organic Synthesis and Application of Bioactive Molecules)

Abstract

:
As a new approach, pyrrolo[1,2-a]pyrazines were synthesized through the cyclization of 2-formylpyrrole-based enaminones in the presence of ammonium acetate. The enaminones were prepared with a straightforward method, reacting the corresponding alkyl 2-(2-formyl-1H-pyrrol-1-yl)acetates, 2-(2-formyl-1H-pyrrol-1-yl)acetonitrile, and 2-(2-formyl-1H-pyrrol-1-yl)acetophenones with DMFDMA. Analogous enaminones elaborated from alkyl (E)-3-(1H-pyrrol-2-yl)acrylates were treated with a Lewis acid to afford indolizines. The antifungal activity of the series of substituted pyrroles, pyrrole-based enaminones, pyrrolo[1,2-a]pyrazines, and indolizines was evaluated on six Candida spp., including two multidrug-resistant ones. Compared to the reference drugs, most test compounds produced a more robust antifungal effect. Docking analysis suggests that the inhibition of yeast growth was probably mediated by the interaction of the compounds with the catalytic site of HMGR of the Candida species.

Graphical Abstract

1. Introduction

Pyrrolo[1,2-a]pyrazines and indolizines, including their partial or complete saturated analogues, belong to the diverse family of pyrrole-fused aza-bridged heterocyclic compounds, which are abundant in nature. They are known for their strong pharmacological activity. Pyrrolo[1,2-a]pyrazines and structural analogues show a wide range of biological effects [1,2], as illustrated by reports on their anxiolytic [3], anticancer [4,5], and antifungal activity [1,6], as well as their use as insect feeding deterrents [7] and potent and selective non-competitive antagonists of mGluR5 [8]. Antibiotic activity has only been found for the hexahydropyrrolo[1,2-a]pyrazine-1,4-dione analogues, which provides a strong effect against gram-negative bacteria [9,10,11]. Indolizines, on the other hand, have potential as anticancer, herbicidal, anti-inflammatory, antiparasitic, antioxidant, antihistaminic, anticonvulsant, antiviral, and analgesic agents [12,13,14,15,16,17]. In contrast to pyrrolo[1,2-a]pyrazines, antibiotic activity is more widely distributed among indolizines [12,13], in particular against Mycobacterium tuberculosis [18,19]. However, the antifungal effect of indolizines is modest [12]. Although the mechanism of action is not clearly established for either heterocycle, it is probably associated with the nitrogen-bridged heterocyclic scaffold [1,12].
Although pyrrolo[1,2-a]pyrazines have received less attention from researchers than indolizines, their broad spectrum of activity has stimulated the discovery of versatile synthetic strategies [1,2]. One of the most efficient approaches involves pyrrole-containing substrates, which undergo a cyclization process to build the pyrazine ring [20,21,22,23,24] (Scheme 1a). Indolizines have been prepared by following an analogous methodology, starting from functionalized pyrroles as the building block and completing the synthesis by a proper annulation to build the six-membered ring [25,26,27,28,29] (Scheme 1b). Our group has recently described effective procedures for constructing the scaffold of both indolizines and pyrrolo[1,2-a]pyrazines by appropriately substituting pyrrole-based precursors [30] (Scheme 1c).
The capacity of indolizines [31] and pyrrolo[1,2-a]pyrazines [1,6] to treat drug-resistant fungal infections is particularly attractive. Due to the pharmacological value of these heterocycles, the development of short and highly efficient synthetic methods should be a priority.
The aim of the current contribution was to design and test new synthetic approaches for synthesizing pyrrolo[1,2-a]pyrazines and indolizines. For the elaboration of pyrrolo[1,2-a]pyrazines, the potential of the annulation of 2-formylpyrrole-based enaminones (13) in the presence of ammonium acetate was evaluated. On the other hand, analogous enaminones were exposed to Lewis acids to prepare indolizines (Scheme 1d). All precursors and the corresponding products were assessed for their in vitro antifungal effect on six strains of Candida species. The most active compounds were analyzed in silico by docking simulations to examine their pharmacological profile as antifungal agents at the active site of the 3-hydroxy-methyl-glutaryl-CoA reductase (HMGR) enzyme of the six Candida spp. HMGR is the rate-limiting enzyme for the formation of ergosterol in Candida species, which is generated through the mevalonate pathway. Ergosterol is an essential component in the yeast cell membrane.

2. Results and Discussion

2.1. Chemistry

The design of pyrrole-containing compounds as potential antifungal agents was based on the evidence that benzofuran [32,33], benzothiophene [34], and indole [35,36] frames (including those of several natural products) can be built through intramolecular cyclization of 2-phenoxy-, 2-thiophenoxy-, and 2-anilino-3-(dimethylamino)propenoates, respectively. The latter derivatives have generically been called enaminones and proven to be versatile substrates in heterocyclic chemistry [37,38].
In the current investigation, the nitrogen atom of the 2-formylpyrrole ring was incorporated into the C-2 position of the alkyl (Z)-3-(dimethylamino)propenoate-equivalent (1, Y = O, Z = CO2R, Scheme 1d), the (Z)-1-aryl-3-(dimethylamino)prop-2-en-1-one-equivalent (2, Y = O, Z = COAr, Scheme 1d), and the (Z)-3-(dimethylamino)acrylonitrile-equivalent (3, Y = O, Z = CN) (Scheme 1d), followed by the cyclization process in the presence of ammonium acetate to provide the corresponding pyrrolo[1,2-a]pyrazines 4. It was assumed that 2-formylpyrrole (6) can serve as a building block for the divergent synthesis of substituted pyrrolizines [30,39] and pyrrolo[1,2-a]pyrazines [30] to furnish the desired pyrrole-fused aza-bridged heterocyclic derivatives.
Thus, the alkylation of 6 with alkyl bromoacetates 7a and 7b under basic conditions produced the series of N-substituted pyrroles 8a [40] and 8b in good yields (Scheme 2). Similarly, the reaction of 6 with the series of bromoacetophenones 7cf generated 8cf, and the reaction of 6 with bromoacetonitrile (7g) gave pyrrole 8g. The thermal treatment of 8ag with N,N-dimethylformamide dimethyl acetal (DMFDMA) afforded the series of enaminones 1ab and 2ad in acceptable yields as a single Z isomer, as well as enaminone 3a as an inseparable mixture of Z/E (54:46) isomers (Scheme 2).
For the conversion of 2-formylpyrrole-based enaminones 1ab, 2ad, and 3a into the series of 4-substituted pyrrolo[1,2-a]pyrazines 4ag, a combination of ammonium acetate (as the source of nitrogen) and DBU (as the base) was employed as previously described [30]. However, the yields were low. Similar low yields were obtained with sodium hydride, potassium tert-butoxide, or cesium carbonate as the base. The reaction was improved when applying lithium carbonate as the base and DMF as the solvent at variable temperatures and reaction times (Table 1).
As an alternative route, a multicomponent reaction was explored for the preparation of pyrrolo[1,2-a]pyrazines 4, starting from substituted pyrroles 8a, 8c, and 8g in the presence of DMFDMA and ammonium acetate (Scheme 3). Thus, pyrroles 8a and 8c were reacted with an excess of DMFDMA (5.0 mol eq.) and ammonium acetate (3.0 mol eq.) to provide the corresponding pyrrolo[1,2-a]pyrazines 4a and 4c in 57% and 80% yields, respectively. When comparing this one-step process to that of the previously described two-step procedure, the current overall yield was lower for 4a (57% vs. 74%) and higher for 4c (80% vs. 46%). The reduction of 4c with NaBH4 resulted in alcohol 4h in moderate yield. The reaction of 8g with DMFDMA generated a complex mixture of products that included the starting material.
Considering the important biological properties of brominated pyrroles [40,41] and pyrrolo[1,2-a]pyrazines [5], the brominated derivatives 8h [40], 1c, and 4i were conceived as potential antifungal compounds. The bromination of 8a with NBS (1.0 mol eq.) afforded 8h as a single regioisomer in high yield (Scheme 3). Treatment of the latter with DMFDMA gave enaminone 1c, which was reacted with ammonium acetate to deliver 7-bromopyrrolo[1,2-a]pyrazine 4i in good yield.
In the same context, pyrrolo[1,2-a]pyrazine 4a was brominated with an equimolar quantity of NBS to yield 6-bromopyrrolo[1,2-a]pyrazine 4j (Scheme 4), and the reaction of 4a with two mol equivalents of NBS furnished 6,7-dibromopyrrolo[1,2-a]pyrazine 4k.
With the aim of expanding the scope of the methodology, the synthesis of 4,6-disubstituted pyrrolo[1,2-a]pyrazine 4l was undertaken. Starting from 6, alkyl 3-(pyrrol-2-yl)acrylate 9a was obtained by a Horner–Wadsworth–Emmons reaction, followed by N-alkylation with 7a to give pyrrole 10a, as reported [42] (Scheme 5). The latter was formylated under the usual Vilsmeier–Haack conditions to give rise to the respective 5-formyl analog 8i. Treatment of 8i with DMFDMA generated a complex mixture of products, from which it was possible to isolate the single Z isomer of enaminone 11 in low yield. Finally, the amination of the latter with ammonium acetate delivered the desired pyrrolo[1,2-a]pyrazine 4l in modest yield, in contrast to the high yields for 4ag (Table 1).
It was decided to start with similar enaminones to test a new approach for the construction of indolizines. Therefore, 9a or 9b were reacted with the corresponding methyl or ethyl bromoacetates 7a and 7b under basic conditions to afford pyrroles 10ac [42], which were reacted with DMFDMA to convert them into enaminones 12ac (Scheme 6). Unfortunately, enaminone 12b was formed in a mixture of methyl esters as a result of transesterification with the methoxy ions in the reaction medium. Hence, this compound was more efficiently obtained in modest yield when using Bredereck’s reagent under MW irradiation.
With the aim of achieving the direct annulation of 12ac to the corresponding indolizines 5ac, the reaction was promoted by diverse Lewis acids. By treating 12a with ZnI2 or CuCl in methylene chloride as the solvent at 0 °C, the starting material was recovered. An increase in the temperature led to the decomposition of the reaction mixture. When utilizing POCl3 in DMF at 0 °C, the yield of 5a was 28%, and the conversion of 12b to 5b with this Lewis acid was insignificant. The transformation of 12c to 5c with iodine [36] provided a low yield (18%). Finally, a 1.0 M nitrobenzene solution of AlCl3 in methylene chloride as the solvent at room temperature efficiently furnished the series 5ac in variable yields (Scheme 6). Disappointingly, when this strategy was started by reacting pyrrole 9a with bromoacetophenones 7cf, the conversion into the corresponding N-alkyl pyrroles did not proceed efficiently. The structure of each product obtained in all the synthetic schemes was fully established by 1H and 13C NMR, assisted by 2D experiments (ROESY, HSQC, and HMBC) and HRMS.

2.2. Antifungal Activity

According to previous studies, brominated and non-brominated pyrrole-containing compounds structurally related to derivatives 8a, 8h, and 1c inhibit yeast growth and the synthesis of ergosterol by Candida glabrata [40]. Therefore, selected compounds of the present series, consisting of pyrrole-based compounds (8ai and 10ac), enaminones (1ac, 2ad, 11, and 12ac), pyrrolo[1,2-a]pyrazines (4al), and indolizines (5ac), were assessed at their minimum inhibitory concentration (MIC) against six Candida spp.: C. albicans, C. glabrata, C. dubliniensis, C. krusei, C. auris, and C. haemulonii. The latter two species are multidrug-resistant. Most of the compounds evaluated afforded substantial yeast growth inhibition. Table 2 summarizes the lowest MIC50–70 values in regard to the inhibition of the Candida spp. by the test compounds and reference drugs (fluconazole, simvastatin, and atorvastatin). Simvastatin and atorvastatin are well-known inhibitors of human HMGR (hHMGR) and are proposed as inhibitors of HMGR of Candida spp. [40,43,44]. Fluconazole is a reference inhibitor of the enzyme C-14 alpha demethylase of lanosterol (CYP51) that participates in the synthesis of ergosterol [45]. Interestingly, simvastatin and atorvastatin showed a more efficient profile than fluconazole for some of the yeasts, including the two multidrug-resistant species.
Inhibition of yeast growth was observed for all the test compounds, which had lower MICs than the three reference drugs in all cases. Nevertheless, the majority of the MIC50–70 values for the compounds on C. auris and C. haemulonii were higher than those obtained on C. albicans, C. glabrata, C. dubliniensis, and C. krusei. This can perhaps be explained by the strong resistance of C. auris and C. haemulonii to a variety of traditional antifungal drugs [46,47].
Pyrrole derivatives are reported to inhibit the growth of C. albicans, C. glabrata, C. parapsilosis, C. tropicalis, Cryptococcus neoformans, Saccharomyces cerevisiae, Aspergillus fumigatus, A. niger, Geotrichum candidum, Syncephalastrum racemosum, and dermatophytes (Trichophyton rubrum, T. mentagrophytes, and Microsporum gypseum), exhibiting in almost all the cases MICs higher than those determined presently [48,49,50,51,52]. These results suggest that the N-alkyl group attached to the pyrrole ring of derivatives 8ag improves the antifungal effect (especially for N-alkyl 2-formylpyrroles 8a, 8c, and 8g), as seems to be demonstrated for the pyrrole-based drug atorvastatin. Low to modest antifungal activity has also been observed for pyrrole- and/or pyridine-containing fused-bis-heterocyclic compounds [53,54,55].
The few reports on enaminone-containing compounds have evidenced their antibacterial activity against Escherichia coli, Bacillus subtilis, and Salmonella typhi [56] as well as antifungal activity against C. albicans, in each case showing a modest inhibition [57]. In the current contribution, the enaminones 1a, 1c, 2ad, 11, and 12ac exhibited good inhibition of the six Candida spp. (Table 2).
Pyrrole-based compounds 8i and 1012 bear the acrylate moiety as another potentially active pharmacophore group, as does pyrrolo[1,2-a]pyrazine 4l, one of the most active derivatives against the six Candida species. Some acrylate-based heterocyclic derivatives have been proposed as antifungals due to their growth inhibition of Aspergillus, Geotrichum, and various other pathogenic plant fungi [58,59].
Pyrrolo[1,2-a]pyrazines 4al were highly active agents against all the Candida spp. herein tested, particularly derivatives 4b, 4g, and 4l. The conjugated C-4 carbonyl group does not seem to be a key pharmacophore, given that the antifungal activity is comparable for alcohol 4h and precursor 4c. Moderate activity was previously found for analogous heterocycles against Penicillium chrysogenum AUMC 530-15, four species of Aspergillus, Fusarium solani AUMC 2690-6, and Trichothecium roseum AUMC 7410-2 [60]. Interestingly, the brominated pyrrolo[1,2-a]pyrazines 4ik were more active than their corresponding non-halogenated 4a.
The indolizines 5ac also significantly inhibited the growth of the majority of the Candida spp., in agreement with recent reports on other analogous compounds [61]. However, these results are in contrast with a series of 7-cyano-1,2-diphenylindolizines, which were not active on C. krusei [18].

2.3. Molecular Docking Study

Molecular docking is a computational tool used to explore the interaction, binding mode (e.g., protein–protein), and affinity between two molecules, generally an organic compound and an endogenous receptor (protein or nucleic acid) of a cell [62,63,64]. Because docking simulations serve to explore the molecular recognition between the active site of a certain therapeutic target (an enzyme) and an organic molecule (ligand), they are widely used to design and develop new drugs that may improve the treatment for diseases or for infections by microorganisms, such as a pathogenic fungus. The binding interaction between two molecules depends on the hydrophilic, electrostatic, and hydrophobic intermolecular forces that take place. The strength of such interactions is expressed in binding energy (ΔG, kcal/mol). A more negative value of ΔG is related to a more stable enzyme–ligand complex and a greater biological activity.
To explore the binding interactions of the test compounds on HMGR, the following compounds were selected among the most active derivatives: enaminone-containing pyrroles 1a, 2a, and 2c, pyrrolo[1,2-a]pyrazines 4b, 4g, and 4l, indolizine 5a, N-alkyl 2-formylpyrroles 8a, 8c, and 8g, and pyrrole-based alkyl acrylates 10a and 12a. The molecular docking study was carried out on the active site of the HMGR enzyme of C. albicans (CaHMGR), C. glabrata (CgHMGR), C. dubliniensis (CdHMGR), C. krusei (CkHMGR), C. auris (CauHMGR), and C. haemulonii (ChaHMGR). The models of the six HMGRs were built (Figure S77, Supplementary Materials) and overlapped with human HMGR, noting a close structural identity between the 3D structures. The high quality of the models employed in the study is evidenced by the fact that over 90% of the amino acid residues were in permitted areas of the Ramachandran diagram (Figures S78–S83, Supplementary Materials), as previously reported for CgHMGR [40,44,65].
The binding energy values of the reference compound atorvastatin were in all cases lower than those for simvastatin (Table 3). A similar pattern of binding energy values was observed for the entire series of compounds. For example, enaminones 2a and 2c and the N-alkyl pyrrole 8c showed more negative values than simvastatin for all the species of Candida spp. herein tested. The pyrrolo[1,2-a]pyrazine 4l had better binding energy values than simvastatin on five of the yeasts. In contrast to the experimental antifungal effect, enaminone 1a only exhibited better binding energy values than simvastatin on CaHMGR and CkHMGR. Overall, the binding energy values found for these series of compounds were better than those previously described for other derivatives proposed as inhibitors of CgHMGR [40,44].
To predict the binding mode of the twelve selected compounds at the active site of the HMGRs, the three most prevalent species (C. albicans, C. glabrata, and C. auris) were selected. The hydrophilic and hydrophobic interactions and the amino acid residues involved are summarized in Table 4 for C. albicans, and in Tables S1 and S2 for C. glabrata, and C. auris, respectively. Likewise, the interactions are illustrated in Figure 1 for C. albicans and in Figures S84 and S85 for C. glabrata and C. auris. Regarding their binding modes to the HMGRs of the three Candida spp., the twelve test compounds share key hydrophilic and hydrophobic interactions with simvastatin and atorvastatin. The residues participating in binding are the following: Glu96, Lys228, and Asp304 in CaHMGR; Glu93, Lys227, and Asp303 in CgHMGR; and Glu96, Lys229, and Asp305 in CauHMGR. Considering the binding interactions of either of the test compounds, at least one of these residues is shared with the binding interactions of the reference compounds, suggesting that all the test compounds and simvastatin and atorvastatin may have a similar mechanism of action. These types of interactions have been previously reported in other works for the HMGR of H. sapiens and CgHMGR [40,44,66].
Hydrophilic interactions involving a conventional hydrogen bond are predominant for the carbonyl groups of compounds 1a, 2a, 2c, and 8a with the amino acids Asp227, Lys228, and Asp304 of CaHMGR as well as with the amino acids Thr92 and Glu93 of CgHMGR. Similar interactions are observed for indolizine 5a and enaminone 13a with residues Gln303, Asn195, and Thr295 of CaHMGR. For more than one of the compounds assessed on CauHMGR, the principal interactions are both conventional hydrogen bond and carbon hydrogen with Thr94, Thr95, Lys229, Asn292, Thr295, and Asp305. In addition, the amino acids that predominate in the hydrophobic interactions with CaHMGR are Glu96 (π-anion), Met192 (π-sigma or π-alkyl), and Met194 (π-sigma or π-alkyl). Compounds 2a and 8g show π-alkyl interactions with Met189 in CgHMGR, and 1a, 2a, 2c, 4l, 8c, and simvastatin display π-alkyl interactions with Pro306 in CauHMGR. The presence of the aromatic rings in the test compounds plays an important role in stabilizing the p interactions with the enzyme residues. According to the results, the test compounds can be considered as inhibitors of the HMGR enzymes of the Candida spp. herein evaluated, because they bind to amino acid residues of the catalytic site and share certain hydrophilic and hydrophobic interactions with simvastatin and atorvastatin.

3. Materials and Methods

3.1. General Information

Melting points were determined on a Krüss KSP 1N (KRÜSS GmbH, Hamburg, Germany) capillary melting point apparatus. IR spectra were recorded on FT-IR 2000 Perkin-Elmer (PerkinElmer, Waltham, MA, USA) and Bruker Vertex 70 (ATR-FT) (Bruker Corporation, Billerica, MA, USA) spectrophotometers. 1H and 13C NMR spectra were recorded on the following instruments: Varian Mercury (300 MHz) (Varian, Inc., Palo Alto, CA, USA), Bruker Ascend 400 (400 MHz) (Bruker Corporation, Billerica, MA, USA), Varian VNMR System (500 MHz) (Varian, Inc., Palo Alto, CA, USA), Bruker 600Avance III (600 MHz), and Bruker Avance III HD (750 MHz) (Bruker Corporation, Billerica, MA, USA), with CDCl3 as the solvent and TMS or CDCl3 as the internal standards. Signal assignments were based on 2D NMR spectra (HSQC, HMBC, and ROESY). High-resolution mass spectra (HRMS) (in electron impact mode) were obtained on Jeol JMS-GCMateII and Jeol JMS T100-LC AccuTOF DART (JEOL, Ltd., Tokyo, Japan) spectrometers. MW irradiation was emitted from a CEM MW reactor (CEM Corporation, Matthews, NC, USA). A Multi-Therm Benchmark, Model H5000-HC (Benchmark Scientific, Inc., Sayreville, NJ, USA) was utilized as a heating and cooling shaker in enzymatic stability assays. Yeast growth was quantified in a Multiskan™ GO microplate spectrophotometer (Thermo Fischer Scientific, Waltham, MA, USA) at 620 nm. Analytical thin-layer chromatography was carried out on 0.25 plates coated with silica gel 60 F254 (E. Merck, Darmstadt, Germany), which were visualized by a long- and short-wavelength UV lamp. Flash column chromatography was performed over silica gel (230–400 mesh, Natland International Co., Morrisville, NC, USA). Commercial reagents were used as received, and anhydrous solvents were obtained by a distillation process. Compounds 1a and 8a, 9a, 9b, and 10b were prepared as reported [40,42].

3.2. Chemistry

  • Ethyl 2-(2-formyl-1H-pyrrol-1-yl) acetate (8b). At 0 °C and under N2 atmosphere, NaH (60%, 0.051 g, 1.27 mmol) was added to a solution of 6 (0.100 g, 1.05 mmol) and anhydrous DMF (2.0 mL), and the mixture was stirred for 20 min. Then, 7b (0.210 g, 1.26 mmol) was added, and the solution was stirred at room temperature (rt) for 4 h. The reaction mixture was extracted with hexane/EtOAc (1:1, 15.0 mL). The organic layer was washed with brine (5.0 mL × 3) and dried (Na2SO4), and the solvent was removed under vacuum. The residue was purified by column chromatography over silica gel (hexane/EtOAc, 9:1) to obtain 8b (0.179 g, 94%) as a pale violet oil. Rf 0.64 (hexane/EtOAc, 1:1). IR (film): ῡ 3130, 2993, 2978, 2798, 1735, 1656, 1482, 1402, 1315, 1218, 1027, 752 cm−1. 1H NMR (400 MHz, CDCl3): δ 1.28 (t, J = 7.2 Hz, 3H, CO2CH2CH3), 4.22 (q, J = 7.2 Hz, 2H, CO2CH2CH3), 5.05 (s, 2H, CH2), 6.29 (dd, J = 4.0, 2.4 Hz, 1H, H-4′), 6.90–6.93 (m, 1H, H-5′), 6.98 (dd, J = 4.0, 1.6 Hz, 1H, H-3′), 9.52 (d, J = 0.8 Hz, 1H, CHO). 13C NMR (100 MHz, CDCl3): δ 14.1 (CO2CH2CH3), 50.2 (CH2), 61.6 (CO2CH2CH3), 110.2 (C-4′), 124.6 (C-3′) 131.7 (C-2′), 132.0 (C-5′), 168.3 (CO2CH2CH3), 179.7 (CHO). HRMS (EI): m/z [M+] calcd. for C9H11NO3: 181.0739; found: 181.0733.
  • 1-(2-Oxo-2-phenylethyl)-1H-pyrrole-2-carbaldehyde (8c). Following the method described for 8b, a mixture of 6 (0.100 g, 1.05 mmol), NaH (60%, 0.505 g, 1.26 mmol), and 7c (0.251 g, 1.26 mmol) furnished 8c (0.200 g, 89%) as white needles. Rf 0.19 (hexane/EtOAc, 9:1); mp 114–115 °C. IR (film): ῡ 3112, 2938, 2812, 1702, 1648, 1402, 1366, 1330, 1222, 1080, 1019, 1005, 748, 690 cm−1. 1H NMR (300 MHz, CDCl3): δ 5.81 (s, 2H, CH2), 6.36 (dd, J = 4.2, 2.5 Hz, 1H, H-4), 6.94–6.98 (m, 1H, H-5), 7.04 (dd, J = 4.2, 1.5 Hz, 1H, H-3), 7.48–7.55 (m, 2H, H-3″), 7.59–7.67 (m, 1H, H-4″), 7.98–8.03 (m, H-2″), 9.51 (d, J = 0.9 Hz, 1H, CHO). 13C NMR (187.5 MHz, CDCl3): δ 54.7 (CH2), 110.3 (C-4), 124.7 (C-3), 128.0 (C-2″), 128.9 (C-3″), 131.6 (C-2), 132.5 (C-5), 133.8 (C-4″), 134.8 (C-1″), 179.8 (CHO), 192.9 (CO). HRMS (EI): m/z [M+] calcd. for C13H11NO2: 213.0790; found: 213.0790.
  • 1-(2-(3-Methoxyphenyl)-2-oxoethyl)-1H-pyrrole-2-carbaldehyde (8d). Following the method described for 8b, a mixture of 6 (0.060 g, 0.63 mmol), NaH (60%, 0.030 g, 0.76 mmol), and 7c (0.174 g, 0.76 mmol) afforded 8d (0.070 g, 45%) as white needles. Rf 0.48 (hexane/EtOAc, 1:1); mp 106–107 °C. IR (KBr): ῡ 2939, 2799, 1694, 1649, 1591, 1403, 1260, 1192, 1024, 857, 754 cm−1. 1H NMR (750 MHz, CDCl3): δ 3.85 (s, 3H, CH3O), 5.79 (s, 2H, CH2), 6.35 (dd, J = 3.9, 2.6 Hz, 1H, H-4), 6.95 (br s, 1H, H-5), 7.03 (dd, J = 3.9, 1.5 Hz, 1H, H-3), 7.16 (ddd, J = 8.3, 2.6, 1.1 Hz, H-4″), 7.42 (t, J = 8.3 Hz, 1H, H-5″), 7.50 (dd, J = 2.6, 1.5 Hz, 1H, H-2″), 7.58 (dt, J = 7.5, 1.5 Hz, 1H, H-6″), 9.51 (d, J = 0.8 Hz, 1H, CHO). 13C NMR (187.5 MHz, CDCl3): δ 54.9 (CH2), 55.5 (CH3O), 110.3 (C-4), 112.4 (C-2″), 120.4 (C-4″), 120.5 (C-6″), 124.8 (C-3), 129.9 (C-5″), 131.6 (C-2), 132.5 (C-5), 136.1 (C-1″), 160.0 (C-3″), 179.8 (CHO), 192.8 (CO). HRMS (EI): m/z [M+] calcd. for C14H13NO3: 243.0895; found: 243.0891.
  • 1-(2-(4-Methoxyphenyl)-2-oxoethyl)-1H-pyrrole-2-carbaldehyde (8e). Following the method described for 8b, a mixture of 6 (0.200 g, 2.10 mmol), NaH (60%, 0.100 g, 2.52 mmol), and 7e (0.577 g, 2.52 mmol) provided 8e (0.413 g, 72%) as white needles. Rf 0.70 (hexane/EtOAc, 1:1); mp 118–119 °C. IR (KBr): ῡ 2939, 1652, 1601, 1406, 1366, 1226, 1178, 1024, 846, 754 cm−1. 1H NMR (600 MHz, CDCl3): δ 3.88 (s,1H, CH3O), 5.77 (s, 2H, CH2), 6.35 (dd, J = 3.9, 2.4 Hz, 1H, H-4), 6.95 (br s, 1H, H-5), 6.96–6.99 (m, 2H, H-3″), 7.02 (dd, J = 3.9, 1.8 Hz, 1H, H-3), 7.97–8.00 (m, 2H, H-2″), 9.50 (d, J = 0.6 Hz, 1H, CHO). 13C NMR (150 MHz, CDCl3): δ 54.3 (CH2), 55.5 (CH3O), 110.2 (C-4), 114.1 (C-3″), 124.7 (C-3), 127.8 (C-1″), 130.4 (C-2″), 131.6 (C-2), 132.6 (C-5), 164.1 (C-4″), 179.8 (CHO), 191.3 (CO). HRMS (EI): m/z [M+] calcd. for C14H13NO3: 243.0895; found: 243.0885.
  • 1-(2-(3,4-Dimethoxyphenyl)-2-oxoethyl)-1H-pyrrole-2-carbaldehyde (8f). Following the method described for 8b, a mixture of 6 (0.030 g, 0.32 mmol), NaH (60%, 0.015 g, 0.38 mmol), and 7f (0.098 g, 0.38 mmol) gave 8f (0.070 g, 81%) as white needles. Rf 0.13 (hexane/EtOAc, 1:1); 148–149 °C. IR (film): ῡ 3011, 1707, 1356, 1219, 1417, 762 cm−1. 1H NMR (600 MHz, CDCl3): δ 3.93 (s, 3H, CH3O), 3.97 (s, 3H, CH3O), 5.79 (s, 2H, CH2), 6.35 (dd, J = 3.9, 2.7 Hz, 1H, H-4), 6.94 (d, J = 8.1 Hz, 1H, H-5″), 6.96 (br s, 1H, H-5), 7.03 (dd, J = 3.9, 1.8 Hz, 1H, H-3), 7.53 (d, J = 2.1 Hz, 1H, H-2″), 7.66 (dd, J = 8.1, 2.1 Hz, 1H, H-6″), 9.52 (d, J = 0.6 Hz, 1H, CHO). 13C NMR (150 MHz, CDCl3): δ 54.2 (CH2), 56.0 (CH3O), 56.1 (CH3O), 110.2 (C-2″, C-4), 110.3 (C-5″), 122.6 (C-6″), 124.8 (C-3), 128.0 (C-1″), 131.7 (C-2), 132.6 (C-5), 149.3 (C-3″), 154.0 (C-4″), 179.9 (CHO), 191.5 (CO). HRMS (EI): m/z [M+] calcd. for C15H15NO4: 273.1001; found: 273.1002.
  • 2-(2-Formyl-1H-pyrrol-1-yl)acetonitrile (8g). Following the method described for 8b, a mixture of 6 (0.050 g, 0.53 mmol), NaH (60%, 0.025 g, 0.63 mmol), and 7g (0.076 g, 0.63 mmol) produced 8g (0.036 g, 51%) as pale violet oil. Rf 0.64 (hexane/EtOAc, 1:1). IR (film): ῡ 3113, 2864, 2253, 1652, 1475, 1406, 1362, 1308, 1222, 1028, 748 cm−1. 1H NMR (750 MHz, CDCl3): δ 5.35 (s, 2H, CH2), 6.34 (dd, J = 3.9, 2.6 Hz, 1H, H-4′), 7.00 (dd, J = 3.9, 1.5 Hz, 1H, H-3′), 7.08 (br s, 1H, H-5′), 9.57 (d, J = 0.8 Hz, 1H, CHO). 13C NMR (187.5 MHz, CDCl3): δ 36.2 (CH2), 111.5 (C-4′), 114.5 (CN), 125.2 (C-3′), 130.7 (C-5′), 131.0 (C-2′), 179.9 (CHO). HRMS (EI): m/z [M+] calcd. for C7H6N2O: 134.0480; found: 134.0477.
  • Ethyl (Z)-3-(dimethylamino)-2-(2-formyl-1H-pyrrol-1-yl)acrylate (1b). In a threaded ACE glass pressure tube equipped with a magnetic stirring bar and sealed with a Teflon screw cap, a mixture of 8b (0.100 g, 0.55 mmol) and DMFDMA (0.329 g, 2.75 mmol) in anhydrous DMF (2.0 mL) was heated at 100 °C for 20 h. A mixture of CH2Cl2/toluene (10.0 mL/1.0 mL) was added, and the solvent was removed under vacuum. The residue was purified by column chromatography over silica gel (hexane/EtOAc, 7:3), resulting in 1b (0.057 g, 44%) as a yellow oil. Rf 0.16 (hexane/EtOAc, 1:1). IR (film): ῡ 2934, 1663, 1618, 1303, 1209, 1078, 749 cm−1. 1H NMR (750 MHz, CDCl3): δ 1.16 (t, J = 6.8 Hz, 3H, CO2CH2CH3), 2.65 (br, 6H, N(CH3)2), 4.04–4.16 (m, 2H, CO2CH2CH3), 6.29 (dd, J = 3.8, 2.3 Hz, H-4′), 6.82 (br t, J = 2.3 Hz, 1H, H-5′), 7.01 (dd, J = 3.8, 1.5 Hz, H-3′), 7.51 (s, 1H, H-3), 9.56 (s, 1H, CHO). 13C NMR (187.5 MHz, CDCl3): δ 14.5 (CO2CH2CH3), 34.9 (N(CH3)2), 60.1 (CO2CH2CH3), 97.9 (C-2), 110.2 (C-4′), 120.9 (C-3′), 134.2 (C-5′), 134.9 (C-2′), 146.0 (C-3), 166.9 (CO2Et), 179.8 (CHO). HRMS (EI): m/z [M+] calcd. for C12H16N2O3: 236.1161; found: 236.1159.
  • (Z)-1-(1-(Dimethylamino)-3-oxo-3-phenylprop-1-en-2-yl)-1H-pyrrole-2-carbaldehyde (2a). Following the method described for 1b, a mixture of 8c (0.030 g, 0.14 mmol) and DMFDMA (0.084 g, 0.70 mmol) in anhydrous DMF (1.0 mL) delivered 2a (0.020 g, 53%) as a yellow oil. Rf 0.16 (hexane/EtOAc, 1:1). IR (film): ῡ 3097, 2928, 1637, 1543, 1413, 1388, 1323, 1095, 950, 878, 766, 708 cm−1. 1H NMR (750 MHz, CDCl3): δ 2.68 (br, 6H, N(CH3)2), 6.33 (dd, J = 3.9, 2.6 Hz, 1H, H-4), 6.89 (br s, 1H, H-5), 7.02 (dd, J = 3.9, 1.5 Hz, 1H, H-3), 7.31–7.36 (m, 3H, H-1′, H-3″), 7.37–7.40 (m, 1H, H-4″), 7.48–7.52 (m, 2H, H-2″), 9.60 (d, J = 0.9 Hz, 1H, CHO). 13C NMR (187.5 MHz, CDCl3): δ 29.6 (br, N(CH3)2), 110.6 (C-2′, C-4), 122.6 (C-3), 127.90 (C-2″), 127.93 (C-3″), 129.9 (C-4″), 134.39 (C-2), 134.43 (C-5), 139.9 (C-1″), 150.2 (C-1′), 179.4 (CHO), 190.9 (CO). HRMS (EI): m/z [M+] calcd. for C16H16N2O2: 268.1212; found: 268.1213.
  • (Z)-1-(1-(Dimethylamino)-3-(3-methoxyphenyl)-3-oxoprop-1-en-2-yl)-1H-pyrrole-2-carbaldehyde (2b). Following the method described for 1b, a mixture of 8d (0.050 g, 0.21 mmol) and DMFDMA (0.125 g, 1.05 mmol) in anhydrous DMF (1.0 mL) formed 2b (0.035 g, 56%) as a pale violet oil. Rf 0.45 (hexane/EtOAc, 1:1). IR (film): ῡ 2925, 1663, 1588, 1424, 1322, 1042, 748 cm−1. 1H NMR (500 MHz, CDCl3): δ 2.70 (br, 6H, N(CH3)2), 3.79 (s, 3H, CH3O), 6.31–6.34 (m, 1H, H-4), 6.88 (br s, 1H, H-5), 6.92 (dm, J = 8.0 Hz, 1H, H-4″), 7.01–7.04 (m, 2H, H-2″, H-3), 7.06–7.10 (m, 1H, H-6″), 7.23 (t, J = 8.0 Hz, 1H, H-5″), 7.37 (br s, 1H, H-1′), 9.60 (s, 1H, CHO). 13C NMR (125 MHz, CDCl3): δ 36.5 (br, N(CH3)2), 55.2 (CH3O), 110.6 (C-4), 111.1 (C-2′), 112.7 (C-2″), 116.3 (C-4″), 120.3 (C-6″), 122.5 (C-3), 128.9 (C-5″), 129.5 (C-2), 134.4 (C-5), 141.1 (C-1″), 150.4 (C-1′), 159.2 (C-3″), 179.4 (CHO), 190.6 (CO). HRMS (EI): m/z [M+] calcd. for C17H18N2O3: 298.1317; found: 298.1311.
  • (Z)-1-(1-(Dimethylamino)-3-(4-methoxyphenyl)-3-oxoprop-1-en-2-yl)-1H-pyrrole-2-carbaldehyde (2c). Following the method described for 1b, a mixture of 8e (0.100 g, 0.41 mmol) and DMFDMA (0.245 g, 2.05 mmol) in anhydrous DMF (1.0 mL) provided 2c (0.111 g, 90%) as a yellow oil. Rf 0.16 (hexane/EtOAc, 1:1). IR (film): ῡ 2939, 1663, 1600, 1584, 1560, 1386, 1325, 1243, 1096, 1021, 840, 765, 738 cm−1. 1H NMR (500 MHz, CDCl3): δ 2.67 (br, 6H, N(CH3)2), 3.79 (s, 3H, CH3O), 6.31 (dd, J = 4.2, 2.4 Hz, 1H, H-4), 6.80 (d, J = 8.4 Hz, 2H, H-3″), 6.87 (br s, 1H, H-5), 7.01 (dd, J = 4.2, 1.8 Hz, 1H, H-3), 7.35 (s, 1H, H-1′), 7.45 (d, J = 8.4 Hz, 1H, H-2″), 9.56 (s, 1H, CHO). 13C NMR (125 MHz, CDCl3): δ 34.8 (br, N(CH3)2), 55.2 (CH3O), 110.1 (C-2′), 110.6 (C-4), 113.2 (C-3″), 122.3 (C-3), 130.0 (C-2″), 132.1 (C-1″), 134.3 (C-2), 134.4 (C-5), 149.7 (C-1′), 161.2 (C-4″), 179.5 (CHO), 189.7 (CO). HRMS (EI): m/z [M+] calcd. for C17H18N2O3: 298.1317; found: 298.1314.
  • (Z)-1-(3-(3,4-Dimethoxyphenyl)-1-(dimethylamino)-3-oxoprop-1-en-2-yl)-1H-pyrrole-2-carbaldehyde (2d). Following the method described for 1b, a mixture of 8f (0.050 g, 0.18 mmol) and DMFDMA (0.109 g, 0.90 mmol) in anhydrous DMF (1.0 mL) afforded 2d (0.050 g, 83%) as a brown solid. Rf 0.03 (hexane/EtOAc, 1:1); 138–140 °C. IR (film): ῡ 2929, 1738, 1639, 1564, 1509, 1410, 1311, 1096, 1018, 748 cm−1. 1H NMR (600 MHz, CDCl3): δ 2.69 (br, 6H, N(CH3)2), 3.82 (CH3O), 3.87 (CH3O), 6.31 (dd, J = 4.0, 2.1 Hz, 1H, H-4), 6.76 (d, J = 8.1 Hz, 1H, H-5″), 6.87 (br s, 1H, H-5), 7.02 (dd, J = 4.0, 1.8 Hz, 1H, H-3), 7.04 (br s, 1H, H-2″), 7.09 (br d, J = 8.1 Hz, 1H, H-6″), 7.43 (s, 1H, H-1′), 9.58 (CHO). 13C NMR (150 MHz, CDCl3): δ 42.0 (br, N(CH3)2), 55.8 (CH3O), 55.9 (CH3O), 110.00 (C-5″), 110.02 (C-2′), 110.6 (C-4), 111.2 (C-2″), 121.7 (C-6″), 122.2 (C-3), 132.2 (C-1″), 134.3 (C-5), 134.5 (C-2), 148.4 (C-3″), 149.6 (C-1′), 150.1 (C-4″), 179.4 (CHO), 189.4 (CO). HRMS (EI): m/z [M+] calcd. for C18H20N2O4: 328.1423; found: 328.1426.
  • (Z)-3-(Dimethylamino)-2-(2-formyl-1H-pyrrol-1-yl)acrylonitrile (3a). (E)-3-(Dimethylamino)-2-(2-formyl-1H-pyrrol-1-yl)acrylonitrile (3a’). Following the method described for 1b, a mixture of 8g (0.077 g, 0.57 mmol) and DMFDMA (0.339 g, 2.85 mmol) in anhydrous DMF (2.0 mL) generated an inseparable mixture of 3a/3a’ (54:46, 0.030 g, 28%) as a pale violet oil. Rf 0.16 (hexane/EtOAc, 1:1). IR (film): ῡ 3095, 2922, 2803, 2184, 1650, 1365, 1130, 772 cm−1. 1H NMR (600 MHz, CDCl3): δ 2.63 (br, 6H, N(CH3)2), 3.16 (s, 6H, N(CH3)2), 6.25 (dd, J = 4.2, 2.4 Hz, 1H, H-4′), 6.31 (dd, J = 4.2, 2.4 Hz, 1H, H-4′), 6.68 (s, 1H, H-3), 6.73 (s, 1H, H-3), 6.87–6.89 (m, 1H, H-5′), 6.91–6.93 (m, 1H, H-5′), 6.96 (dd, J = 4.2, 1.8 Hz, 1H, H-3′), 6.99 (dd, J = 4.2, 1.8 Hz, 1H, H-3′), 9.63 (s, 1H, CHO), 9.66 (s, 1H, CHO). 13C NMR (150 MHz, CDCl3): δ 42.3 (br, N(CH3)2), 78.6 (C-2), 78.8 (C-2), 110.7 (C-4′), 111.2 (C-4′), 118.5 (CN), 120.24 (CN), 122.5 (2C-3′), 133.0 (C-5′), 133.2 (C-2′), 133.8 (C-5′), 134.5 (C-2′), 147.4 (C-3), 151.0 (C-3), 178.9 (CHO), 179.2 (CHO). HRMS (EI): m/z [M+] calcd. for C10H11N3O: 189.0902; found: 189.0900.
  • Methyl pyrrolo[1,2-a]pyrazine-4-carboxylate (4a). Method A: In a threaded ACE glass pressure tube equipped with a magnetic stirring bar and sealed with a Teflon screw cap, a mixture of NH4OAc (0.052 g, 0.675 mmol) and Li2CO3 (0.050 g, 0.675 mmol) in anhydrous DMF (1.0 mL) was stirred at rt for 20 min. Then, 1a (0.050 g, 0.225 mmol) was added, and the solution was heated at 70 °C for 4 h. The mixture was diluted with hexane/EtOAc (1:1, 15 mL) and washed with brine (5.0 mL × 3). The organic layer was dried (Na2SO4), and the solvent was removed under vacuum. The residue was purified by column chromatography over silica gel (hexane/EtOAc, 8:2) to give 4a (0.036 g, 90%) as a yellow solid. Rf 0.58 (hexane/EtOAc, 1:1); mp 93–95 °C.
  • Method B: In a threaded ACE glass pressure tube equipped with a magnetic stirring bar and sealed with a Teflon screw cap, a mixture of 8a (0.030 g, 0.18 mmol), DMFDMA (0.114 g, 0.90 mmol), and NH4OAc (0.042 g, 0.54 mmol) was heated at 100 °C for 24 h. The mixture was suspended in CH2Cl2/PhMe (10:1, 11 mL), and the solvent was removed under vacuum. The residue was purified by column chromatography over silica gel (hexane/EtOAc, 1:1) to furnish 4a (0.018 g, 57%) as a yellow solid. Rf 0.58 (hexane/EtOAc, 1:1); mp 93–95 °C. IR (film): ῡ 3168, 2954, 1720, 1621, 1441, 1430, 1306, 1217, 1203, 1177, 1112, 764, 744 cm−1. 1H NMR (500 MHz, CDCl3): δ 4.01 (s, 3H, CO2CH3), 7.02 (s, 2H, H-7, H-8), 8.46 (s, 1H, H-3), 8.74 (s, 1H, H-6), 8.92 (s, 1H, H-1). 13C NMR (125 MHz, CDCl3): δ 52.4 (CO2CH3), 106.7 (C-8), 116.6 (C-7), 118.3 (C-4), 118.9 (C-6), 129.9 (C-8a), 134.8 (C-3), 148.1 (C-1), 163.3 (CO2CH3). HRMS (EI): m/z [M+] calcd. for C9H8N2O2: 176.0586; found: 176.0592.
  • Ethyl pyrrolo[1,2-a]pyrazine-4-carboxylate (4b). Following method A described for 4a, a mixture of 1b (0.030 g, 0.13 mmol), NH4OAc (0.030 g, 0.39 mmol), and Li2CO3 (0.028 g, 0.39 mmol) was heated at 80 °C for 3 h to obtain 4b (0.016 g, 66%) as a yellow solid. Rf 0.67 (hexane/EtOAc, 1:1); mp 68–70 °C. IR (film): ῡ 3177, 3097, 2848, 1699, 1427, 1287, 1178, 1088, 1033, 734 cm−1. 1H NMR (750 MHz, CDCl3): δ 1.45 (t, J = 7.4 Hz, CO2CH2CH3), 4.47 (q, J = 7.4 Hz, CO2CH2CH3), 7.01–7.03 (m, 2H, H-7, H-8), 8.46 (s, 1H, H-3), 8.73–8.74 (m, 1H, H-6), 8.91 (s, 1H, H-1). 13C NMR (187.5 MHz, CDCl3): δ 14.3 (CO2CH2CH3), 61.6 (CO2CH2CH3), 106.6 (C-8), 116.5 (C-7), 118.9 (C-6), 129.9 (C-8a), 134.6 (C-3), 134.8 (C-4), 148.0 (C-1), 162.8 (CO2CH2CH3). HRMS (EI): m/z [M+] calcd. for C10H10N2O2: 190.0742; found: 190.0742.
  • Phenyl(pyrrolo[1,2-a]pyrazin-4-yl)methanone (4c). Following method A described for 4a, a mixture of 2a (0.050 g, 0.187 mmol), NH4OAc (0.043 g, 0.56 mmol), and Li2CO3 (0.041 g, 0.56 mmol) was heated at 80 °C for 3 h to give 4c (0.036 g, 87%) as a yellow solid. Rf 0.48 (hexane/EtOAc, 1:1); mp 61–62 °C. Following method B described for 4a, a mixture of 8c (0.030 g, 0.141 mmol), DMFDMA (0.084 g, 0.70 mmol), and NH4OAc (0.054 g, 0.70 mmol) produced 4c (0.025 g, 80%) as a yellow solid. Rf 0.48 (hexane/EtOAc, 1:1); mp 61–62 °C. IR (film): ῡ 3101, 3040, 2924, 1633, 1467, 1290, 1203, 1052, 882, 741 cm−1. 1H NMR (750 MHz, CDCl3): δ 7.10 (d, J = 1.5 Hz, 2H, H-7′, H-8′), 7.53 (t, J = 7.5 Hz, 2H, H-3″), 7.63 (t, J = 7.5 Hz, 1H, H-4″), 7.81 (br d, J = 7.5 Hz, 2H, H-2″), 8.10 (s, 1H, H-3′), 8.92 (br m, 1H, H-6′), 8.94 (s, 1H, H-1′). 13C NMR (187.5 MHz, CDCl3): δ 107.4 (C-8′), 117.1 (C-7′), 119.6 (C-6′), 124.5 (C-8a’), 128.5 (C-3″), 129.6 (C-2″), 130.1 (C-4′), 132.5 (C-4″), 137.8 (C-1″), 139.0 (C-3′), 148 (C-1′), 190.9 (CO). HRMS (EI): m/z [M+] calcd. for C14H10N2O: 222.0793; found: 222.0794.
  • (3-Methoxyphenyl)(pyrrolo[1,2-a]pyrazin-4-yl)methanone (4d). Following method A described for 4a, a mixture of 2b (0.030 g, 0.10 mmol), NH4OAc (0.023 g, 0.30 mmol), and Li2CO3 (0.022 g, 0.30 mmol) provided 4d (0.021 g, 83%) as a yellow solid. Rf 0.39 (hexane/EtOAc, 1:1); mp 88–89 °C. IR (film): ῡ 2970, 1734, 1594, 1424, 1298, 1250, 1171, 1028, 854, 741 cm−1. 1H NMR (600 MHz, CDCl3): δ 3.88 (s, 3H, CH3O), 7.09 (br s, 2H, H-7′, H-8′), 7.17 (dd, J = 8.0, 2.6 Hz, 1H, H-4″), 7.34 (br d, J = 1.2 Hz, 1H, H-2″), 7.36 (br d, J = 8.0 Hz, 1H, H-6″), 7.43 (t, J = 8.0 Hz, 1H, H-5″), 8.13 (s, 1H, H-3′), 8.91 (s, 1H, H-6′), 8.94 (s, 1H, H-1′). 13C NMR (150 MHz, CDCl3): δ 55.4 (CH3O), 107.5 (C-8′), 114.2 (C-2″), 117.2 (C-7′), 118.9 (C-4″), 119.6 (C-6′), 122.2 (C-6″), 124.6 (C-8a’), 129.5 (C-5″), 130.1 (C-4′), 138.9 (C-3′), 139.0 (C-1″), 147.9 (C-1′), 159.7 (C-3″), 190.6 (CO). HRMS (EI): m/z [M+] calcd. for C15H12N2O2: 252.0899; found: 252.0900.
  • (4-Methoxyphenyl)(pyrrolo[1,2-a]pyrazin-4-yl)methanone (4e). Following method A described for 4a, a mixture of 2c (0.050 g, 0.168 mmol), NH4OAc (0.039 g, 0.50 mmol), and Li2CO3 (0.037 g, 0.50 mmol) formed 4e (0.036 g, 84%) as a yellow solid. Rf 0.29 (hexane/EtOAc, 1:1); mp 138–139 °C. IR (KBr): ῡ 2928, 1739, 1590, 1507, 1301, 1265, 1167, 1109, 1030, 845, 759, 719 cm−1. 1H NMR (600 MHz, CDCl3): δ 3.91 (s, 3H, CH3O), 7.00–7.03 (m, 2H, H-3″), 7.05 (br s, 1H, H-8′), 7.06 (br s, 1H, H-7′), 7.84–7.87 (m, 2H, H-2″), 8.05 (br s, 1H, H-3′), 8.74 (s, 1H, H-6′), 8.92 (s, 1H, H-1′). 13C NMR (150 MHz, CDCl3): δ 55.6 (CH3O), 106.9 (C-8′), 113.9 (C-3″), 116.8 (C-7′), 119.0 (C-6′), 124.8 (C-8a’), 130.0 (C-4′), 130.1 (C-1″), 132.2 (C-2″), 137.4 (C-3′), 147.7 (C-1′), 163.6 (C-4″), 189.3 (CO). HRMS (EI): m/z [M+] calcd. for C15H12N2O2: 252.0899; found: 252.0899.
  • (3,4-Dimethoxyphenyl)(pyrrolo[1,2-a]pyrazin-4-yl)methanone (4f). Following method A described for 4a, a mixture of 2d (0.079 g, 0.24 mmol), NH4OAc (0.056 g, 0.72 mmol), and Li2CO3 (0.053 g, 0.72 mmol) afforded 4f (0.041 g, 60%) as a yellow solid. Rf 0.16 (hexane/EtOAc, 1:1); mp 148–149 °C. IR (KBr): ῡ 3158, 2943, 1632, 1594, 1509, 1414, 1260, 1171, 1014, 772 cm−1. 1H NMR (750 MHz, CDCl3): δ 3.95 (s, 3H, CH3O), 3.97 (s, 3H, CH3O), 6.94 (d, J = 8.8 Hz, 1H, H-5″), 7.04–7.06 (m, 2H, H-7′, H-8′), 7.44–7.47 (m, H-2″, H-6″), 8.06 (s, 1H, H-3′), 8.69 (br s, 1H, H-6′), 8.91 (s, 1H, H-1′). 13C NMR (187.5 MHz, CDCl3): δ 56.0 (CH3O), 56.1 (CH3O), 106.9 (C-8′), 110.0 (C-5″), 111.8 (C-2″), 116.8 (C-7′), 118.9 (C-6′), 124.7 (C-8a’), 124.8 (C-6″), 129.9 (C-4′), 130.1 (C-1″), 137.2 (C-3′), 147.6 (C-1′), 149.2 (C-3″), 153.4 (C-4″), 189.2 (CO). HRMS (EI): m/z [M+] calcd. for C16H14N2O3: 282.1005; found: 282.1004.
  • Pyrrolo[1,2-a]pyrazine-4-carbonitrile (4g). Following method A described for 4a, a mixture of 3a (0.032 g, 0.169 mmol), NH4OAc (0.039 g, 0.51 mmol), and Li2CO3 (0.038 g, 0.51 mmol) furnished 4g (0.013 g, 53%) as a brown solid. Rf 0.55 (hexane/EtOAc, 1:1); mp 128–130 °C. IR (film): ῡ 3010, 2600, 1741, 1366, 1212 cm−1. 1H NMR (600 MHz, CDCl3): δ 7.13 (s, 2H, H-7, H-8), 7.84 (s, 1H, H-6), 8.08 (br s, 1H, H-3), 9.00 (br s, 1H, H-1). 13C NMR (100 MHz, CDCl3): δ 104.5 (CN), 107.9 (C-8), 112.8 (C-4), 116.6 (C-6), 117.0 (C-7), 127.9 (C-8a), 136.3 (C-3), 148.1 (C-1). HRMS (EI): m/z [M+] calcd. for C8H5N3: 143.0483; found: 143.0485.
  • Phenyl(pyrrolo[1,2-a]pyrazin-4-yl)methanol (4h). In a round-bottom flask equipped with a magnetic stirring bar, NaBH4 (0.051 g, 1.33 mmol) was slowly added to a solution of 4c (0.030 g, 0.13 mmol) in anhydrous THF (5.0 mL). After heating the reaction mixture at 60 °C for 2 h, MeOH (8.0 mL) was added dropwise and stirred at rt for 5.0 min. Then, a saturated aqueous solution of NH4Cl (8.0 mL) was added and stirred at rt for 20 min. The mixture was extracted with hexane/EtOAc (1:1, 15 mL) and brine (3 × 5.0 mL). The organic layer was dried (Na2SO4), and the solvent was removed under vacuum. The residue was purified by column chromatography over silica gel (hexane/EtOAc, 7:3), resulting in 4h (0.014 g, 48%) as a yellow solid. Rf 0.10 (hexane/EtOAc, 1:1); mp 150–152 °C. IR (film): ῡ 3042, 2820, 1618, 1451, 1318, 1048, 727, 704 cm−1. 1H NMR (600 MHz, CDCl3): δ 3.10 (br, 1H, OH), 5.98 (s, 1H, CHOH), 6.80 (s, 2H, H-7′, H-8′), 7.30–7.39 (m, 4H, H-6′, H-3″, H-4″), 7.43 (br s, 1H, H-3′), 7.44–7.46 (m, 2H, H-2″), 8.58 (s, 1H, H-1′). 13C NMR (187.5 MHz, CDCl3): δ 71.9 (CHOH), 104.5 (C-8′), 114.6 (C-6′), 115.3 (C-7′), 125.5 (C-3′), 126.6 (C-2″), 128.5 (C-4″), 128.83 (C-3″), 128.88 (C-8a’), 130.2 (C-4′), 138.6 (C-1″), 144.3 (C-1′). HRMS (EI): m/z [M+] calcd. for C14H12N2O: 224.0950; found: 224.0948.
  • Methyl 2-(4-bromo-2-formyl-1H-pyrrol-1-yl)acetate (8h). In a round-bottom flask at 0 °C with a magnetic stirring bar, a solution of NBS (0.080 g, 0.45 mmol) in anhydrous CH2Cl2 (5.0 mL) was added dropwise to a solution of 8a (0.05 g, 0.30 mmol) in anhydrous DMF (5.0 mL). The mixture was stirred at rt for 2.0 h before adding a 10% aqueous solution of NaHSO3 (1.0 mL). It was then washed with brine (3 × 5.0 mL), the organic phase was dried (Na2SO4), and the solvent was removed under vacuum. The residue was purified by column chromatography over silica gel (hexane/EtOAc, 8:2) to afford 8h (0.066 g, 90%) as a violet oil. Rf 0.74 (hexane/EtOAc, 1:1). IR (film): ῡ 2922, 1692, 1661, 1393, 1215, 998, 768 cm−1. 1H NMR (500 MHz, CDCl3): δ 3.76 (s, 3H, CO2CH3), 5.01 (s, 2H, CH2), 6.90 (br s, 1H, H-5′), 6.95 (d, J = 2.0 Hz, 1H, H-3′), 9.45 (d, J = 0.5 Hz, 1H, CHO). 13C NMR (125 MHz, CDCl3): δ 50.1 (CH2), 52.6 (CO2CH3), 97.5 (C-4′), 125.2 (C-3′) 131.2 (C-5′), 131.6 (C-2′), 168.2 (CO2CH3), 179.3 (CHO). HRMS (EI): m/z [M+] calcd. for C8H8BrNO3: 244.9688; found: 244.9680.
  • Methyl (Z)-2-(4-bromo-2-formyl-1H-pyrrol-1-yl)-3-(dimethylamino)acrylate (1c). Following the method described for 1b, a mixture of 8h (0.050 g, 0.20 mmol) and DMFDMA (0.121 g, 1.02 mmol) in anhydrous DMF (1.0 mL) gave 1c (0.041 g, 66%) as a pale violet oil. Rf 0.12 (hexane/EtOAc, 7:3). IR (film): ῡ 2921, 1666, 1621, 1212, 1084, 922, 755 cm−1. 1H NMR (600 MHz, CDCl3): δ 2.67 (br, 6H, N(CH3)2), 3.62 (s, 3H, CO2CH3), 6.81 (br s, 1H, H-5′), 6.99 (br s, 1H, H-3′), 7.50 (s, 1H, H-3), 9.49 (s, 1H, CHO). 13C NMR (125 MHz, CDCl3): δ 34.7 (N(CH3)2), 51.6 (CO2CH3), 96.6 (C-2), 98.2 (C-4′), 121.6 (C-3′), 132.9 (C-5′), 134.8 (C-2′), 146.4 (C-3), 166.9 (CO2CH3), 179.1 (CHO). HRMS (EI): m/z [M+] calcd. for C11H13BrN2O3: 300.0110; found: 300.0108.
  • Methyl 7-bromopyrrolo[1,2-a]pyrazine-4-carboxylate (4i). Following method A described for 4a, a mixture of 1c (0.030 g, 0.10 mmol), NH4OAc (0.031 g, 0.40 mmol), and Li2CO3 (0.029 g, 0.40 mmol) provided 4i (0.017 g, 74%) as a yellow solid. Rf 0.38 (hexane/EtOAc, 1:1); mp 93–95 °C. IR (film): ῡ 3173, 3126, 2957, 1720, 1442, 1308, 1200, 1175 cm−1. 1H NMR (500 MHz, CDCl3): δ 4.01 (s, 3H, CO2CH3), 7.10 (br s, 1H, H-8), 8.45 (br s, 1H, H-3), 8.78 (s, 1H, H-6), 8.87 (br s, 1H, H-1). 13C NMR (125 MHz, CDCl3): δ 52.8 (CO2CH3), 107.2 (C-7), 109.2 (C-8), 117.7 (C-4), 119.5 (C-6), 129.6 (C-8a), 133.7 (C-3), 145.9 (C-1), 162.5 (CO2CH3). HRMS (EI): m/z [M+] calcd. for C9H7BrN2O2: 253.9691; found: 253.9681.
  • Methyl 6-bromopyrrolo[1,2-a]pyrazine-4-carboxylate (4j). In a round-bottom flask at 0 °C with a magnetic stirring bar, a solution of NBS (0.040 g, 0.23 mmol) in anhydrous CH2Cl2 (5.0 mL) was added dropwise to a solution of 4a (0.040 g, 0.23 mmol) in anhydrous CH2Cl2 (5.0 mL). The mixture was stirred at rt for 2.0 h before adding a 10% aqueous solution of NaHSO3 (1.0 mL). It was then washed with brine (3 × 5.0 mL), the organic phase was dried (Na2SO4), and the solvent was removed under vacuum. The residue was purified by column chromatography over silica gel (hexane/EtOAc, 1:1) to obtain 4j (0.032 g, 55%) as a pale orange solid. Rf 0.32 (hexane/EtOAc, 1:1); mp 115–117 °C. IR (film): ῡ 3144, 2953, 1724, 1417, 1294, 1175, 957, 857, 773 cm−1. 1H NMR (600 MHz, CDCl3): δ 4.00 (s, 3H, CO2CH3), 7.03 (d, J = 2.4 Hz, 1H, H-8), 8.46 (s, 1H, H-3), 8.67 (dd, 1H, J = 2.4, 0.6 Hz, H-7), 8.92 (br s, 1H, H-1). 13C NMR (150 MHz, CDCl3): δ 52.6 (CO2CH3), 94.3 (C-6), 118.5 (C-8), 118.8 (C-7), 127.1 (C-4), 129.5 (C-8a), 135.2 (C-3), 147.2 (C-1), 162.8 (CO2CH3). HRMS (EI): m/z [M+] calcd. for C9H7BrN2O2: 253.9691; found: 253.9701.
  • Methyl 6,7-dibromopyrrolo[1,2-a]pyrazine-4-carboxylate (4k). Following the method described for 4j, a mixture of 4a (0.030 g, 0.17 mmol) and NBS (0.063 g, 0.36 mmol) produced 4k (0.023 g, 40%) as an orange solid. Rf 0.61 (hexane/EtOAc, 1:1); mp 106–108 °C. IR (film): ῡ 3123, 2952, 2918, 2850, 1733, 1601, 1464, 1420, 1312, 1294, 1200, 1172, 1095, 862, 753 cm −1. 1H NMR (500 MHz, CDCl3): δ 4.02 (s, 3H, CO2CH3), 7.04 (s, 1H, H-8), 8.00 (s, 1H, H-3), 8.80 (s, 1H, H-1). 13C NMR (125 MHz, CDCl3): δ 53.0 (CO2CH3), 95.0 (C-6), 99.6 (C-7), 121.1 (C-4), 122.0 (C-8), 128.7 (C-8a), 133.1 (C-3), 145.8 (C-1), 161.6 (CO2CH3). HRMS (EI): m/z [M+] calcd. for C9H6N2O2Br2: 331.8796; found: 331.8792.
  • Methyl (E)-3-(1-(2-methoxy-2-oxoethyl)-1H-pyrrol-2-yl)acrylate (10a). At 0 °C and under N2 atmosphere, NaH (60%, 0.032 g, 0.80 mmol) was added to a solution of 9a (0.100 g, 0.66 mmol) in anhydrous DMF (2.0 mL), and the mixture was stirred for 15 min. Then, 7a (0.124 g, 0.80 mmol) was added, and the solution was stirred at rt for 3 h. The reaction mixture was extracted with a hexane/EtOAc (1:1, 15.0 mL), the organic layer was washed with brine (5.0 mL × 3) and dried (Na2SO4), and the solvent was removed under vacuum. The residue was purified by column chromatography over silica gel (hexane/EtOAc, 9:1), resulting in 10a (0.088 g, 60%) as a white solid. Rf 0.32 (hexane/EtOAc, 7:3); mp 116–118 °C. IR (film): ῡ 2960, 1753, 1688, 1623, 1467, 1294, 1171, 1080, 987, 745 cm−1. 1H NMR (600 MHz, CDCl3): δ 3.76 (s, 3H, =CHCO2CH3), 3.77 (s, 3H, CH2CO2CH3), 4.75 (s, 2H, CH2), 6.15 (d, J = 15.6 Hz, 1H, H-2), 6.24–6.26 (m, 1H, H-4′), 6.72 (dd, J = 3.8, 1.4 Hz, 1H, H-3′), 6.78 (br t, J = 1.4 Hz, 1H, H-5′), 7.43 (d, J = 15.6 Hz, 1H, H-3). 13C NMR (150 MHz, CDCl3): δ 48.2 (CH2), 51.5 (CH2CO2CH3), 52.7 (=CHCO2CH3), 110.4 (C-4′), 112.3 (C-3′), 113.5 (C-2), 126.8 (C-5′), 129.4 (C-2′), 131.5 (C-3), 167.9 (=CHCO2CH3), 168.4 (CH2CO2CH3). HRMS (EI): m/z [M+] calcd. for C11H13NO4: 223.0845; found: 223.0845.
  • Methyl (E)-3-(5-formyl-1-(2-methoxy-2-oxoethyl)-1H-pyrrol-2-yl)acrylate (8i). At 0 °C and under N2 atmosphere, POCl3 (0.220 g, 1.44 mmol) was added to DMF (0.105 g, 1.44 mmol), and the mixture was stirred for 10 min. Subsequently, 10a (0.200 g, 0.90 mmol) in anhydrous CH2Cl2 (6.0 mL) was added dropwise, and the solution was stirred at 0 °C for 3 h. The reaction mixture was quenched with a 2N aqueous solution of NaOH until pH 8.0, and then CH2Cl2 (21.0 mL) was added. The mixture was washed with brine (7.0 mL × 3), the organic layer was dried (Na2SO4), and the solvent was removed under vacuum. The residue was purified by column chromatography over silica gel (hexane/EtOAc, 9:1) to furnish 8i (0.142 g, 63%) as a pale brown solid. Rf 0.67 (hexane/EtOAc, 1:1); mp 108–110 °C. IR (film): ῡ 2960, 1741, 1704, 1663, 1198, 1168, 980, 778 cm−1. 1H NMR (500 MHz, CDCl3): δ 3.75 (s, 3H, CH2CO2CH3), 3.77 (s, 3H, =CHCO2CH3), 5.24 (s, 2H, CH2), 6.41 (d, J = 15.8 Hz, 1H, H-2), 6.70 (d, J = 4.0 Hz, 1H, H-3′), 6.97 (d, J = 4.0 Hz, 1H, H-4′), 7.41 (d, J = 15.8 Hz, 1H, H-3), 9.53 (CHO). 13C NMR (125 MHz, CDCl3) δ 46.3 (CH2), 52.0 (CO2CH3), 52.8 (CO2CH3), 111.4 (C-3′), 121.1 (C-2), 124.6 (C-4′), 129.7 (C-3), 133.7 (C-5′), 137.7 (C-2′), 166.7 (=CHCO2CH3), 168.3 (CH2CO2CH3), 180.2 (CHO). HRMS (EI): m/z [M+] calcd. for C12H13NO5: 251.0794; found: 251.0785.
  • Methyl (Z)-3-(dimethylamino)-2-(2-formyl-5-((E)-3-methoxy-3-oxoprop-1-en-1-yl)-1H-pyrrol-1-yl)acrylate (11). Following the method described for 1b, a mixture of 8i (0.090 g, 0.36 mmol) and DMFDMA (0.213 g, 1.79 mmol) in anhydrous DMF (1.0 mL) afforded 11 (0.031 g, 28%) as a pale violet oil. Rf 0.16 (hexane/EtOAc, 1:1). IR (film): ῡ 2957, 1619, 1435, 1164, 1103, 1037, 762 cm−1. 1H NMR (750 MHz, CDCl3): δ 2.46 (br, 6H, N(CH3)2), 3.63 (s, 3H, CO2CH3-1), 3.78 (s, 3H, =CHCO2CH3), 6.41 (d, J = 16.1 Hz, 1H, H-2″), 6.74 (d, J = 3.8 Hz, 1H, H-4′), 7.05 (d, J = 3.8 Hz, 1H, H-3′) 7.41 (d, J = 16.1 Hz, 1H, H-1″), 7.67 (s, 1H, H-3), 9.61 (s, 1H, CHO). 13C NMR (187.5 MHz, CDCl3): δ 36.4 (br, N(CH3)2), 51.6 (CO2CH3-1), 51.8 (=CHCO2CH3), 93.4 (C-2), 111.2 (C-4′), 119.5 (C-2″), 120.6 (C-3′), 131.6 (C-1″), 136.8 (C-2′), 139.0 (C-5′), 147.4 (C-3), 166.8 (CO2CH3-1), 167.0 (=CHCO2CH3), 180.0 (CHO). HRMS (EI): m/z [M+] calcd. for C15H18N2O5: 306.1216; found: 306.1214.
  • Methyl (E)-6-(3-methoxy-3-oxoprop-1-en-1-yl)pyrrolo[1,2-a]pyrazine-4-carboxylate (4l). Following method A described for 4a, a mixture of 11 (0.030 g, 0.10 mmol), NH4OAc (0.023 g, 0.30 mmol), and Li2CO3 (0.022 g, 0.30 mmol) was heated at 70 °C for 7 h to give 4l (0.015 g, 61%) as a yellow solid. Rf 0.25 (hexane/EtOAc, 1:1); mp 100–102 °C. IR (film): ῡ 2917, 2852, 1717, 1623, 1717, 1623, 1427, 1330, 1262, 1171, 1099, 1066 cm−1. 1H NMR (600 MHz, CDCl3): δ 3.82 (s, 3H, =CHCO2CH3), 4.04 (s, 3H, CO2CH3-4), 6.39 (d, J = 15.6 Hz, 1H, H-2′), 7.18 (d, J = 4.2 Hz, 1H, H-8), 7.35 (d, J = 4.2 Hz, 1H, H-7), 7.70 (d, J = 15.6 Hz, 1H, H-1′), 8.24 (s, 1H, H-3), 8.93 (br s, 1H, H-1). 13C NMR (187.5 MHz, CDCl3): δ 51.8 (=CHCO2CH3), 53.0 (CO2CH3-4), 108.5 (C-8), 115.9 (C-2′), 118.1 (C-7), 120.9 (C-4), 126.9 (C-6), 132.01 (C-8a), 133.3 (C-1′), 135.2 (C-3), 147.9 (C-1), 163.3 (CO2CH3-4), 167.1 (=CHCO2CH3). HRMS (EI): m/z [M+] calcd. for C13H12N2O4: 260.0797; found: 260.0795.
  • Ethyl (E)-3-(1-(2-methoxy-2-oxoethyl)-1H-pyrrol-2-yl)acrylate (10c). Following the method described for 10a, a mixture of 9b (0.100 g, 0.60 mmol), NaH (60%, 0.029 g, 0.72 mmol), and 7a (0.110 g, 0.72 mmol) formed 10c (0.10 g, 70%) as a pale brown solid. Rf 0.67 (hexane/EtOAc, 1:1); mp 56–57 °C. IR (film): ῡ 2997, 2949, 1742, 1692, 1626, 1474, 1290, 1171, 1080, 983, 745 cm−1. 1H NMR (400 MHz, CDCl3): δ 1.31 (t, J = 7.2 Hz, 3H, CO2CH2CH3), 3.77 (s, 3H, CO2CH3), 4.22 (q, J = 7.2 Hz, 2H, CO2CH2CH3), 4.75 (s, 2H, CH2), 6.16 (d, J = 15.6 Hz, 1H, H-2), 6.26–6.26 (ddd, J = 4.0, 3.0, 0.8 Hz, 1H, H-4′), 6.71 (dd, J = 4.0, 1.4 Hz, 1H, H-3′), 6.78 (dd, J = 3.0, 1.4 Hz, 1H, H-5′), 7.44 (d, J = 15.6 Hz, 1H, H-3). 13C NMR (100 MHz, CDCl3): δ 14.3 (CO2CH2CH3), 48.2 (CH2), 52.7 (CO2CH3), 60.2 (CO2CH2CH3), 110.4 (C-4′), 112.1 (C-3′), 114.0 (C-2), 126.7 (C-5′), 129.4 (C-2′), 131.3 (C-3), 167.5 (CO2CH2CH3), 168.4 (CO2CH3). HRMS (EI): m/z [M+] calcd. for C12H15NO4: 237.1001; found: 237.1001.
  • Methyl (Z)-3-(dimethylamino)-2-(2-((E)-3-methoxy-3-oxoprop-1-en-1-yl)-1H-pyrrol-1-yl)acrylate (12a). Following the method described for 1b, a mixture of 10a (0.119 g, 0.53 mmol) and DMFDMA (0.318 g, 2.67 mmol) in anhydrous DMF (1.0 mL) provided 12a (0.134 g, 90%) as a pale violet oil. Rf 0.33 (hexane/EtOAc, 7:3). IR (film): ῡ 2949, 1690, 1615, 1308, 1208, 1164, 1103, 1079, 1034 cm−1. 1H NMR (400 MHz, CDCl3): δ 2.70 (br, 6H, N(CH3)2), 3.62 (s, 3H, =CHCO2CH3), 3.73 (s, 3H, CO2CH3-1), 6.15 (d, J = 15.8 Hz, 1H, H-2″), 6.23 (ddd, J = 3.7, 2.4, 0.4 Hz, 1H, H-4′), 6.78 (dd, J = 3.7, 1.5 Hz, 1H, H-3′), 6.71 (dd, J = 2.4, 1.5 Hz, H-5′), 7.37 (d, J = 15.8 Hz, 1H, H-1″), 7.57 (s, 1H, H-3). 13C NMR (100 MHz, CDCl3): δ 35.2 (N(CH3)2), 46.7 (N(CH3)2), 51.3 (CO2CH3-1), 51.5 (=CHCO2CH3), 96.3 (C-2), 110.1 (C-4′), 111.7 (C-3′), 112.5 (C-2″), 130.1 (C-5′), 132.3 (C-2′), 133.6 (C-1″), 147.1 (C-3), 167.6 (CO2CH3-1), 168.2 (=CHCO2CH3). HRMS (EI): m/z [M+] calcd. for C14H18N2O4: 278.1267; found: 278.1263.
  • Ethyl (Z)-3-(dimethylamino)-2-(2-((E)-3-ethoxy-3-oxoprop-1-en-1-yl)-1H-pyrrol-1-yl)acrylate (12b). In a MW glass vial equipped with a magnetic stirring bar and sealed with a cap, a mixture of 10b (0.300 g, 1.20 mmol) and tert-butoxy bis(dimethylamino)methane (0.625 g, 3.60 mmol) was heated at 125 °C for 2.0 h under MW irradiation (200 W) and a N2 atmosphere. The crude mixture was suspended and stirred in CH2Cl2/toluene (10:1, 11 mL), and the solvent was removed under vacuum. The residue was purified by column chromatography over silica gel (hexane/EtOAc, 7:3), resulting in 12b (0.150 g, 41%) as a pale brown oil. Rf 0.52 (hexane/EtOAc, 1:1). IR (film): ῡ 2977, 1690, 1615, 1301, 1212, 1161, 1079, 1034 cm−1. 1H NMR (500 MHz, CDCl3): δ 1.16 (t, J = 7.0 Hz, 3H, CO2CH2CH3), 1.29 (t, J = 7.0 Hz, 3H, CO2CH2CH3), 2.29 (br, 3H, N(CH3)2), 3.01 (br, 3H, N(CH3)2), 4.05–4.17 (m, 2H, CO2CH2CH3), 4.17–4.23 (m, 2H, CO2CH2CH3), 6.15 (d, J = 15.5 Hz, 1H, H-2″), 6.23 (dd, J = 4.0, 2.5 Hz, 1H, H-4′), 6.67 (dd, J = 4.0, 1.5 Hz, 1H, H-3′), 6.71 (dd, J = 2.5, 1.5 Hz, 1H, H-5′), 7.37 (d, J = 15.5 Hz, 1H, H-1″), 7.56 (s, 1H, H-3). 13C NMR (125 MHz, CDCl3): δ 14.4 (CO2CH2CH3), 14.5 (CO2CH2CH3), 38.6 (N(CH3)2), 59.98 (CO2CH2CH3), 60.03 (CO2CH2CH3), 96.3 (C-2), 110.0 (C-4′), 111.6 (C-3′), 112.7 (C-2″), 130.1 (C-5′), 132.4 (C-2′), 133.4 (C-1″), 146.9 (C-3), 167.2 (CO2CH2CH3-1), 168.9 (=CHCO2CH3). HRMS (EI): m/z [M+] calcd. for C16H22N2O4: 306.1580; found: 306.1573.
  • Methyl (Z)-3-(dimethylamino)-2-(2-((E)-3-ethoxy-3-oxoprop-1-en-1-yl)-1H-pyrrol-1-yl)acrylate (12c). Following the method described for 1b, a mixture of 10c (0.060 g, 0.25 mmol) and DMFDMA (0.151 g, 1.25 mmol) in anhydrous DMF (2.0 mL) afforded 12c (0.068 g, 92%) as a pale violet oil. Rf 0.48 (hexane/EtOAc, 1:1). IR (film): ῡ 2923, 1697, 1618, 1308, 1212, 1158, 1079, 1308, 1034 cm−1. 1H NMR (500 MHz, CDCl3): δ 1.29 (t, J = 7.2 Hz, 3H, CO2CH2CH3), 2.26 (br, 3H, N(CH3)2), 3.05 (br, 3H, N(CH3)2), 3.63 (s, 3H, CO2CH3), 4.20 (q, J = 7.2 Hz, 2H, CO2CH2CH3), 6.15 (d, J = 16.0 Hz, 1H, H-2″), 6.24 (br d, J = 3.3 Hz, 1H, H-4′), 6.68 (br d, J = 3.3 Hz, 1H, H-3′), 6.71 (br t, J = 1.7 Hz, 1H, H-5′), 7.38 (d, J = 16.0 Hz, 1H, H-1″), 7.57 (s, 1H, H-3). 13C NMR (125 MHz, CDCl3): δ 14.4 (CO2CH2CH3), 35.0 (N(CH3)2), 47.4 (N(CH3)2), 51.5 (CO2CH3), 60.1 (CO2CH2CH3), 96.3 (C-2), 110.1 (C-4′), 111.6 (C-3′), 113.0 (C-2″), 130.1 (C-5′), 132.4 (C-2′), 133.3 (C-1″), 147.1 (C-3), 167.7 (CO2CH3), 167.8 (CO2CH2CH3). HRMS (EI): m/z [M+] calcd. for C15H20N2O4: 292.1423; found: 292.1414.
  • Dimethyl indolizine-5,7-dicarboxylate (5a). At rt and under N2 atmosphere, a solution of AlCl3 (0.043 g, 0.321 mmol) in nitrobenzene (1.0 M) was added to a solution of 12a (0.030 g, 0.17 mmol) in anhydrous CH2Cl2 (5.0 mL). The mixture was stirred at rt for 2 h before adding CH2Cl2 (15 mL). It was then washed with brine (5.0 mL × 3) and dried (Na2SO4), and the solvent was removed under vacuum. The residue was purified by column chromatography over silica gel (hexane/EtOAc, 9:1) to give 5a (0.024 g, 66%) as a yellow solid. Rf 0.80 (hexane/EtOAc, 1:1); mp 110–112 °C. IR (film): ῡ 2919, 1707, 1625, 1434, 1232, 1195, 1161, 1082, 758, 730 cm−1. 1H NMR (750 MHz, CDCl3): δ 3.94 (s, 3H, CO2CH3-5), 4.00 (s, 3H, CO2CH3-7), 6.95 (dd, J = 3.8, 0.9 Hz, 1H, H-1), 7.02 (dd, J = 3.8, 1.9 Hz, 1H, H-2), 8.17 (br d, J = 1.5 Hz, 1H, H-6), 8.41 (br d, J = 1.5 Hz, 1H, H-8), 8.84 (br s, 1H, H-3). 13C NMR (187.5 MHz, CDCl3): δ 52.2 (CO2CH3-7), 52.4 (CO2CH3-5), 107.6 (C-1), 116.0 (C-5), 116.7 (C-2), 118.0 (C-6), 119.4 (C-3), 123.0 (C-7), 127.2 (C-8), 133.5 (C-8a), 163.1 (CO2CH3-5), 165.9 (CO2CH3-7). HRMS (EI): m/z [M+] calcd. for C12H11NO4: 233.0688; found: 233.0687.
  • Diethyl indolizine-5,7-dicarboxylate (5b). Following the method described for 5a, a mixture of 12b (0.060 g, 0.20 mmol) and AlCl3 (0.078 g, 0.39 mmol) in anhydrous CH2Cl2 (2.0 mL) was stirred at rt for 2h. After the further addition of AlCl3 (0.078 g, 0.39 mmol), the mixture was stirred for 2 h to obtain 5b (0.016 g, 31%) as a yellow solid. Rf 0.81 (hexane/EtOAc, 7:3); mp 55–57 °C. IR (film): ῡ 2923, 2851, 1707, 1226, 1198, 1182, 1024 cm−1. 1H NMR (500 MHz, CDCl3): δ 1.42 (t, J = 7.0 Hz, 3H, CO2CH2CH3), 1.45 (t, J = 7.0 Hz, 3H, CO2CH2CH3), 4.39 (q, J = 7.0 Hz, 2H, CO2CH2CH3), 4.46 (q, J = 7.0 Hz, 2H, CO2CH2CH3), 6.94 (br d, J = 4.0 Hz, 1H, H-1), 7.02 (dd, J = 4.0, 3.0 Hz, 1H, H-2), 8.17 (d, J = 1.5 Hz, 1H, H-6), 8.41 (d, J = 1.5 Hz, 1H, H-8), 8.85 (br s, 1H, H-3). 13C NMR (125 MHz, CDCl3): δ 14.33 (CO2CH2CH3), 14.41 (CO2CH2CH3), 61.1 (CO2CH2CH3), 61.6 (CO2CH2CH3), 107.4 (C-1), 116.4 (C-5), 116.6 (C-2), 117.8 (C-6), 119.4 (C-3), 123.2 (C-7), 127.0 (C-8), 133.5 (C-8a), 162.7 (CO2Et), 165.6 (CO2Et). HRMS (EI): m/z [M+] calcd. for C12H11NO4: 233.0688; found: 233.0687.
  • 7-Ethyl 5-methyl indolizine-5,7-dicarboxylate (5c). Following the method described for 5a, a mixture of 12c (0.030 g, 0.10 mmol) and AlCl3 (0.028 g, 0.20 mmol) in anhydrous CH2Cl2 (2.0 mL) furnished 5c (0.013 g, 52%) as a yellow solid. Rf 0.87 (hexane/EtOAc, 1:1); mp 133–135 °C. IR (film): ῡ 2987, 1704, 1256, 1232, 1198, 1168, 1018, 751 cm−1. 1H NMR (600 MHz, CDCl3): δ 1.42 (t, J = 7.2 Hz, 3H, CO2CH2CH3), 4.00 (s, 3H, CO2CH3), 4.40 (q, J = 7.2 Hz, 2H, CO2CH2CH3), 6.94 (br d, J = 4.2 Hz, 1H, H-1), 7.02 (dd, J = 4.2, 2.0 Hz, 1H, H-2), 8.16 (br d, J = 1.8 Hz, H-6), 8.41 (s, 1H, H-8), 8.84 (br s, 1H, H-3). 13C NMR (150 MHz, CDCl3): δ 14.4 (CO2CH2CH3), 52.4 (CO2CH3), 61.1 (CO2CH2CH3), 107.5 (C-1), 116.4 (C-5), 116.6 (C-2), 118.1 (C-6), 119.4 (C-3), 123.0 (C-7), 127.1 (C-8), 133.5 (C-8a), 163.1 (CO2CH3), 165.5 (CO2CH2CH3). HRMS (EI): m/z [M+] calcd. for C13H13NO4: 247.0845; found: 247.0840.

3.3. Evaluation of Antifungal Activity

The pyrrole derivatives herein synthesized were submitted to the CLSI M27-A3 microdilution method, and an evaluation of the antifungal sensitivity of the various concentrations was carried out against Candida spp. (C. albicans ATCC 10231, C. glabrata CBS138, C. dubliniensis CD36, C. krusei ATCC 14423, C. auris Monterrey, and C. haemulonii ENCB87) [67]. The compounds were used at 187–0.01 μg/mL. The references were fluconazole, simvastatin, and atorvastatin. The inoculum of Candida spp. was adjusted in a spectrophotometer to 620 nm. Subsequently, a 1:1000 dilution was made with RPMI medium. The 96-well microplates were inoculated with 75 µL of yeast suspension and 75 µL of the concentration of the compound to be tested. RPMI served as the sterility control, and DMSO in the absence of an antifungal agent as the growth control. The microplates were incubated at 37 °C for 24 h, and growth was quantified in a microplate spectrophotometer at 620 nm. The values of yeast growth are expressed as the average of three independent assays.

3.4. Docking Studies

The sequences of the HMGR enzyme from each Candida spp., including Candida albicans (CaHMGR), C. glabrata (CgHMGR), C. dubliniensis (CdHMGR), C. krusei (CkHMGR), C. auris (CauHMGR), and C. haemulonii (ChaHMGR), were downloaded from the NCBI database [68]. The HMGR sequences were processed by the homology modeling technique to generate 3D models on the Modeller program version 10.2 [69]. The crystallized structure of the catalytic portion of human HMG-CoA reductase with simvastatin (PDB: 1HW9), deposited in the protein data bank (PDB) [70], served as a template. These 3D models were validated with the Procheck program [71] before being utilized in the docking studies, which were run on Autodock4 with the series of enaminones, N-alkyl pyrroles, pyrrolo[1,2-a]pyrazines, 2-methyl acrylates, and indolizines. The 2D structure of each ligand was sketched in the chemical editor ChemDraw [72] and converted to 3D in mol2 format with the Open Babel GUI program [73]. The structures of the reference compounds (simvastatin and atorvastatin) were downloaded from ZINC 20 [74] and optimized with Gaussian 16W software [75]. The instruction files were prepared in AutoDock Tools [76], setting a grid box of 80 × 62 × 58 Å, centered at: X = 23.123, Y = 9.113, and Z = 1.802, with a grid spacing of 0.375 Å3. In Autodock, the Lamarckian Genetic Algorithm was chosen, considering 100 docking trials, a population size of 150, a maximum number of energy evaluations of 25,000, a maximum number of generations of 27,000, and a mutation rate of 0.02. After docking, the coupling with the lowest binding energy (expressed in kcal/mol) was selected and analyzed with Biovia Discovery Studio 2017 R2 software [77].

4. Conclusions

New synthetic strategies are described for the preparation of pyrrolo[1,2-a]pyrazines and indolizines. The design of these approaches is centered on the cyclization of pyrrole-based enaminone precursors through treatment with ammonium acetate to afford pyrrolo[1,2-a]pyrazines, and on the application of a Lewis acid to enaminone precursors to furnish indolizines. All the intermediates and precursors were synthesized starting from 2-formylpyrrole (6), following short and efficient routes. The stereoselective introduction of the (Z)-enaminone moiety was achieved by reacting the N-alkyl pyrrole with DMFDMA under conventional heating or with Bredereck’s reagent under MW irradiation. The following series of the intermediates and cyclization products were: enaminone-containing pyrroles 1a, 1c, and 2ad, pyrrolo[1,2-a]pyrazines 4al, indolizines 5ac, N-alkyl 2-formylpyrroles 8ah, and pyrrole-based alkyl acrylates 8i, 10ac, 11, and 12ac, which were evaluated for their in vitro growth inhibition of six Candida spp. (C. albicans, C. glabrata, C. dubliniensis, C. krusei, C. auris, and C. haemulonii). The majority of these compounds demonstrated good inhibition of yeast growth. Docking simulations were carried out between the twelve most active compounds and the three most prevalent Candida spp. (C. albicans, C. glabrata, and C. auris). Insights were provided into the possible mechanism of action of the compounds evaluated. Polar and non-polar interactions between the functional groups of the derivatives and the active site of the HMGR enzyme of the three selected yeasts are involved, which have also been established for simvastatin and atorvastatin (the reference drugs). The lead compounds bear plausible pharmacophore groups responsible for the antifungal activity, which was found against all six Candida spp. analyzed, including the two multidrug-resistant species. Hence, the structural requirement is presently established in order that the most active analogues can be used in future studies on drug design and development.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28207223/s1. Figures S1–S76: 1H and 13C NMR spectra of all synthesized compounds; Figure S77: Acquisition and evaluation of the models of the HMGRs of the Candida spp; Figures S78–S83: Ramachandran plots of HMGRs; Tables S1 and S2: Data of the interactions between the simvastatin, atorvastatin, and compounds 1a, 2a, 2c, 4b, 4g, 4l, 5a, 8a, 8c, 8g, 10a, and 12a at the active site of the enzyme HMGR of C. glabrata (CgHMGR) and C. auris (CauHMGR); Figures S84 and 85: Representation of the interactions between simvastatin, atorvastatin, 1a, 2a, 2c, 4b, 4g, 4l, 5a, 8a, 8c, 8g, 10a, and 12a at the active site of CgHMGR and CauHMGR, respectively.

Author Contributions

Conceptualization, J.T.; writing—original draft preparation, J.T.; methodology, D.M.-S., C.H.E., D.A.-P., E.B. and L.V.-T.; software, O.G.-G.; formal analysis, F.D. and J.T., study investigation, D.M.-S., D.A.-P. and E.B.; data curation, O.G.-G., C.H.E. and E.B.; interpretation, D.M.-S., D.A.-P. and C.H.E.; supervision, J.T., F.D. and O.G.-G. All authors contributed to the writing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The research was supported by the Consejo Nacional de Ciencia y Tecnología (CONACYT, Mexico) (grant A1-S-17131) and SIP/IPN (grants 20200227, 20210700, 20221003, 20221599, 20221255, 20220742, and 20220900).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the Supplementary Materials.

Acknowledgments

We thank Elvia Becerra, Eder I. Martínez, Julio C. López, and the CNMN-IPN for their assistance in spectrometric measurements, and Bruce A. Larsen for proofreading. D.M.-S., C.H.E., and E.B. are grateful to CONACYT for graduate scholarships awarded and to SIP-IPN (PIFI), and to the Ludwig K. Hellweg Foundation for scholarship complements and postdoctoral fellowships. O.G.-G., L.V.-T., F.D. and J.T. are fellows of the Estímulos al Desempeño de los Investigadores (EDI)-IPN and Comisión de Operación y Fomento de Actividades Académicas (COFAA)-IPN programs.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not available.

References

  1. Dehnavi, F.; Alizadeh, S.R.; Ebrahimzadeh, M.A. Pyrrolopyrazine derivatives: Synthetic approaches and biological activities. Med. Chem. Res. 2021, 30, 1981–2006. [Google Scholar] [CrossRef]
  2. Karmakar, A.; Ramalingam, S.; Basha, M.; Indasi, G.K.; Belema, M.; Meanwell, N.A.; Dhar, T.G.M.; Rampulla, R.; Mathur, A.; Gupta, A.; et al. Facile access to 1,4-disubstituted pyrrolo[1,2-a]pyrazines from a-aminoacetonitriles. Synthesis 2020, 52, 441–449. [Google Scholar] [CrossRef]
  3. Mokrov, G.V.; Deeva, O.A.; Gudasheva, T.A.; Yarkov, S.A.; Yarkova, M.A.; Seredenin, S.B. Design, synthesis and anxiolytic-like activity of 1-arylpyrrolo[1,2-a]pyrazine-3-carboxamides. Bioorg. Med. Chem. 2015, 23, 3368–3378. [Google Scholar]
  4. Kim, J.; Park, M.; Choi, J.; Singh, D.K.; Kwon, H.J.; Kim, S.H.; Kim, I. Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives. Bioorg. Med. Chem. Lett. 2019, 29, 1350–1356. [Google Scholar] [CrossRef]
  5. Seo, Y.; Lee, J.H.; Park, S.; Namkung, W.; Kim, I. Expansion of chemical space based on a pyrrolo[1,2-a]pyrazine core: Synthesis and its anticancer activity in prostate cancer and breast cancer cells. Eur. J. Med. Chem. 2020, 188, 111988. [Google Scholar] [CrossRef] [PubMed]
  6. Vijay, K.; Devi, T.S.; Sree, K.K.; Elgorban, A.M.; Kumar, P.; Govarthanan, M.; Kavitha, T. In vitro screening and in silico prediction of antifungal metabolites from rhizobacterium Achromobacter kerstersii JKP9. Arch. Microbiol. 2020, 10, 2855–2864. [Google Scholar] [CrossRef] [PubMed]
  7. Ito, S.; Yamamoto, Y.; Nishikawa, T. A concise synthesis of peramine, a metabolite of endophytic fungi. Biosci. Biotech. Bioch. 2018, 82, 2053–2058. [Google Scholar] [CrossRef]
  8. Micheli, F.; Bertani, B.; Bozzoli, A.; Crippa, L.; Cavanni, P.; Di Fabio, R.; Donati, D.; Marzorati, P.; Merlo, G.; Paio, A.; et al. Phenylethynyl-pyrrolo[1,2-a]pyrazine: A new potent and selective tool in the mGluR5 antagonists arena. Bioorg. Med. Chem. Lett. 2008, 18, 1804–1809. [Google Scholar] [CrossRef] [PubMed]
  9. Melo, I.S.; Santos, S.N.; Rosa, L.H.; Parma, M.M.; Silva, L.J.; Queiroz, S.C.N.; Pellizari, V.H. Isolation and biological activities of an endophytic Mortierella alpina strain from the Antarctic moss Schistidium antarctici. Extremophiles 2014, 18, 15–23. [Google Scholar] [CrossRef]
  10. Rajivgandhi, G.; Vijayan, R.; Maruthupandy, M.; Vaseeharan, B.; Manoharan, N. Antibiofilm effect of Nocardiopsis sp. GRG1 (KT235640) compound against biofilm forming Gram negative bacteria on UTIs. Microb. Pathog. 2018, 118, 190–198. [Google Scholar] [CrossRef]
  11. Kiran, G.S.; Priyadharsini, S.; Sajayan, A.; Ravindran, A.; Selvin, J. An antibiotic agent pyrrolo[1,2-a]pyrazine-1,4-dione,hexahydro isolated from a marine bacteria Bacillus tequilensis MSI45 effectively controls multi-drug resistant Staphylococcus aureus. RSC Adv. 2018, 8, 17837. [Google Scholar] [CrossRef] [PubMed]
  12. Sharma, V.; Kumar, V. Indolizine: A biologically active moiety. Med. Chem. Res. 2014, 23, 3593–3606. [Google Scholar] [CrossRef]
  13. Singh, G.S.; Mmatli, E.E. Recent progress in synthesis and bioactivity studies of indolizines. Eur. J. Med. Chem. 2011, 46, 5237–5257. [Google Scholar] [CrossRef]
  14. Park, S.; Kim, E.H.; Kim, J.; Kim, S.H.; Kim, I. Biological evaluation of indolizine-chalcone hybrids as new anticancer agents. Eur. J. Med. Chem. 2018, 144, 435–443. [Google Scholar] [CrossRef]
  15. Huang, W.; Zuo, T.; Jin, H.; Liu, Z.; Yang, Z.; Yu, X.; Zhang, L.; Zhang, L. Design, synthesis, and biological evaluation of indolizine derivatives as HIV-1 VIF-ElonginC interaction inhibitors. Mol. Divers. 2013, 17, 221–243. [Google Scholar] [CrossRef]
  16. Dawood, K.M.; Abdel-Gawad, H.; Ellithey, M.; Mohamed, H.A.; Hegazi, B. Synthesis, anticonvulsant, and anti-inflammatory activities of some new benzofuran-based heterocycles. Arch. Pharm. Chem. Life Sci. 2006, 339, 133–140. [Google Scholar] [CrossRef] [PubMed]
  17. Gundersen, L.-L.; Malterud, K.E.; Negussie, A.H.; Rise, F.; Teklu, S.; Østby, O.B. Indolizines as novel potent inhibitors of 15-lipoxygenase. Bioorg. Med. Chem. 2003, 11, 5409–5415. [Google Scholar] [CrossRef]
  18. Gundersen, L.-L.; Charnock, C.; Negussie, A.H.; Rise, F.; Teklu, S. Synthesis of indolizine derivatives with selective antibacterial activity against Mycobacterium tuberculosis. Eur. J. Pharm. Sci. 2007, 30, 26–35. [Google Scholar] [CrossRef]
  19. Venugopala, K.N.; Chandrashekharappa, S.; Pillay, M.; Abdallah, H.H.; Mahomoodally, F.M.; Bhandary, S.; Chopra, D.; Attimarad, M.; Aldhubiad, B.E.; Nair, A.B.; et al. Computational, crystallographic studies, cytotoxicity and anti-tubercular activity of substituted 7-methoxy-indolizine analogues. PLoS ONE 2019, 14, e0217270. [Google Scholar] [CrossRef]
  20. Park, S.; Jung, Y.; Kim, I. Diversity-oriented decoration of pyrrolo[1,2-a]pyrazines. Tetrahedron 2014, 70, 7534–7550. [Google Scholar] [CrossRef]
  21. Sari, O.; Seybek, A.F.; Kaya, S.; Menges, N.; Erdem, S.S.; Balci, M. Mechanistic insights into the reaction of N-propargylated pyrrole- and indole-carbaldehyde with ammonia, alkyl amines, and branched amines: A synthetic and theoretical investigation. Eur. J. Org. Chem. 2019, 2019, 5261–5274. [Google Scholar] [CrossRef]
  22. Alfonsi, M.; Dell’Acqua, M.; Facoetti, D.; Arcadi, A.; Abbiati, G.; Rossi, E. Microwave-promoted synthesis of N-heterocycles by tandem imination/annulation of γ- and δ-ketoalkynes in the presence of ammonia. Eur. J. Org. Chem. 2009, 2009, 2852–2862. [Google Scholar] [CrossRef]
  23. Beccalli, E.M.; Broggini, G.; Martinelli, M.; Paladino, G. Pd-catalyzed intramolecular cyclization of pyrrolo-2-carboxamides: Regiodivergent routes to pyrrolo-pyrazines and pyrrolo-pyridines. Tetrahedron 2005, 61, 1077–1082. [Google Scholar] [CrossRef]
  24. Sobenina, L.N.; Sagitova, E.F.; Ushakov, I.A.; Trofimov, B.A. Transition-metal-free synthesis of pyrrolo[1,2-a]pyrazines via intramolecular cyclization of N-propargyl(pyrrolyl)enaminones. Synthesis 2017, 49, 4065–4081. [Google Scholar] [CrossRef]
  25. Kim, M.; Jung, Y.; Kim, I. Domino Knoevenagel condensation/intramolecular aldol cyclization route to diverse indolizines with densely functionalized pyridine units. J. Org. Chem. 2013, 78, 10395–10404. [Google Scholar] [CrossRef]
  26. Ge, Y.-Q.; Jia, J.; Yang, H.; Zhao, G.-L.; Zhan, F.-X.; Wang, J. A facile approach to indolizines via tandem reaction. Heterocycles 2009, 78, 725–736. [Google Scholar] [CrossRef]
  27. Lee, J.H.; Kim, I. Cycloaromatization approach to polysubstituted indolizines from 2-acetylpyrroles: Decoration of the pyridine unit. J. Org. Chem. 2013, 78, 1283–1288. [Google Scholar] [CrossRef] [PubMed]
  28. Park, S.; Kim, I. Electron-withdrawing group effect in aryl group of allyl bromides for the successful synthesis of indolizines via a novel [3+3] annulation approach. Tetrahedron 2015, 71, 1982–1991. [Google Scholar] [CrossRef]
  29. Zhong, W.; Zhu, H.; Zou, H. ‘One-pot’ cascade approach to 5,6-dihydroindolizines and indolizines from pyrrole-2-carbaldehydes and nitroethylenes. Tetrahedron 2017, 73, 3181–3187. [Google Scholar] [CrossRef]
  30. Escalante, C.H.; Carmona-Hernández, F.A.; Hernández-López, A.; Martínez-Mora, E.I.; Delgado, F.; Tamariz, J. Cascade synthesis of indolizines and pyrrolo[1,2-a]pyrazines from 2-formyl-1-propargylpyrroles. Org. Biomol. Chem. 2022, 20, 396–409. [Google Scholar] [CrossRef]
  31. Hazra, A.; Mondal, S.; Maity, A.; Naskar, S.; Saha, P.; Paira, R.; Sahu, K.B.; Paira, P.; Ghosh, S.; Sinha, C.; et al. Amberlite-IRA-402 (OH) ion exchange resin mediated synthesis of indolizines, pyrrolo[1,2-a]quinolines and isoquinolines: Antibacterial and antifungal evaluation of the products. Eur. J. Med. Chem. 2011, 46, 2132–2140. [Google Scholar] [CrossRef]
  32. Cruz, M.C.; Tamariz, J. An efficient synthesis of benzofurans and their application in the preparation of natural products of the genus Calea. Tetrahedron 2005, 61, 10061–10072. [Google Scholar] [CrossRef]
  33. Correa, C.; Cruz, M.C.; Jiménez, F.; Zepeda, L.G.; Tamariz, J. A new synthetic route of 2-aroyl- and 2-benzyl-benzofurans and their application in the total synthesis of a metabolite isolated from Dorstenia gigas. Aust. J. Chem. 2008, 61, 991–999. [Google Scholar] [CrossRef]
  34. Labarrios, E.; Jerezano, A.; Jiménez, F.; Cruz, M.C.; Delgado, F.; Zepeda, L.G.; Tamariz, J. Efficient synthetic approach to substituted benzo[b]furans and benzo[b]thiophenes by iodine-promoted cyclization of enaminones. J. Heterocycl. Chem. 2014, 51, 954–971. [Google Scholar] [CrossRef]
  35. Cruz, M.C.; Jiménez, F.; Delgado, F.; Tamariz, J. Regioselective and versatile synthesis of indoles via intramolecular Friedel-Crafts heteroannulation of enaminones. Synlett 2006, 2006, 749–755. [Google Scholar] [CrossRef]
  36. Jerezano, A.V.; Labarrios, E.M.; Jiménez, F.E.; Cruz, M.C.; Pazos, D.C.; Gutiérrez, R.U.; Delgado, F.; Tamariz, J. Iodine-mediated one-pot synthesis of indoles and 3-dimethylaminoindoles via annulation of enaminones. Arkivoc 2014, 2014, 18–53. [Google Scholar] [CrossRef]
  37. Stanovnik, B.; Svete, J. Alkyl 2-substituted 3-(dimethylamino)propenoates and related compounds—versatile reagents in heterocyclic chemistry. Synlett 2000, 2000, 1077–1091. [Google Scholar] [CrossRef]
  38. Gottam, S.; Gaddam, V.; Kanaparthy, S. Gold-catalyzed reactions using N-propargyl β-enaminones. Arkivoc 2022, 2002, 1–18. [Google Scholar] [CrossRef]
  39. Martínez-Mora, E.I.; Caracas, M.A.; Escalante, C.H.; Espinoza-Hicks, C.; Quiroz-Florentino, H.; Delgado, F.; Tamariz, J. 2-Formylpyrroles as building blocks in a divergent synthesis of pyrrolizines. Synthesis 2016, 48, 1055–1068. [Google Scholar] [CrossRef]
  40. Madrigal-Aguilar, D.A.; Gonzalez-Silva, A.; Rosales-Acosta, B.; Bautista-Crescencio, C.; Ortiz-Álvarez, J.; Escalante, C.H.; Sánchez-Navarrete, J.; Hernández-Rodríguez, C.; Chamorro-Cevallos, G.; Tamariz, J.; et al. Antifungal activity of fibrate-based compounds and substituted pyrroles that inhibit the enzyme 3-hydroxy-methyl-glutaryl-CoA reductase of Candida glabrata (CgHMGR), thus decreasing yeast viability and ergosterol synthesis. Microbiol. Spectr. 2022, 10, e01642-21. [Google Scholar] [CrossRef]
  41. Wang, M.-Z.; Xu, H.; Liu, T.-W.; Feng, Q.; Yu, S.-J.; Wang, S.-H.; Li, Z.-M. Design, synthesis and antifungal activities of novel pyrrole alkaloid analogs. Eur. J. Med. Chem. 2011, 46, 1463–1472. [Google Scholar] [CrossRef]
  42. Martínez-Mora, E.I.; Caracas, M.A.; Escalante, C.H.; Madrigal, D.A.; Quiroz-Florentino, H.; Delgado, F.; Tamariz, J. Divergent and selective functionalization of 2-formylpyrrole and its application in the total synthesis of the aglycone alkaloid pyrrolemarumine. J. Mex. Chem. Soc. 2016, 60, 23–33. [Google Scholar] [CrossRef]
  43. Pedersen, T.R.; Tobert, J.A. Simvastatin: A review. Expert Opin. Pharmacother. 2004, 5, 2583–2596. [Google Scholar] [CrossRef]
  44. Andrade-Pavón, D.; Ortiz-Álvarez, J.; Sánchez-Sandoval, E.; Tamariz, J.; Hernández-Rodríguez, C.; Ibarra, J.A.; Villa-Tanaca, L. Inhibition of recombinant enzyme 3-hydroxy-3-methylglutaryl-CoA reductase from Candida glabrata by α-asarone-based synthetic compounds as antifungal agents. J. Biotechnol. 2019, 292, 64–67. [Google Scholar] [CrossRef] [PubMed]
  45. Shi, N.; Zheng, Q.; Zhang, H. Molecular dynamics investigations of binding mechanism for triazoles inhibitors to CYP51. Front. Mol. Biosci. 2020, 7, 586540. [Google Scholar] [CrossRef] [PubMed]
  46. Ademe, M.; Girma, F. Candida auris: From multidrug resistance to pan-resistant strains. Infect. Drug Resist. 2020, 13, 1287–1294. [Google Scholar] [CrossRef] [PubMed]
  47. Ben-Ami, R.; Berman, J.; Novikov, A.; Bash, E.; Shachor-Meyouhas, Y.; Zakin, S.; Maor, Y.; Tarabia, J.; Schechner, V.; Adler, A.; et al. Multidrug-resistant Candida haemulonii and C. auris, Tel Aviv, Israel. Emerg. Infect. Dis. 2017, 23, 195–203. [Google Scholar] [CrossRef]
  48. Di Santo, R.; Tafi, A.; Costi, R.; Botta, M.; Artico, M.; Corelli, F.; Forte, M.; Caporuscio, F.; Angiolella, L.; Palamara, A.T. Antifungal agents. 11. N-Substituted derivatives of 1-[(aryl)(4-aryl-1H-pyrrol-3-yl)methyl]-1H-imidazole:  Synthesis, anti-Candida activity, and QSAR studies. J. Med. Chem. 2005, 48, 5140–5153. [Google Scholar] [CrossRef]
  49. Onnis, V.; De Logu, A.; Cocco, M.T.; Fadda, R.; Meleddu, R.; Congiu, C. 2-Acylhydrazino-5-arylpyrrole derivatives: Synthesis and antifungal activity evaluation. Eur. J. Med. Chem. 2009, 44, 1288–1295. [Google Scholar] [CrossRef] [PubMed]
  50. Sortino, M.; Garibotto, F.; Filho, V.C.; Gupta, M.; Enriz, R.; Zacchino, S. Antifungal, cytotoxic and SAR studies of a series of N-alkyl, N-aryl and N-alkylphenyl-1,4-pyrrolediones and related compounds. Bioorg. Med. Chem. 2011, 19, 2823–2834. [Google Scholar] [CrossRef]
  51. Yurttaş, L.; Özkay, Y.; Kaplancıklı, Z.A.; Tunalı, Y.; Karaca, H. Synthesis and antimicrobial activity of some new hydrazone-bridged thiazole-pyrrole derivatives. J. Enzym. Inhib. Med. Chem. 2013, 28, 830–835. [Google Scholar] [CrossRef] [PubMed]
  52. Kheder, N.A. Hydrazonoyl chlorides as precursors for synthesis of novel bis-pyrrole derivatives. Molecules 2016, 21, 326. [Google Scholar] [CrossRef] [PubMed]
  53. Hilmy, K.M.H.; Khalifa, M.M.A.; Hawata, M.A.A.; Keshk, R.M.A.; El-Torgman, A.A. Synthesis of new pyrrolo[2,3-d]pyrimidine derivatives as antibacterial and antifungal agents. Eur. J. Med. Chem. 2010, 45, 5243–5250. [Google Scholar] [CrossRef] [PubMed]
  54. Jose, G.; Kumara, T.H.S.; Sowmya, H.B.V.; Sriram, D.; Row, T.N.G.; Hosamani, A.A.; More, S.S.; Janardhan, B.; Harish, B.G.; Telkar, S.; et al. Synthesis, molecular docking, antimycobacterial and antimicrobial evaluation of new pyrrolo[3,2-c]pyridine Mannich bases. Eur. J. Med. Chem. 2017, 131, 275–288. [Google Scholar] [CrossRef]
  55. Wójcicka, A.; Becan, L.; Junka, A.; Bartoszewicz, M.; Secewicz, A.; Trynda, J.; Wietrzyk, J. Synthesis and biological activity of novel 6-phenyl-1H-pyrrolo[3,4-c]pyridine-1,3-dione derivatives. Acta Pol. Pharm. 2017, 74, 435–443. [Google Scholar] [PubMed]
  56. Bangalore, P.K.; Pedapati, R.K.; Pranathi, A.N.; Batchu, U.R.; Misra, S.; Estharala, M.; Sriram, D.; Kantevari, S. Aryl-n-hexanamide linked enaminones of usnic acid as promising antimicrobial agents. Mol. Divers. 2023, 27, 811–836. [Google Scholar] [CrossRef]
  57. Mendieta-Moctezuma, A.; Rugerio-Escalona, C.; Villa-Ruano, N.; Gutiérrez, R.U.; Jiménez-Montejo, F.E.; Fragoso-Vázquez, M.J.; Correa-Basurto, J.; Cruz-López, M.C.; Delgado, F.; Tamariz, J. Synthesis and biological evaluation of novel chromonyl enaminones as α-glucosidase inhibitors. Med. Chem. Res. 2019, 28, 831–848. [Google Scholar] [CrossRef]
  58. Shen, Y.; Sun, Y.; Sang, Z.; Sun, C.; Dai, Y.; Deng, Y. Synthesis, characterization, antibacterial and antifungal evaluation of novel monosaccharide esters. Molecules 2012, 17, 8661–8673. [Google Scholar] [CrossRef]
  59. Bao, L.; Wang, S.; Song, D.; Wang, J.; Cao, X.; Ke, S. Synthesis and bio-evaluation of natural butenolides-acrylate conjugates. Molecules 2019, 24, 1304. [Google Scholar] [CrossRef]
  60. Farghaly, A.-R.; El-Kashef, H. Pyrazoles and pyrazolo[4,3-e]pyrrolo[1,2-a]pyrazines, I. Synthesis and antimicrobial activity. Monatsh. Chem. 2005, 136, 217–227. [Google Scholar] [CrossRef]
  61. Faghih-Mirzaei, E.; Seifi, M.; Abaszadeh, M.; Zomorodian, K.; Helali, H. Design, synthesis, biological evaluation and molecular modeling study of novel indolizine-1-carbonitrile derivatives as potential anti-microbial agents. Iran. J. Pharm. Res. 2018, 17, 883–895. [Google Scholar] [PubMed]
  62. Meng, X.Y.; Zhang, H.X.; Mezei, M.; Cui, M. Molecular docking: A powerful approach for structure-based drug discovery. Curr. Comput. Aided Drug Des. 2011, 7, 146–157. [Google Scholar] [CrossRef]
  63. Feng, Y.; Yan, Y.; He, J.; Tao, H.; Wu, Q.; Huang, S.-Y. Docking and scoring for nucleic acid-ligand interactions: Principles and current status. Drug Discov. Today 2022, 27, 838–847. [Google Scholar] [CrossRef]
  64. Vakser, I.A. Protein-protein docking: From interaction to interactome. Biophys. J. 2014, 107, 1785–1793. [Google Scholar] [CrossRef] [PubMed]
  65. Andrade-Pavón, D.; Cuevas-Hernández, R.I.; Trujillo-Ferrara, J.G.; Hernández-Rodríguez, C.; Ibarra, J.A.; Villa-Tanaca, L. Recombinant 3-hydroxy 3-methyl glutaryl-CoA reductase from Candida glabrata (Rec-CgHMGR) obtained by heterologous expression, as a novel therapeutic target model for testing synthetic drugs. Appl. Biochem. Biotechnol. 2017, 182, 1478–1490. [Google Scholar] [CrossRef] [PubMed]
  66. Pucheta, A.; Mendieta, A.; Madrigal, D.A.; Hernández-Benitez, R.I.; Romero, L.; Garduño-Siciliano, L.; Rugerio-Escalona, C.; Cruz-López, M.C.; Jiménez, F.; Ramírez-Villalva, A.; et al. Synthesis and biological activity of fibrate-based acyl- and alkyl-phenoxyacetic methyl esters and 1,2-dihydroquinolines. Med. Chem. Res. 2020, 29, 459–478. [Google Scholar] [CrossRef]
  67. Cuenca-Estrella, M.; Lee-Yang, W.; Ciblak, M.A.; Arthington-Skaggs, B.A.; Mellado, E.; Warnock, D.W.; Rodríguez-Tudela, J.L. Comparative evaluation of NCCLS M27-A and EUCAST broth microdilution procedures for antifungal susceptibility testing of Candida species. Antimicrob. Agents Chemother. 2002, 46, 3644–4647. [Google Scholar] [CrossRef]
  68. Sharma, S.; Ciufo, S.; Starchenko, E.; Darji, D.; Chlumsky, L.; Karsch-Mizrachi, I.; Schoch, C.L. The NCBI BioCollections Database. Database 2018, 2018, 1–8. [Google Scholar] [CrossRef]
  69. Webb, B.; Sali, A. Comparative protein structure modeling using Modeller. Curr. Protoc. Protein Sci. 2016, 86, 2.9.1–2.9.37. [Google Scholar] [CrossRef]
  70. Berman, H.M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.N.; Weissig, H.; Shindyalov, I.N.; Bourne, P.E. The protein data bank. Nucleic Acids Res. 2000, 28, 235–242. [Google Scholar] [CrossRef]
  71. Laskowski, R.A.; MacArthur, M.W.; Moss, D.S.; Thornton, J.M. PROCHECK: A program to check the stereochemical quality of protein structures. J. Appl. Cryst. 1993, 26, 283–291. [Google Scholar] [CrossRef]
  72. Cousins, K.R. Computer review of ChemDraw Ultra 12.0. J. Am. Chem. Soc. 2011, 133, 8388. [Google Scholar] [CrossRef] [PubMed]
  73. O’Boyle, N.M.; Banck, M.; James, C.A.; Morley, C.; Vandermeersch, T.; Hutchison, G.R. Open Babel: An open chemical toolbox. J. Cheminform. 2011, 3, 33. [Google Scholar] [CrossRef] [PubMed]
  74. Irwin, J.J.; Tang, K.G.; Young, Y.; Dandarchuluun, C.; Wong, B.R.; Khurelbaatar, M.; Moroz, Y.S.; Mayfield, J.; Sayle, R.A. ZINC20—A free ultra large-scale chemical database for ligand discovery. J. Chem. Inf. Model. 2020, 60, 6065–6073. [Google Scholar] [CrossRef]
  75. Frisch, M.J.; Trucks, G.W.; Schlegel, H.B.; Scuseria, G.E.; Robb, M.A.; Cheeseman, J.R.; Montgomery, J.A., Jr.; Vreven, T.; Kudin, K.N.; Burant, J.C.; et al. Gaussian 98, version A.6; Gaussian, Inc.: Wallingford, CT, USA, 2004. [Google Scholar]
  76. Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J. Comput. Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef]
  77. Dassault Systèmes BIOVIA. Discovery Studio Modeling Environment; Release 2017 R2 Client; Dassault Systèmes: San Diego, CA, USA, 2017; Available online: https://discover.3ds.com/discovery-studio-visualizer-download (accessed on 8 October 2023).
Scheme 1. Previously reported synthetic procedures: (a) a Pd(II)-promoted vinyl cyclization to achieve pyrrolo[1,2-a]pyrazines; (b) a condensation process to afford indolizines; and (c) the use of 2-formylpyrroles as the starting material to obtain pyrrolo[1,2-a]pyrazines and indolizines. (d) With the current strategy, pyrrole-based enaminones (13) were the building block for the preparation of pyrrolo[1,2-a]pyrazines (4) and indolizines (5) [23,25,30].
Scheme 1. Previously reported synthetic procedures: (a) a Pd(II)-promoted vinyl cyclization to achieve pyrrolo[1,2-a]pyrazines; (b) a condensation process to afford indolizines; and (c) the use of 2-formylpyrroles as the starting material to obtain pyrrolo[1,2-a]pyrazines and indolizines. (d) With the current strategy, pyrrole-based enaminones (13) were the building block for the preparation of pyrrolo[1,2-a]pyrazines (4) and indolizines (5) [23,25,30].
Molecules 28 07223 sch001
Scheme 2. Preparation of 2-formylpyrrole-based enaminones 1ab, 2ad, and 3a.
Scheme 2. Preparation of 2-formylpyrrole-based enaminones 1ab, 2ad, and 3a.
Molecules 28 07223 sch002
Scheme 3. Preparation of pyrrolo[1,2-a]pyrazines 4a and 4c via a multicomponent procedure, and of 4h and 4i by the transformation of 4c and 8h, respectively.
Scheme 3. Preparation of pyrrolo[1,2-a]pyrazines 4a and 4c via a multicomponent procedure, and of 4h and 4i by the transformation of 4c and 8h, respectively.
Molecules 28 07223 sch003
Scheme 4. Preparation of pyrrolo[1,2-a]pyrazines 4j and 4k by bromination of 4a.
Scheme 4. Preparation of pyrrolo[1,2-a]pyrazines 4j and 4k by bromination of 4a.
Molecules 28 07223 sch004
Scheme 5. Preparation of pyrrolo[1,2-a]pyrazine 4l by transformation of pyrrole 9a.
Scheme 5. Preparation of pyrrolo[1,2-a]pyrazine 4l by transformation of pyrrole 9a.
Molecules 28 07223 sch005
Scheme 6. Conversion of the series 10ac into indolizines 5ac.
Scheme 6. Conversion of the series 10ac into indolizines 5ac.
Molecules 28 07223 sch006
Figure 1. Representation of the interactions between the active site of HMGR of C. albicans and simvastatin, atorvastatin, and enaminone-containing pyrroles 1a, 2a, and 2c, pyrrolo[1,2-a]pyrazines 4b, 4g, and 4l, indolizine 5a, N-alkyl 2-formylpyrroles 8a, 8c, and 8g, and pyrrole-based alkyl acrylates 10a and 12a. The 3D model shows the amino acid residues involved in ligand binding to the active site of the enzyme. In the 2D model, the following interactions are depicted with dotted lines: conventional hydrogen bond (dark green), carbon hydrogen (light green), π–sigma (purple), π–π T-shaped and π–π stacked (fuchsia), π–alkyl and alkyl (pink), π–anion (orange), and π–cation (yellow). The amino acids are represented in pink circles (for basic amino acids), orange (for acid), cyan (for polars), and yellow (for non-polar).
Figure 1. Representation of the interactions between the active site of HMGR of C. albicans and simvastatin, atorvastatin, and enaminone-containing pyrroles 1a, 2a, and 2c, pyrrolo[1,2-a]pyrazines 4b, 4g, and 4l, indolizine 5a, N-alkyl 2-formylpyrroles 8a, 8c, and 8g, and pyrrole-based alkyl acrylates 10a and 12a. The 3D model shows the amino acid residues involved in ligand binding to the active site of the enzyme. In the 2D model, the following interactions are depicted with dotted lines: conventional hydrogen bond (dark green), carbon hydrogen (light green), π–sigma (purple), π–π T-shaped and π–π stacked (fuchsia), π–alkyl and alkyl (pink), π–anion (orange), and π–cation (yellow). The amino acids are represented in pink circles (for basic amino acids), orange (for acid), cyan (for polars), and yellow (for non-polar).
Molecules 28 07223 g001aMolecules 28 07223 g001bMolecules 28 07223 g001cMolecules 28 07223 g001dMolecules 28 07223 g001e
Table 1. Reaction conditions and yields for the preparation of 4-substituted pyrrolo[1,2-a]pyrazines 4ag a.
Table 1. Reaction conditions and yields for the preparation of 4-substituted pyrrolo[1,2-a]pyrazines 4ag a.
Molecules 28 07223 i001
Entry1–3ZT (°C)t (h)4 (%) b
11aCO2Me7044a (90)
21bCO2Et8034b (66)
32aCOPh8034c (87)
42bCOC6H4-3-OMe7044d (83)
52cCOC6H4-4-OMe7044e (84)
62dCOC6H3-3,4-(OMe)27044f (60)
73aCN7044g (53)
a The reactions were carried out with the enaminones 13 (1.0 mol eq.), NH4OAc (3.0 mol eq.), and Li2CO3 (3.0 mol eq.) in anh. DMF. b After purification by column chromatography.
Table 2. MIC50 and MIC70 values for the inhibition of Candida spp. by the following compounds: enaminone-containing pyrroles 1a, 1c, and 2ad, pyrrolo[1,2-a]pyrazines 4al, indolizines 5ac, N-alkyl 2-formylpyrroles 8a–h, and pyrrole-based alkyl acrylates 8i, 10ac, 11, and 12ac a.
Table 2. MIC50 and MIC70 values for the inhibition of Candida spp. by the following compounds: enaminone-containing pyrroles 1a, 1c, and 2ad, pyrrolo[1,2-a]pyrazines 4al, indolizines 5ac, N-alkyl 2-formylpyrroles 8a–h, and pyrrole-based alkyl acrylates 8i, 10ac, 11, and 12ac a.
CompoundC. albicansC. glabrataC. dubliniensisC. kruseiC. aurisC. haemulonii
MIC50MIC70MIC50MIC70MIC50MIC70MIC50MIC70MIC50MIC70MIC50MIC70
µg/mLµg/mLµg/mLµg/mLµg/mLµg/mL
fluconazole1.401.805.607.201.401.805.607.20>44.8>57.6>44.8>57.6
simvastatin1.251.7515.0021.001.251.7540.0056.0010.0014.0020.0028.00
atorvastatin3.775.271.712.392.773.874.806.7215.321.428.0011.20
1a0.110.150.090.120.070.090.050.074.175.834.456.23
1c4.205.880.650.914.005.600.360.502.593.622.002.80
2a0.130.180.090.120.060.080.030.045.577.793.745.23
2b0.130.180.110.150.070.090.050.075.317.435.207.28
2c0.100.140.070.090.060.080.060.084.506.304.456.23
2d0.120.160.120.160.090.120.040.064.686.5512.4817.47
4a0.180.251.562.180.731.020.200.285.858.1911.7016.30
4b0.150.210.110.150.070.090.090.123.074.293.905.46
4c0.330.460.150.210.090.120.010.021.462.0418.7226.20
4d0.450.630.120.160.160.220.180.361.622.2511.7016.38
4e0.330.460.130.180.360.500.180.361.462.047.8010.92
4f0.180.250.160.220.090.120.360.5010.6314.8811.7016.38
4g0.070.090.180.250.090.120.140.196.158.616.889.63
4h0.230.320.120.160.090.120.120.162.203.084.926.88
4i0.080.110.150.210.010.020.280.3911.6816.3511.6816.35
4j0.220.300.110.150.220.040.050.0711.6816.3511.6816.35
4k0.100.140.080.110.070.090.180.363.905.4618.7226.20
4l0.040.050.110.150.060.080.040.065.577.794.926.88
5a0.250.170.120.160.120.160.120.164.005.603.124.36
5b0.500.701.822.541.582.214.005.603.104.342.553.57
5c0.500.702.002.802.453.430.450.632.603.643.244.53
8a0.100.140.240.330.030.040.060.081.462.047.2010.08
8b0.190.260.120.160.140.190.060.082.964.1418.7226.20
8c0.070.090.100.140.070.090.040.068.3511.6911.7016.38
8d0.200.280.030.040.050.070.050.076.158.6111.7016.38
8e0.280.390.090.120.060.080.040.069.0012.6010.6314.88
8f0.200.280.050.070.050.070.050.074.686.5510.6314.88
8g0.120.160.080.110.160.220.040.065.577.7916.7123.39
8h4.005.600.821.142.433.401.001.402.383.332.273.17
8i0.140.190.450.630.320.440.200.286.889.6310.6314.88
10a0.500.700.500.700.550.770.881.233.574.994.005.60
10b0.580.810.260.364.005.604.666.522.823.942.383.33
10c3.745.231.421.981.081.511.021.422.563.582.303.22
110.180.250.841.170.731.020.160.224.876.815.858.19
12a0.100.140.130.180.100.700.140.194.005.602.002.80
12b0.120.170.911.272.142.990.881.202.803.922.273.17
12c1.772.470.851.192.383.331.301.822.853.992.703.78
a MIC50 is the 50% inhibition of yeast growth. MIC70 is the 70% inhibition of yeast growth.
Table 3. Binding energy values (DG, kcal/mol) at the active site of the HMGR enzymes of six Candida species for simvastatin and atorvastatin and for the test compounds: enaminone-containing pyrroles 1a, 2a, and 2c, pyrrolo[1,2-a]pyrazines 4b, 4g, and 4l, indolizine 5a, N-alkyl 2-formylpyrroles 8a, 8c, and 8g, and pyrrole-based alkyl acrylates 10a and 12a a.
Table 3. Binding energy values (DG, kcal/mol) at the active site of the HMGR enzymes of six Candida species for simvastatin and atorvastatin and for the test compounds: enaminone-containing pyrroles 1a, 2a, and 2c, pyrrolo[1,2-a]pyrazines 4b, 4g, and 4l, indolizine 5a, N-alkyl 2-formylpyrroles 8a, 8c, and 8g, and pyrrole-based alkyl acrylates 10a and 12a a.
CompoundHMGR Enzymes of Candida spp.
C. albicansC. glabrataC. dubliniensisC. kruseiC. aurisC. haemulonii
simvastatin−6.12−6.30−6.57−6.06−6.51−6.18
atorvastatin−3.82−4.63−4.64−4.97−2.14−4.66
1a−6.21−5.26−5.42−6.55−5.47−5.34
2a−7.19−7.48−8.00−8.73−8.34−6.89
2c−7.14−6.34−8.05−6.88−7.98−7.35
4b−6.41−5.53−5.74−7.02−5.69−7.01
4g−5.74−5.18−5.18−6.07−5.18−6.22
4l−7.70−6.44−6.48−7.58−7.31−6.43
5a−7.22−6.65−6.73−7.04−6.71−5.57
8a−5.33−4.74−4.49−5.55−5.55−5.92
8c−8.24−6.77−6.81−8.32−7.06−7.49
8g−5.21−5.00−4.56−5.28−4.93−5.58
10a−6.42−5.88−5.92−6.52−6.11−4.93
12a−7.09−7.20−7.15−5.42−6.70−5.50
a C. albicans (CaHMGR), C. glabrata (CgHMGR), C. dubliniensis (CdHMGR), C. krusei (CkHMGR), C. auris (CauHMGR), and C. haemulonii (ChaHMGR).
Table 4. Data on the interactions of enaminones 1a, 2a, and 2c, pyrrolo[1,2-a]pyrazines 4b, 4g, and 4l, indolizine 5a, N-alkyl 2-formylpyrroles 8a, 8c, and 8g, and pyrrole-based alkyl acrylates 10a and 12a at the active site of the HMGR enzyme of C. albicans (CaHMGR).
Table 4. Data on the interactions of enaminones 1a, 2a, and 2c, pyrrolo[1,2-a]pyrazines 4b, 4g, and 4l, indolizine 5a, N-alkyl 2-formylpyrroles 8a, 8c, and 8g, and pyrrole-based alkyl acrylates 10a and 12a at the active site of the HMGR enzyme of C. albicans (CaHMGR).
CompoundResidues of the Enzyme Interacting with
the Ligand
Polar InteractionsHydrophobic
Interactions
simvastatinAla62, Thr95, Glu96, Gly97, Cys98, Arg127, Met192, Met194, Asn195, Asp227, Lys228, Gly302, Gln303, Asp304, Gln307, Gly343, Gly344 Asp227, Lys228,
Asp304
-
atorvastatinThr95, Glu96, Gly97, Met192, Met194, Asn195, Asp227, Lys228, Asn292, His289, Gly302, Gln303, Asp304, Gln307, Gly342, Gly343, Gly344, Leu389Asn195, Asp304Glu96, Met192, Met194, Lys228, Asp304
1aAla62, Cys63, Thr95, Glu96, Gly97, Met192, Gly302, Gln303, Asp304, Pro305, Gly343, Gly344 Asp304Cys63, Glu96
2aGlu96, Leu99, Arg127, Met194, Asn195, Ser221, Asp227, Lys228, Lys272, Ala288, His289, Ans292, Leu389Asp227, Lys228,
Asn292
Glu96, Met194, His289, Leu389
2cThr95, Glu96, Met192, Met194, Asn195, Asp227, Lys228, Gln303, Asp304, Pro305, Gly344Lys228Met192, Met194
4bThr95, Glu96, Met192, Gly302, Gln303, Asp304, Gln307, Glu337, Val338, Gly339, Ile341, Gly342, Gly343, Gly344, Thr345Gly339, Ile341, Gly344Met192, Asp304, Gly342, Gly343
4gLeu73, Ala93, Thr94, Thr95, Glu96, Thr295, Ala 296, Leu299, Gln303, Asp304, Pro305Thr95Ala296, Pro305
4lLeu73, Ala93, Thr94, Thr95, Glu96, Leu99, Asn292, Thr295, Ala296, Leu299, Gly302, Gln303, Asp304, pro305, Gly344Ala93, Thr94Leu73, Thr94, Ala296, Leu299, Pro305
5aThr95, Glu96, Met192, Met194, Asn195, Gly302, Gln303, Asp304, Gly339, Ile341, Gly342, Gly343, Gly344, Thr345Thr95, Asn195, Gly302, Gly343, Gly344Met192, Gly339, Gly342, Thr345
8aThr95, Glu96, Met192, Gly302, Gln303, Asp304, Gln307, Glu337, Val338, Gly339, Ile341 Gly342, Gly343, Gly344, Thr345Asp304, Thr95, Val338, Gly344Met192
8cThr95, Glu96, Met192, Met194, Asn195, Lys228, Gly302, Gln303, Asp304, Gly339, Ile341 Gly342, Gly343, Gly344, Thr345Thr95, Gly344Glu96, Met192
8gThr94, Thr95, Ala93, Glu96, Leu99, Asn292, Thr295, Ala296, Gln303, Pro305Thr94, Thr95, Thr295Ala296, Pro305
10aLeu73, Ala93, Thr94, Thr95, Glu96, Leu99, Asn292, Thr295, Ala296, Leu299, Gln303, Asp304, Pro305, Ala306Asn292, Thr295, Asp304Leu73, Thr94, Ala296, Leu299, Pro305
12aAla62, Thr95, Glu96, Gly97, Met192, Gly193, Met194, Asn195, Gly302, Gln303, Asp304, Ile341, Gly342, Gly343, Gly344Ala62, Gly97, Ile341Met192, Met194
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Miranda-Sánchez, D.; Escalante, C.H.; Andrade-Pavón, D.; Gómez-García, O.; Barrera, E.; Villa-Tanaca, L.; Delgado, F.; Tamariz, J. Pyrrole-Based Enaminones as Building Blocks for the Synthesis of Indolizines and Pyrrolo[1,2-a]pyrazines Showing Potent Antifungal Activity. Molecules 2023, 28, 7223. https://doi.org/10.3390/molecules28207223

AMA Style

Miranda-Sánchez D, Escalante CH, Andrade-Pavón D, Gómez-García O, Barrera E, Villa-Tanaca L, Delgado F, Tamariz J. Pyrrole-Based Enaminones as Building Blocks for the Synthesis of Indolizines and Pyrrolo[1,2-a]pyrazines Showing Potent Antifungal Activity. Molecules. 2023; 28(20):7223. https://doi.org/10.3390/molecules28207223

Chicago/Turabian Style

Miranda-Sánchez, Diter, Carlos H. Escalante, Dulce Andrade-Pavón, Omar Gómez-García, Edson Barrera, Lourdes Villa-Tanaca, Francisco Delgado, and Joaquín Tamariz. 2023. "Pyrrole-Based Enaminones as Building Blocks for the Synthesis of Indolizines and Pyrrolo[1,2-a]pyrazines Showing Potent Antifungal Activity" Molecules 28, no. 20: 7223. https://doi.org/10.3390/molecules28207223

Article Metrics

Back to TopTop