Next Article in Journal
Xanthium spinosum L. Extracts Inhibit Breast Cancer in Mice by Apoptosis Induction and Immune System Modulation
Next Article in Special Issue
Over 40 Years of Fosmidomycin Drug Research: A Comprehensive Review and Future Opportunities
Previous Article in Journal
Immunomodulatory, Anticancer, and Antimicrobial Effects of Rice Bran Grown in Iraq: An In Vitro and In Vivo Study
Previous Article in Special Issue
Adherent Bacteria and Parasiticidal Secretion Products of Human Cervicovaginal Microbiota-Associated Lactobacillus gasseri Confer Non-Identical Cell Protection against Trichomonas vaginalis-Induced Cell Detachment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

New Derivatives of the Multi-Stage Active Malaria Box Compound MMV030666 and Their Antiplasmodial Potencies

1
Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Schubertstraße 1, 8010 Graz, Austria
2
Pharmacognosy, Institute of Pharmaceutical Sciences, University of Graz, Beethovenstraße 8, 8010 Graz, Austria
3
Swiss Tropical and Public Health Institute, Kreuzstraße 2, Allschwil, CH-4123 Basel, Switzerland
4
Faculty of Philosophy and Natural Sciences, Swiss Tropical and Public Health Institute Petersplatz 1, University of Basel, CH-4003 Basel, Switzerland
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2022, 15(12), 1503; https://doi.org/10.3390/ph15121503
Submission received: 8 November 2022 / Revised: 29 November 2022 / Accepted: 30 November 2022 / Published: 2 December 2022

Abstract

:
MMV’s Malaria Box compound MMV030666 shows multi-stage activity against various strains of Plasmodium falciparum and lacks resistance development. To evaluate the importance of its diarylether partial structure, diarylthioethers and diphenylamines with varying substitution patterns were prepared. A number of evident structure-activity relationships were revealed. Physicochemical and pharmacokinetic parameters were determined experimentally (passive permeability) or calculated. Compared to the lead compound a diarylthioether was more active and less cytotoxic resulting in an excellent selectivity index of 850. In addition, pharmacokinetic and physicochemical parameters were improved.

Graphical Abstract

1. Introduction

With an estimated 241 million cases and 627,000 deaths in 2020 the hard-earned reduction in malaria casualties fell victim to disruptions in prevention and care due to the COVID-19 pandemic. Furthermore, the number of malaria endemic countries rose from 26 in 2000 to 47 in 2020 [1]. Malaria belongs to the infectious diseases and is caused by eukaryotic, single-celled protozoans of the species Plasmodium. Various strains are human pathogens with Plasmodium falciparum being the most dangerous and deadly [2]. Emerging resistance to the gold standard in malaria therapy, the artemisinins and their partner drugs in the WHO African region are a serious cause for concern, as these countries are among those most affected [3,4,5]. To this day, double or triple Artemisinin-based combination therapies show acceptable efficacy, nonetheless alternative treatments and orally applicable drugs with new modes of action are urgently needed to successfully fight the malaria parasite [6,7,8]. Vaccine development is a difficult task to undertake due to the parasite’s complex life cycle and multiple possible targets. RTS,S/AS01, the first vaccine against any parasitic diseases, shows promising features and activities, however only partial efficacy [9,10,11].
The Medicines for Malaria Venture (MMV) is one of many foundations that made it their business to find new strategies to combat the increasing risk of untreatable malaria. Therefore, they published results of a huge screening project, the so-called Malaria Box, a collection of 400 drug- and probe-like compounds with activities against various strains of Plasmodia [12,13,14]. One of these compounds, the 2-phenoxybenzamide 1, exhibits multi-stage activity against sexual, asexual and liver stages of P. falciparum and lacks resistance development in sub-lethal doses. It shows a metabolomic profile resembling atovaquone. On the one hand, it disrupts the mitochondrial electron transport-chain by inhibiting the dihydroorotate-dehydrogenase and the cytochrome bc1 complex, on the other hand, an interaction with the digestive vacuole was detected [15,16].
Within our first study, we reported the retrosynthetic preparation of the lead compound 1 via a multi-step synthesis. Furthermore, a series of derivatives was prepared to gain first insights into structure-activity relationships. Thereby, the importance of the 2-phenoxy-group, as well as the beneficial effects of its 4-fluoro substituent on the antiplasmodial activity were determined. A shift of the N-Boc-piperazinyl group from 2′ to 4′ position of the benzanilide of the benzamide further increased the activity of compounds. Replacement of the N-Boc-group with a N-pivaloyl-group enhanced the acid-stability of compounds, whereby only a slight loss in activity was detected (Figure 1) [17]. This paper deals with the preparation of diversely substituted diarylethers, diarylthioethers and diphenylamines to gain further insights into structure-activity relationships.

2. Results and Discussion

2.1. Chemistry

New derivatives of the lead structure 1 were obtained by firstly synthesizing the corresponding carboxylic acid as well as anilino derivatives and subsequently coupling these partial structures to the desired benzamides. Preparation of the substituted benzoic acids started by treating the respective anthranilic acid with sodium nitrite under acidic conditions yielding the diazonium salts. Subsequent Sandmeyer-like reaction with an aqueous potassium iodide solution gave the desired 2-iodo-benzoic acid derivatives 5, 6, 7 and 8 as light brown solids in mostly high yields [18]. In our last study, only 2-phenoxybenzamides were prepared. In order to evaluate the importance of the diarylether partial structure, corresponding diarylthioethers and diphenylamines were prepared. In the course of a copper-catalyzed Ullmann-like ether synthesis, the obtained iodo-benzoic acids 5, 6, 7 and 8 were coupled with phenols or anilines, respectively, giving diarylethers (912), a diarylthioether (13) as well as diphenylamines (14, 15) (Figure 2) [19]. Coupling was confirmed by the appearance of additional proton signals in the 1H NMR spectrum.
In order to obtain the 2- and 4-substituted derivatives of aniline 16, 17, 18 and 19, 1-fluoro-2-nitrobenzene and 1-fluoro-4-nitrobenzene were firstly treated with N-Boc-piperazine and potassium carbonate in dry DMSO in the course of a nucleophilic aromatic substitution yielding compounds 20 and 21 in high yields [20]. Their N-Boc-group was cleaved using trifluoroacetic acid in dry dichloromethane giving 1-(2-nitrophenyl)piperazine 22 and 1-(4-nitrophenyl)piperazine 23 [21]. Treatment of these piperazine derivatives with triethylamine and pivaloyl chloride in dry dichloromethane yielded the N-pivaloylpiperazinyl analogs 24 and 25 [22]. Subsequent reduction of the nitro group of compounds 20, 21, 24 and 25 with palladium in an atmosphere of hydrogen at the parr-apparatus gave the desired aromatic amines 16, 17, 18 and 19 (Figure 3) [23]. Successful reduction of the nitro group was detected in the 1H NMR spectrum by a shift of the aromatic protons to lower frequencies as well as by the appearance of a NH2-signal.
Amide synthesis of carboxylic acids and anilino derivatives was achieved using a combination of 2-chloro-N-methylpyridinium iodide (Mukaiyama reagent) and diisopropylethylamine (DIPEA) in dry dichloromethane (Figure 4) [24]. Efficient amide bond formation was detected in the 1H NMR spectrum. The NH resonance was shifted 6 ppm downfield.
Coupling of the benzoic acid derivatives 915 with the 2-substituted anilino derivatives 16 and 18 gave the N-[2-(4-N-Boc-piperazinyl)phenyl]benzamides and the N-[2-(4-N-pivaloyl-piperazinyl)phenyl]benzamides 2635. Reaction of the carboxylic acids with the 4-substituted aniline analogs 17 and 19 on the other hand, yielded the N-[4-(4-N-Boc-piperazinyl)phenyl]benzamides 3642, as well as the N-[4-(4-N-pivaloylpiperazinyl)phenyl]benzamides 4345.
In order to evaluate the influence of an amino-group compared to electron withdrawing trifluoromethyl or nitro groups in ring position 3 of the benzamide, compounds 46 and 47 were prepared. The nitro group of 28 and 38 was reduced with palladium in an atmosphere of hydrogen at the parr-apparatus yielding compounds 46 and 47 (Figure 5) [23]. The reduction was detected by the appearance of a singlet for two amino protons in the 1H NMR spectrum.

2.2. Antiplasmodial Activity and Cytotoxicity

All newly prepared compounds were tested for their in vitro activity against the chloroquine sensitive strain NF54 of P. falciparum. Cytotoxicity was determined using L-6 cells (rat skeletal myofibroblasts). Chloroquine and podophyllotoxin were used as standards. Results obtained are summarized in Table 1.
MMVs Malaria Box compound 1 exhibits sub micromolar antiplasmodial activity (PfNF54 IC50 = 0.4134 µM) and promising selectivity (S.I. = 316.9). Apart from the lead compound, its para-substituted analog 2 as well as their N-pivaloyl analogs 3 and 4 served as comparisons for the newly synthesized derivatives.
Replacing the 3-trifluoromethyl group of 1 with other electron-withdrawing groups or hydrogen yielded compounds 2628 with slightly decreased antiplasmodial activities (PfNF54 IC50 = 0.5908–0.6496 µM) and selectivity indices (S.I. = 153.9–245.5). However, their amino analog 46 showed reduced activity (PfNF54 IC50 = 1.010 µM) and selectivity (S.I. = 126.0). A stark loss of activities was observed in the groups of para-substituted analogs as well as in both N-pivaloyl groups. Substitution of the 3-trifluoromethyl group of 2 (PfNF54 IC50 = 0.2690 µM; S.I. = 461.0) by above-mentioned groups gave compounds 3638 and 47 with distinctly reduced activity (PfNF54 IC50 = 1.432–4.072 µM) and selectivity (S.I. = 8.676–59.88). The substitution of the 3-trifluoromethyl group of the N-pivaloyl analogs 3 (PfNF54 IC50 = 0.6172 µM; S.I. = 299.7) and 4 (PfNF54 IC50 = 0.5790 µM; S.I. = 171.8) by electron-withdrawing groups or hydrogen gave compounds 3335 (PfNF54 IC50 = 2.138–2.613 µM; S.I. = 7.275–43.75) and 4345 (PfNF54 IC50 = 1.498–5.615 µM; S.I. = 32.84–96.41), respectively, with markedly decreased antiplasmodial activities and selectivities.
Replacing the 2-(4-fluorophenoxy) group of 1 with a phenoxy group yielded compound 30 with moderate activity (PfNF54 IC50 = 1.131 µM) and selectivity (S.I. = 19.87). In comparison the anilino analogs 31 and 32 showed only slightly decreased activity (PfNF54 IC50 = 0.6142–0.6364 µM) and selectivity (S.I. = 193.8–209.6). However, the most promising variation in the pattern was the substitution with a 4-(fluorophenyl)sulfanyl group. Compound 29 exhibited distinctly improved selectivity (PfNF54 IC50 = 0.1946 µM) and activity (S.I. = 850.5). The same changes were observed for the para-substituted analogs. The 2-phenoxy derivate 40 (PfNF54 IC50 = 3.094 µM; S.I. = 31.78) was moderately active, but the anilino derivates 41 and 42 (PfNF54 IC50 = 0.5389–0.6336 µM; S.I. = 53.29–174.8) were only slightly less active than 2. Again, the 4-(fluorophenyl)sulfanyl derivate 39 (PfNF54 IC50 = 0.1494 µM; S.I. = 212.8) was the most active.
In summary, highest antiplasmodial activity and selectivity was observed for compounds with a trifluoromethyl group in ring position 3 and a 4-(fluorophenyl)sulfanyl or a 4-fluorophenoxy substituent in ring position 2 of the benzamide. The anilino moiety should be substituted by a 4-bocpiperazinyl group in ring positions 2 or 4.

2.3. Physicochemical and Pharmacokinetic Properties

In addition to in vitro activity and cytotoxicity tests, some key pharmacokinetic parameters were calculated (log P, log D, LE) or determined experimentally (Pe). Results obtained are summarized in Table 2. The log P and log D7.4 values of compounds range from 4.73 up to 7.68. Ligand efficiency (LE) is an important parameter in early drug development. It is defined as the maximum in vitro binding affinity achievable by ligands, that is 1.5 kcal per mole per heavy atom (HA, non-hydrogen atom). The higher the LE value, the higher the binding affinity [25,26]. The calculated values range from 0.200 up to 0.236 kcal/mol/HA. Compounds 29 and 39 with the most promising antiplasmodial activities also exhibit highest ligand efficiencies of 0.230 and 0.234 kcal/mol/HA, respectively.
The parallel artificial membrane permeability assay (PAMPA) is a fast and easy high-throughput assay to determine passive permeability of compounds through semi-permeable membranes (for example the blood-brain-barrier) without the influence of efflux pumps or transporter molecules. Permeability is defined using hydrochlorothiazide (Pe = 0.09 × 10−6 cm/s) and caffeine (Pe = 8.00 × 10−6 cm/s) as standards. Compounds with a permeability higher than 1.5 × 10−6 cm/s are considered to be highly permeable. Permeability through a semi-permeable membrane could be detected for all compounds except 26, 27, 33 and 44 due to insufficient solubility in DMSO and methanol. Compounds 29 and 39 with the highest antiplasmodial activity also exhibit very promising permeability of 4.31 and 3.77 × 10−6 cm/s, respectively. The unsubstituted 2-phenoxybenzamides 30 (Pe = 8.52 × 10−6 cm/s) and 40 (Pe = 5.57 × 10−6 cm/s) as well as the N-pivaloylpiperazinyl derivatives 34 (Pe = 6.92 × 10−6 cm/s) and 45 (Pe = 5.13 × 10−6 cm/s) exhibit the by far highest permeabilities.

3. Materials and Methods

3.1. Instrumentation and Chemicals

IR spectra were acquired using a Bruker Alpha Platinum ATR FTIR spectrometer (Bruker, Ettlingen, Germany) (preparation of KBr discs). HRMS: Q Exactive Hybrid Quadrupole-Orbitrap mass spectrometer (Thermo Fisher Scientific, Waltham, MA, USA) run by Thermo Q Exactive 2.9 (Thermo Fisher Scientific, Waltham, MA, USA) and Thermo XcaliburTM Software Version 4.4 (Thermo Fisher Scientific, Waltham, MA, USA). The structures of all new compounds were determined by one- and two-dimensional NMR spectroscopy using a Bruker Avance Neo 400 MHz spectrometer, 5 mm tubes and TMS as internal standard. Shifts in 1H NMR (400 MHz) and 13C NMR (100 MHz) are reported in ppm; 1H- and 13C-resonances were assigned using 1H,1H- and 1H,13C-correlation spectra and are numbered as given in Figure 4. Signal multiplicities are abbreviated as follows: br, broad; d, doublet; dd, doublet of doublets; ddd, doublet of doublet of doublets; dt, doublet of triplets; m, multiplet; q, quartet; t, triplet; td, triplet of doublet; s, singlet. Melting points were determined using an Electrothermal IA 9200 melting point apparatus (Fisher Scientific, Birmingham, UK).
Materials: thin layer chromatography (TLC): TLC plates silica gel 60 (F254 (Merck); column chromatography (CC): silica gel 60 (Merck 70–230 mesh, pore diameter 60 Å), flash silica gel (VWR 230–400 mesh, pore diameter 60 Å or Merck 230–400 mesh, pore diameter 60 Å); PAMPA: 96-well pre-coated Corning Gentest PAMPA plate system (Corning, Glendale, AZ, USA), 96-well UV-star Microplates (Greiner Bio-One, Kremsmünster, Austria), SpectraMax M3 UV plate-reader (Molecular Devices, San Jose, CA, USA), 1H NMR and 13C NMR spectra of new compounds are available in the Supplementary Materials Section (Figures S1–S27).

3.2. Syntheses

3.2.1. General Procedure for the Synthesis of Compounds 58

The corresponding anthranilic acid (6.00 mmol) was dissolved in (DMSO) (11 mL) and the solution was ice-cooled. Upon adding 11 mL of 30% aq sulfuric acid (H2SO4), the reaction mixture was stirred at 0 °C for 5 min. After that, the ice bath was removed and sodium nitrate (13.28 mmol) was added. The reaction mixture was stirred at room temperature for 2 h. Subsequently, a solution of KI (10.92 mmol) in 5 mL of demineralized water was added dropwise with a syringe via a septum. The mixture was stirred for another hour before adding a second portion of KI (6.00 mmol) dissolved in 3 mL of aqua dest. After stirring for 1 h at ambient temperature, 50 mL of ethyl acetate was added. The aqueous and organic phases were separated. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and the solvent was removed in vacuo. The respective residues were purified by recrystallization from water.
2-Iodo-3-(trifluoromethyl)benzoic acid (5): The reaction of 3-(trifluoromethyl)anthranilic acid (2.11 g (10.33 mmol)) dissolved in DMSO (17 mL) and 30% aq H2SO4 (17 mL) with NaNO2 (1.54 g (22.35 mmol)) and KI (4.74 g (28.54 mmol)) in water (17 mL) gave the raw benzoic acid. It was purified by recrystallization from water (10 mL) giving compound 5 as brownish solid (2.97 g (91%)). m.P. 134 °C. NMR data were in accordance with literature data [27].
2-Iodobenzoic acid (6): The reaction of anthranilic acid (831 mg (6.06 mmol)) dissolved in DMSO (11 mL) and 30% aq H2SO4 (11 mL) with NaNO2 (921 mg (13.35 mmol)) and KI (2.83 g (17.02 mmol)) in water (8 mL) gave the raw iodobenzoic acid. It was purified by recrystallization from water (7 mL) yielding compound 6 as brown solid (1.48 g (99%)). m.P. 160 °C. NMR data were in accordance with literature data [28].
3-Fluoro-2-iodobenzoic acid (7): The reaction of 2-amino-3-fluorobenzoic acid (313 mg (2.02 mmol)) dissolved in DMSO (4 mL) and 30% aq H2SO4 (4 mL) with NaNO2 (309 mg (4.48 mmol)) and KI (939 mg (5.66 mmol)) in water (3 mL) yielded the raw product. It was purified by recrystallization from water (4 mL) giving compound 7 as brown solid (245 mg (46%)). m.P. 153 °C. NMR data were in accordance with literature data [29].
2-Iodo-3-nitrobenzoic acid (8): The reaction of 2-amino-3-nitrobenzoic acid (558 mg (3.06 mmol)) dissolved in DMSO (6 mL) and 30% aq H2SO4 (6 mL) with NaNO2 (460 mg (6.67 mmol)) and KI (1.42 g (8.55 mmol)) in water (4 mL) gave the raw product. It was purified by recrystallization from water (5 mL) yielding compound 8 as brown solid 799 mg (89%)). m.P. 207 °C. NMR data were in accordance with literature data [30].

3.2.2. General Procedure for the Synthesis of Compounds 915

The corresponding 2-iodobenzoic acid derivative (4.00 mmol) was dissolved in dry DMF (32 mL). The respective phenol, thiophenol or aniline (4.20 mmol), catalytic amounts of copper (0.53 mmol) and copper (I) iodide (0.18 mmol), 1,8-diazabicyclo[5.4.0]undec-7-ene (12.00 mmol) and dry pyridine (0.80 mmol) were added. The reaction mixture was refluxed in an oil bath at 160 °C for 2–24 h. Then, it was cooled to ambient temperature and acidified with 2 N HCl to a pH of 1. Ice and dichloromethane were added. Phases were separated. The aqueous phase was extracted twice with dichloromethane. The organic phases were combined, washed with water and brine, dried over anhydrous sodium sulfate and filtered. The solvent was evaporated in vacuo. The crude products were subsequently purified by column chromatography.
2-(4-Fluorophenoxy)benzoic acid (9): The reaction of compound 6 (688 mg (2.77 mmol)) with 4-fluorophenol (334 mg (2.98 mmol)), copper (31 mg (0.49 mmol)), copper (I) iodide (37 mg (0.19 mmol)), DBU (1.27 g (8.32 mmol)) and dry pyridine (44 mg (0.56 mmol)) in dry DMF (23 mL) for 2 h gave the crude product. It was purified by column chromatography (flash silica gel, cyclohexane (CH)/ethyl acetate (EtAc)/EtOH/AcOH) 9:1 (3:1:0.08)) yielding compound 9 as white amorphous solid (386 mg (60%)). NMR data were in accordance with literature data [30].
3-Fluoro-2-(4-fluorophenoxy)benzoic acid (10): To a solution of compound 7 (595 mg (2.24 mmol)) in dry DMF (18 mL), 4-fluorophenol ((263 mg (2.35 mmol)), copper (19 mg (0.30 mmol)), copper (I) iodide (19 mg (0.10 mmol)), DBU (1.02 g (6.72 mmol)) and dry pyridine (36 mg (0.45 mmol)) were added. The reaction mixture was refluxed for 2 h giving the crude product. It was purified by column chromatography (silica gel, CH2Cl2/MeOH/AcOH 75:1:1) yielding compound 10 as yellow amorphous solid (240 mg (43%)). m.P. 134 °C. IR = 3427, 1703, 1504, 1468, 1268, 1222, 1190, 770; 1H NMR (CDCl3) δ = 6.84–6.88 (m, 2H, 2′-H, 6′-H), 6.95–7.01 (m, 2H, 3′-H, 5′-H), 7.29 (td, J = 8.1, 4.8 Hz, 1H, 5-H), 7.40 (ddd, J = 10.0, 8.3, 1.7 Hz, 1H, 4-H), 7.86 (dt, J = 7.9, 1.5 Hz, 1H, 6-H); 13C NMR (CDCl3) δ = 116.09 (d, J = 23.6 Hz, C-3′, C-5′), 116.72 (d, J = 8.2 Hz, C-2′, C-6′), 122.02 (d, J = 18.7 Hz, C-4), 125.44 (C-1), 125.69 (d, J = 7.5 Hz, C-5), 127.75 (d, J = 3.6 Hz, C-6), 143.08 (q, J = 13.1 Hz, C-2), 153.92 (t, J = 2.3 Hz, C-1′), 155.63 (d, J = 252 Hz, C-3), 158.44 (d, J = 241 Hz, C-4′), 167.73 (C=O); HRMS (ESI−) calcd for C13H7F2O3 [M − H]: 249.0363; found: 249.0364.
2-(4-Fluorophenoxy)-3-nitrobenzoic acid (11): The reaction of compound 8 (2.46 g (8.41 mmol)) with 4-fluorophenol (1.00 g (8.92 mmol)), copper (83 mg (1.31 mmol)), copper (I) iodide (76 mg (0.40 mmol)), DBU (3.84 g (25.23 mmol) and dry pyridine (132 mg (1.67 mmol)) in dry DMF (68 mL) for 2 h gave the crude product. It was purified by column chromatography (silica gel, CH2Cl2/MeOH/AcOH 149:1:1) yielding compound 11 as pale orange amorphous solid (1.52 g (65%)). IR = 3077, 1707, 1539, 1504, 1447, 1359, 1309, 1249, 1221, 1180, 1096, 829, 782, 695; 1H NMR (CDCl3) δ = 6.74–6.78 (m, 2H, 2′-H, 6′-H), 6.93–7.00 (m, 2H, 3′-H, 5′-H), 7.48 (t, J = 8.0 Hz, 1H, 5-H), 8.12 (dd, J = 8.1, 1.8 Hz, 1H, 4-H), 8.25 (dd, J = 7.9, 1.8 Hz, 1H, 6-H); 13C NMR (CDCl3) δ = 116.15 (d, J = 23.6 Hz, C-3′, C-5′), 116.65 (d, J = 8.4 Hz, C-2′, C-6′), 125.45 (C-5), 126.23 (C-1), 130.06 (C-4), 136.67 (C-6), 144.92 (C-3), 148.06 (C-2), 154.04 (d, J = 2.4 Hz, C-1′), 158.41 (d, J = 241 Hz, C-4′), 167.94 (C=O); HRMS (ESI−): calcd for C13H7FNO5 [M − H]: 276.0308; found: 276.0308.
2-Phenoxybenzoic acid (12): The reaction of compound 6 (992 mg (4.00 mmol)) with phenol (395 mg (4.2 mmol)), copper (34 mg (0.53 mmol)), copper (I) iodide (34 mg (0.18 mmol)), DBU (1.83 g (12.00 mmol)) and dry pyridine (63 mg (0.80 mmol)) in dry DMF (32 mL) gave the crude product. It was purified by column chromatography (silica gel, CH/EtAc/EtOH/AcOH 160:30:9.2:0.8) yielding compound 12 as white amorphous solid (554 mg (65%)). NMR data were in accordance with literature data [30].
2-[(4-Fluorophenyl)sulfanyl]-3-(trifluoromethyl)benzoic acid (13): To a solution of compound 5 (1.90 g (6.02 mmol)) in dry DMF (48 mL), 4-fluorothiophenol (806 mg (6.29 mmol)), copper (76 mg (1.20 mmol)), copper (I) iodide (60 mg (0.32 mmol)), DBU (2.74 g (17.99 mmol)) and dry pyridine (94 mg (1.19 mmol)) were added. The reaction mixture was refluxed for 24 h giving the crude product. It was purified by column chromatography (flash silica gel, CH2Cl2/MeOH/AcOH 149:1:1) yielding compound 13 as light brown amorphous solid (199 mg (21%)). IR = 3066, 1714, 1582, 1492, 1315, 1274, 1231, 1209, 1135, 827, 676; 1H NMR (DMSO-d6) δ = 7.04–7.14 (m, 4H, 2′-H, 3′-H, 5′-H, 6′-H), 7.75 (t, J = 7.8 Hz, 1H, 5-H), 7.88 (d, J = 7.8 Hz, 1H, 6-H), 7.99 (d, J = 7.8 Hz, 1H, 4-H), 13.74 (br, 1H, OH); 13C NMR (DMSO-d6) δ = 116.11 (d, J = 22.3 Hz, C-3′, C-5′), 123.28 (q, J = 274 Hz, CF3), 127.83 (C-2), 128.72 (q, J = 5.7 Hz, C-4), 130.00 (d, J = 8.2 Hz, C-2′, C-6′), 130.67 (C-5), 132.12 (d, J = 3.1 Hz, C-1′), 132.52 (C-6), 133.04 (q, J = 29.1 Hz, C-3), 143.31 (C-1), 160.78 (d, J = 244 Hz, C-4′), 167.86 (C=O); HRMS (ESI−): calcd for C14H7F4O2S [M − H]: 315.0109; found: 315.0114.
2-Anilino-3-(trifluoromethyl)benzoic acid (14): Reaction of compound 5 (1.26 g (4.00 mmol)) with aniline (391 mg (4.20 mmol)), copper (34 mg (0.53 mmol)), copper (I) iodide (34 mg (0.18 mmol)), DBU (1.83 mmol (12.00 mmol)) and dry pyridine (63 mg (0.80 mmol)) in dry DMF (32 mL) gave the crude product. It was purified by column chromatography (silica gel, CH2Cl2/MeOH/AcOH 29:1:0.3) yielding compound 14 as pale brown amorphous solid (231 mg (21%)). IR = 3344, 1674, 1594, 1497, 1453, 1317, 1253, 1136, 1090, 757, 665; 1H NMR (CDCl3) δ = 6.75–6.77 (m, 2H, 2′-H, 6′-H), 6.94–6.98 (m, 1H, 4′-H), 7.18–7.24 (m, 2H, 3′-H, 5′-H), 7.36 (t, J = 7.9 Hz, 1H, 5-H), 7.89 (dd, J = 7.9, 1.6 Hz, 1H, 4-H), 8.30 (dd, J = 7.9, 1.6 Hz, 1H, 6-H); 13C NMR (CDCl3) δ = 117.77 (C-2′, C-6′), 122.69 (C-4′), 123.49 (q, J = 274 Hz, CF3), 124.13 (C-5), 125.63 (C-1), 125.84 (q, J = 30.2 Hz, C-3), 129.32 (C-3′, C-5′), 132.55 (q, J = 5.2 Hz, C-4), 136.07 (C-6), 141.70 (br, C-2), 144.78 (C-1′), 168.62 (C=O); HRMS (ESI+): calcd for C14H11F3NO2+ [M + H]+: 282.0736; found: 282.0729.
2-(4-Fluoroanilino)-3-(trifluoromethyl)benzoic acid (15): Reaction of compound 5 (2.28 g (7.23 mmol)) with 4-fluoroaniline (707 mg (7.60 mmol)), copper (60 mg (0.96 mmol)), copper (I) iodide (62 mg (0.33 mmol)), DBU (3.31 g (21.00 mmol)) and dry pyridine (115 mg (1.45 mmol)) in dry DMF (58 mL) for 24 h gave the crude product. It was purified by column chromatography (flash silica gel, CH2Cl2/MeOH/AcOH 29:1:0.2) yielding compound 15 as yellow amorphous solid (564 mg (26%)). IR = 3330, 1671, 1594, 1508, 1445, 1308, 1264, 1219, 1171, 1128, 1090, 763, 685; 1H NMR (CDCl3) δ = 6.75–6.79 (m, 2H, 2′-H, 6′-H), 6.89–6.95 (m, 2H, 3′-H, 5′-H), 7.28 (t, J = 7.9 Hz, 1H, 5-H), 7.87 (dd, J = 7.9, 1.7 Hz, 1H, 4-H), 8.25 (dd, J = 7.9, 1.7 Hz, 1H, 6-H); 13C NMR (CDCl3) δ = 115.88 (d, J = 22.8 Hz, C-3′, C-5′), 120.36 (d, J = 8.0 Hz, C-2′, C-6′), 123.06 (C-5), 123.48 (q, J = 274 Hz, CF3), 123.79 (C-1), 124.78 (q, J = 30.3 Hz, C-3), 133.03 (q, J = 5.3 Hz, C-4), 136.12 (C-6), 140.93 (br, C-1′), 143.14 (C-2), 158.83 (d, J = 242 Hz, C-4′), 169.57 (C=O); HRMS (ESI+): calcd for C14H10F4NO2+ [M + H]+: 300.0642; found: 300.0635.

3.2.3. General Procedure for the Synthesis of Compounds 20 and 21

N-Boc-piperazine (14.00 mmol) and potassium carbonate (14 mmol) were suspended in dry DMSO (40 mL). The corresponding fluoronitrobenzene (7.00 mmol) was added and the suspension was refluxed at 80 °C for 72 h. The reaction mixture was cooled to ambient temperature and acidified with 2 N HCl to a pH of 1. Phases were separated. The aqueous phase was extracted with diethyl ether. The organic phases were combined, washed with ice water and brine, dried over anhydrous sodium sulfate and filtered. The solvent was evaporated in vacuo yielding the pure products.
tert-Butyl 4-(2-nitrophenyl)piperazine-1-carboxylate (20): The 1-fluoro-2-nitrobenzene (1.00 g (7.10 mmol)) was refluxed with N-Boc-piperazine (2.64 g (14.20 mmol)) and potassium carbonate (1.96 g (14.20 mmol)) in dry DMSO giving compound 20 as orange oil (2.07 g (95%)). It was used without further purification. NMR data were in accordance with literature data [23].
tert-Butyl 4-(4-nitrophenyl)piperazine-1-carboxylate (21): The reaction of 1-fluoro-4-nitrobenzene (988 mg (7.00 mmol)) with N-Boc-piperazine (2.69 g (14.44 mmol)) and potassium carbonate (1.94 g (14.02 mmol)) in dry DMSO yielded compound 21 as orange amorphous solid (2.07 g (96%)) which was used without further purification. NMR data were in accordance with literature data [31].

3.2.4. General Procedure for the Synthesis of Compounds 22 and 23

The corresponding N-Boc-piperazinyl derivative (1.00 mmol) was dissolved in dry dichloromethane (10 mL) and cooled to 0 °C in an ice bath. A solution of trifluoroacetic acid (30 mmol) in dry dichloromethane (3 mL) was added via a dropping funnel. After that, the ice bath was removed and the reaction mixture was stirred at room temperature for 24 h. Subsequently, the solvent and access trifluoroacetic acid were evaporated in vacuo. The residue was suspended in a solution of potassium carbonate (20 mmol) in water (6 mL). The aqueous suspension was extracted five times with dichloromethane/propan-2-ol 3:1. The combined organic phases were dried over anhydrous sodium sulfate and filtered. The solvent was evaporated in vacuo yielding the desired compounds as pure products.
1-(2-Nitrophenyl)piperazine (22): Compound 20 (2.24 g (7.30 mmol)) reacted with trifluoroacetic acid (5.00 g (43.80mmol)) in dry dichloromethane (95 mL) giving the protonated form of compound 22. A solution of potassium carbonate (6.06 g (43.80 mmol)) in water (88 mL) was added to obtain compound 22 as orange oil (1.36 g (90%)) which was used without further purification. NMR data were in accordance with literature data [32].
1-(4-Nitrophenyl)piperazine (23): The reaction of compound 21 (1.02 g (3.33 mmol)) with trifluoroacetic acid (4.45 g (39.00 mmol)) in dry dichloromethane (42 mL) gave the protonated product 23. Work-up with an aqueous solution of potassium carbonate (2.70 g (19.56 mmol)) in 40 mL of water yielded compound 23 as brownish amorphous solid (683 mg (99%)) which was used without further purification. NMR data were in accordance with literature data [33].

3.2.5. General Procedure for the Synthesis of Compounds 24 and 25

The corresponding piperazinyl derivative (2.00 mmol) was dissolved in dry dichloromethane (8 mL) and cooled to 0 °C in an ice bath. Dry triethylamine (3.00 mmol) was added dropwise. Pivaloyl chloride (2.10 mmol) was added with a syringe via a septum. The ice bath was removed and the reaction mixture was stirred at ambient temperature for 24 h. After that, water (30 mL) was added. The aqueous and organic phases were separated. The organic phase was washed with 2 N HCl, 8% aq NaHCO3 and brine, dried over anhydrous sodium sulfate and filtered. The solvent was evaporated in vacuo yielding the pivaloylpiperazine derivatives which were used without further purification.
2,2-Dimethyl-1-[4-(2-nitrophenyl)piperazin-1-yl]propan-1-one (24): The reaction of the piperazinyl derivative 22 (555 mg (2.68 mmol)) with dry triethylamine (815 mg (8.05 mmol)) and pivaloyl chloride (340 mg (2.82 mmol)) in dry dichloromethane (11 mL) yielded compound 24 as yellow amorphous solid (322 mg (69%)) which was used without further purification. NMR data were in accordance with literature data [17].
2,2-Dimethyl-1-[4-(4-nitrophenyl)piperazin-1-yl]propan-1-one (25): The reaction of compound 23 (414 mg (2.00 mmol)) with dry triethylamine (607 mg (6.00 mmol)) and pivaloyl chloride (253 mg (2.10 mmol)) in dry dichloromethane (8 mL) gave the pivaloylpiperazinyl derivative 25 as orange amorphous solid (513 mg (88%)) which was used without further purification. NMR data were in accordance with literature data [17].

3.2.6. General Procedure for the Synthesis of Compounds 1619, 46 and 47

To a solution of 15% (m/m) palladium on activated carbon in dry methanol (100 mL) the corresponding nitro compound (2.00 mmol) was added. Reduction of the nitro group was performed in an atmosphere of hydrogen (50 psi) at the parr apparatus at ambient temperature for 24 h. The reaction mixture was filtered and the solvent was removed in vacuo yielding the desired anilino-derivatives that were either purified by column chromatography or used without further purification.
tert-Butyl 4-(2-aminophenyl)piperazine-1-carboxylate (16): The nitro group of compound 20 (3.67 g (11.93 mmol)) was reduced with PdC (560 mg) in dry methanol (100 mL) to obtain the raw anilino derivative. It was purified by column chromatography (silica gel, CH2Cl2/MeOH 79:1) yielding compound 16 as pale brown amorphous solid (1.75 g (53%)). NMR data were in accordance with literature data [23].
tert-Butyl 4-(4-aminophenyl)piperazine-1-carboxylate (17): The reaction of compound 21 (1.98 g (6.45 mmol)) with PdC (299 mg) in dry methanol (100 mL) yielded the anilino derivative 17 as dark red oil (1.66 g (93%)) which was used without further purification. NMR data were in accordance with literature data [34].
1-[4-(2-Aminophenyl)piperazin-1-yl]-2,2-dimethylpropan-1-one (18): The nitro group of compound 24 (555 mg (1.90 mmol)) was reduced with PdC (111 mg) in dry methanol (90 mL) to obtain compound 18 as grey amorphous solid (367 mg (74%)) which was used without further purification. NMR data were in accordance with literature data [17].
1-[4-(4-Aminophenyl)piperazin-1-yl]-2,2-dimethylpropan-1-one (19): The nitro group of compound 25 (410 mg (1.41 mmol)) was reduced with PdC (69 mg) in dry methanol (100 mL) yielding the anilino derivative 19 as dark red oil (346 mg (94%)) which was used without further purification. NMR data were in accordance with literature data [17].
tert-Butyl 4-{2-[3-amino-2-(4-fluorophenoxy)benzamido]phenyl}piperazine-1-carboxylate (46): The reaction of compound 28 (294 mg (0.55 mmol)) with PdC (42 mg) in dry methanol (100 mL) gave the pure product 46 as white amorphous solid (258 mg (93%)). IR = 3354, 1692, 1619, 1498, 1452, 1365, 1249, 1191, 765; 1H NMR (CDCl3) δ = 1.50 (s, 9H, (CH3)3), 2.81–2.84 (m, 4H, N(CH2)2), 3.63 (br, 4H, N(CH2)2), 3.85 (s, 2H, NH2), 6.81–6.84 (m, 2H, 2′-H, 6′-H), 6.88–6.92 (m, 2H, 3′-H, 5′-H), 6.97 (dd, J = 8.0, 1.5 Hz, 1H, 4-H), 7.02–7.07 (m, 1H, 4″-H), 7.09–7.15 (m, 2H, 3″-H, 5″-H), 7.20 (t, J = 7.9 Hz, 1H, 5-H), 7.42 (dd, J = 7.8, 1.5 Hz, 1H, 6-H), 8.42 (d, J = 8.1 Hz, 1H, 6″-H), 9.70 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.42 ((CH3)3), 44.05 (N(CH2)2), 52.12 (N(CH2)2), 80.07 (CMe3), 116.26 (d, J = 8.1 Hz, C-2′, C-6′), 116.46 (d, J = 23.6 Hz, C-3′, C-5′), 119.58 (C-4), 120.14 (C-3″), 120.26 (C-6″), 120.32 (C-6), 124.00 (C-4″), 125.54 (C-5″), 126.66 (C-5), 133.49 (C-1″), 138.49 (C-2), 140.26 (C-3), 141.23 (C-2″), 152.77 (d, J = 2.4 Hz, C-1′), 154.72 (N(C=O)O), 158.33 (d, J = 243 Hz, C-4′), 163.28 ((C=O)NH); HRMS (ESI+): calcd for C28H32FN4O4+ [M + H]+: 507.2408; found: 507.2416.
tert-Butyl 4-{4-[3-amino-2-(4-fluorophenoxy)benzamido]phenyl}piperazine-1-carboxylate (47): The nitro group of compound 38 (157mg (0.29 mmol)) was reduced with PdC (22 mg) in dry methanol (80 mL) yielding the pure compound 47 as pale yellow amorphous solid (142 mg (96%)). IR = 3396, 1687, 1516, 1499, 1474, 1417, 1366, 1323, 1231, 1196, 830, 767; 1H NMR (CDCl3) δ = 1.48 (s, 9H, (CH3)3), 3.04–3.08 (m, 4H, N(CH2)2), 3.54–3.57 (m, 4H, N(CH2)2), 3.80 (s, 2H, NH2), 6.84–6.91 (m, 4H, 2′-H, 3″-H, 5″-H, 6′-H), 6.94–7.00 (m, 3H, 3′-H, 4-H, 5′-H), 7.20 (t, J = 7.8 Hz, 1H, 5-H), 7.35 (d, J = 8.6 Hz, 2H, 2″-H, 6″-H), 7.53 (d, J = 7.7 Hz, 1H, 6-H), 8.71 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.40 ((CH3)3), 43.57 (N(CH2)2), 49.75 (N(CH2)2), 79.89 (CMe3), 115.90 (d, J = 8.2 Hz, C-2′, C-6′), 116.70 (d, J = 23.6 Hz, C-3′, C-5′), 117.16 (C-3″, C-5″), 119.72 (C-4), 121.02 (C-6), 121.43 (C-2″, C-6″), 126.73 (C-5), 128.79 (C-1), 130.83 (C-1″), 138.01 (C-2), 148.30 (C-4″), 152.26 (d, J = 2.4 Hz, C-1′), 154.68 (N(C=O)O), 158.51 (d, J = 242 Hz, C-4′), 162.72 ((C=O)NH); HRMS (ESI−): calcd for C28H30FN4O4 [M − H]: 505.2251; found: 505.2251.

3.2.7. General Procedure for the Synthesis of Compounds 2645

The corresponding benzoic acid derivative (1.00 mmol) and the anilino derivative (1.00 mmol) were dissolved in dry dichloromethane (30 mL) and cooled in an ice bath to 0 °C. The Mukaiyama reagent (1.75 mmol) and DIPEA (5.00 mmol) were added. The reaction mixture was stirred at room temperature for 24–48 h. After that, 20% aq ammonium chloride (50 mL) was added. Phases were separated. The aqueous phase was extracted twice with EtAc. The organic phases were combined, washed with 8% aq NaHCO3 and brine, dried over anhydrous sodium sulfate and filtered. The solvent was evaporated in vacuo yielding the raw products that were subsequently purified.
tert-Butyl 4-{2-[2-(4-fluorophenoxy)benzamido]phenyl}piperazine-1-carboxylate (26): Reaction of the carboxylic acid 9 (238 mg (1.02 mmol) with the amine 16 (281 mg (1.01 mmol)), 2-chloro-N-methylpyridinium iodide (451 mg (1.77 mmol) and DIPEA (646 mg (5.00 mmol)) in dry dichloromethane (30 mL) gave the raw benzamide. It was purified by column chromatography (silica gel, CH/EtAc 6:1) yielding compound 26 as pale orange amorphous solid (363 mg (73%)). IR = 3318, 2971, 1694, 1662, 1589, 1516, 1501, 1453, 1395, 1364, 1309, 1277, 1203, 1114, 854, 766; 1H NMR (CDCl3) δ = 1.48 (s, 9H, (CH3)3), 2.75–2.79 (m, 4H, N(CH2)2), 3.32 (br, 4H, N(CH2)2), 6.79 (br d, J = 8.3 Hz, 1H, 3H), 7.06–7.13 (m, 6H, 2′-H, 3′-H, 3″-H, 4″-H, 5′-H, 6′-H), 7.17–7.25 (m, 2H, 5-H, 5″-H), 7.41 (td, J = 7.8, 1.7 Hz, 1H, 4-H), 8.34 (dd, J = 7.9, 1.7 Hz, 1H, 6-H), 8.60 (d, J = 8.1 Hz, 1H, 6″-H), 10.45 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.40 ((CH3)3), 43.73 (N(CH2)2), 52.21 (N(CH2)2), 79.90 (CMe3), 117.00 (d, J = 23.5 Hz, C-3′, C-5′), 117.32 (C-3), 120.35 (C-3″), 121.04 (C-6″), 122.39 (d, J = 8.4 Hz, C-2′, C-6′), 123.57 (C-5), 124.08 (C-1), 124.13 (C-4″), 125.59 (C-5″), 132.69 (C-6), 133.04 (C-4), 134.01 (C-1″), 141.99 (C-2″), 151.10 (d, J = 2.8 Hz, C-1′), 154.80 (N(C=O)O), 156.17 (N), 159.83 (d, J = 245 Hz, C-4′), 162.77 ((C=O)NH); HRMS (ESI+) calcd for C28H31FN3O4+ [M + H]+: 492.2299; found: 492.2301.
tert-Butyl 4-{4-[3-fluoro-2-(4-fluorophenoxy)benzamido]phenyl}piperazin-1-carboxylate (27): Reaction of the carboxylic acid 10 (100 mg (0.40 mmol)) with the amine 16 (111 mg (0.40 mmol)), 2-chloro-N-methylpyridinium iodide (179 mg (0.70 mmol)) and DIPEA (259 mg (2.00 mmol)) in dry dichloromethane (12 mL) gave the crude product. It was purified by column chromatography (flash silica gel, petroleum ether/EtAc 5.5:3) yielding compound 27 as white amorphous solid (62 mg (30%)). IR = 3441, 1692, 1668, 1501, 1455, 1267, 1178, 765; 1H NMR (CDCl3) δ = 1.50 (s, 9H, (CH3)3), 2.77–2.81 (m, 4H, N(CH2)2), 3.54 (br, 4H, N(CH2)2), 6.95 (br s, 2H, 3′-H, 5′-H), 6.97 (br s, 2H, 2′-H, 6′-H), 7.06–7.14 (m, 2H, 3″-H, 4″-H), 7.18 (br td, J = 7.4, 2.0 Hz, 1H, 5″-H), 7.29–7.35 (m, 2H, 4-H, 5-H), 8.00–8.02 (m, 1H, 6-H), 8.50 (d, J = 8.1 Hz, 1H, 6″-H), 10.07 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.43 ((CH3)3), 43.97 (N(CH2)2), 52.22 (N(CH2)2), 80.05 (CMe3), 116.42 (d, J = 23.7 Hz, C-3′, C-5′), 117.99 (d, J = 8.2 Hz, C-2′, C-6′), 120.36 (d, J = 18.6 Hz, C-4), 120.40 (C-3″), 120.75 (C-6″), 124.47 (C-4″), 125.58 (C-5″), 126.08 (d, J = 7.7 Hz, C-5), 126.87 (d, J = 3.5 Hz, C-6), 130.14 (C-1), 133.40 (C-1″), 141.44 (d, J = 12.6 Hz, C-2), 141.75 (C-2″), 153.52 (t, J = 2.1 Hz, C-1′), 154.72 (N(C=O)O), 155.11 (d, J = 252 Hz, C-3), 158.91 (d, J = 243 Hz, C-4′), 161.73 (d, J = 3.1 Hz, ((C=O)NH)); HRMS (ESI+): calcd for C28H30F2N3O4+ [M + H]+: 510.2199; found: 510.2191.
tert-Butyl 4-{2-[2-(4-fluorophenoxy)-3-nitrobenzamido]phenyl}piperazine-1-carboxylate (28): Reaction of the carboxylic acid 11 (567 mg (2.05 mmol)) with the aniline 16 (555 mg (2.00 mmol)), 2-chloro-N-methylpyridinium iodide (895 mg (3.50 mmol)) and DIPEA (1292 mg (10.00 mmol)) in dry dichloromethane (60 mL) gave the raw product. It was purified by column chromatography (silica gel, CH/EtAc 3:1) yielding compound 28 as pale green amorphous solid (634 mg (59%)). IR = 3321, 1688, 1594, 1523, 1498, 1452, 1422, 1364, 1249, 1177, 1130, 837, 775; 1H NMR (CDCl3) δ = 1.50 (s, 9H, (CH3)3), 2.78–2.81 (m, 4H, N(CH2)2), 3.58 (br, 4H, N(CH2)2), 6.79–6.83 (m, 2H, 2′-H, 6′-H), 6.90–6.95 (m, 2H, 3′-H, 5′-H), 7.08–7.18 (m, 3H, 3″-H, 4″-H, 5″-H), 7.54 (t, J = 8.0 Hz, 1H, 5-H), 8.07 (dd, J = 8.0, 1.4 Hz, 1H, 4-H), 8.35–8.41 (m, 2H, 6-H, 6″-H), 9.81 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.41 ((CH3)3), 44.20 (N(CH2)2), 52.29 (N(CH2)2), 80.15 (CMe3), 116.57 (d, J = 23.8 Hz, C-3′, C-5′), 117.36 (d, J = 8.3 Hz, C-2′, C-6′), 120.24 (C-6″), 120.61 (C-3″), 124.76 (C-4″), 125.76 (C-5″), 126.17 (C-5), 128.63 (C-4), 132.12 (C-1), 133.00 (C-1″), 136.07 (C-6), 141.48 (C-2″), 143.87 (C-3), 145.56 (C-2), 153.09 (d, J = 2.6 Hz, C-1′), 154.60 (N(C=O)O), 158.83 (d, J = 243 Hz, C-4′), 160.95 ((C=O)NH); HRMS (ESI+) calcd for C28H30FN4O6+ [M + H]+: 537.2149; found: 537.2155.
tert-Butyl 4-(2-{2-[(4-fluorophenyl)sulfanyl]-3-(trifluoromethyl)benz-amido}phenyl) piperazine-1-carboxylate (29): Reaction of the carboxylic acid 13 (235 mg (0.74 mmol)) with the amine 16 (212 mg (0.76 mmol)), 2-chloro-N-methylpyridinium iodide (339 mg (1.33 mmol)) and DIPEA (479 mg (3.70 mmol)) in dry dichloromethane (22 mL) for 48 h gave the raw product. It was purified by column chromatography (silica gel, CH/EtAc 4:1) yielding compound 29 as white amorphous solid (189 mg (44%)). IR = 3331, 2976, 1693, 1590, 1516, 1490, 1418, 1366, 1313, 1231, 1171, 828, 761, 687; 1H NMR (CDCl3) δ = 1.48 (s, 9H, (CH3)3), 2.78–2.81 (m, 4H, N(CH2)2), 3.47 (br, 4H, N(CH2)2), 6.75–6.79 (m, 2H, 3′-H, 5′-H), 7.05–7.13 (m, 3H, 2′-H, 4″-H, 6′-H), 7.15–7.22 (m, 2H, 3″-H, 5″-H), 7.58 (t, J = 7.7 Hz, 1H, 5-H), 7.75 (dd, J = 7.7, 1.4 Hz, 1H, 6-H), 7.89 (dd, J = 7.7, 1.4 Hz, 1H, 4-H), 8.34 (dd, J = 8.1, 1.5 Hz, 1H, 6″-H), 9.01 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.39 ((CH3)3), 44.14 (N(CH2)2), 52.37 (N(CH2)2), 80.11 (CMe3), 116.16 (d, J = 22.2 Hz, C-3′, C-5′), 119.24 (C-6″), 120.86 (C-3″), 123.26 (q, J = 274 Hz, CF3), 124.32 (C-4″), 126.04 (C-5″), 128.48 (q, J = 5.7 Hz, C-4), 129.40 (C-5), 130.77 (d, J = 3.3 Hz, C-1′), 131.21 (C-2), 132.50 (d, J = 8.3 Hz, C-2′, C-6′), 132.75 (C-6), 133.35 (C-1″), 134.27 (q, J = 29.7 Hz, C-3), 140.89 (C-2″), 144.33 (C-1), 154.62 (N(C=O)O), 161.98 (d, J = 248 Hz, C-4′), 164.81 ((C=O)-NH); HRMS (ESI+): calcd for C29H30F4N3O3S+ [M + H]+: 576.1938; found: 576.1925.
tert-Butyl 4-[2-(2-phenoxybenzamido)phenyl]piperazin-1-carboxylate (30): Reaction of the carboxylic acid 12 (263 mg (1.23 mmol)) with the amine 16 (341 mg (1.23 mmol)), 2-chloro-N-methylpyridinium iodide (549 mg (2.15 mmol)) and DIPEA (795 mg (6.15 mmol)) in dry dichloromethane (37 mL) gave the crude product. It was purified by column chromatography (silica gel, CH/EtAc 3:1) yielding compound 30 as white amorphous solid (192 mg (33%)). IR = 3313, 2970, 1695, 1656, 1591, 1513, 1452, 1393, 1363, 1308, 1276, 1215, 1161, 1114, 762; 1H NMR (CDCl3) δ = 1.48 (s, 9H, ((CH3)3)), 2.74–2.78 (m, 4H, N(CH2)2), 3.30 (br, 4H, N(CH2)2), 6.83 (d, J = 8.2 Hz, 1H, 3-H), 7.05–7.15 (m, 4H, 2′-H, 3″-H, 4″-H, 6′-H), 7.17–7.26 (m, 3H, 4′-H, 5-H, 5″-H), 7.36–7.43 (m, 3H, 3′-H, 4-H, 5′-H), 8.36 (dd, J = 7.9, 1.9 Hz, 1H, 6-H), 8.60 (d, J = 8.0 Hz, 1H, 6″-H), 10.51 (br s, 1H, NH); 13C NMR (CDCl3) δ = 28.42 ((CH3)3), 43.56 (N(CH2)2), 52.18 (N(CH2)2), 79.73 (CMe3), 117.74 (C-3), 120.30 (C-3″), 120.86 (C-2′, C-6′), 121.10 (C-6″), 123.42 (C-5), 124.07 (C-4″), 124.08 (C-1), 125.31 (C-4′), 125.48 (C-5″), 130.27 (C-3′, C-5′), 132.58 (C-6), 132.98 (C-4), 134.03 (C-1″), 142.09 (C-2″), 154.79 (N(C=O)O), 155.29 (C-1′), 156.08 (C-2), 162.88 ((C=O)NH); HRMS (ESI+): calcd for C28H32N3O4+ [M + H]+: 474.2387; found: 474.2376.
tert-Butyl 4-{2-[2-anilino-3-(trifluoromethyl)benzamido]phenyl}piperazin-1-carboxylate (31): Reaction of the carboxylic acid 14 (376 mg (1.34 mmol)) with the amine 16 (370 mg (1.34 mmol)), 2-chloro-N-methylpyridinium iodide (598 mg (2.34 mmol)) and DIPEA (862 mg (6.67 mmol)) in dry dichloromethane (40 mL) gave the crude product. It was purified by column chromatography (flash silica gel, CH2Cl2/MeOH 149:1) obtaining compound 31 as pale yellow oil (50 mg (7%)). IR = 3302, 1693, 1649, 1594, 1518, 1453, 1366, 1323, 1251, 1171, 1132, 748, 691; 1H NMR (CDCl3) δ = 1.50 (s, 9H, (CH3)3), 2.73–2.76 (m, 4H, N(CH2)2), 3.55 (br, 4H, N(CH2)2), 6.23 (br s, 1H, NH), 6.70–6.74 (m, 2H, 2′-H, 6′-H), 6.78–6.83 (m, 1H, 4′-H), 6.99–7.03 (m, 1H, 4″-H), 7.05–7.11 (m, 4H, 3′-H, 3″-H, 5′-H, 5″-H), 7.39 (br t, J = 7.9 Hz, 1H, 5-H), 7.83 (dd, J = 7.9, 1.7 Hz, 1H, 4-H), 8.08 (dd, J = 7.9, 1.7 Hz, 1H, 6-H), 8.21 (br d, J = 7.6 Hz, 1H, 6″-H), 9.79 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.41 ((CH3)3), 44.34 (N(CH2)2), 52.16 (N(CH2)2), 80.18 (CMe3), 116.39 (C-2′, C-6′), 119.76 (C-6″), 120.34 (C-3″), 121.93 (C-4′), 123.95 (q, J = 274 Hz, CF3), 124.05 (C-4″), 124.46 (C-5), 125.09 (q, J = 29.2 Hz, C-3), 125.66 (C-5″), 129.28 (C-3′, C-5′), 129.73 (q, J = 5.3 Hz, C-4), 132.87 (C-1), 133.20 (C-1″), 134.27 (C-6), 138.44 (q, J = 1.4 Hz, C-2), 141.13 (C-2″), 144.57 (C-1′), 154.64 (N(C=O)O), 164.07 ((C=O)NH); HRMS (ESI+): calcd for C29H32F3N4O3+ [M + H]+: 541.2421; found: 541.2409.
tert-Butyl 4-{2-[2-(4-fluoroanilino)-3-(trifluoromethyl)benzamido]phenyl}piperazin-1-carboxylate (32): Reaction of the carboxylic acid 15 (444 mg (1.48 mmol)) with the amine 16 (418 mg (1.50 mmol)), 2-chloro-N-methylpyridinium iodide (662 mg (2.58 mmol)) and DIPEA (957 mg (7.40 mmol)) in dry dichloromethane (45 mL) gave the crude product. It was purified by column chromatography (flash silica gel, CH2Cl2/MeOH 79+1) yielding compound 32 as pale yellow amorphous solid (22 mg (4%)). IR = 3328, 2977, 1693, 1591, 1508, 1453, 1366, 1319, 1169, 1034, 1001, 912, 824, 760, 690; 1H NMR (CDCl3) δ = 1.49 (s, 9H, (CH3)3), 2.75–2.78 (m, 4H, N(CH2)2), 3.54 (br, 4H, N(CH2)2), 6.26 (br s, 1H, NH), 6.67–6.71 (m, 2H, 2′-H, 6′-H), 6.76–6.81 (m, 2H, 3′-H, 5′-H), 7.01–7.12 (m, 3H, 3″-H, 4″-H, 5″-H), 7.37 (br t, J = 7.8 Hz, 1H, 5-H), 7.82 (dd, J = 7.8 Hz, 1H, 4-H), 8.03 (dd, J = 7.8, Hz, 1H, 6-H), 8.20 (d, J = 7.9 Hz, 1H, 6″-H), 9.68 (br s, 1H, NH); 13C NMR (CDCl3) δ = 28.40 ((CH3)3), 44.24 (N(CH2)2), 52.17 (N(CH2)2), 80.23 (CMe3), 115.90 (d, J = 22.9 Hz, C-3′, C-5′), 118.54 (d, J = 7.9 Hz, C-2′, C-6′), 119.64 (C-6″), 120.34 (C-3″), 123.93 (q, J = 273 Hz, CF3), 124.18 (C-5), 124.22 (C-4″), 124.69 (q, J = 29.4 Hz, C-3), 125.75 (C-5″), 129.80 (q, J = 5.2 Hz, C-4), 132.24 (C-1), 133.04 (C-1″), 134.13 (C-6), 139.10 (C-2), 140.78 (d, J = 2.6 Hz, C-1′), 141.05 (C-2″), 154.63 (N(C=O)O), 158.25 (d, J = 241 Hz, C-4′), 164.12 ((C=O)NH); HRMS (ESI+): calcd for C29H31F4N4O3+ [M + H]+: 559.2327; found: 559.2313.
N-{2-[4-(2,2-Dimethylpropanoyl)piperazin-1-yl]phenyl}-2-(4-fluorophenoxy)benzamid (33): Reaction of the carboxylic acid 9 (232 mg (1.00 mmol)) with the amine 18 (261 mg (1.00 mmol)), 2-chloro-N-methylpyridinium iodide (447 mg (1.75 mmol)) and DIPEA (646 mg (5.00 mmol)) in dry dichloromethane (30 mL) gave the crude product. It was purified by column chromatography (silica gel, CH/EtAc 3:1) yielding compound 33 as white amorphous solid (340 mg (72%)). IR = 3307, 1622, 1589, 1500, 1451, 1308, 1211, 1016, 750; 1H NMR (CDCl3) δ = 1.25 (s, 9H, (CH3)3), 2.78–2.82 (m, 4H, N(CH2)2), 3.51 (br, 4H, N(CH2)2), 6.79 (dd, J = 8.3, 1.1 Hz, 1H, 3-H), 7.07–7.14 (m, 6H, 2′-H, 3′-H, 3″-H, 4″-H, 5′-H, 6′-H), 7.19–7.27 (m, 2H, 5-H, 5″-H), 7.41 (ddd, J = 8.3, 7.3, 1.9 Hz, 1H, 4-H), 8.36 (dd, J = 7.9, 1.8 Hz, 1H, 6-H), 8.63 (dd, J = 8.3, 1.4 Hz, 1H, 6″-H), 10.50 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.31 ((CH3)3), 38.59 (CMe3), 45.05 (N(CH2)2), 52.50 (N(CH2)2), 117.08 (d, J = 24.0 Hz, C-3′, C-5′), 117.10 (C-3), 120.49 (C-3″), 121.07 (C-6″), 122.60 (d, J = 8.4 Hz, C-2′, C-6′), 123.57 (C-5), 123.86 (C-1), 124.14 (C-4″), 125.81 (C-5″), 132.78 (C-6), 133.10 (C-4), 134.13 (C-1″), 141.51 (C-2″), 151.00 (d, J = 2.9 Hz, C-1′), 156.23 (C-2), 159.89 (d, J = 246 Hz, C-4′), 162.75 ((C=O)NH), 176.42 (C=O); HRMS (ESI+): calcd for C28H31FN3O3+ [M + H]+: 476.2344; found: 476.2332.
N-{2-[4-(2,2-Dimethylpropanoyl)piperazin-1-yl]phenyl}-2-(4-fluorophenoxy)-3-nitrobenzamid (34): Reaction of the carboxylic acid 11 (150 mg (0.54 mmol)) with the amine 18 (141 mg (0.54 mmol)), 2-chloro-N-methylpyridinium iodide (243 mg (0.95 mmol)) and DIPEA (349 mg (2.70 mmol)) in dry dichloromethane (16 mL) gave the crude product. It was purified by column chromatography (flash silica gel, CH2Cl2/CH/EtAc 3:2:1) yielding compound 34 as pale green amorphous solid (172 mg (61%)). IR = 3441, 1630, 1519, 1499, 1449, 1361, 1184, 775; 1H NMR (CDCl3) δ = 1.33 (s, 9H, (CH3)3), 2.81–2.85 (m, 4H, N(CH2)2), 3.79 (br, 4H, N(CH2)2), 6.80–6.84 (m, 2H, 2′-H, 6′-H), 6.91–6.96 (m, 2H, 3′-H, 5′-H), 7.08–7.19 (m, 3H, 3″-H, 4″-H, 5″-H), 7.55 (t, J = 8.0 Hz, 1H, 5-H), 8.06 (dd, J = 8.0, 1.8 Hz, 1H, 4-H), 8.37–8.42 (m, 2H, 6-H, 6″-H), 9.84 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.43 ((CH3)3), 38.71 (CMe3), 45.48 (N(CH2)2), 52.55 (N(CH2)2), 116.64 (d, J = 23.8 Hz, C-3′, C-5′), 117.49 (d, J = 8.2 Hz, C-2′, C-6′), 120.37 (C-6″), 120.64 (C-3″), 124.81 (C-4″), 125.93 (C-5″), 126.20 (C-5), 128.67 (C-4), 131.98 (C-1), 133.04 (C-1″), 136.13 (C-6), 141.08 (C-2″), 143.92 (C-3), 145.63 (C-2), 153.04 (d, J = 2.6 Hz, C-1′), 158.89 (d, J = 244 Hz, C-4′), 160.93 ((C=O)NH), 176.48 (C=O); HRMS (ESI+): calcd for C28H30FN4O5+ [M + H]+: 521.2195; found: 521.2183.
N-{2-[4-(2,2-Dimethylpropanoyl)piperazin-1-yl]phenyl}-3-fluoro-2-(4-fluorophenoxy)benzamid (35): Reaction of the carboxylic acid 10 (100 mg (0.40 mmol)) with the amine 18 (105 mg (0.40 mmol)), 2-chloro-N-methylpyridinium iodide (179 mg (0.70 mmol)) and DIPEA (259 mg (2.00 mmol)) in dry dichloromethane (12 mL) gave the raw product. It was purified by column chromatography (flash silica gel, CH/EtAc 2.75:1) yielding compound 35 as white amorphous solid (114 mg (58%)). IR = 3331, 2928, 1670, 1631, 1579, 1499, 1454, 1267, 1184, 1015, 765; 1H NMR (CDCl3) δ = 1.31 (s, 9H, (CH3)3), 2.80–2.83 (m, 4H, N(CH2)2), 3.73 (br, 4H, N(CH2)2), 6.96–6.99 (m, 4H, 2′-H, 3′-H, 5′-H, 6′-H), 7.07–7.14 (m, 2H, 3″-H, 4″-H), 7.17–7.21 (m, 1H, 5″-H), 7.31–7.37 (m, 2H, 4-H, 5-H), 8.02–8.05 (m, 1H, 6-H), 8.52 (br d, J = 8.0 Hz, 1H, 6″-H), 10.11 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.42 ((CH3)3), 38.70 (CMe3), 45.32 (N(CH2)2), 52.49 (N(CH2)2), 116.46 (d, J = 23.7 Hz, C-3′, C-5′), 118.23 (d, J = 8.4 Hz, C-2′, C-6′), 120.42 (d, J = 18.8 Hz, C-4), 120.49 (C-3″), 120.83 (C-6″), 124.50 (C-4″), 125.78 (C-5″), 126.07 (d, J = 7.6 Hz, C-5), 126.96 (d, J = 3.3 Hz, C-6), 129.93 (C-1), 133.49 (C-1″), 141.32 (C-2″), 141.59 (d, J = 12.4 Hz, C-2), 153.50 (t, J = 2.2 Hz, C-1′), 155.08 (d, J = 252 Hz, C-3), 157.05 (d, J = 243 Hz, C-4′), 161.71 (d, J = 3.2 Hz, (C=O)NH), 176.50 (C=O); HRMS (ESI+): calcd for C28H30F2N3O3+ [M + H]+: 494.2250; found: 494.2236.
tert-Butyl 4-{4-[2-(4-fluorophenoxy)benzamido]phenyl}piperazine-1-carboxylate (36): Reaction of the carboxylic acid 9 (240 mg (1.03 mmol)) with the amine 17 (281 mg (1.01 mmol)), 2-chloro-N-methylpyridinium iodide (449 mg (1.76 mmol) and DIPEA (646 mg (5.00 mmol)) in dry dichloromethane (30 mL) gave the raw product. It was purified by column chromatography (silica gel, CH/EtAc 2:1) yielding compound 36 as pale brown amorphous solid (136 mg (27%)). IR = 3376, 1689, 1662, 1597, 1538, 1501, 1476, 1449, 1414, 1365, 1317, 1282, 1233, 1202, 1119, 924, 866, 763; 1H NMR (CDCl3) δ = 1.48 (s, 9H, (CH3)3), 3.06–3.09 (m, 4H, N(CH2)2), 3.56–3.59 (m, 4H, N(CH2)2), 6.83 (d, J = 8.1 Hz, 1H, 3-H), 6.89–6.92 (m, 2H, 3″-H, 5″-H), 7.06–7.13 (m, 4H, 2′-H, 3′-H, 4′-H, 5′-H), 7.22–7.26 (m, 1H, 5-H), 7.41 (td, J = 8.1, 1.7 Hz, 1H, 4-H), 7.50–7.53 (m, 2H, 2″-H, 6″-H), 8.32 (dd, J = 7.9, 1.7 Hz, 1H, 6-H), 9.42 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.41 ((CH3)3), 43.77 (N(CH2)2), 49.84 (N(CH2)2), 79.86 (CMe3), 116.94 (d, J = 23.6 Hz, C-3′, C-5′), 117.23 (C-3″, C-5″), 117.80 (C-3), 121.14 (d, J = 8.4 Hz, C-2′, C-6′), 121.68 (C-2″, C-6″), 123.96 (C-5), 124.07 (C-1), 131.09 (C-1″), 132.49 (C-6), 132.95 (C-4), 148.31 (C-4″), 151.07 (d, J = 2.7 Hz, C-1′), 154.66 (N(C=O)O), 155.42 (C-2), 159.64 (d, J = 244 Hz, C-4′), 162.33 ((C=O)NH); HRMS (ESI+) calcd for C28H31FN3O4+ [M + H]+: 492.2299; found: 492.2302.
tert-Butyl 4-{4-[3-fluoro-2-(4-fluorophenoxy)benzamido]phenyl}piperazine-1-carb-oxylate (37): Reaction of the carboxylic acid 10 (106 mg (0.42 mmol)) with the amine 17 (119 mg (0.43 mmol)), 2-chloro-N-methylpyridinium iodide (191 mg (0.75 mmol)) and DIPEA (273 mg (2.12 mmol)) in dry dichloromethane (13 mL) gave the raw product. It was purified by column chromatography (silica gel, CH2Cl2/EtAc 9:1) yielding compound 37 as light brown amorphous solid (92 mg (43%)). IR = 3276, 1688, 1654, 1601, 1517, 1501, 1468, 1415, 1365, 1326, 1266, 1232, 1183, 1122, 1002, 927, 881, 834, 768; 1H NMR (CDCl3) δ = 1.48 (s, 9H, (CH3)3), 3.06–3.09 (m, 4H, N(CH2)2), 3.55–3.58 (m, 4H, N(CH2)2), 6.87 (d, J = 8.9 Hz, 2H, 3″-H, 5″-H), 6.93–6.97 (m, 2H, 2′-H, 6′-H), 6.99–7.04 (m, 2H, 3′-H, 5′-H), 7.31–7.37 (m, 2H, 4-H, 5-H), 7.42 (d, J = 8.9 Hz, 2H, 2″-H, 6″-H), 8.05 (dd, J = 7.9, 1.9 Hz, 1H, 6-H), 9.02 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.41 ((CH3)3), 43.52 (N(CH2)2), 49.68 (N(CH2)2), 79.88 (CMe3), 116.59 (d, J = 23.7 Hz, C-3′, C-5′), 116.72 (d, J = 8.3 Hz, C-2′, C-6′), 117.12 (C-3″, C-5″), 120.24 (d, J = 18.4 Hz, C-4), 121.56 (C-2″, C-6″), 126.24 (d, J = 7.6 Hz, C-5), 127.08 (d, J = 3.3 Hz, C-6), 129.39 (C-1), 130.52 (C-1″), 140.40 (d, J = 12.8 Hz, C-2), 148.53 (C-4″), 153.00 (t, J = 2.2 Hz, C-1′), 154.66 (N(C=O)O), 154.98 (d, J = 252 Hz, C-3), 158.84 (d, J = 243 Hz, C-4′), 161.17 (d, J = 3.1 Hz, ((C=O)NH)); HRMS (ESI+): calcd for C28H30F2N3O4+ [M + H]+: 510.2199; found: 510.2190.
tert-Butyl 4-{4-[2-(4-fluorophenoxy)-3-nitrobenzamido]phenyl}piperazine-1-carb-oxylate (38): Reaction of the carboxylic acid 11 (562 mg (2.03 mmol)) with the amine 17 (557 mg (2.01 mmol)), 2-chloro-N-methylpyridinium iodide (898 mg (3.51 mmol)) and DIPEA (1292 mg (10.00 mmol)) in dry dichloromethane (60 mL) gave the raw product. It was purified by column chromatography (silica gel, CH2Cl2/acetonitrile 12:1) yielding compound 38 as yellow amorphous solid (415 mg (39%)). IR = 3422, 1672, 1534, 1500, 1418, 1365, 1229, 1174, 828, 776; 1H NMR (CDCl3) δ = 1.48 (s, 9H, (CH3)3), 3.06–3.10 (m, 4H, N(CH2)2), 3.54–3.58 (m, 4H, N(CH2)2), 6.83–6.88 (m, 4H, 2′-H, 3″-H, 5″-H, 6′-H), 6.96–7.01 (m, 2H, 3′-H, 5′-H), 7.32 (d, J = 8.8 Hz, 2H, 2″-H, 6″-H), 7.54 (t, J = 8.0 Hz, 1H, 5-H), 8.07 (dd, J = 8.1, 1.7 Hz, 1H, 4-H), 8.43 (dd, J = 7.9, 1.7 Hz, 1H, 6-H), 8.66 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.40 ((CH3)3), 43.48 (N(CH2)2), 49.51 (N(CH2)2), 79.93 (CMe3), 116.59 (d, J = 8.3 Hz, C-2′, C-6′), 116.81 (d, J = 22.9 Hz, C-3′, C-5′), 117.00 (C-3″, C-5″), 121.70 (C-2″, C-6″), 126.30 (C-5), 128.60 (C-4), 129.80 (C-1″), 131.13 (C-1), 136.57 (C-6), 143.82 (C-3), 144.96 (C-2), 148.80 (C-4″), 152.85 (d, J = 2.5 Hz, C-1′), 154.65 (N(C=O)O), 158.89 (d, J = 243 Hz, C-4′), 160.47 ((C=O)NH); HRMS (ESI−) calcd for C28H28FN4O6 [M − H]: 535.1993; found: 535.1989.
tert-Butyl 4-(4-{2-[(4-fluorophenyl)sulfanyl]-3-(trifluoromethyl)benzamido}phenyl) piperazine-1-carboxylate (39): Reaction of the carboxylic acid 13 (212 mg (0.67 mmol)) with the amine 17 (190 mg (0.69 mmol)), 2-chloro-N-methylpyridinium iodide (300 mg (1.17 mmol)) and DIPEA (433 mg (3.35 mmol)) in dry dichloromethane (20 mL) gave the raw product. It was purified by column chromatography (silica gel, CH2Cl2/MeOH 79:1) yielding compound 39 as pale brown amorphous solid (66 mg (17%)). IR = 3424, 1656, 1518, 1423, 1313, 1231, 1128, 830; 1H NMR (CDCl3) δ = 1.49 (s, 9H, (CH3)3), 3.09–3.12 (m, 4H, N(CH2)2), 3.57–3.60 (m, 4H, N(CH2)2), 6.82–6.90 (m, 4H, 3′-H, 3″-H, 5′-H, 5″-H), 7.06–7.10 (m, 2H, 2′-H, 6′-H), 7.26–7.30 (m, 2H, 2″-H, 6″-H), 7.58 (t, J = 7.8 Hz, 1H, 5-H), 7.70 (s, 1H, NH), 7.84 (d, J = 7.8 Hz, 1H, 6-H), 7.89 (d, J = 7.8 Hz, 1H, 4-H); 13C NMR (CDCl3) δ = 28.42 ((CH3)3), 43.40 (N(CH2)2), 49.67 (N(CH2)2), 79.94 (CMe3), 116.38 (d, J = 22.2 Hz, C-3′, C-5′), 117.08 (C-3″, C-5″), 120.96 (C-2″, C-6″), 123.25 (q, J = 274 Hz, CF3), 128.55 (q, J = 5.7 Hz, C-4), 129.50 (C-5), 130.25 (C-1″), 130.36 (C-2), 130.92 (d, J = 3.2 Hz, C-1′), 131.66 (d, J = 8.1 Hz, C-2′, C-6′), 133.67 (C-6), 134.32 (q, J = 29.5 Hz, C-3), 143.80 (C-1), 148.53 (C-4″), 154.68 (N(C=O)O), 161.95 (d, J = 248 Hz, C-4′), 164.48 ((C=O)NH); HRMS (ESI+): calcd for C29H30F4N3O3S+ [M + H]+: 576.1938; found: 576.1927.
tert-Butyl 4-[4-(2-phenoxybenzamido)phenyl]piperazin-1-carboxylate (40): Reaction of the carboxylic acid 12 (291 mg (1.36 mmol)) with the amine 17 (378 mg (1.36 mmol)), 2-chloro-N-methylpyridinium iodide (608 mg (2.38 mmol)) and DIPEA (879 mg (6.80 mmol)) in dry dichloromethane (40 mL) gave the crude product. It was purified by column chromatography (silica gel, CH/EtAc 2.5:1) yielding compound 40 as white amorphous solid (262 mg (40%)). IR = 3372, 1691, 1662, 1598, 1537, 1514, 1475, 1449, 1415, 1364, 1317, 1282, 1217, 1163, 1119, 923, 829, 753; 1H NMR (CDCl3) δ = 1.48 (s, 9H, ((CH3)3)), 3.06–3.09 (m, 4H, N(CH2)2), 3.55–3.58 (m, 4H, N(CH2)2), 6.87–6.91 (m, 3H, 3-H, 3″-H, 5″-H), 7.11 (d, J = 7.9 Hz, 2H, 2′-H, 6′-H), 7.20–7.27 (m, 2H, 4′-H, 5-H), 7.40–7.44 (m, 3H, 3′-H, 4-H, 5′-H), 7.51 (d, J = 8.9 Hz, 2H, 2″-H, 6″-H), 8.33 (d, J = 7.9, 1.8 Hz, 1H, 6-H), 9.51 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.41 ((CH3)3), 43.52 (N(CH2)2), 49.86 (N(CH2)2), 79.85 (CMe3), 117.23 (C-3″, C-5″), 118.44 (C-3), 119.46 (C-2′, C-6′), 121.66 (C-2″, C-6″), 123.95 (C-5), 124.24 (C-1), 124.85 (C-4′), 130.27 (C-3′, C-5′), 131.20 (C-1″), 132.41 (C-6), 132.90 (C-4), 148.24 (C-4″), 154.67 (N(C=O)O), 155.14 (C-2), 155.36 (C-1′), 162.40 ((C=O)NH); HRMS (ESI+): calcd for C28H32N3O4+ [M + H]+: 474.2387; found: 474.2376.
tert-Butyl 4-{4-[2-anilino-3-(trifluoromethyl)benzamido]phenyl}piperazin-1-carboxylate (41): Reaction of the carboxylic acid 14 (230 mg (0.82 mmol)) with the amine 17 (227 mg (0.82 mmol)), 2-chloro-N-methylpyridinium iodide (367 mg (1.44 mmol)) and DIPEA (530 mg (4.10 mmol)) in dry dichloromethane (25 mL) gave the crude product. It was purified by column chromatography (flash silica gel, CH/EtAc 3:1) yielding compound 41 as pale brown amorphous solid (44 mg (10%)). IR = 3320, 1705, 1640, 1597, 1528, 1453, 1420, 1316, 1232, 1167, 1131, 753; 1H NMR (CDCl3) δ = 1.47 (s, 9H, (CH3)3), 3.02–3.05 (m, 4H, N(CH2)2), 3.52–3.55 (m, 4H, N(CH2)2), 6.09 (br s, 1H, NH), 6.75–6.79 (m, 4H, 2′-H, 3″-H, 5″-H, 6′-H), 6.89–6.93 (m, 1H, 4′-H), 7.05–7.09 (m, 2H, 2″-H, 6″-H), 7.17–7.22 (m, 2H, 3′-H, 5′-H), 7.44 (td, J = 7.9 Hz, 1H, 5-H), 7.84 (dd, J = 7.9, 1.6 Hz, 1H, 4-H), 8.34 (dd, J = 7.9, 1.6 Hz, 1H, 6-H), 9.42 (br s, 1H, NH); 13C NMR (CDCl3) δ = 28.41 ((CH3)3), 43.53 (N(CH2)2), 49.65 (N(CH2)2), 79.88 (CMe3), 115.95 (C-2′, C-6′), 116.93 (C-3″, C-5″), 121.23 (q, J = 274 Hz, CF3), 122.18 (C-2″, C-4′, C-6″), 125.37 (C-5), 125.53 (q, J = 29.2 Hz, C-3), 129.71 (C-3′, C-5′), 129.80 (q, J = 5.5 Hz, C-4), 130.08 (C-1″), 132.57 (C-1), 135.49 (C-6), 137.41 (q, J = 1.3 Hz, C-2), 144.60 (C-1′), 148.48 (C-4″), 154.68 (N(C=O)O), 163.11 ((C=O)NH); HRMS (ESI+): calcd for C29H32F3N4O3+ [M + H]+: 541.2421; found: 541.2410.
tert-Butyl 4-{4-[2-(fluoroanilino)-3-(trifluoromethyl)benzamido]phenyl}piperazin-1-carboxylate (42): Reaction of the carboxylic acid 15 (299 mg (1.00 mmol)) with the carboxylic acid 17 (277 mg (1.00 mmol)), 2-chloro-N-methylpyridinium iodide (447 mg (1.75 mmol)) and DIPEA (646 mg (5.00 mmol)) in dry dichloromethane (30 mL) gave the crude product. It was purified by column chromatography (flash silica gel, CH2Cl2/MeOH 79:1) yielding compound 42 as pale brown amorphous solid (124 mg (22%)). IR = 3427, 1665, 1508, 1454, 1315, 1229, 1167, 824; 1H NMR (CDCl3) δ = 1.48 (s, 9H, (CH3)3), 3.04–3.07 (m, 4H, N(CH2)2), 3.53–3.56 (m, 4H, N(CH2)2), 6.05 (br s, 1H, NH), 6.72–6.76 (m, 2H, 2′-H, 6′-H), 6.79–6.81 (m, 2H, 3″-H, 5″-H), 6.85–6.90 (m, 2H, 3′-H, 5′-H), 7.13 (d, J = 8.9 Hz, 2H, 2″-H, 6″-H), 7.42 (t, J = 7.9 Hz, 1H, 5-H), 7.83 (dd, J = 7.9, 1.6 Hz, 1H, 4-H), 8.28 (dd, J = 7.9, 1.6 Hz, 1H, 6-H), 9.24 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.41 ((CH3)3), 43.38 (N(CH2)2), 49.62 (N(CH2)2), 79.89 (CMe3), 116.22 (d, J = 22.9 Hz, C-3′, C-5′), 116.97 (C-3″, C-5″), 117.80 (d, J = 7.9 Hz, C-2′, C-6′), 121.89 (C-2″, C-6″), 123.94 (q, J = 273 Hz, CF3), 125.13 (q, J = 29.2 Hz, C-3), 125.16 (C-5), 129.79 (q, J = 5.2 Hz, C-4), 130.00 (C-1″), 132.16 (C-1), 135.45 (C-6), 137.86 (q, J = 1.8 Hz, C-2), 140.74 (d, J = 2.5 Hz, C-1′), 148.53 (C-4″), 154.68 (N(C=O)O), 158.34 (d, J = 241 Hz, C-4′), 163.13 ((C=O)NH); HRMS (ESI+): calcd for C29H31F4N4O3+ [M + H]+: 559.2327; found: 559.2311.
N-{4-[4-(2,2-Dimethylpropanoyl)piperazin-1-yl]phenyl}-2-(4-fluorophenoxy)benzamid (43): Reaction of the carboxylic acid 9 (232 mg (1.00 mmol)) with the amine 19 (261 mg (1.00 mmol)), 2-chloro-N-methylpyridinium iodide (447 mg (1.75 mmol)) and DIPEA (646 mg (5.00 mmol)) in dry dichloromethane (30 mL) gave the crude product. It was purified by column chromatography (silica gel, CH/EtAc 1:1) yielding compound 43 as pale yellow amorphous solid (158 mg (33%)). IR = 3385, 1654, 1614, 1518, 1504, 1477, 1419, 1321, 1205, 857; 1H NMR (CDCl3) δ = 1.31 (s, 9H, (CH3)3), 3.10–3.13 (m, 4H, N(CH2)2), 3.78–3.81 (m, 4H, N(CH2)2), 6.83 (d, J = 7.8 Hz, 1H, 3-H), 6.90 (d, J = 9.0 Hz, 2H, 3″-H, 5″-H), 7.06–7.14 (m, 4H, 2′-H, 3′-H, 5′-H, 6′-H), 7.23–7.28 (m, 1H, 5-H), 7.42 (ddd, J = 9.0, 7.9, 1.8 Hz, 1H, 4-H), 7.53 (d, J = 9.0 Hz, 2H, 2″-H, 6″-H), 8.32 (dd, J = 7.9, 1.9 Hz, 1H, 6-H), 9.42 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.42 ((CH3)3), 38.65 (CMe3), 44.97 (N(CH2)2), 49.98 (N(CH2)2), 116.95 (d, J = 23.6 Hz, C-3′, C-5′), 117.01 (C-3″, C-5″), 117.80 (C-3), 121.15 (d, J = 8.3 Hz, C-2′, C-6′), 121.69 (C-2″, C-6″), 123.97 (C-5), 124.03 (C-1), 131.20 (C-1″), 132.49 (C-6), 132.98 (C-4), 147.98 (C-4″), 151.06 (d, J = 2.6 Hz, C-1′), 155.43 (C-2), 159.65 (d, J = 244 Hz, C-4′), 162.35 (N(C=O)O), 176.37 ((C=O)NH); HRMS (ESI+): calcd for C28H31FN3O3+ [M + H]+: 476.2344; found: 476.2332.
N-{4-[4-(2,2-Dimethylpropanoyl)piperazin-1-yl]phenyl}-2-(4-fluorophenoxy)-3-nitrobenzamid (44): Reaction of the carboxylic acid 11 (140 mg (0.51 mmol)) with the amine 19 (133 mg (0.51 mmol)), 2-chloro-N-methylpyridinium iodide (227 mg (0.89 mmol)) and DIPEA (300 mg (2.55 mmol)) in dry dichloromethane (15 mL) gave the crude product. It was purified by column chromatography (silica gel, EtAc/CH 3:1) yielding compound 44 as orange amorphous solid (194 mg (73%)). IR = 3423, 1677, 1604, 1524, 1501, 1363, 1323, 1189, 836, 776; 1H NMR (CDCl3) δ = 1.31 (s, 9H, (CH3)3), 3.10–3.13 (m, 4H, N(CH2)2), 3.77–3.80 (m, 4H, N(CH2)2), 6.83–6.87 (m, 4H, 2′-H, 3″-H, 5″-H, 6′-H), 6.96–7.01 (m, 2H, 3′-H, 5′-H), 7.33 (d, J = 9.0 Hz, 2H, 2″-H, 6″-H), 7.55 (t, J = 8.0 Hz, 1H, 5-H), 8.08 (dd, J = 8.0, 1.8 Hz, 1H, 4-H), 8.44 (dd, J = 8.0, 1.8 Hz, 1H, 6-H), 8.66 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.41 ((CH3)3), 38.66 (CMe3), 44.91 (N(CH2)2), 49.68 (N(CH2)2), 116.60 (d, J = 8.3 Hz, C-2′, C-6′), 116.81 (C-3″, C-5″), 116.82 (d, J = 23.8 Hz, C-3′, C-5′), 121.71 (C-2″, C-6″), 126.32 (C-5), 128.63 (C-4), 129.93 (C-1″), 131.12 (C-1), 136.58 (C-6), 143.84 (C-3), 144.97 (C-2), 148.52 (C-4″), 152.86 (d, J = 2.6 Hz, C-1′), 158.91 (d, J = 244 Hz, C-4′), 160.49 ((C=O)NH), 176.38 (C=O); HRMS (ESI+): calcd for C28H30FN4O5+ [M + H]+: 521.2195; found: 521.2183.
N-{4-[4-(2,2-Dimethylpropanoyl)piperazin-1-yl]phenyl}-3-fluoro-2-(4-fluorophenoxy)benzamid (45): Reaction of the carboxylic acid 10 (100 mg (0.40 mmol)) with the amine 19 (105 mg (0.40 mmol)), 2-chloro-N-methylpyridinium iodide (179 mg (0.70 mmol)) and DIPEA (259 mg (2.00 mmol) in dry dichloromethane (12 mL) gave the crude product. It was purified by column chromatography (flash silica gel, CH/CH2Cl2/EtAc 1:0.3:1.25) yielding compound 45 as pale yellow amorphous solid (122 mg (63%)). IR = 3405, 1623, 1502, 1462, 1417, 1320, 1270, 1232, 1188, 1016, 828, 767; 1H NMR (CDCl3) δ = 1.31 (s, 9H, (CH3)3), 3.09–3.12 (m, 4H, N(CH2)2), 3.77–3.81 (m, 4H, N(CH2)2), 6.88 (d, J = 9.0 Hz, 2H, 3″-H, 5″-H), 6.93–6.97 (m, 2H, 2′-H, 6′-H), 6.99–7.04 (m, 2H, 3′-H, 5′-H), 7.30–7.38 (m, 2H, 4-H, 5-H), 7.42 (d, J = 9.0 Hz, 2H, 2″-H, 6″-H), 8.04–8.07 (m, 1H, 6-H), 9.03 (s, 1H, NH); 13C NMR (CDCl3) δ = 28.41 ((CH3)3), 38.65 (CMe3), 44.93 (N(CH2)2), 49.84 (N(CH2)2), 116.61 (d, J = 23.8 Hz, C-3′, C-5′), 116.72 (d, J = 8.3 Hz, C-2′, C-6′), 116.93 (C-3″, C-5″), 120.29 (d, J = 18.6 Hz, C-4), 121.55 (C-2″, C-6″), 126.27 (d, J = 7.7 Hz, C-5), 127.10 (d, J = 3.3 Hz, C-6), 129.32 (C-1), 130.63 (C-1″), 140.40 (d, J = 13.1 Hz, C-2), 148.22 (C-4″), 152.96 (t, J = 2.2 Hz, C-1′), 154.98 (d, J = 252 Hz, C-3), 158.85 (d, J = 243 Hz, C-4′), 161.17 (d, J = 3.2 Hz, (C=O)NH), 176.36 (C=O); HRMS (ESI+): calcd for C28H30F2N3O3+ [M + H]+: 494.2250; found: 494.2234.

3.3. Biological Tests

3.3.1. In Vitro Microplate Assay against P. falciparum NF54

The in vitro activity of compounds against erythrocytic stages of the drug sensitive NF54 strain of P. falciparum originating from Thailand was determined using a 3H-hypoxanthine incorporation assay [35,36,37]. Compounds were dissolved in DMSO at 10 mg/mL and further diluted in medium before adding to parasite cultures that were incubated in RPMI 1640 medium without hypoxanthine, supplemented with HEPES (5.94 g/L), NaHCO3 (2.1 g/L), neomycin (100 U/mL), AlbumaxR (5 g/L) and washed human red blood cells A+ at 2.5% haematocrit (0.3% parasitaemia). Serial drug dilutions of eleven 3-fold dilutions steps covering a range from 100 to 0.002 µg/mL were prepared. The 96-well plates were incubated in a humidified atmosphere at 37 °C; 4% CO2, 3% O2, 93% N2. After 48 h of incubation time, 0.05 mL of 3H-hypoxanthine (=0.5 µCi) was added to each well of the plate. The plates were incubated for further 24 h under the same conditions. Plates were then harvested using a BetaplateTM cell harvester (Wallac, Zurich, Switzerland). Red blood cells were transferred onto a glass fiber filter and then washed with distilled water. The dried filters were inserted into a plastic foil with 10 mL of scintillation fluid and counted in a BetaplateTM liquid scintillation counter (Wallac, Zurich, Switzerland). IC50 values were calculated from sigmoidal inhibition curves by linear regression using Microsoft Excel [38]. Chloroquine (Sigma C6628) was used as control.

3.3.2. In Vitro Cytotoxicity with L-6 Cells

The cytotoxicity assays were performed using 96-well microtiter plates, each well containing 4000 L-6 cells (a primary cell line derived from rat skeletal myofibroblasts, ATCC CRL-1458TM) in 0.1 mL of RPMI 1640 medium supplemented with 1% glutamine (200 mM) and 10% fetal bovine serum [39,40]. Serial drug dilutions of eleven 3-fold dilution steps covering a range from 100 to 0.002 µg/mL were prepared. After 70 h of incubation, the plates were inspected under an inverted microscope to assure growth of the controls and sterile conditions. Then, 0.01 mL resazurin solution (resazurin, 12.5 mg in 100 mL double-distilled water) was added to each well and the plates were incubated for another 2 h. The plates were read with a Spectramax Gemini XS microplate fluorometer (Molecular Devices Cooperation, Sunnyvale, CA, USA) using an excitation wavelength of 536 nm and an emission wavelength of 588 nm. IC50 values were calculated by linear regression from the sigmoidal dose inhibition curves using SoftmaxPro software (Molecular Devices Cooperation, Sunnyvale, CA, USA) [38]. Podophyllotoxin (Sigma P4405) was used as control.

3.3.3. Parallel Artificial Membrane Permeability Assay

With the high-throughput PAMPA the newly synthesized compounds were tested for their passive permeability through cell membranes without the influence of efflux pumps or transporter proteins. The assay was performed using a Corning® GentestTM Pre-coated PAMPA Plate System with 96-well polystyrene plates. The bottom of the acceptor plate consists of a porous membrane, whereby the pores are lined with a lipid-oil-lipid triple layer. Stock solutions of each test compound at 10 mM were prepared in DMSO or methanol and diluted with phosphate-buffered saline (PBS at a pH of 7.4) to a final concentration of 200 µM. Hydrochlorothiazide (Pe = 0.9 nm/s) and caffeine (Pe = 80 nm/s) were used as standards. The donor plate (bottom plate) was filled with the compound solutions, whereby all compounds were tested in quadruplicates. Each well of the acceptor plate (top plate) was filled with PBS buffer. Donor and acceptor plates were combined and incubated at room temperature for 5 h. After that, the plates were separated and 150 µL of each well of both plates were transferred to 96-well UV plates (Greiner Bio-One). Absorption at different wavelengths covering a range from 200 to 300 nm was measured using a SpectraMax M3 UV plate reader. By measuring serial dilutions of five dilution steps covering a range from 200 to 12.5 µM, a calibration curve was prepared. The plates were analyzed at the wavelength where the R2 value of the calibration curve was higher than 0.99 [41]. Effective permeability Pe of each test compound was calculated using the following Equations (1)–(3):
P e = ln [ 1 c A ( t ) c e q u ] S ( 1 V D + 1 V A ) t
where:
  • Pe—effective permeability;
  • S—filter area (0.3 cm2);
  • VD—donor well volume (0.3 mL);
  • VA—acceptor well volume (0.2 mL);
  • t—incubation time (18,000 s);
  • cA(t)acceptor well compound concentration at time t;
  • cequ—equilibrium concentration.
c e q u = [ c D ( t ) V D + c A ( t ) V A ] ( V D + V A )
where:
  • VD—donor well volume (0.3 mL);
  • VA—acceptor well volume (0.2 mL);
  • cA(t)acceptor well compound concentration at time t;
  • cD(t)donor well compound concentration at time t.
Recovery of compounds from donor and acceptor wells (mass retention) was calculated as shown in the equation below. Data were only accepted when recovery exceeded 70%.
R = 1 [ c D ( t ) V D + c A ( t ) V A ] ( c 0 V D )
where:
  • R—mass retention (%);
  • VD—donor well volume (0.3 mL);
  • VA—acceptor well volume (0.2 mL);
  • cA(t)acceptor well compound concentration at time t;
  • cD(t)donor well compound concentration at time t;
  • c0—initial donor well compound concentration (200 µM).

3.3.4. Ligand Efficiency (LE)

Ligand efficiency was calculated as shown in the following Equation (4):
L E = 1.37 H A p I C 50
where:
  • LE—ligand efficiency;
  • HA—number of heavy atoms;
  • pIC50—negative logarithm of IC50.

4. Conclusions

This paper deals with the synthesis of derivates of MMV’s Malaria Box compound 1, which exhibits multi-stage activity against different strains of P. falciparum and lack of resistance development. It is a 2-(4-fluorophenoxy)-3-(trifluoromethyl)benzanilide with a N-bocpiperazinyl group in ortho position of the anilide nitrogen. The first series focused on the derivatization of the anilino moiety showing the positive influence of a N-bocpiperazinyl group or a 4-pivaloylpiperazinyl group in ortho or para position, which became partial structure of all of the new compounds. The 3-(trifluoromethyl) group was replaced by hydrogen, fluoro, amino or nitrogen substituents, but turned out to be the preferable substitution. The 2-(4-fluorophenoxy) moiety was replaced by an anilino, a 4-fluoroanilino or a (4-fluorophenyl)sulfanyl substituent. The latter was partial structure of 29 which exhibits a N-bocpiperazinyl group in 2′-position of the benzanilide. Compared to 1 it showed improved activity, selectivity and passive permeability (Figure 6).

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph15121503/s1, Figures S1–S27: 1H- and 13C-NMR spectra for compounds 10, 11, 1315 and 2647.

Author Contributions

Conceptualization, T.H. and R.W. (Robert Weis); methodology, T.H. and R.W. (Robert Weis); investigation, T.H., R.W. (Robin Wallner), J.D., W.S., E.-M.P.-W., M.K., P.M. and R.W. (Robert Weis); data curation, T.H., R.W. (Robin Wallner), J.D., W.S., E.-M.P.-W., M.K., P.M. and R.W. (Robert Weis); writing—original draft preparation, T.H. and R.W. (Robert Weis); writing—review and editing, T.H. and R.W. (Robert Weis); supervision, R.W. (Robert Weis); project administration: T.H. and R.W. (Robert Weis). All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article and supplementary material.

Acknowledgments

The Authors acknowledge Open Access Funding by the University of Graz. NAWI Graz is acknowledged for supporting Central Lab Plant, Environmental and Microbial Metabolomics.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. World Malaria Report 2021; World Health Organization: Geneva, Switzerland, 2021. [Google Scholar]
  2. Ashley, E.A.; Phyo, A.P.; Woodrow, C.J. Malaria. Lancet 2018, 391, 1608–1621. [Google Scholar] [CrossRef]
  3. Hanboonkunupakarn, B.; White, N.J. The threat of antimalarial drug resistance. Trop. Dis. Travel Med. Vaccines 2016, 2, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Nkumama, I.N.; O’Meara, W.P.; Osier, F.H. Changes in Malaria Epidemiology in Africa and New Challenges for Elimination. Trends Parasitol. 2016, 33, 128–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Balikagala, B.; Fukuda, N.; Ikeda, M.; Katuro, O.T.; Tachibana, S.-I.; Yamauchi, M.; Opio, W.; Emoto, S.; Anywar, D.A.; Kimura, E.; et al. Evidence of Artemisinin-Resistant Malaria in Africa. N. Engl. J. Med. 2021, 385, 1163–1171. [Google Scholar] [CrossRef]
  6. van der Pluijm, R.W.; Amaratunga, C.; Dhorda, M.; Dondorp, A.M. Triple Artemisinin-Based Combination Therapies for Malaria—A New Paradigm? Trends Parasitol. 2020, 37, 15–24. [Google Scholar] [CrossRef]
  7. Tindana, P.; de Haan, F.; Amaratunga, C.; Dhorda, M.; van der Pluijm, R.W.; Dondorp, A.M.; Cheah, P.Y. Deploying triple artemisinin-based combination therapy (TACT) for malaria treatment in Africa: Ethical and practical considerations. Malar. J. 2021, 20, 1–7. [Google Scholar] [CrossRef]
  8. Peto, T.J.; Tripura, R.; Callery, J.J.; Lek, D.; Nghia, H.D.T.; Nguon, C.; Thuong, N.T.H.; van der Pluijm, R.; Dung, N.T.P.; Sokha, M.; et al. Triple therapy with artemether-lumefantrine plus amodiaquine versus artemether-lumefantrine alone for artemisinin-resistant, uncomplicated falciparum malaria: An open-label, randomized, multicentre trial. Lancet Infect Dis. 2022, 22, 867–878. [Google Scholar] [CrossRef]
  9. Beeson, J.G.; Kurtovic, L.; Dobaño, C.; Opi, D.H.; Chan, J.-A.; Feng, G.; Good, M.F.; Reiling, L.; Boyle, M.J. Challenges and strategies for developing efficacious and long-lasting malaria vaccines. Sci. Transl. Med. 2019, 11, eaau1458. [Google Scholar] [CrossRef]
  10. Laurens, M.B. RTS, s/AS01 vaccine (MosquirixTM): An overview. Hum. Vaccines Immunother. 2020, 16, 480–489. [Google Scholar] [CrossRef]
  11. Duffy, P.E. Making a good malaria vaccine better. Trends Parasitol. 2021, 38, 9–10. [Google Scholar] [CrossRef]
  12. Van Voorhis, W.C.; Adams, J.H.; Adelfio, R.; Ahyong, V.; Akabas, M.H.; Alano, P.; Alday, A.; Resto, Y.A.; Alsibaee, A.; Alzualde, A.; et al. Open Source Drug Discovery with the Malaria Box Compound Collection for Neglected Diseases and Beyond. PLoS Pathog. 2016, 12, e1005763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Guiguemde, W.A.; Shelat, A.; Garcia-Bustos, J.; Diagana, T.T.; Gamo, F.-J.; Guy, R.K. Global Phenotypic Screening for Antimalarials. Chem. Biol. 2012, 19, 116–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Corey, V.C.; Lukens, A.K.; Istvan, E.S.; Lee, M.; Franco, V.; Magistrado, P.; Coburn-Flynn, O.; Sakata-Kato, T.; Fuchs, O.; Gnädig, N.F.; et al. A broad analysis of resistance development in the malaria parasite. Nat. Commun. 2016, 7, 11901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Murithi, J.M.; Owen, E.; Istvan, E.S.; Lee, M.; Ottilie, S.; Chibale, K.; Goldberg, D.E.; Winzeler, E.; Llinás, M.; Fidock, D.A.; et al. Combining Stage Specificity and Metabolomic Profiling to Advance Antimalarial Drug Discovery. Cell Chem. Biol. 2019, 27, 158–171.e3. [Google Scholar] [CrossRef] [Green Version]
  16. Carolino, K.; Winzeler, E. The antimalarial resistome-finding new drug targets and their modes of action. Curr. Opin. Microbiol. 2020, 57, 49–55. [Google Scholar] [CrossRef]
  17. Hermann, T.; Hochegger, P.; Dolensky, J.; Seebacher, W.; Pferschy-Wenzig, E.-M.; Saf, R.; Kaiser, M.; Mäser, P.; Weis, R. Synthesis and Structure-Activity Relationships of New 2-Phenoxybenzamides with Antiplasmodial Activity. Pharmaceuticals 2021, 14, 1109. [Google Scholar] [CrossRef]
  18. Debets, M.F.; Prins, J.S.; Merkx, D.; van Berkel, S.S.; van Delft, F.L.; van Hest, J.C.M.; Rutjes, F.P.J.T. Synthesis of DIBAC analogues with excellent SPAAC rate constants. Org. Biomol. Chem. 2014, 12, 5031–5037. [Google Scholar] [CrossRef]
  19. Hossian, A.; Jana, R. Carboxyl radical-assisted 1,5-aryl migration through Smiles rearrangement. Org. Biomol. Chem. 2016, 14, 9768–9779. [Google Scholar] [CrossRef]
  20. Neuville, L.; Zhu, J. Solution phase combinatorial synthesis of arylpiperazines. Tetrahedron Lett. 1997, 38, 4091–4094. [Google Scholar] [CrossRef]
  21. Zaragoza, F.; Stephensen, H.; Knudsen, S.M.; Pridal, L.; Wulff, A.B.S.; Rimvall, K. 1-Alkyl-4-acylpiperazines as a New Class of Imidazole-Free Histamine H3 Receptor Antagonists. J. Med. Chem. 2004, 47, 2833–2838. [Google Scholar] [CrossRef]
  22. Akbar, A.; McNeil, N.M.R.; Albert, M.R.; Ta, V.; Adhikary, G.; Bourgeois, K.; Eckert, R.L.; Keillor, J.W. Structure–Activity Relationships of Potent, Targeted Covalent Inhibitors That Abolish Both the Transamidation and GTP Binding Activities of Human Tissue Transglutaminase. J. Med. Chem. 2017, 60, 7910–7927. [Google Scholar] [CrossRef] [PubMed]
  23. Zupancic, B. Synthesis of Vortioxetine via (2-(Piperazine-1-yl)Phenylaniline Intermediates; LEK Pharmaceuticals, D.D.: Ljubljana, Slovenia, 2015. [Google Scholar]
  24. Ren, Q.; Dai, L.; Zhang, H.; Tan, W.; Xu, Z.; Ye, T. Total Synthesis of Largazole. Synlett 2008, 2008, 2379–2383. [Google Scholar] [CrossRef] [Green Version]
  25. Hopkins, A.L.; Groom, C.R.; Alex, A. Ligand efficiency: A useful metric for lead selection. Drug Discov. Today 2004, 9, 430–431. [Google Scholar] [CrossRef]
  26. Hopkins, A.L.; Keserü, G.M.; Leeson, P.D.; Rees, D.C.; Reynolds, C.H. The role of ligand efficiency metrics in drug discovery. Nat. Rev. Drug Discov. 2014, 13, 105–121. [Google Scholar] [CrossRef]
  27. Perez-Medrano, A.; Nelson, D.; Carroll, W.; Kort, M.; Gregg, R.; Voight, E.; Jarvis, M.; Kowaluk, E. Abbott Laboratories The Use of Selective P2x7 Receptor Antagonists; WO2006/86229; Abbott Laboratories; Abbott Park: Chicago, IL, USA, 2006. [Google Scholar]
  28. Huang, H.; Zhang, G.; Chen, Y. Dual Hypervalent Iodine(III) Reagents and Photoredox Catalysis Enable Decarboxylative Ynonylation under Mild Conditions. Angew. Chem. 2015, 127, 7983–7987. [Google Scholar] [CrossRef]
  29. Weis, E.; Johansson, M.J.; Martín-Matute, B. Ir III -Catalyzed Selective ortho -Monoiodination of Benzoic Acids with Unbiased C−H Bonds. Chem. A Eur. J. 2020, 26, 10185–10190. [Google Scholar] [CrossRef]
  30. Gonzalez-Gomez, J.C.; Ramirez, N.P.; Lana-Villarreal, T.; Bonete, P. A photoredox-neutral Smiles rearrangement of 2-aryloxybenzoic acids. Org. Biomol. Chem. 2017, 15, 9680–9684. [Google Scholar] [CrossRef] [Green Version]
  31. Wang, C.; Wang, X.; Li, Y.; Wang, T.; Huang, Z.; Qin, Z.; Yang, S.; Xiang, R.; Fan, Y. Design and optimization of orally spleen tyrosine kinase (SYK) inhibitors for treatment of solid tumor. Bioorg. Chem. 2019, 95, 103547. [Google Scholar] [CrossRef]
  32. Lagu, B.; Tian, D.; Nagarathnam, D.; Marzabadi, M.R.; Wong, W.C.; Miao, S.W.; Zhang, F.; Sun, W.; Chiu, G.; Fang, J.; et al. Design and Synthesis of Novel α1a Adrenoceptor-Selective Antagonists. 3. Approaches To Eliminate Opioid Agonist Metabolites by Using Substituted Phenylpiperazine Side Chains. J. Med. Chem. 1999, 42, 4794–4803. [Google Scholar] [CrossRef]
  33. Russell, C.; Lin, A.J.S.; Hains, P.; Simone, M.I.; Robinson, P.J.; McCluskey, A. An integrated flow and microwave approach to a broad spectrum protein kinase inhibitor. RSC Adv. 2015, 5, 93433–93437. [Google Scholar] [CrossRef]
  34. Player, M.R.; Hunag, H.; Hutta, D.A. Janssen Pharmaceutica nv. 5-oxo-5,8-Dihydro-Pyrido-Pyrimidines as Inhibitors of C-Fms Kinase; Janssen Pharmaceuticaln: Beerse, Belgium, 2007. [Google Scholar]
  35. Desjardins, R.; Canfield, C.J.; Haynes, J.D.; Chulay, J.D. Quantitative assessment of antimalarial activity in vitro by a semiautomated microdilution technique. Antimicrob. Agents Chemother. 1979, 16, 710–718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Matile, H.; Richard, J.; Pink, L. Plasmodium Falciparum Malaria Parasite Cultures and Their Use in Immunology; Immunological methods; Academic Press: Cambridge, MA, USA, 1990; pp. 221–234. [Google Scholar] [CrossRef]
  37. Ponnudurai, T.; Leeuwenberg, A.D.; Meuwissen, J.H. Chloroquine sensitivity of isolates of Plasmodium falciparum adapted to in vitro culture. Trop. Geogr. Med. 1981, 33, 50–54. [Google Scholar] [PubMed]
  38. Huber, W.; Koella, J.C. A comparison of three methods of estimating EC50 in studies of drug resistance of malaria parasites. Acta Trop. 1993, 55, 257–261. [Google Scholar] [CrossRef] [PubMed]
  39. Page, B.; Page, M.; Noel, C. A New Fluorometric Assay for Cytotoxicity Measurements In-Vitro. Int. J. Oncol. 1993, 3, 473–476. [Google Scholar] [CrossRef]
  40. Ahmed, S.A.; Gogal, R.M.; Walsh, J.E. A new rapid and simple non-radioactive assay to monitor and determine the proliferation of lymphocytes: An alternative to [3H]thymidine incorporation assay. J. Immunol. Methods 1994, 170, 211–224. [Google Scholar] [CrossRef]
  41. Chen, X.; Murawski, A.; Patel, K.; Crespi, C.L.; Balimane, P.V. A Novel Design of Artificial Membrane for Improving the PAMPA Model. Pharm. Res. 2008, 25, 1511–1520. [Google Scholar] [CrossRef]
Figure 1. Structure-activity relationships of the lead compound 1 and its first series of derivatives [17].
Figure 1. Structure-activity relationships of the lead compound 1 and its first series of derivatives [17].
Pharmaceuticals 15 01503 g001
Figure 2. Preparation of the 2- and 3-substituted benzoic acid derivatives 915. Reagents and conditions: (a) (1) H2SO4 30%, dimethylsulfoxide (DMSO), 0 °C, 5 min; (2) NaNO2, rt, 2 h; (3) KI, H2O, rt, 1 h; (4) KI, H2O, rt, 1 h; (b) corresponding phenol or aniline, Cu, CuI, 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU), dry pyridine, dry DMF, 160 °C, 2 h or 24 h.
Figure 2. Preparation of the 2- and 3-substituted benzoic acid derivatives 915. Reagents and conditions: (a) (1) H2SO4 30%, dimethylsulfoxide (DMSO), 0 °C, 5 min; (2) NaNO2, rt, 2 h; (3) KI, H2O, rt, 1 h; (4) KI, H2O, rt, 1 h; (b) corresponding phenol or aniline, Cu, CuI, 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU), dry pyridine, dry DMF, 160 °C, 2 h or 24 h.
Pharmaceuticals 15 01503 g002
Figure 3. Preparation of anilino derivatives 1619. Reagents and conditions: (a) K2CO3, N-Boc-piperazine, dry DMSO, 80 °C, 72 h; (b) 15% (m/m) palladium on activated carbon, H2, methanol, rt, 24 h; (c) (1) dry CH2Cl2, 0 °C, 5 min; (2) trifluoroacetic acid, dry CH2Cl2, rt, 24 h; (d) pivaloyl chloride, triethylamine, dry CH2Cl2, rt, 24 h; (e) 15% (m/m) palladium on activated carbon, H2, methanol rt, 24 h.
Figure 3. Preparation of anilino derivatives 1619. Reagents and conditions: (a) K2CO3, N-Boc-piperazine, dry DMSO, 80 °C, 72 h; (b) 15% (m/m) palladium on activated carbon, H2, methanol, rt, 24 h; (c) (1) dry CH2Cl2, 0 °C, 5 min; (2) trifluoroacetic acid, dry CH2Cl2, rt, 24 h; (d) pivaloyl chloride, triethylamine, dry CH2Cl2, rt, 24 h; (e) 15% (m/m) palladium on activated carbon, H2, methanol rt, 24 h.
Pharmaceuticals 15 01503 g003
Figure 4. Preparation of compounds 2645. Reagents and conditions: (a) (1) amine 16, dry CH2Cl2, 0 °C, 5 min; (2) 2-chloro-N-methylpyridinium iodide, DIPEA, rt, 24 h (compounds 2628, 3032) or (1) amine 16, dry CH2Cl2, 0 °C, 5 min; (2) 2-chloro-N-methylpyridinium iodide, DIPEA, rt, 48 h (compound 29) or (1) amine 18, dry CH2Cl2, 0 °C, 5 min; (2) 2-chloro-N-methylpyridinium iodide, DIPEA, rt, 24 h (compounds 3335); (b) (1) amine 17, dry CH2Cl2, 0 °C, 5 min; (2) 2-chloro-N-methylpyridinium iodide, DIPEA, rt, 24 h (compounds 3642) or (1) amine 19, dry CH2Cl2, 0 °C, 5 min; (2) 2-chloro-N-methyl-pyridinium iodide, diisopropylethylamine, rt, 24 h (compounds 4345).
Figure 4. Preparation of compounds 2645. Reagents and conditions: (a) (1) amine 16, dry CH2Cl2, 0 °C, 5 min; (2) 2-chloro-N-methylpyridinium iodide, DIPEA, rt, 24 h (compounds 2628, 3032) or (1) amine 16, dry CH2Cl2, 0 °C, 5 min; (2) 2-chloro-N-methylpyridinium iodide, DIPEA, rt, 48 h (compound 29) or (1) amine 18, dry CH2Cl2, 0 °C, 5 min; (2) 2-chloro-N-methylpyridinium iodide, DIPEA, rt, 24 h (compounds 3335); (b) (1) amine 17, dry CH2Cl2, 0 °C, 5 min; (2) 2-chloro-N-methylpyridinium iodide, DIPEA, rt, 24 h (compounds 3642) or (1) amine 19, dry CH2Cl2, 0 °C, 5 min; (2) 2-chloro-N-methyl-pyridinium iodide, diisopropylethylamine, rt, 24 h (compounds 4345).
Pharmaceuticals 15 01503 g004
Figure 5. Preparation of compounds 46 and 47. Reagents and conditions: (a) 15% (m/m) palladium on activated carbon, H2, methanol, rt, 24 h.
Figure 5. Preparation of compounds 46 and 47. Reagents and conditions: (a) 15% (m/m) palladium on activated carbon, H2, methanol, rt, 24 h.
Pharmaceuticals 15 01503 g005
Figure 6. Structure−activity relationships of compounds 1 and 29.
Figure 6. Structure−activity relationships of compounds 1 and 29.
Pharmaceuticals 15 01503 g006
Table 1. Activities of compounds 2647 against P. falciparum NF54 and L-6 cells, expressed as IC50 (µM) a.
Table 1. Activities of compounds 2647 against P. falciparum NF54 and L-6 cells, expressed as IC50 (µM) a.
CompoundPfNF54 b
IC50 (µM)
S.I. =
IC50 (Cyt.)/IC50
(PfNF54)
Cytotoxicity
L-6 Cells
IC50 (µM)
10.4134316.9131.0
20.2690461.0124.0
30.6172299.7185.0
40.5795171.999.62
260.6266245.5153.8
270.6496237.5154.3
280.5908153.990.39
290.1946850.5165.5
301.13119.8722.47
310.6142209.6128.8
320.6364193.8123.4
332.14517.8438.27
342.61343.75114.3
352.1387.27515.55
363.39759.88203.4
374.0728.67635.33
381.43249.4670.82
390.1494212.831.79
403.09431.7898.30
410.633653.2933.76
420.5389174.894.17
435.61532.84184.4
441.49896.41144.5
451.79360.90109.2
461.010126.0127.3
473.08351.38158.4
CQ0.009967290.92
POD 0.012
CQ = chloroquine; POD = podophyllotoxin; a IC50 values represent the average of four determinations (two determinations of two independent experiments); b sensitive to chloroquine.
Table 2. Key physicochemical parameters and passive permeability values of compounds 2647.
Table 2. Key physicochemical parameters and passive permeability values of compounds 2647.
Compoundlog P/log D7.4 aLE
(kcal/mol/HA)
Pe b
(10−6 cm/s)
16.440.2192.37
26.440.2250.09
36.570.2180.23
45.560.2360.24
265.560.236n.d.
275.710.229n.d.
285.500.2001.68
297.130.2303.77
305.420.2338.52
317.540.2180.65
327.680.2120.57
335.690.222n.d.
345.630.2016.92
355.830.2160.00
365.560.2082.84
375.710.2000.70
385.500.2191.53
397.130.2344.31
405.420.2165.57
417.540.2181.32
427.680.2150.20
435.690.2060.02
445.630.210n.d.
455.830.2195.13
464.730.2221.44
474.730.2040.62
Hydrochlorothiazide 0.09
Caffeine 8.00
a log P and log D were calculated using the ChemAxon software JChem for Excel 14.9.1500.912 (2014) (Chemaxon, Budapest, Hungary); b determined by PAMPA, n.d. could not be determined.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hermann, T.; Wallner, R.; Dolensky, J.; Seebacher, W.; Pferschy-Wenzig, E.-M.; Kaiser, M.; Mäser, P.; Weis, R. New Derivatives of the Multi-Stage Active Malaria Box Compound MMV030666 and Their Antiplasmodial Potencies. Pharmaceuticals 2022, 15, 1503. https://doi.org/10.3390/ph15121503

AMA Style

Hermann T, Wallner R, Dolensky J, Seebacher W, Pferschy-Wenzig E-M, Kaiser M, Mäser P, Weis R. New Derivatives of the Multi-Stage Active Malaria Box Compound MMV030666 and Their Antiplasmodial Potencies. Pharmaceuticals. 2022; 15(12):1503. https://doi.org/10.3390/ph15121503

Chicago/Turabian Style

Hermann, Theresa, Robin Wallner, Johanna Dolensky, Werner Seebacher, Eva-Maria Pferschy-Wenzig, Marcel Kaiser, Pascal Mäser, and Robert Weis. 2022. "New Derivatives of the Multi-Stage Active Malaria Box Compound MMV030666 and Their Antiplasmodial Potencies" Pharmaceuticals 15, no. 12: 1503. https://doi.org/10.3390/ph15121503

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop