Open Access Article
Angeliki-Sofia Foscolos
ab,
Richard L. Athertonc,
Maria Billiad,
Markos-Orestis Georgiadisae,
Nuno Santarémf,
Anabela Cordeiro da Silvafg,
Martin C. Taylorc,
John M. Kellyc,
Theodora Calogeropoulouh,
Andrew Tsotinis
a,
Thomas Mavromoustakosd and
Ioannis P. Papanastasiou
*a
aSchool of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopoli-Zografou, 15771 Athens, Greece. E-mail: papanastasiou@pharm.uoa.gr
bInstitute of Nanoscience & Nanotechnology, NCSR “Demokritos”, 15341 Athens, Greece
cDepartment of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E7HT, UK
dFaculty of Chemistry, Department of Organic Chemistry, National and Kapodistrian University of Athens, Panepistimiopoli-Zografou, 15771 Athens, Greece
eCenter for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
fHost-Parasite Interaction Group, i3S, Institute for Research and Innovation in Health, University of Porto, Porto 4200-135, Portugal
gLaboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
hInstitute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
First published on 6th November 2025
In this article, we report the design, synthesis, and biological evaluation of a new series of nitroheterocyclic aromatic adamantane amides targeting trypanosomes. These compounds feature diverse substituents on the adamantane scaffold, variations in side chain linker length, and a range of nitroheterocyclic moieties. This work represents a continuation of our previous efforts, with a particular focus on elucidating the structural and functional role of the linker connecting the phenyladamantane core to the nitroheterocyclic ring. The structure–activity relationship data underscore the importance of strategic modifications in enhancing the pharmacological profile of these compounds against trypanosome parasites. Further modifications are recommended to optimize the physicochemical properties of the current derivatives to improve intracellular targeting of trypanosomatids, an important clinical stage in their life cycle.
Several of the currently available trypanocidal agents are nitroaromatic compounds: nifurtimox (Nfx) and benznidazole (Bzn) are the main treatments for CD, while nifurtimox is also used in combination therapy for the second stage of HAT.3 More recently, fexinidazole (Fxd) has replaced suramin and melarsoprol, as the first-line treatment, for individuals aged 6 years and older, who weigh 20 kg or more.4
Nifurtimox and benznidazole are prodrugs that undergo activation via a NADH-dependent, mitochondrially localized, bacterial-like type I nitroreductase (NTR).5 Moreover, NTR plays a crucial role in the activation of fexinidazole and its metabolites in L. donovani.6 Notably, the bacterial-like type I NTR, which bioactivates these nitroaromatic drugs, lacks homologs in mammals—a characteristic that contributes to their selective activity against trypanosomatids. The successful use of nitroaromatic drugs in treating trypanosomatid diseases has further encouraged the investigation of nitro derivatives for their chemotherapeutic potential, despite ongoing safety concerns.7
Based on the promising findings of our initial work on N-[4-(1-adamantyl)phenylalkyl]-5-nitrofuran-2-carboxamides 1a–c against T. brucei and T. cruzi,8 we report herein the synthesis and the trypanocidal evaluation of the nitroheterocyclic aromatic adamantane amides, 2a–c, 3a–c, 4a–c, 5a,b, 6a–f and 7a–c (Fig. 1).
As a structural variation of the N-[4-(1-adamantyl)phenylalkyl]-5-nitrofuran-2-carboxamides 1a,b, we synthesized the congeneric C-2 adducts, 2a–c. Both nitroimidazoles and bicyclic nitroimidazoles are well known for their antitrypanosomal activity.9 Building on the benznidazole scaffold, we also synthesized C-1 and C-2 adamantane hybrids, the reverse amides 3a–c and 4a–c, respectively. The (CH2)n spacer length between the adamantane cage and the aromatic ring varies from 0 to 2 methylene units in this series of derivatives. Additionally, we explored the effect of the spacer length between the nitroheterocyclic moiety and the amido group, producing adducts 5a and b. In addition, based on previous reports on the antitrypanosomal activity of nitro-triazole derivatives,10,11 we replaced the (2-nitro-1H-imidazol-1-yl) functional group with the (3-nitro-1H-1,2,4-triazol-1-yl) moiety in the adducts 6a–f, to further probe how the spacer length affects the amide bond's positioning relative to both ends of the side chain. Lastly, we compared the (1-adamantyl)anilinamides with different heterocyclic rings (7a and 7b) or different aromatic substitutions (para-7b vs. ortho-7c).
Both amines 10 and 12 (ref. 12) were converted into the desired N-substituted carboxamides 2a–b via amidation with 5-nitro-furoic acid, using 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HBTU) in the presence of diisopropylethylamine (DIPEA).
The synthesis of the N-[4-(2-adamantyl)phenethyl]-5-nitrofuran-2-carboxamide (2c) is outlined in Scheme 2. The reduction of the phenylacetonitrile 13 (ref. 13) to the corresponding amine was a key step in this process. Several reduction protocols were tested, including the use of lithium aluminum hydride and catalytic hydrogenation in the presence of RANEY®-Ni (vide SI). However, both approaches failed to produce the desired amine 14. After considerable experimentation, the desired reduction was effected with borane dimethylsulfide, affording the ethanamine 14, which was then converted into the desired carboxamide 2c, via the previously described method.
![]() | ||
Scheme 2 Reagents and conditions: a) BMS 2 M in toluene, THF, RT for 24 h then 50 °C for 5 h, 81%, b) 5-nitro-2-furoic acid, HBTU, DIPEA, DCM/DMF 1 : 1 (v/v), RT, 24 h, 76%. | ||
The 2-nitroimidazole acetamides 3a–c and 4a–c were obtained by the bromoacetylating the corresponding amines 10,12 12, and 14–17,14,15 using bromoacetyl chloride in a biphasic chloroform/water system in the presence of sodium carbonate. The resulting bromoacetamides 18–23 were then coupled with the sodium salt of 2-nitroimidazole (prepared from 2-nitroimidazole and sodium hydride), as demonstrated in Scheme 3.
The N-alkyl-2-nitroimidazole acetamides 5a–b were prepared by coupling the appropriate amines 24, 25 (ref. 16 and 17) with 2-[4-(1-adamantyl)phenyl]acetyl chloride,14 as shown in Scheme 4.
![]() | ||
| Scheme 4 Reagents and conditions: a) 2-[4-(1-adamantyl)phenyl]acetyl chloride, Et3N, DCM, RT, 24 h, 62% (5a), 58% (5b). | ||
The 3-nitrotriazole amides 6a–f were synthesized as depicted in Scheme 5. Alkylation of 3-nitro-1H-1,2,4-triazole (26) with N-(2-bromoethyl)phthalimide or N-(3-bromopropyl)phthalimide led to the corresponding diones 27, 28, which upon hydrazinolysis afforded the desired amines 29, 30, respectively. Interestingly, the amine 29 was obtained in 80% yield, compared to the reported 69%.18
These amines were then coupled with the appropriate carboxylic acids 4-(1-adamantyl)benzoic acid,19 2-[4-(1-adamantyl)phenyl]acetic acid,14 and 3-[4-(1-adamantyl)phenyl]propanoic acid,14 as previously shown, to give the desired benzamides 6a–f.
The synthesis of the (1-adamantyl)anilinamides 7a–c is illustrated in Scheme 6.
The N-substituted-5-nitrofuran-2-carboxamide 7a was synthesized via amidation of the 4-(1-adamantyl)aniline (15)14 with 5-nitrofuroic acid, using the protocol described for carboxamides 2a–c. Aniline 15 and 2-(1-adamantyl)aniline 31 (ref. 20) were bromoacylated to the corresponding amides 18, 33. The latter acetamides led to the desired anilamides 7a–c, as shown above.
| cmpd | T. brucei IC50a (μM) | T. brucei IC90a (μM) | L6† cells IC50 (μM) | S.I.b |
|---|---|---|---|---|
| a IC50 and IC90: concentrations that inhibit growth by 50 and 90%, respectively.b Selectivity index: ratio of IC50 values obtained with L6 cells and T. brucei.c Fexinidazole was tested under the same conditions as for the reported derivatives. | ||||
| 2a | 0.13 ± 0.04 | 0.33 ± 0.02 | 0.47 ± 0.07 | 4 |
| 2b | 0.017 ± 0.003 | 0.031 ± 0.001 | 1.6 ± 0.1 | 95 |
| 2c | 0.042 ± 0.008 | 0.073 ± 0.008 | 1.8 ± 0.2 | 42 |
| 6a | 0.23 ± 0.05 | 0.36 ± 0.03 | 28 ± 7 | 120 |
| 6b | 0.42 ± 0.07 | 1.8 ± 0.2 | 30 ± 2 | 72 |
| 6c | 0.20 ± 0.02 | 0.32 ± 0.01 | 23 ± 2 | 114 |
| 6d | 0.18 ± 0.05 | 0.31 ± 0.04 | 22 ± 1 | 123 |
| 6e | 0.24 ± 0.01 | 0.40 ± 0.01 | 23 ± 3 | 96 |
| 6f | 0.14 ± 0.02 | 0.28 ± 0.01 | 19 ± 2 | 134 |
| 7a | 0.091 ± 0.005 | 0.22 ± 0.01 | <0.03 | |
| 7b | 0.20 ± 0.07 | 0.91 ± 0.19 | 7.1 ± 0.3 | 35 |
| 7c | >25 | >25 | — | — |
| Nfx | 4.4 ± 0.7 (ref. 21) | — | 32. ± 0.1 (ref. 21) | 7 |
| Fxd | 2.4 ± 0.2c | — | >333 (ref. 22) | |
| cmpd | T. cruzi (epimastigotes) IC50a (μM) | T. cruzi (epimastigotes) IC90a (μM) | T. cruzi (intracellular amastigotes) IC50a (μM) | T. cruzi (intracellular amastigotes) IC90a (μM) | L6† cells IC50 (μM) | S.I.b (epimastigotes) IC50/Tc IC50 | S.I.b (intracellular) IC50/Tc IC50 |
|---|---|---|---|---|---|---|---|
| a IC50 and IC90: concentrations that inhibit growth by 50 and 90%, respectively.b Selectivity index: ratio of IC50 values obtained with L6 cells and T. cruzi.c Benznidazole was tested under the same conditions as for the reported derivatives. | |||||||
| 2a | 0.55 ± 0.05 | 1.2 ± 0.03 | 0.47 ± 0.07 | — | |||
| 2b | 0.56 ± 0.02 | 1.2 ± 0.02 | 1.6 ± 0.1 | 3 | |||
| 2c | 0.28 ± 0.08 | 1.2 ± 0.08 | 1.8 ± 0.2 | 6 | |||
| 6a | 0.54 ± 0.02 | 1.2 ± 0.01 | 4.88 ± 0.51 | 21.4 ± 3.4 | 28 ± 7 | 51 | 6 |
| 6b | 0.20 ± 0.03 | 0.66 ± 0.04 | 1.83 ± 0.70 | 4.29 ± 0.31 | 30 ± 2 | 154 | 16 |
| 6c | 0.45 ± 0.02 | 1.2 ± 0.02 | 1.58 ± 0.07 | 3.23 ± 0.85 | 23 ± 2 | 50 | 15 |
| 6d | 0.45 ± 0.01 | 1.1 ± 0.02 | 3.73 ± 0.44 | 8.42 ± 0.44 | 22 ± 1 | 49 | 6 |
| 6e | 0.14 ± 0.01 | 0.27 ± 0.02 | 2.48 ± 0.31 | 7.08 ± 0.47 | 23 ± 3 | 168 | 9 |
| 6f | 0.44 ± 0.02 | 0.91 ± 0.01 | 2.44 ± 0.57 | 8.23 ± 1.78 | 19 ± 2 | 43 | 8 |
| 7a | >0.025 | >0.025 | <0.03 | ||||
| 7b | — | — | 7.1 ± 0.3 | — | |||
| 7c | >25 | >25 | — | — | |||
| Nfx | 3.1 ± 0.5 (ref. 23) | — | 32. ± 0.1 (ref. 21) | 10 | |||
| Bzn | 5.4 ± 0.2c | — | 1.04 ± 0.32 | 2.54 ± 0.81 | 510 ± 22 (ref. 21) | 95 | 490 |
| cpmd | L. infantum (promastigotes) | L. infantum (intracellular amastigotes) | THP-1 CC50 (μM) | SI (promastigotes) CC50/Li IC50 | SI (intracellular amastigotes) CC50/Li ia IC50 | |
|---|---|---|---|---|---|---|
| IC50 (μM), 95% CI | % of growth inhibition at 10 μM ± ST DEV (μM, n ≥ 3, SD) | IC50 (μM), 95% CI | ||||
| Na: not active (average activity less than 40%); Nt: not tested. | ||||||
| 1a | 3.46 (2.56 to 4.57) | Nt | CC50 > 100 | SI > 29 | ||
| 1b | 0.66 (0.51 to 0.83) | Na | 100 > CC50 > 50 | 151 > SI > 76 | ||
| 2a | 0.12 (0.094 to 0.16) | Na | 6.25 > CC50 > 1.56 | 52 > SI > 26 | ||
| 2b | 0.09 (0.094 to 0.16) | 84.6 ± 15.6 | 3.97 (3.11 to 5.05) | 25 > CC50 > 12.5 | 272 > SI > 136 | 6 > SI > 3 |
| 2c | 0.05 (0.05 to 0.06) | Na | 12.5 > CC50 > 3.13 | 232 > SI > 58 | ||
| 3a | 0.70 (0.50 to 0.96) | Na | 100 > CC50 > 25 | 143 > SI > 36 | ||
| 3b | 13.81 (12.14 to 15.66) | Na | 100 > CC50 > 50 | 7 > SI > 4 | ||
| 3c | 22.13 (19.69 to 24.92) | Na | 100 > CC50 > 50 | 4 > SI > 2 | ||
| 4b | >40 | Na | CC50 > 100 | — | ||
| 4c | 37.87 (30.96 to 49.04) | Na | CC50 > 50 | SI > 1 | ||
| 5a | 3.48 (2.84 to 4.24) | Na | 50 > CC50 > 25 | 3 > SI > 14 | ||
| 5b | 4.81 (2.84 to 7.42) | Na | 100 > CC50 > 50 | 21 > SI > 10 | ||
| 6a | 0.74 (0.59 to 0.92) | Na | 50 > CC50 > 25 | 135 > SI > 34 | ||
| 6b | 0.49 (0.43 to 0.57) | 50.1 ± 23.0 | 100 > CC50 > 50 | 204 > SI > 102 | ||
| 6c | 0.42 (0.35 to 0.50) | 47.6 ± 16.3 | CC50 > 100 | SI > 238 | ||
| 6d | 0.27 (0.25 to 0.29) | Na | 100 > CC50 > 50 | 370 > SI > 185 | ||
| 6e | 0.31 (0.26 to 0.36) | 59.5 ± 12.0 | 6.93 (5.62 to 8.55) | 100 > CC50 > 50 | 322 > SI > 161 | 14 > SI > 7 |
| 6f | 0.39 (0.36 to0.43) | Na | 100 > CC50 > 50 | 256 > SI > 128 | ||
| 7a | 0.76 (0.53 to 1.06) | Na | 25 > CC50 > 6.25 | 33 > SI > 8 | ||
| MF | 11.93 (11.36 to 12.54) | 92 ± 5.0 | 2.65 (2.33 to 3.01) | 29.99 (24.57 to 36.52) | 2.5 | 11.3 |
From the results shown in Tables 1 and 2, it becomes apparent that among the 5-nitrofuran-2-carboxamides 2a–c, the derivatives bearing C-2 substituents on the adamantane ring exhibit higher activity compared to their C-1 substituted counterparts, 1a–c.8 The benzylamide derivative 2b and the phenylacetamide analogue 2c have IC50 values of 17 nM and 42 nM, respectively, against T. brucei. However, this trend is reversed against T. cruzi epimastigotes, where phenylacetamide 2c exhibits twice the activity of benzylamide 2b, with both being active in the sub-micromolar range. Conversely, their C-1 substituted congeners display, in general, activity, in a micromolar scale, with the exception of the anilamide 7a, which has an IC50 value of 91 nM against T. brucei, although this is likely due to toxicity, since it is also highly deleterious to the mammalian cells. Replacing the 5-nitrofuran unit at the terminal position of the tested amides with 2-nitroimidazole did not give the expected results, as derivatives 3a–c, 4a–c, and 5a,b were inactive (>25 μM). Similarly, inverting the amide bond in the lateral chain linking the 4-(1-adamantyl)phenyl scaffold to the 2-nitroimidazole moiety, had no effect on the antitrypanosomal activity, as the corresponding compounds 5a and 5b remained inactive. However, the replacement of the 2-nitro(1H-imidazol-1-yl) moiety with the 3-nitro(1H-1,2,4-triazol-1-yl) ring significantly enhanced the pharmacological activity of the tested compounds. A comparison of the triazole analogues reveals that the propyldiamino linker in the side chain induces stronger antitrypanosomal action than the ethyldiamino linker, as compounds 6d–f are more potent than 6a–c. However, both homologous series follow the same structure–activity relationship (SAR) pattern: when the carbonyl group is removed from the phenolic ring, the antitrypanosomal activity increases. The propanamides 6c and 6f are more active than the corresponding acetamides 6b and 6e, as well as the benzamides 6a and 6d. Within the acetamide and benzamide series, the trend is reversed, with benzamides 6a and 6d being more active than acetamides 6b and 6e, though the latter exhibit a better selectivity index (SI). The less cytotoxic derivatives were tested against intracellular T. cruzi amastigotes., their activity was found to be in the good to moderate range. Regarding the activity against intracellular amastigotes, the ethanediamino derivatives 6b and 6c show higher activity compared to the other nitrotriazole analogues.
The nitrofuramide 2b displays nanomolar-scale activity, and is 140–260 fold more potent against T. brucei than the reference drugs fexinidazole and nifurtimox. Regarding the anilamides 7a–c, the short side chain of 7a reduces trypanocidal activity. Comparing the two aromatic isomers 7b and 7c, it is evident that the steric hindrance in the ortho-substituted 7c also reduces activity.
The results shown in Table 3, with respect to L. infantum, point towards the same pharmacological profile for the adamantane nitroheterocyclic amides. Most of the tested molecules were active at sub micromolar potency against promastigotes. The C-2 adamantane 5-nitrofuran-2-carboxamides substituted analogues, 2a–c, exhibit higher activity than their C-1 substituted congeners 1a,b.8 High anti-parasitic potency of 2a–c was also observed for T. brucei (Table 2). However, 2a–c were more toxic to the THP-1 cells used as hosts for the intracellular infections, with 2a being the most cytotoxic molecule tested. Replacing the 2-nitrofuran-1-yl moiety (in derivatives 1a,b, 2a–b) with the 2-nitro-(1H-imidazol-1-yl) ring (derivatives 3a–c, 4a–c, 5a,b) did not improve antileishmanial activity. Notably, the 2-nitroimidazole reverse amide 5a was six-fold more potent than the corresponding 2-nitroimidazole amide 3c with the same linker length. Once again, the 3-nitro-1H-1,2,4-triazole adducts were more potent than the 2-nitro-1H-imidazole compounds. However, in this case, the length of the diamine spacer between the amide bond and the heterocycle is not a determining factor for antileishmanial activity, as the propanediamino adducts 6e–f were only slightly more potent than the ethanediamino derivatives 6a–c. Among the tested compounds, nitrofuramide 2c was the most potent against L. infantum promastigotes, whilst nitrotriazole 6d exhibited the best balance between promastigote activity and cytotoxicity. Nitrofuramide 2b has an equipotent SI to the nitrotriazole 6d, but exhibits better antileishmanial activity, close to that of the derivative 2c, the most potent compound tested against promastigotes. The nitrofuramide 2b was also active against L. infantum amastigotes, although the IC50 value (3.97 μM) was higher. The toxicity issues of the C-2 adamantane 5-nitrofuran-2-carboxamides substituted analogues, however, do not result in a favourable SI (3 to 6). Only three other molecules presented significant activity against intracellular amastigotes (6b, 6c and 6e). In fact, 6e was the most promising molecule with a predicted SI between 7 and 14. It is noteworthy, that 6e was among the most potent tested against T. cruzi (both epimastigotes and amastigotes) and the molecule with the best SI (Table 3). These indices facilitate the development of useful structure–activity relationships.
In addition to the biological evaluation, an in silico assessment of the compounds' drug-likeness and predicted toxicity was conducted to further explore their potential as drug candidates. Drug-likeness and toxicity predictions for the synthetic nitro amides were performed using the SwissADME and ProTox-3.0 platforms, respectively.
The physicochemical properties, drug-likeness, and toxicological profiles of the adamantylamide compounds were evaluated to provide a comprehensive understanding of their potential. These compounds display a lipophilic backbone while adhering to Lipinski's rule of five. Due to their high lipophilicity, the compounds exhibit low ESOL values, while maintaining favorable properties in terms of rotatable bonds, hydrogen bond donors and acceptors, and surface area (TSA). Minor variations based on structural differences were observed. All derivatives show excellent drug-likeness, as supported by the biological assays.
While pharmacologically promising, the nitro group (–NO2) in their backbone places them within the PAINS (Pan assay interference compounds) functional groups. Additionally, the presence of a long aliphatic carbon chain may increase toxicity, as such molecules are more likely to bind multiple targets. According to the Protox 3 platform, without accounting for potential prodrug activity, this structure predicts potential respiratory, toxicity, carcinogenicity, and mutagenicity. However, considering the presence of the nitro group and its potential for activation via metabolic processes, these predictions may not fully reflect the compound's in vivo behavior.
In a previous study,24 we have synthesized both nitro- and non-nitro furan derivatives, and the lack of activity observed in the non-nitro compounds indicated that the presence of the nitro group on the furan ring is essential for conferring trypanocidal activity. Based on this indirect evidence regarding the mechanism of action, along with the rational design of our compounds inspired by benznidazole, we hypothesize that the current derivatives act through a similar pathway to that of known commercial nitroheterocyclic drugs—namely, activation by parasite-specific nitroreductase enzymes.
Summary tables and a heatmap of the results for each compound are provided in the SI.
:
mScarlet25 culture form trypomastigotes (1
:
1 multiplicity of infection). After 16 hours, extracellular parasites were removed by washing in growth medium (×3), test compounds were added, and the plates incubated for 5 days. Fluorescence intensities were then determined using a BMG FLUOstar Omega plate reader (ex 545 nm, em 590 nm), and the data analysed using GraphPad Prism 9.0 software. The values were expressed as IC50/90 ± SD and are the mean of triplicate experiments.
Leishmania infantum Luciferase-expressing L. infantum (strain MHOM/MA/67/ITMAP-263) axenic amastigotes were cultured in MAA/20 medium at 37 °C with 5% CO2, as described by Sereno et al. 1998.26 The parasites were maintained in 5 mL T25 ventilated flasks and subcultured every seven days at a concentration of 1 × 106 cells per mL.
The THP-1 cell line, a human leukemia line (ATCC® TIB-202™), was cultured in RPMI-1640 medium supplemented with 10% heat-inactivated fetal bovine serum (FBS), 2 mM. L-Glutamine, 100 IU mL−1 penicillin/streptomycin, and 20 mM HEPES. The cells were maintained in a humidified incubator at 37 °C with 5% CO2. Subculturing was performed every three days, using 20 mL of media at a concentration of 2 × 105 cells per mL in a T75 flask. All cell culture reagents were purchased from Lonza Bioscience (Morrisville, NC).
The efficacy of the compounds against L. infantum promastigotes was evaluated using a resazurin-based assay. Parasites were added to 100 μl of serial dilutions of the compounds in supplemented complete medium at a cell density of 5 × 105 ml−1. As a quality control, a dose–response curve with the antileishmanial drug miltefosine was included in all assays. The final volume of each assay was 200 μl per well, and each condition was tested in duplicate. After a 72-hour incubation under specific conditions for each parasite, 20 μl of a 0.5 mM resazurin solution was added, and the plates were incubated for an additional 4 hours under the same conditions. Fluorescence was measured at 544 nm for excitation and 590 nm for emission using a Synergy 2 Multi-Mode Reader (Biotek, Winooski, VT, USA). Results were expressed as the percentage of parasite growth inhibition compared to controls (untreated parasites) and represent the average of at least three independent experiments. The effect was evaluated by determining the IC50 value (the concentration required to inhibit 50% growth), calculated using non-linear regression curves via GraphPad Prism version 8.1.1 for Windows (GraphPad Software, San Diego, CA, USA).
:
10 for four hours at 37 °C in a 5% CO2 atmosphere. After this incubation, non-internalized parasites were washed away, and various concentrations of test compounds were added in a final volume of 100 μL. A dose–response curve for miltefosine was included in all assays to serve as a quality control. Each experimental condition was tested in quadruplicate. After 72 hours of incubation, the medium was replaced with 100 μL of PBS, and 25 μL of Glo-lysis buffer from the Steady-Glo Luciferase Assay System (Promega, Madison, WI, USA) was added. The plates were then agitated at 100 rpm for 10 minutes at room temperature. Following this, 30 μL of the Steady-Glo reagent (Promega, Madison, WI, USA) was added, and the mixture was incubated for 15 minutes in the dark under the same conditions. A total of 140 μL from each well was transferred to white-bottom 96-well plates, and luminescence intensity was measured using a Synergy 2 Multi-Mode Reader (Biotek, Winooski, VT, USA). The antileishmanial effect was assessed by comparing the results with those from non-treated infected cells. The IC50 value were determined through non-linear regression analysis using GraphPad Prism version 8.1.1 for Windows (GraphPad Software, San Diego, CA, USA). Results represent the average of at least three independent experiments.
The cytotoxicity of the compounds on THP-1-derived macrophages was evaluated using the colorimetric MTT assay (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide), as described in Magoulas et al. (2021).28 THP-1 cells were suspended in complete RPMI medium at a density of 1 × 106 cells per mL, and 100 μL of this suspension was seeded into each well of a 96-well plate. The cells were differentiated into macrophages by adding 40 ng mL−1 of phorbol-myristate 13-acetate (PMA, Sigma, Saint Louis, MI, USA) for 24 hours, followed by replacement with fresh medium for an additional 24 hours. Subsequently, the cells were incubated with 100 μL of various compounds, diluted in complete RPMI medium, at concentrations ranging from 100 to 12.5 μM. Each condition was tested in triplicate. After 72 hours of incubation at 37 °C with 5% CO2, the medium was removed, and 200 μL of a 0.5 mg mL−1 MTT solution, diluted in RPMI, was added to each well. The plates were incubated for an additional 4 hours. Following this incubation, 160 μL of the medium was removed and replaced with 160 μL of 2-propanol. Absorbance was measured at 570 nm using a Synergy 2 Multi-Mode Reader (Biotek, Winooski, VT, USA). Cytotoxicity was determined by presenting the CC50 (the concentration of the drug that reduces cell viability by 50%) interval or by calculating the CC50 value through non-linear regression analysis using GraphPad Prism version 8.1.1 for Windows (GraphPad Software, San Diego, CA, USA). The results represent the average of at least three independent experiments.
N): 2133.5 cm−1, v(C
O) 1701.2 cm−1.
The product is a mixture of rotamers A and B (Fig. 2).
1H-NMR (CDCl3), δ (ppm): (rotamer A): 1.56–1.59 (∼d, 2H, 4,9-Heq), 1.64–1.79 (complex m, 6H, 4,9-Hax, 5,6,7-H), 1.85–2.04 (complex m, 4H, 8,10-H), 2.49 (s, 1H, 1-H), 2.62–2.65 (∼d, 1H, 3-H), 2.88 (s, 1H, 2-H), 5.93 (br.s, 2H, NH2), 7.42–7.44 (d, 2H, J ≈ 8.7 Hz, 3,5-Har), 7.76–7.78 (d, 2H, J ≈ 8.7 Hz, 2,6-Har); 13C-NMR (100 MHz, DMSO-d6), δ (ppm): 26.79 (7-C), 27.81 (5-C), 30.21 (1-C), 32.12 (4,9-C), 33.84 (3-C), 35.12 (6-C), 39.19 (8,10-C), 47.11 (2-C), 127.28 (3,5-Car), 127.42 (2,6-Car), 130.32 (4-Car), 149.32 (1-Car), 169.71 (C
O).
1H-NMR (CDCl3), δ (ppm): (rotamer B): 1.56–1.59 (∼d, 2H, 4,9-Heq), 1.64–1.79 (complex m, 6H, 4,9-Hax, 5,6,7-H), 1.85–2.04 (complex m, 4H, 8,10-H), 2.49 (s, 1H, 1-H), 2.62–2.65 (∼d, 1H, 3-H), 3.03 (s, 1H, 2-H), 5.93 (br.s, 2H, NH2), 7.62–7.64 (d, 2H, J ≈ 8.7 Hz, 3,5-Har), 7.82–7.84 (d, 2H, J ≈ 8.7 Hz, 2,6-Har); 13C-NMR (100 MHz, DMSO-d6), δ (ppm): 27.19 (7-C), 28.07 (5-C), 31.19 (1-C), 32.12 (4,9-C), 33.84 (3-C), 37.30 (2-C), 37.87 (6-C), 39.19 (8,10-C), 125.59 (3,5-Car), 128.03 (2,6-Car), 132.43 (4-Car), 148.35 (1-Car), 169.71 (C
O).
:
anhydrous DCM (1
:
1, 10 mL), the following reagents were sequentially added: 5-nitro-furoic acid (143 mg, 0.91 mmol), HBTU (345 mg, 0.91 mmol), and DIPEA (0.5 ml, 343 mg, 2.65 mmol). The mixture was stirred at room temperature for 24 hours under an argon atmosphere. The reaction mixture was then extracted with ethyl acetate, and the combined organic layers were washed with water, and brine, dried over Na2SO4, and evaporated in vacuo. The residue was purified by column chromatography using a gradient elution of methanol in ethyl acetate (0–5%) to afford 7a as a yellow-orange solid (210 mg, 76%).
O). Anal. calcd for C21H22N2O4: C, 68.84; H, 6.05; N, 7.65; found: C, 68.51; H, 6.38; N, 7.83.The product is a mixture of E and Z conformers. E > Z.
1H-NMR (400 MHz, CDCl3), δ (ppm): (conformer E): 1H-NMR (400 MHz, CDCl3), δ (ppm): 1.55–1.58 (d, 2H, J ≈ 12 Hz 4,9-Heq), 1.77–1.84 (m, 5H, 4,9-Hax, 5,6-H), 1.92–2.02 (m, 5H, 7,8,10-H), 2.46 (s, 2H, 1,3-H), 3.00 (s, 1H, 2-H), 4.60–4.62 (d, 2H, J ≈ 6 Hz, α-H), 6.83 (s, 1H, NH), 7.28–7.31 (m, 3H, 3-Hf, 3,5-Har), 7.35–7.37 (m, 3H, 4-Hf, 2,6-Har); 13C-NMR (100 MHz, CDCl3), δ (ppm): 27.88 (7-C), 28.15 (5-C), 31.20 (1,3-C), 32.09 (4,9-C), 37.98 (6-C), 39.25 (8,10-C), 43.54 (α-C), 46.80 (2-C), 112.57 (4-Cf), 116.21 (3-Cf), 127.59 (2,6-Car), 128.16 (3,5-Car), 133.73 (1-Car), 144.66 (2-Cf), 148.16 (4-Car), 151.41 (5-Cf), 156.20 (C
O).
1H-NMR (CDCl3), δ (ppm): (400 MHz) (conformer Z): 1H-NMR (400 MHz, CDCl3), δ (ppm): 1.55–1.58 (d, 2H, J ≈ 12 Hz 4,9-Heq), 1.77–1.84 (m, 5H, 4,9-Hax, 5,6-H), 1.92–2.02 (m, 5H, 7,8,10-H), 2.46 (s, 2H, 1,3-H), 3.00 (s, 1H, 2-H), 4.64–4.65 (∼d, 2H, α-H), 6.83 (s, 1H, NH), 7.28–7.31 (m, 1H, 3-Hf), 7.35–7.37 (m, 1H, 4-Hf), 7.46–7.48 (∼d, 2H, 3,5-Har), 7.53–7.55 (∼d, 2H, 2,6-Har); 13C-NMR (100 MHz, CDCl3), δ (ppm): 27.88 (7-C), 28.15 (5-C), 31.20 (1,3-C), 32.09 (4,9-C), 37.98 (6-C), 39.25 (8,10-C), 43.40 (α-C), 46.80 (2-C), 112.57 (4-Cf), 116.21 (3-Cf), 126.24 (2,6-Car), 127.84 (3,5-Car), 133.73 (1-Car), 144.66 (2-Cf), 148.16 (4-Car), 151.41 (5-Cf), 156.20 (C
O). Anal. calcd for C22H24N2O4: C, 69.46; H, 6.36; N, 7.36; found: C, 69.27; H, 6.49; N, 7.11.
1H-NMR (600 MHz, CDCl3), δ (ppm): 1.55–1.57 (d, 2H, J ≈ 13 Hz, 4,9-Heq), 1.77–1.85 (m, 5H, 4,9-Hax, 5,6-H), 1.93–2.02 (m, 5H, 7,8,10-H), 2.46 (s, 2H, 1,3-H), 2.91–2.93 (t, 2H, J ≈ 7.1 Hz, β-CH2), 2.99 (s, 1H, 2-H), 3.70–3.73 (d, 2H, J ≈ 6.8 Hz, α-H), 6.61 (s, 1H, NH), 7.28–7.31 (d, 2H, J ≈ 7.6 Hz, 3,5-Har), 7.23–7.24 (d, 1H, J ≈ 4 Hz, 3-Hf), 7.32–7.34 (m, 3H, 4-Hf, 2,6-Har); 13C-NMR (150 MHz, CDCl3), δ (ppm): 27.97 (7-C), 28.22 (5-C), 31.26 (1,3-C), 32.13 (4,9-C), 35.20 (β-C), 38.06 (6-C), 39.33 (8,10-C), 43.54 (α-C), 46.80 (2-C), 112.47 (4-Cf), 115.88 (3-Cf), 127.49 (3,5-Car), 128.64 (2,6-Car), 134.90 (1-Car), 143.26 (2-Cf), 148.32 (4-Car), 151.36 (5-Cf), 156.35 (C
O); anal. calcd for C23H26N2O4: C, 70.03; H, 6.64; N, 7.10; found: C, 70.17; H, 6.38; N, 6.99.
O); anal. calcd for C21H22N2O4: C, 68.84; H, 6.05; N, 7.65; found: C, 69.03; H, 6.23; N, 7.44.
O); anal. calcd for C21H24N4O3: C, 66.30; H, 6.36; N, 14.73; found: C, 66.46; H, 6.09; N, 14.49.The product is a mixture of E and Z conformers. E/Z: 1.8.
1H-NMR (400 MHz, CDCl3), δ (ppm): (conformer E): 1.73–1.81 (∼q, 6H, 4,6,10-H), 1.89 (br.s, 6H, 2,8,9-H), 2.10 (s, 3H, 3,5,7-H), 4.42–4.44 (d, 2H, J ≈ 5.6 Hz, α-CH2), 5.00 (s, 2H, α′-CH2), 6.22 (br.s, 1H, NH), 7.13–7.16 (m, 2H, 2,6-Har), 7.20–7.22 (d, J ≈ 8.2 Hz, 4-Him), 7.25–7.30 (m, 2H, 3,5-Har), 7.33–7.35 (d, J ≈ 8.2 Hz, 5-Him); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.02 (3,5,7-C), 36.38 (1-C), 36.85 (4,6,10-C), 43.99 (2,8,9-C), 44.58 (α-C), 52.43 (α′-C), 124.72 (3,5-Car), 125.57 (5-Cim), 127.31 (4-Cim), 128.71 (2,6-Car), 136.81 (1-Car), 144.84 (2-Cim), 152.41 (4-Car), 164.59 (C
O).
1H-NMR (400 MHz, CDCl3), δ (ppm): (conformer E): 1.73–1.81 (∼q, 6H, 4,6,10-H), 1.89 (br.s, 6H, 2,8,9-H), 2.10 (s, 3H, 3,5,7-H), 4.42–4.47 (m, 2H, α-CH2), 5.00–5.01 (∼d, 2H, α′-CH2), 6.22 (br.s, 1H, NH), 7.06–7.08 (∼d, 1H, 4-Him), 7.13–7.16 (m, 2H, 3,5-Har), 7.20–7.35 (very complex m, 3H, 5-Him, 2,6-Har); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.02 (3,5,7-C), 36.38 (1-C), 36.85 (4,6,10-C), 43.99 (2,8,9-C), 43.94 (α-C), 52.37 (α′-C), 124.72 (2,6-Car), 125.19 (5-Cim), 127.86 (4-Cim), 128.78 (2,6-Car), 134.18 (1-Car), 144.84 (2-Cim), 151.34 (4-Car), 164.59 (C
O). Anal. calcd for C22H26N4O3: C, 66.99; H, 6.64; N, 14.20; found: C, 67.16; H, 6.97; N, 14.09.
The product is a mixture of E and Z conformers. E/Z: 1.6.
1H-NMR (400 MHz, CDCl3), δ (ppm): (conformer E): 1.73–1.81 (∼q, 6H, 4,6,10-H), 1.90 (br.s, 6H, 2,8,9-H), 2.09 (s, 3H, 3,5,7-H), 2.80–2.85 (m, 2H, β-CH2), 2.93 (s, 2H, α′-CH2), 3.51–3.57 (m, 2H, α-CH2), 7.02–7.04 (d, 1H, J ≈ 6.6 Hz, 4-Him), 7.14–7.31 (very complex m, 6H, NH, 5-Him, 3,5-Har, 2,6-Har); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.07 (3,5,7-C), 35.33 (1-C), 36.18 (β-C), 36.92 (4,6,10-C), 40.31 (α-C), 43.34 (2,8,9-C), 63.25 (α′-C), 123.13 (3,5-Car), 125.14 (5-Cim), 125.57 (4-Cim), 128.60 (2,6-Car), 138.73 (1-Car), 149.72 (2-Cim), 151.84 (4-Car), 170.57 (C
O).
1H-NMR (400 MHz, CDCl3), δ (ppm): (conformer Z): 1.73–1.81 (∼q, 6H, 4,6,10-H), 1.90 (br.s, 6H, 2,8,9-H), 2.09 (s, 3H, 3,5,7-H), 2.80–2.85 (m, 2H, β-CH2), 2.92 (s, 2H, α′-CH2), 3.51–3.57 (m, 2H, α-CH2), 7.02–7.04 (d, 1H, J ≈ 6.6 Hz, 4-Him), 7.14–7.31 (very complex m, 6H, NH, 5-Him, 3,5-Har, 2,6-Har); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.07 (3,5,7-C), 35.33 (1-C), 36.05 (β-C), 36.92 (4,6,10-C), 40.16 (α-C), 43.34 (2,8,9-C), 63.19 (α′-C), 123.13 (3,5-Car), 125.14 (5-Cim), 125.97 (4-Cim), 128.47 (2,6-Car), 136.03 (1-Car), 149.72 (2-Cim), 151.84 (4-Car), 170.57 (C
O). Anal. calcd for C23H28N4O3: C, 67.63; H, 6.91; N, 13.72; found: C, 67.36; H, 7.13; N, 13.44.
O); anal. calcd for C21H24N4O3: C, 66.30; H, 6.36; N, 14.73; found: C, 66.38; H, 6.21; N, 14.66.
O); anal. calcd for C22H26N4O3: C, 66.99; H, 6.64; N, 14.20; found: C, 66.75; H, 6.72; N, 14.47.
O); anal. calcd for C23H28N4O3: C, 67.63; H, 6.91; N, 13.72; found: C, 67.67; H, 6.84; N, 13.91.The product is a mixture of E and Z conformers. E/Z: 10/3.
1H-NMR (CDCl3), δ (ppm): (conformer E) (400 MHz, CDCl3), δ (ppm): 1.65–1.74 (q, 6H, J ≈ 11 Hz 4,6,10-H), 1.82 (br.s, 6H, 2,8,9-H), 2.03 (s, 3H, 3,5,7-H), 3.49 (s, 2H, α-CH2), 3.56–3.59 (∼d, 2H, α′-CH2), 4.44–4.47 (t, 2H, J ≈ 5.6 Hz, β′-CH2), 6.04 (br.s, 1H, NH), 6.70–6.71 (∼d, 1H, 5-Him), 6.87–6.89 (∼d, 1H, 4-Him), 7.19–7.27 (m, 4H, 2,3,5,6-Har); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.00 (3,5,7-C), 36.36 (1-C), 36.83 (4,6,10-C), 39.83 (α′-C), 43.30 (2,8,9-C), 44.06 (α-C), 49.14 (β′-C), 124.29 (3,5-Car), 125.78 (2,6-Car), 127.26 (5-Cim), 128.23 (4-Cim), 134.29 (1-Car), 144.57 (2-Cim), 152.57 (4-Car), 172.48 (C
O).
1H-NMR (CDCl3), δ (ppm): (conformer Z) (400 MHz, CDCl3), δ (ppm): 1.65–1.74 (q, 6H, J ≈ 11 Hz 4,6,10-H), 1.82 (br.s, 6H, 2,8,9-H), 2.03 (s, 3H, 3,5,7-H), 3.46 (s, 2H, α-CH2), 3.56–3.59 (∼d, 2H, α′-CH2), 4.44–4.47 (t, 2H, J ≈ 5.6 Hz, β′-CH2), 6.04 (br.s, 1H, NH), 6.70–6.71 (∼d, 1H, 5-Him), 6.87–6.89 (∼d, 1H, 4-Him), 6.96–6.98 (∼d, 2H, 3,5- Har), 7.08–7.11 (∼d, 2H, 2,6-Har); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.01 (3,5,7-C), 36.20 (1-C), 36.84 (4,6,10-C), 39.83 (α′-C), 43.30 (2,8,9-C), 44.06 (α-C), 49.14 (β′-C), 126.51 (3,5-Car), 127.26 (5-Cim), 128.23 (4-Cim), 129.15 (2,6-Car), 131.55 (1-Car), 144.57 (2-Cim), 150.95 (4-Car), 172.48 (C
O). Anal. calcd for C23H28N4O3: C, 67.63; H, 6.91; N, 13.72; found: C, 67.58; H, 7.13; N, 13.58.
The product is a mixture of E and Z conformers. E/Z: 3/1.
1H-NMR (CDCl3), δ (ppm): (conformer E) (400 MHz, CDCl3), δ (ppm): 1.72–1.81 (∼q, 6H, 4,6,10-H), 1.90 (br.s, 6H, 2,8,9-H), 1.96–1.99 (m, 2H, β′-CH2), 2.09 (s, 3H, 3,5,7-H), 3.25–3.30 (q, 2H, J ≈ 6.2 Hz, α′-CH2), 3.59 (s, 2H, α-CH2), 4.33–4.36 (t, 2H, J ≈ 6.7 Hz, γ′-CH2), 5.61 (br.s, 1H, NH), 7.11 (s, 1H, 5-Him), 7.17–7.21 (m, 2H, 3,5-Har), 7.25–7.26 (∼d, 1H, 4-Him), 7.31–7.33 (m, 2H, 2,6-Har); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.01 (3,5,7-C), 31.16 (β′-C), 36.22 (1-C), 36.40 (α′-C), 36.84 (4,6,10-C), 43.30 (2,8,9-C), 44.32 (α-C), 47.77 (γ′-C), 124.39 (2,6-Car), 125.93 (3,5-Car), 126.26 (4-Cim), 128.64 (5-Cim), 134.34 (1-Car), 144.27 (2-Cim), 152.72 (4-Car), 172.21 (C
O).
1H-NMR (CDCl3), δ (ppm): (conformer E) (400 MHz, CDCl3), δ (ppm): 1.72–1.81 (∼q, 6H, 4,6,10-H), 1.90 (br.s, 6H, 2,8,9-H), 1.96–1.99 (m, 2H, β′-CH2), 2.09 (s, 3H, 3,5,7-H), 3.25–3.30 (q, 2H, J ≈ 6.2 Hz, α′-CH2), 3.56 (s, 2H, α-CH2), 4.33–4.36 (t, 2H, J ≈ 6.7 Hz, γ′-CH2), 5.61 (br.s, 1H, NH), 7.08–7.11 (m, 2H, 3.5-Har),7.11 (s, 1H, 5-Him), 7.25–7.26 (∼d, 1H, 4-Him), 7.35–7.37 (m, 2H, 2,6-Har); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.01 (3,5,7-C), 31.16 (β′-C), 36.22 (1-C), 36.40 (α′-C), 36.84 (4,6,10-C), 43.30 (2,8,9-C), 44.32 (α-C), 47.77 (γ′-C), 126.26 (4-Cim), 126.61 (3,5-Car), 128.64 (5-Cim), 129.32 (2,6-Car), 134.34 (1-Car), 144.27 (2-Cim), 152.72 (4-Car), 172.21 (C
O). Anal. calcd for C24H30N4O3: C, 68.22; H, 7.16; N, 13.26; found: C, 68.16; H, 7.44; N, 13.45.
:
anhydrous DCM (1
:
1, 10 mL), the following reagents were sequentially added: 4-(1-adamantyl)-benzoic acid19 (318 mg, 1.2 mmol), HBTU (470 mg, 1.2 mmol), and DIPEA (467 mg, 3.7 mmol, 0.6 ml). The mixture was stirred at room temperature for 24 hours under an argon atmosphere. The reaction mixture was then extracted with ethyl acetate, and the combined organic layers were washed with water and brine, dried over Na2SO4, and evaporated in vacuo. The residue was purified by column chromatography using a gradient elution of methanol in ethyl acetate (0–20%) to afford benzamide 6a as a white crystalline solid. (290 mg, 71%).
The product is a mixture of E and Z conformers. E/Z: 9/1.
1H-NMR (CDCl3), δ (ppm): (conformer E) (600 MHz, CDCl3), δ (ppm):1.73–1.81 (q, 6H, J ≈ 21.6 Hz 4,6,10-H), 1.90 (br.s, 6H, 2,8,9-H), 2.10 (s, 3H, 3,5,7-H), 3.92–3.95 (q, 2H, J ≈ 5.5 Hz, α′-CH2), 4.58–4.59 (t, 2H, J ≈ 5.5 Hz, β′-CH2), 6.67 (br.s, 1H, NH), 7.39–7.41 (d, 2H, J ≈ 8.1 Hz, 3,5-Har), 7.65–7.66 (d, 2H, J ≈ 8.1 Hz, 2,6-Har), 8.15 (s, 1H, 5-Ht); 13C-NMR (150 MHz, CDCl3), δ (ppm): 28.96 (3,5,7-C), 31.71 (1-C), 36.80 (4,6,10-C), 39.69 (α′-C), 43.10 (2,8,9-C), 50.44 (β′-C), 125.52 (3,5-Car), 126.97 (2,6-Car), 130.41 (1-Car), 145.91 (5-Ct), 156.11 (4-Car), 163.27 (3-Ct), 168.55 (C
O).
1H-NMR (CDCl3), δ (ppm): (conformer Z) (600 MHz, CDCl3), δ (ppm): 1.73–1.81 (q, 6H, J ≈ 21.6 Hz 4,6,10-H), 1.90 (br.s, 6H, 2,8,9-H), 2.10 (s, 3H, 3,5,7-H), 3.92–3.95 (q, 2H, J ≈ 5.5 Hz, α′-CH2), 4.58–4.59 (t, 2H, J ≈ 5.5 Hz, β′-CH2), 6.72 (br.s, 1H, NH), 7.39–7.41 (d, 2H, J ≈ 8.1 Hz, 3,5-Har), 7.65–7.66 (d, 2H, J ≈ 8.1 Hz, 2,6-Har), 8.17 (s, 1H, 5-Ht); 13C-NMR (150 MHz, CDCl3), δ (ppm): 28.96 (3,5,7-C), 31.71 (1-C), 36.80 (4,6,10-C), 39.69 (α′-C), 43.10 (2,8,9-C), 50.44 (β′-C), 125.52 (3,5-Car), 126.97 (2,6-Car), 130.41 (1-Car), 145.91 (5-Ct), 156.11 (4-Car), 163.27 (3-Ct), 168.55 (C
O). Anal. calcd for C21H25N5O3: C, 63.78; H, 6.37; N, 17.71; found: C, 63.66; H, 6.59; N, 17.63.
The product is a mixture of E and Z conformers. E/Z: 4/1.
1H-NMR (CDCl3), δ (ppm): (conformer E) (400 MHz, CDCl3), δ (ppm): 1.72–1.81 (m, 6H, 4,6,10-H), 1.88 (br.s, 6H, 2,8,9-H), 2.09 (s, 3H, 3,5,7-H), 3.50 (s, 2H, α-CH2), 3.67–3.71 (q, 2H, J ≈ 5.5 Hz, α′-CH2), 4.41–4.44 (t, 2H, J ≈ 5.7 Hz, β′-CH2), 5.67 (br.s, 1H, NH), 7.07–7.09 (d, 2H, J ≈ 8.2 Hz, 3,5-Har), 7.30–7.32 (d, 2H, J ≈ 8.2 Hz, 2,6-Har), 7.98 (s, 1H, 5-Ht); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.01 (3,5,7-C), 36.20 (1-C), 36.84 (4,6,10-C), 39.12 (α′-C), 43.20 (2,8,9-C), 43.26 (α-C), 49.99 (β′-C), 125.97 (3,5-Car), 129.07 (2,6-Car), 131.06 (1-Car), 145.70 (5-Ct), 151.30 (4-Car), 163.15 (3-Ct), 172.63 (C
O).
1H-NMR (CDCl3), δ (ppm): (conformer Z) (400 MHz, CDCl3), δ (ppm): 1.72–1.81 (m, 6H, 4,6,10-H), 1.88 (br.s, 6H, 2,8,9-H), 2.09 (s, 3H, 3,5,7-H), 3.53 (s, 2H, α-CH2), 3.67–3.71 (q, 2H, J ≈ 5.5 Hz, α′-CH2), 4.41–4.44 (t, 2H, J ≈ 5.7 Hz, β′-CH2), 5.67 (br.s, 1H, NH), 7.07–7.09 (d, 2H, J ≈ 8.2 Hz, 3,5-Har), 7.30–7.32 (d, 2H, J ≈ 8.2 Hz, 2,6-Har), 7.92 (s, 1H, 5-Ht); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.01 (3,5,7-C), 36.20 (1-C), 36.84 (4,6,10-C), 39.12 (α′-C), 43.20 (2,8,9-C), 43.26 (α-C), 49.99 (β′-C), 125.97 (3,5-Car), 129.07 (2,6-Car), 131.06 (1-Car), 145.70 (5-Ct), 151.30 (4-Car), 163.15 (3-Ct), 172.63 (C
O). Anal. calcd for C22H27N5O3: C, 64.53; H, 6.65; N, 17.10; found: C, 64.72; H, 6.34; N, 17.04.
The product is a mixture of E and Z conformers. E/Z: 6/1.
1H-NMR (400 MHz, CDCl3), δ (ppm): (conformer E) 1.71–1.80 (q, 6H, J ≈ 8.47 Hz, 4,6,10-H), 1.88 (br.s, 6H, 2,8,9-H), 2.08 (br.s, 3H, 3,5,7-H), 2.46–2.49 (t, 2H, J ≈ 7.4 Hz, α-CH2), 2.86–2.95 (quintet, 2H, J ≈ 7.2 Hz, β-CH2), 3.64–3.69 (q, 2H, J ≈ 5.3 Hz, α′-CH2), 4.31–4.34 (t, 2H, J ≈ 5.5 Hz, β′-CH2), 5.85–5.88 (∼t, 1H, NH), 7.12–7.14 (d, 2H, J ≈ 7.12 Hz, 3,5-Har), 7.28–7.30 (d, 2H, J ≈ 7.12 Hz, 2,6-Har), 7.65 (s, 1H, 5-Ht); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.05 (3,5,7-C), 30.98 (β-C), 36.09 (1-C), 36.87 (4,6,10-C), 37.96 (α-C), 39.16 (α′-C), 43.32 (2,8,9-C), 43.36 (β′-C), 125.28 (3,5-Car), 128.31 (2,6-Car), 137.39 (1-Car), 145.74 (5-Ct), 149.96 (4-Car), 163.15 (3-Ct), 173.33 (C
O).
1H-NMR (400 MHz, CDCl3), δ (ppm): (conformer Z) 1.71–1.80 (q, 6H, J ≈ 8.47 Hz, 4,6,10-H), 1.88 (br.s, 6H, 2,8,9-H), 2.08 (br.s, 3H, 3,5,7-H), 2.46–2.49 (t, 2H, J ≈ 7.4 Hz, α-CH2), 2.86–2.95 (quintet, 2H, J ≈ 7.2 Hz, β-CH2), 3.53–3.61 (m, 2H, α′-CH2), 3.97–4.01 (m, 2H, β′-CH2), 5.85–5.88 (∼t, 1H, NH), 77.12–7.14 (d, 2H, J ≈ 7.12 Hz, 3,5-Har), 7.28–7.30 (d, 2H, J ≈ 7.12 Hz, 2,6-Har), 7.65 (s, 1H, 5-Ht); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.05 (3,5,7-C), 30.98 (β-C), 36.09 (1-C), 36.87 (4,6,10-C), 37.96 (α-C) 39.16 (α′-C), 43.32 (2,8,9-C), 43.36 (β′-C), 125.17 (3,5-Car), 128.20 (2,6-Car), 137.39 (1-Car), 145.74 (5-Ct), 149.96 (4-Car), 163.15 (3-Ct), 173.60 (C
O). Anal. calcd for C23H29N5O3: C, 65.23; H, 6.90; N, 16.54; found: C, 65.51; H, 7.02; N, 16.75.
The product is a mixture of E and Z conformers. E > Z.
1H-NMR (CDCl3), δ (ppm): (conformer E) (400 MHz, CDCl3), δ (ppm): 1.73–1.82 (q, 6H, J ≈ 10.98 Hz, 4,6,10-H), 1.91 (br.s, 6H, 2,8,9-H), 2.11 (br.s, 3H, 3,5,7-H), 2.22–2.29 (quintet, 2H, J ≈ 6.1 Hz, β′-CH2), 3.50–3.54 (q, 2H, J ≈ 6 Hz, α′-CH2), 4.37–4.40 (t, 2H, J ≈ 6.3 Hz, γ′-CH2), 6.51 (br.s, 1H, NH), 7.42–7.44 (d, 2H, J ≈ 8.3 Hz, 3,5-Har), 7.70–7.72 (d, 2H, J ≈ 8.3 Hz, 2,6-Har), 8.47 (s, 1H, 5-Ht); 13C-NMR (100 MHz, CDCl3), δ (ppm): 28.94 (3,5,7-C), 30.62 (β′-C), 31.72 (1-C), 36.69 (α′-C), 36.79 (4,6,10-C), 43.09 (2,8,9-C), 49.15 (γ′-C), 125.42 (3,5-Car), 126.97 (2,6-Car), 130.93 (1-Car), 145.98 (5-Ct), 155.88 (4-Car), 163.04 (3-Ct), 168.40 (C
O).
1H-NMR (CDCl3), δ (ppm): (conformer Z) (400 MHz, CDCl3), δ (ppm): 1.73–1.82 (q, 6H, J ≈ 10.98 Hz, 4,6,10-H), 1.91 (br.s, 6H, 2,8,9-H), 2.11 (br.s, 3H, 3,5,7-H), 2.22–2.29 (quintet, 2H, J ≈ 6.1 Hz, β′-CH2), 3.62–3.68 (m, 2H, α′-CH2), 4.0.12–4.16 (m, 2H, γ′-CH2), 6.51 (br.s, 1H, NH), 7.52–7.54 (d, 2H, J ≈ 8.1 Hz, 3,5-Har), 7.85–7.88 (d, 2H, J ≈ 8.1 Hz, 2,6-Har), 8.50 (s, 1H, 5-Ht); 13C-NMR (100 MHz, CDCl3), δ (ppm): 28.94 (3,5,7-C), 30.62 (β′-C), 31.72 (1-C), 36.69 (α′-C), 36.79 (4,6,10-C), 43.09 (2,8,9-C), 49.15 (γ′-C), 125.42 (3,5-Car), 126.97 (2,6-Car), 130.93 (1-Car), 145.98 (5-Ct), 155.88 (4-Car), 163.04 (3-Ct), 168.40 (C
O). Anal. calcd for C22H27N5O3: C, 64.53; H, 6.65; N, 17.10; found: C, 64.81; H, 6.31; N, 17.13.
The product is a mixture of E and Z conformers. E/Z: 4/1.
1H-NMR (CDCl3), δ (ppm): (conformer E) (600 MHz, CDCl3), δ (ppm): 1.73–1.80 (q, 6H, J ≈ 20.9 Hz, 4,6,10-H), 1.88 (br.s, 6H, 2,8,9-H), 2.08 (br.m, 5H, 3,5,7-H, β′-CH2), 3.23–3.26 (q, 2H, J ≈ 6.4 Hz, α′-CH2), 3.53 (s, 2H, J ≈ 21 Hz, α-CH2), 4.24–4.26 (t, 2H, J ≈ 6.9 Hz, γ′-CH2), 5.79 (br.s, 1H, NH), 7.16–7.18 (d, 2H, J ≈ 8.1 Hz, 3,5-Har), 7.33–7.34 (d, 2H, J ≈ 8.1 Hz, 2,6-Har), 8.35 (s, 1H, 5-Ht); 13C-NMR (100 MHz, CDCl3), δ (ppm): 28.99 (3,5,7-C), 30.23 (β′-C), 31.68 (1-C), 36.18 (α′-C), 36.82 (4,6,10-C), 43.26 (2,8,9-C), 43.36 (α-C), 48.94 (γ′-C), 125.85 (3,5-Car), 129.20 (2,6-Car), 131.50 (1-Car), 145.89 (5-Ct), 150.99 (4-Car), 162.94 (3-Ct), 172.51 (C
O).
1H-NMR (CDCl3), δ (ppm): (conformer Z) (600 MHz, CDCl3), δ (ppm): 1.73–1.80 (q, 6H, J ≈ 20.9 Hz, 4,6,10-H), 1.88 (br.s, 6H, 2,8,9-H), 2.08 (br.m, 5H, 3,5,7-H, β′-CH2), 3.23–3.26 (q, 2H, J ≈ 6.4 Hz, α′-CH2), 3.57 (s, 2H, J ≈ 21 Hz, α-CH2), 4.24–4.26 (t, 2H, J ≈ 6.9 Hz, γ′-CH2), 5.79 (br.s, 1H, NH), 7.23 (br.s, 2H, 3,5-Har), 7.29–7.30 (m, 2H, 2,6-Har), 8.33 (s, 1H, 5-Ht); 13C-NMR (100 MHz, CDCl3), δ (ppm): 28.99 (3,5,7-C), 30.23 (β′-C), 31.68 (1-C), 36.18 (α′-C), 36.82 (4,6,10-C), 43.26 (2,8,9-C), 44.19 (α-C), 48.94 (γ′-C), 124.34 (3,5-Car), 126.52 (2,6-Car), 134.31 (1-Car), 145.89 (5-Ct), 152.66 (4-Car), 162.94 (3-Ct), 172.51 (C
O). Anal. calcd for C23H29N5O3: C, 65.23; H, 6.90; N, 16.54; found: C, 65.47; H, 7.09; N, 16.71.
The product is a mixture of E and Z conformers. E/Z: 13/1.
1H-NMR (400 MHz, CDCl3), δ (ppm): (conformer E) 1.69–1.76 (q, 6H, J ≈ 13.9 Hz 4,6,10-H), 1.84 (br.s, 6H, 2,8,9-H), 1.97–2.00 (quintet, 2H, J ≈ 6.1 Hz, β′-CH2), 2.06 (br.s, 3H, 3,5,7-H), 2.50–2.54 (t, 2H, J ≈ 7.6 Hz, α-CH2), 2.93–2.97 (t, 2H, J ≈ 7.6 Hz, β-CH2), 3.18–3.23 (q, 2H, J ≈ 6.1 Hz, α′-CH2), 4.02–4.05 (t, 2H, J ≈ 6.3 Hz, γ′-CH2), 5.76–5.79 (∼t, 1H, NH), 7.15–7.17 (d, 2H, J ≈ 7.12 Hz, 3,5-Har), 7.26–7.28 (d, 2H, J ≈ 7.12 Hz, 2,6-Har), 8.33 (s, 1H, 5-Ht); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.00 (3,5,7-C), 30.31 (β′-C), 31.10 (β-C), 35.68 (α′-C), 36.04 (1-C), 36.83 (4,6,10-C), 38.23 (α-C), 43.35 (2,8,9-C), 48.51 (γ′-C), 125.19 (3,5-Car), 128.24 (2,6-Car), 137.50 (1-Car), 146.03 (5-Ct), 149.85 (4-Car), 162.96 (3-Ct), 173.22 (C
O).
1H-NMR (400 MHz, CDCl3), δ (ppm): (conformer Z) 1.69–1.76 (q, 6H, J ≈ 13.9 Hz 4,6,10-H), 1.86 (br.s, 6H, 2,8,9-H), 2.03 (br.s, 2H, β′-CH2), 2.06 (br.s, 3H, 3,5,7-H), 2.43–2.47 (m, 2H, J ≈ 7.6 Hz, α-CH2), 2.93–2.97 (t, 2H, J ≈ 7.6 Hz, β-CH2), 3.79–3.84 (m, 2H, α′-CH2), 4.10–4.12 (m, 2H, γ′-CH2), 5.76–5.79 (∼t, 1H, NH), 7.15–7.17 (d, 2H, J ≈ 7.12 Hz, 3,5-Har), 7.26–7.28 (d, 2H, J ≈ 7.12 Hz, 2,6-Har), 8.45 (s, 1H, 5-Ht); 13C-NMR (100 MHz, CDCl3), δ (ppm): 29.04 (3,5,7-C), 30.31 (β′-C), 31.10 (β-C), 35.68 (α′-C), 36.04 (1-C), 36.88 (4,6,10-C), 38.23 (α-C), 43.35 (2,8,9-C), 48.51 (γ′-C), 125.05 (3,5-Car), 128.19 (2,6-Car), 137.50 (1-Car), 146.03 (5-Ct), 149.85 (4-Car), 162.96 (3-Ct), 172.25 (C
O). Anal. calcd for C24H31N5O3: C, 65.88; H, 7.14; N, 16.01; found: C, 65.69; H, 7.18; N, 16.17.
The 1H-NMR and 13C-NMR spectra of 6a–f in CDCl3 revealed the presence of mixtures of E- and Z-isomers due to the restricted rotation around the amide bond. The conformational studies are provided in the supporting information.
O); anal. calcd for C20H23N5O3: C, 62.98; H, 6.08; N, 18.36; found: C, 62.76; H, 6.22; N, 18.31.Footnote |
| † HeLa and L6 cells were obtained from the London School of Hygiene and Tropical Medicine (LSHTM) cell line repository. |
| This journal is © The Royal Society of Chemistry 2026 |