Noha Ryada,
Ayman Abo Elmaatyb,
Samy Selim*c,
Mohammed S. Almuhayawid,
Soad K. Al Jaounie,
Mohamed S. Abdel-Azizf,
Arwa Sultan Alqahtanig,
Islam Zaki*hj and
Lina M. A. Abdel Ghanyi
aPharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, P.O. Box 77, Giza, Egypt
bMedicinal Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt
cDepartment of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia. E-mail: sabdulsalam@ju.edu.sa
dDepartment of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
eDepartment of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
fMicrobial Chemistry Department, Biotechnology Research Institute, National Research Centre, Cairo, Egypt
gDepartment of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box, 90950, Riyadh 11623, Saudi Arabia
hPharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt. E-mail: eslam.zaki@pharm.psu.edu.eg
iPharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, P.O. Box 77, Giza, Egypt
jPharmaceutical Organic Chemistry Department, Clinical Pharmacy Program, East Port Said National University, Port Said 42526, Egypt
First published on 25th October 2024
Cancer is the second leading cause of death globally, surpassed only by heart disease. Moreover, bacterial infections remain a significant global health burden, contributing substantially to morbidity and mortality, especially among hospitalized patients. EGFR has emerged as a prime therapeutic target due to its pivotal role in driving uncontrolled cell growth and survival across numerous cancer types. In addition, DNA gyrase represents a promising target for the development of novel antimicrobial agents. Therefore, we aimed to design and synthesize new multi-target quinoline hybrids (7–17e) capable of acting as anti-proliferative and antimicrobial agents by inhibiting EGFR and microbial DNA gyrase, respectively. The inhibitory potential of the synthesized compounds was determined using in vitro and in silico approaches. The antiproliferative activity of the synthesized quinoline-oxadiazole derivatives 7–17e was assessed against two cancer cell lines, namely, hepatocellular carcinoma (HepG2) and breast adenocarcinoma (MCF-7). The assessed compounds 7–17e showed considerable cytotoxic activity activities against HepG2 and MCF-7 with IC50 values of 0.137–0.332 and 0.164–0.583 μg mL−1, respectively, in comparison to erlotinib as the positive control, which showed an IC50 value of 0.308 and 0.512 μg mL−1, respectively. Moreover, an EGFR tyrosine kinase inhibition assay was conducted on the most prominent candidates. The results showed good IC50 values of 0.14 and 0.18 μM for compounds 8c and 12d, respectively, compared to lapatinib (IC50 value of 0.12 μM). Furthermore, the minimum antimicrobial inhibitory concentration was evaluated for the most prominent candidates with S. aureus, E. coli, and C. albicans. Compounds 17b, 17d and 17e displayed the most potent inhibitory activity, exhibiting 4-, 16- and 8-fold more activity, respectively, than the reference neomycin. Hence, we can conclude that the afforded compounds can be used as lead dual anticancer and antimicrobial candidates for future optimization.
Protein kinases (PKs) are essential enzymes that regulate a wide range of critical cellular processes, including metabolism, cell growth, survival, and death. Their pivotal role in cellular signaling pathways has made them prime targets for therapeutic interventions, particularly in cancer research.4 Protein kinases catalyze the transfer of a phosphate group from ATP to specific hydroxyl residues of amino acids, such as serine, threonine, or tyrosine, on target proteins, a process known as phosphorylation. This crucial phosphorylation process regulates a wide range of cellular functions through intricate signaling networks.5 Consequently, aberrant kinase activity, resulting from either hyper activation or mutations, disrupts critical cellular signaling pathways, contributing to the pathogenesis of various diseases, including cancer.6
The epidermal growth factor receptor (EGFR) is a prominent protein kinase that plays a pivotal role in regulating cell proliferation and migration.7 Many solid tumors, such as non-small cell lung cancer,8 hepatocellular carcinoma,9 and breast cancer,4 overexpress EGFR. Recent advancements in cancer therapy have focused on targeting specific molecules that regulate cancer cell growth and survival.10,11 Consequently, EGFR has emerged as a prime therapeutic target due to its pivotal role in driving uncontrolled cell growth and survival across numerous cancer types.12–17
Erlotinib and gefitinib are examples of first-generation EGFR tyrosine kinase inhibitors.18–21 However, their efficacy can be compromised by the development of resistance mechanisms, such as the EGFR-T790M mutation, which diminishes their anticancer potency.22 To address the emergence of resistance associated with first-generation EGFR tyrosine kinase inhibitors, second-generation EGFR tyrosine kinase inhibitors (e.g., pelitinib and neratinib) were developed.23–28 These drugs have equal affinities towards the wild-type EGFR (WT) and mutant EGFR (EGFR-T790M), resulting in rash and diarrhea.29 Hence, the maximal tolerated dose (MTD) displayed by these drugs30,31 has led to the emergence of third-generation irreversible EGFR-tyrosine kinase inhibitors (e.g., osimertinib and olmutinib),32–35 as shown in Fig. 1. Recently, fourth-generation EGFR tyrosine kinase inhibitors (TKIs) have emerged as a novel therapeutic strategy to address the challenge of acquired resistance mediated by the EGFR C797S mutation, and were subjected to further clinical evaluations.36 Fourth-generation EGFR TKIs offer a novel approach to overcoming resistance to EGFR inhibitors by targeting a distinct binding site on the receptor (allosteric inhibitors). This allosteric mechanism of action differentiates them from previous generations of ATP-competitive inhibitors.36 The continuous emergence of resistance mechanisms underscores the urgent need for innovative strategies to develop novel EGFR inhibitors with enhanced efficacy and safety profiles.
Fig. 1 Some FDA-approved quinoline antibiotics and EGFR TK inhibitors, with their different generations as well as their drawbacks. |
However, bacterial infections remain a significant global health burden, contributing substantially to morbidity and mortality, especially among hospitalized patients.37 Despite the availability of numerous antimicrobial agents, their effectiveness is often compromised by the emergence of bacterial resistance, limiting their clinical utility.38 Antibiotic resistance is a pressing global health crisis, contributing to an estimated 700000 deaths annually.39 Without significant advancements in antimicrobial strategies, drug-resistant infections are projected to claim an estimated 10 million lives annually by 2050.39 Hence, to address the growing problem of bacterial infections, scientists are desperately searching for new antibiotics that can effectively fight both common and antibiotic-resistant bacteria. These new agents have attracted significant interest in medicinal chemistry research and offer a promising solution to the urgent need for more effective treatments.37 Bacterial DNA gyrase, an essential type II topoisomerase, plays a critical role in DNA replication and transcription by introducing negative supercoils into DNA.40,41 Given its pivotal role in bacterial survival, DNA gyrase represents a promising target for the development of novel antimicrobial agents. Quinolines (e.g., ciprofloxacin, moxifloxacin, and ofloxacin) have been identified as potent inhibitors of DNA gyrase (Fig. 1). By targeting this essential enzyme, quinolines can effectively disrupt bacterial DNA replication and recombination, leading to cell death.42
Furthermore, quinoline and oxadiazole, privileged scaffolds in medicinal chemistry,43 have been extensively investigated for its diverse biological properties in numerous research endeavors exhibiting a wide range of pharmacological activities, making them a versatile scaffold for drug discovery, including anticancer,44–49 anti-viral,50–52 anti-microbial,53–57 anti-diabetic,58–60 and anti-inflammatory activities.61–63 In particular, the literature revealed that quinoline oxadiazole hybrids were utilized as antimicrobial and/or anti-proliferative agents.42,64,65
Fig. 2 The design rationale of the synthesized compounds (7a–17e) as antiproliferative and antimicrobial agents. |
Compd no. | IC50 (μg mL−1) | |
---|---|---|
HepG-2 | MCF-7 | |
7 | 0.151 | 0.295 |
8a | 0.215 | 0.440 |
8b | 0.139 | 0.225 |
8c | 0.137 | 0.481 |
8d | 0.311 | 0.287 |
8e | 0.157 | 0.179 |
9 | 0.152 | 0.358 |
10 | 0.217 | 0.446 |
11 | 0.141 | 0.227 |
12a | 0.138 | 0.473 |
12b | 0.327 | 0.287 |
12c | 0.158 | 0.239 |
12d | 0.138 | 0.473 |
13 | 0.332 | 0.287 |
14 | 0.159 | 0.217 |
15a | 0.272 | 0.164 |
15b | 0.154 | 0.411 |
17a | 0.141 | 0.300 |
17b | 0.188 | 0.583 |
17c | 0.164 | 0.221 |
17d | 0.254 | 0.406 |
17e | 0.139 | 0.569 |
Erlotinib | 0.308 | 0.512 |
Comp. no. | Gram+ bacteria | Gram− bacteria | Fungi | |
---|---|---|---|---|
S. aureus | E. coli | C. albicans | A. niger | |
7 | 12 | 14 | 13 | 0 |
8a | 0 | 0 | 0 | 0 |
8b | 0 | 0 | 0 | 0 |
8c | 0 | 0 | 0 | 0 |
8d | 0 | 0 | 0 | 0 |
8e | 0 | 0 | 0 | 0 |
9 | 20 | 23 | 24 | 0 |
10 | 25 | 30 | 28 | 0 |
11 | 14 | 17 | 14 | 0 |
12a | 0 | 0 | 0 | 0 |
12b | 0 | 0 | 0 | 0 |
12c | 0 | 0 | 0 | 0 |
12d | 0 | 0 | 0 | 0 |
13 | 0 | 0 | 0 | 0 |
14 | 0 | 0 | 0 | 0 |
15a | 0 | 0 | 0 | 0 |
15b | 0 | 0 | 0 | 0 |
16 | 0 | 0 | 0 | 0 |
17a | 37 | 35 | 37 | 0 |
17b | 33 | 34 | 36 | 0 |
17c | 35 | 36 | 35 | 0 |
17d | 34 | 37 | 36 | 0 |
17e | 29 | 32 | 30 | 0 |
Neomycin | 26 | 24 | 30 | — |
Cycloheximide | 0 | 21 |
Comp. no. | MIC (μg mL−1) | ||
---|---|---|---|
S. aureus | E. coli | C. albicans | |
9 | 39.06 | 625 | 19.53 |
10 | 39.06 | 312.5 | 19.53 |
11 | 78.125 | 625 | 39.06 |
17a | 39.06 | 156.25 | 4.88 |
17b | 19.53 | 312.5 | 9.77 |
17c | 78.125 | 312.5 | 4.88 |
17d | 4.88 | 312.5 | 9.77 |
17e | 9.77 | 312.5 | 4.88 |
Neomycin | 78.125 | 39.06 | 156.25 |
Comp. no. | MBC (μg mL−1) | ||
---|---|---|---|
S. aureus | E. coli | C. albicans | |
9 | 78.125 | 1250 | 39.06 |
10 | 156.25 | 625 | 19.53 |
11 | 312.5 | 625 | 78.125 |
17a | 39.06 | 625 | 4.88 |
17b | 39.06 | 625 | 9.77 |
17c | 312.5 | 312.5 | 9.77 |
17d | 19.53 | 1250 | 39.06 |
17e | 19.53 | 1250 | 9.77 |
Neomycin | 312.5 | 156.25 | 625 |
Comp. no. | MIC of biofilm inhibition (μg mL−1) | ||
---|---|---|---|
S. aureus | E. coli | C. albicans | |
9 | 39.06 | 19.53 | 39.06 |
10 | 78.125 | 9.77 | 19.53 |
11 | 9.77 | 9.77 | 19.53 |
17a | 1250 | 1250 | 1250 |
17b | 1250 | 1250 | 1250 |
17c | 19.53 | 9.77 | 9.77 |
17d | 625 | 1250 | 1250 |
17e | 156.53 | 156.53 | 78.125 |
Neomycin | 312.5 | 156.25 | 625 |
Accordingly, regarding their physicochemical features, except for compounds (8c, 10, 11, 12d, 14, 15a, 15b, and 17e), all synthesized compounds displayed high GIT absorption due to their feasible lipophilicity. Therefore, eligible oral bio-availabilities can be anticipated.73,74 Moreover, except for compound (7), all afforded compounds cannot pass through the blood–brain barrier. Thus, fewer CNS side effects can be assumed. Notably, compounds 7, 8c, 9, 10, 12a, 12c, 13, 14, 15a, 15b, 16, and 17e are not P-glycoprotein (Pgp-) substrates (Tables 6, 7, and 8). Hence, better bioavailability could be assured, as shown in Fig. 7. Moreover, compound 8c did not show inhibition for the common hepatic metabolizing enzymes (CYP1A2, CYP2C19, CYP2C9, CYP2D6, and CYP3A4). Except for compounds 12d, 15a, and 17e, all of the synthesized quinoline derivatives match the Lipinski's rule of five,75 assuring their oral bioavailability. To further evaluate the compounds' bioavailability, we utilized the bioavailability radar tool provided by SwissADME. This visual representation offers a comprehensive assessment of drug-like properties within a hexagonal framework. Compounds falling within the optimal physicochemical space defined by the radar are considered to have favorable oral bioavailability. These radar plots are shown in the supplementary Fig. SI5.†
Comp. 7 | Comp. 8a | Comp. 8b | Comp. 8c | Comp. 8d | Comp. 8e | Comp. 9 | Comp. 10 | ||
---|---|---|---|---|---|---|---|---|---|
Molecular properties | Molar refractivity | 88.17 | 113.61 | 115.63 | 122.43 | 118.01 | 118.01 | 95.42 | 124.80 |
TPSA (Az) | 51.81 | 51.81 | 72.04 | 97.63 | 77.83 | 77.83 | 90.61 | 94.18 | |
logP o/w (WLOGP) | 4.71 | 6.38 | 6.09 | 6.29 | 5.97 | 5.97 | 5.00 | 6.65 | |
Consensus logP o/w | 3.96 | 5.39 | 5.01 | 4.61 | 4.84 | 4.84 | 4.29 | 5.47 | |
Water solubility | MS | PS | PS | PS | PS | PS | MS | PS | |
Pharmacokinetics parameters | GI absorption | High | High | High | Low | High | High | High | Low |
BBB permeant | Yes | No | No | No | No | No | No | No | |
P-gp substrate | No | Yes | Yes | No | Yes | Yes | No | No | |
CYP1A2 inhibitor | Yes | No | No | No | No | No | Yes | No | |
CYP2C19 inhibitor | Yes | Yes | No | No | Yes | Yes | Yes | Yes | |
CYP2C9 inhibitor | No | No | No | No | No | No | Yes | Yes | |
CYP2D6 inhibitor | Yes | No | No | No | Yes | Yes | No | No | |
CYP3A4 inhibitor | No | No | No | No | No | No | Yes | No | |
Drug/lead likeness | Drug likeness (lipinski) | Yes | Yes | Yes | Yes | Yes | Yes | Yes | Yes |
Lead likeness | No | No | No | No | No | No | No | No | |
Toxicity parameters | Ames toxicity | Yes | No | No | No | No | No | Yes | No |
Max. tolerated dose (log mg kg−1 per day) | 0.362 | 0.649 | 0.626 | 0.596 | 0.624 | 0.633 | 0.4 | 0.671 | |
hERG I inhibitor | No | No | No | No | No | No | No | No | |
hERG II inhibitor | No | Yes | Yes | Yes | Yes | Yes | Yes | Yes | |
Oral rat acute toxicity (LD50) (mol kg−1) | 2.229 | 2.347 | 2.77 | 2.677 | 2.993 | 3.074 | 2.512 | 2.786 | |
Oral rat chronic toxicity (LOAEL) (log mg kg−1 bw per day) | 1.029 | 0.486 | 0.615 | 0.413 | 0.49 | 0.466 | 0.869 | 0.518 | |
Hepatotoxicity | Yes | Yes | Yes | Yes | Yes | Yes | No | Yes | |
Minnow toxicity (log mM) | 1.587 | −0.692 | −0.514 | −2.631 | −0.749 | −0.467 | 1.396 | −1.59 |
Comp. 11 | Comp. 12a | Comp. 12b | Comp. 12c | Comp. 12d | Comp. 13 | Comp. 14 | Comp. 15a | ||
---|---|---|---|---|---|---|---|---|---|
Molecular properties | Molar refractivity | 124.38 | 99.89 | 104.70 | 109.03 | 134.31 | 106.47 | 143.92 | 133.73 |
TPSA (Az) | 77.11 | 77.11 | 77.11 | 77.11 | 77.11 | 114.41 | 123.28 | 106.21 | |
LogP o/w (WLOGP) | 6.85 | 5.44 | 5.83 | 5.99 | 7.41 | 4.89 | 6.46 | 6.25 | |
Consensus logP o/w | 5.81 | 4.63 | 4.95 | 5.17 | 6.34 | 4.03 | 5.12 | 5.17 | |
Water solubility | PS | MS | PS | PS | PS | MS | PS | PS | |
Pharmacokinetics parameters | GI absorption | Low | High | High | High | Low | High | Low | Low |
BBB permeant | No | No | No | No | No | No | No | No | |
P-gp substrate | Yes | No | Yes | No | Yes | No | No | No | |
CYP1A2 inhibitor | No | Yes | Yes | Yes | No | Yes | No | No | |
CYP2C19 inhibitor | Yes | Yes | Yes | Yes | Yes | Yes | Yes | Yes | |
CYP2C9 inhibitor | No | Yes | Yes | Yes | No | Yes | Yes | Yes | |
CYP2D6 inhibitor | No | No | No | No | No | No | No | No | |
CYP3A4 inhibitor | Yes | Yes | Yes | No | Yes | No | Yes | Yes | |
Drug/lead likeness | Drug likeness (lipinski) | Yes | Yes | Yes | Yes | No | Yes | Yes | No |
Lead likeness | No | No | No | No | No | No | No | No | |
Toxicity parameters | Ames toxicity | No | Yes | Yes | Yes | No | No | No | No |
Max. tolerated dose (log mg kg−1 per day) | 0.692 | 0.346 | 0.493 | 0.574 | 0.671 | 1.035 | 0.736 | 0.764 | |
hERG I inhibitor | No | No | No | No | No | No | No | No | |
hERG II inhibitor | Yes | Yes | Yes | Yes | Yes | No | Yes | Yes | |
Oral rat acute toxicity (LD50) (mol kg−1) | 2.547 | 2.317 | 2.264 | 2.213 | 2.531 | 2.604 | 3.091 | 3.009 | |
Oral rat chronic toxicity (LOAEL) (log mg kg−1 bw per day) | 0.433 | 0.848 | 0.802 | 0.734 | 0.44 | 0.792 | 0.371 | 0.413 | |
Hepatotoxicity | Yes | No | No | No | Yes | Yes | Yes | Yes | |
Minnow toxicity (log mM) | −2.429 | 0.929 | 0.54 | 0.091 | −4.27 | 1.452 | −1.724 | −2.091 |
Comp. 15b | Comp. 16 | Comp. 17a | Comp. 17b | Comp. 17c | Comp. 17d | Comp. 17e | Erlotinib | ||
---|---|---|---|---|---|---|---|---|---|
Molecular properties | Molar refractivity | 140.22 | 115.60 | 125.47 | 129.19 | 131.10 | 136.00 | 154.43 | 111.40 |
TPSA (Az) | 115.44 | 103.41 | 88.41 | 79.18 | 91.21 | 82.42 | 79.18 | 74.73 | |
LogP o/w (WLOGP) | 6.26 | 5.37 | 4.61 | 5.76 | 3.80 | 4.14 | 8.49 | 3.48 | |
Consensus logP o/w | 5.10 | 4.77 | 4.32 | 5.17 | 4.01 | 4.26 | 6.78 | 3.28 | |
Water solubility | PS | PS | MS | PS | MS | PS | PS | MS | |
Pharmacokinetics parameters | GI absorption | Low | High | High | High | High | High | Low | High |
BBB permeant | No | No | No | No | No | No | No | Yes | |
P-gp substrate | No | No | Yes | Yes | Yes | Yes | No | No | |
CYP1A2 inhibitor | No | Yes | Yes | Yes | Yes | Yes | No | Yes | |
CYP2C19 inhibitor | Yes | Yes | No | Yes | Yes | No | Yes | Yes | |
CYP2C9 inhibitor | Yes | Yes | Yes | Yes | Yes | Yes | No | Yes | |
CYP2D6 inhibitor | Yes | No | No | No | Yes | No | No | Yes | |
CYP3A4 inhibitor | Yes | Yes | Yes | No | Yes | Yes | No | Yes | |
Drug/lead likeness | Drug likeness (lipinski) | Yes | Yes | Yes | Yes | Yes | Yes | No | Yes |
Lead likeness | No | No | No | No | No | No | No | No | |
Toxicity parameters | Ames toxicity | No | Yes | Yes | No | Yes | Yes | No | No |
Max. tolerated dose (log mg kg−1 per day) | 0.761 | 0.537 | 0.276 | 0.31 | 0.415 | 0.434 | 0.494 | 0.355 | |
hERG I inhibitor | No | No | No | No | No | No | No | No | |
hERG II inhibitor | Yes | Yes | Yes | Yes | Yes | Yes | Yes | Yes | |
Oral rat acute toxicity (LD50) (mol kg−1) | 3.049 | 2.406 | 2.767 | 2.725 | 2.752 | 2.755 | 3.034 | 3.058 | |
Oral rat chronic toxicity (LOAEL) (log mg kg−1 bw per day) | 0.345 | 0.609 | 0.574 | 0.557 | 2.595 | 2.477 | −0.569 | 1.558 | |
Hepatotoxicity | Yes | Yes | Yes | No | No | Yes | Yes | Yes | |
Minnow toxicity (log mM) | −1.971 | 1.28 | 1.564 | 1.102 | 1.763 | 1.651 | −3.411 | −1.971 |
Fig. 7 The boiled-egg representation for the synthesized quinoline derivatives (7–17e), as well as erlotinib as a reference control. |
By employing the pkCSM descriptors algorithm protocol, it was revealed that compounds 7, 9, 12a, 12b, 12c, 16, 17a, 17c, and 17d could manifest Ames toxicity. Thus, a possible mutagenicity could be predicted.76 Additionally, all the synthesized quinoline derivatives are non-inhibitors of hERG I. Therefore, a cardiotoxic effect on the human heart's electrical activity cannot be assumed for these compounds.77 However, except for compounds 7 and 13, all synthesized compounds (including erlotinib) exhibit hERG II inhibitory activity, which raises concerns about possible cardiac arrhythmias,78 Notably, except for compounds 9, 12a, 12b, 12c, 17b, and 17c, all of the synthesized compounds are hepatotoxic.
Compound 6 (10 mmol, 3.98 g) was heated at 10 °C above its melting point for 30 min in an oil bath. After cooling the reaction, quinoline-oxadiazole molecule 7 was attained by crystallization from ethanol (70%).
Buff crystals, yield 61%, m.p. 140–142 °C. IR (KBr, cm−1): 3138 (CH aromatic), 2993 (CH aliphatic), 1537 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 7.60–7.66 (m, 3H, Ar-H), 7.76 (t, 1H, Ar-H), 7.99 (d, 1H, J = 8.4 Hz, Ar-H), 8.04 (d, 2H, J = 8.4 Hz, Ar-H), 8.27 (s, 1H, Ar-H), 8.88 (d, 1H, J = 8.4 Hz, Ar-H), 9.57 (s, 1H, oxadiazole-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 121.17, 124.41, 124.78, 125.42, 127.35, 128.37, 129.33(2C), 130.00, 130.65, 132.27(2C), 137.52, 147.26, 147.93, 154.76, 167.19. MS m/z (%): 354.03 (M + 2, 17.31), 353.47 (M + 1, 12.36), 352.46 (M+, 18.72), 302.58 (100). Anal. calcd for C17H10BrN3O (352.19): C, 57.98; H, 2.86; N 11.93; found: C, 58.14; H, 3.02; N, 11.85.
A mixture of acid hydrazide 3 (10 mmol, 3.42 g), carboxylic acid derivatives (10 mmol) and phosphorous oxychloride (5 mL) was heated at 60 °C for 6–8 h, and then allowed to cool at room temperature. After the reaction mixture was added to ice-cold water, a saturated sodium bicarbonate solution was used to neutralize it. The obtained precipitate was crystallized from ethanol (70%) to give the corresponding oxadiazole product 8a–e.
Buff powder, yield 69%, m.p. 90–92 °C. IR (KBr, cm−1): 3070 (CH aromatic), 2954 (CH aliphatic), 1531 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 7.52 (t, 2H, Ar-H), 7.59–7.63 (m, 1H, Ar-H), 7.76–7.90 (m, 3H, Ar-H), 7.92–7.96 (m, 3H, Ar-H), 8.15–8.28 (m, 3H, Ar-H), 8.47 (s, 1H, Ar-H), 8.80 (d, 1H, J = 8 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 117.81, 123.52, 124.58, 125.56, 126.30, 127.29(2C), 128.34(2C), 129.33(2C), 130.65, 131.78, 132.04(2C), 133.39, 137.45, 140.85, 143.92, 147.99, 154.77, 165.31, 166.00. MS m/z (%): 430.40 (M + 2, 31.26), 428.20 (M+, 30.16), 292.89 (100). Anal. calcd for C23H14BrN3O (428.29): C, 64.50; H, 3.29; N 9.81; found: C, 64.32; H, 3.50; N, 10.04.
White powder, yield 63%, m.p. 190–192 °C. IR (KBr, cm−1): 3099 (CH aromatic), 2993 (CH aliphatic), 1543 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 5.75 (s, 1H, OH, D2O exchangeable), 7.47 (d, 2H, J = 8.4 Hz, Ar-H), 7.75–7.95 (m, 4H, Ar-H), 8.23–8.27 (m, 2H, Ar-H), 8.33 (d, 1H, J = 8.8 Hz, Ar-H), 8.39 (d, 2H, J = 7.2 Hz, Ar-H) 8.82 (s, 1H, Ar-H), 9.20 (d, 1H, J = 8.8 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 116.74(2C), 119.15(2C), 122.16, 123.27, 123.89, 124.86, 126.30, 128.33, 129.03(2C), 129.99, 131.05, 132.01(2C), 132.71, 140.52, 143.25, 147.98, 154.77, 163.65, 164.64. MS m/z (%): 446.02 (M + 2, 17.91), 443.82 (M+, 19.02), 325.62 (100). Anal. calcd for C23H14BrN3O2 (444.29): C, 62.18; H, 3.18; N 9.46; found: C, 62.40; H, 3.29; N, 9.58.
White powder, yield 66%, m.p. 288–290 °C. IR (KBr, cm−1): 3086 (CH aromatic), 2947 (CH aliphatic), 1546 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 7.83–7.89 (m, 3H, Ar-H), 7.97 (t, 1H, Ar-H), 8.26 (d, 1H, J = 8.8 Hz, Ar-H), 8.40 (d, 2H, J = 8.8 Hz, Ar-H), 8.50 (d, 2H, J = 8.4 Hz, Ar-H), 8.60 (d, 2H, J = 8.4 Hz, Ar-H), 8.88 (s, 1H, Ar-H), 9.20 (d, 1H, J = 8 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 116.00, 121.86, 123.90, 124.00, 125.29, 126.65(2C), 127.29(2C), 128.33(2C), 128.39, 132.03, 133.39(2C), 133.77, 139.19, 141.33, 142.18, 147.48, 155.44, 166.00, 166.44. MS m/z (%): 475.22 (M + 2, 59.19), 473.60 (M+, 60.06), 44.39 (100). Anal. calcd for C23H13BrN4O3 (473.29): C, 58.37; H, 2.77; N 11.84; found: C, 58.51; H, 2.94; N, 12.07.
Buff powder, yield 62%, m.p. 220–222 °C. IR (KBr, cm−1): 3095 (CH aromatic), 2980 (CH aliphatic), 1573 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 6.74 (d, 2H, J = 8.0 Hz, Ar-H), 7.82 (m, 3H, Ar-H), 7.94–8.28 (m, 5H, Ar-H + NH2 D2O exchangeable), 8.35–8.39 (m, 3H, Ar-H), 8.78 (s, 1H, Ar-H), 9.20 (d, 1H, J = 8.0 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 114.71(2C), 117.47, 122.17, 124.19, 125.93, 126.60, 127.66, 128.34, 128.62(2C), 129.03(2C), 129.68, 130.68, 132.04(2C), 142.20, 147.31, 149.06, 154.76, 164.33, 165.67. MS m/z (%): 445.95 (M + 2, 22.61), 443.72 (M+, 22.39), 400.91 (100). Anal. calcd for C23H15BrN4O (443.30): C, 62.32; H, 3.41; N 12.64; found: C, 62.56; H, 3.64; N, 12.85.
Brown powder, yield 64%, m.p. 177–179 °C, IR (KBr, cm−1): 3091 (CH aromatic), 2924 (CH aliphatic), 1537 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 6.83 (t, 1H, Ar-H), 6.95 (t, 1H, Ar-H), 7.07 (d, 1H, J = 8.0 Hz, Ar-H), 7.35–7.36 (m, 1H, Ar-H), 7.62–7.94 (m, 4H, Ar-H), 8.08 (d, 1H, J = 8.8 Hz, Ar-H), 8.22 (s, 2H, NH2 D2O exchangeable), 8.37 (d, 2H, J = 8.0 Hz, Ar-H), 8.78 (s, 1H, Ar-H), 9.20 (d, 1H, J = 8.0 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 117.12, 118.77, 119.84, 121.48, 122.84, 123.21, 123.88, 125.57, 126.98, 127.96, 129.69(2C), 129.99, 130.67, 132.02(2C), 133.08, 134.73, 141.21, 147.99, 154.76, 165.69, 166.66 MS m/z (%): 445.47 (M + 2, 16.37), 442.91 (M+, 16.05), 237.34 (100). Anal. calcd for C23H15BrN4O (443.30): C, 62.32; H, 3.41; N 12.64; found: C, 62.50; H, 3.57; N, 12.79.
An equimolar amount of acid hydrazide 3 (10 mmol, 3.42 g) and potassium hydroxide (0.56 g) with carbon disulfide (2 mL) in absolute ethanol (20 mL) was heated under reflux for 12 h. After reaction completion, the excess solvent was evaporated, then neutralized with dil. HCl. The crystallized solid was separated from isopropanol to attain compound 9.
Yellow powder, yield 83%, m.p. 260–262 °C. IR (KBr, cm−1): 3155 (NH), 3086 (CH aromatic), 2899 (CH aliphatic), 1543 (CN), 1238 (CS). 1H NMR (400 MHz, DMSO-d6), δ ppm: 7.75–7.86 (m, 3H, Ar-H), 7.88 (t, 1H, Ar-H), 8.15 (d, 1H, J = 8.0 Hz, Ar-H), 8.21 (d, 2H, J = 8.0 Hz, Ar-H), 8.37 (s, 1H, Ar-H), 8.76 (d, 1H, J = 8.0 Hz, Ar-H), 11.13 (s, 1H, SH, D2O exchangeable). 13C NMR (100 MHz, DMSO-d6), δ ppm: 110.33, 118.37, 122.15, 123.88, 125.33, 127.61, 128.80(2C), 129.36, 130.55, 131.10(2C), 132.49, 136.54, 141.41, 148.67, 154.66. MS m/z (%): 386.53 (M + 2, 19.17), 384.51 (M+, 20.25), 293.51 (100). Anal. calcd for C17H10BrN3OS (384.25): C, 53.14; H, 2.62; N 10.94; found: C, 53.41; H, 2.86; N, 11.17.
Benzoyl chloride (10 mmol, 1.40 mL) was added dropwise to a well-stirred solution of 1,3,4-oxadiazole-2-thiol 9 (10 mmol, 3.84 g) in dioxane (20 mL), and the reaction mixture was stirred at ambient temperature for the entire night. After adding 10% sodium carbonate solution (25 mL) to the reaction mixture, the resulting solid was crystallized from ethanol to get compound 10.
Buff powder, yield 70%, m.p. 220–222 °C. IR (KBr, cm−1): 3061 (CH aromatic), 2920 (CH aliphatic), 1707 (CO), 1589 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 7.50 (t, 2H, Ar-H), 7.61 (t, 1H, Ar-H), 7.76–7.81 (m, 3H, Ar-H), 7.90–7.96 (m, 3H, Ar-H), 8.19 (d, 1H, J = 8.0 Hz, Ar-H), 8.27 (d, 2H, J = 8 Hz, Ar-H), 8.45 (s, 1H, Ar-H), 8.80 (d, 1H, J = 8 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 118.48, 122.55, 123.91, 124.58, 125.26, 127.96, 128.37(2C), 128.65(2C), 129.33, 130.38, 130.68, 131.05, 131.74(2C), 132.71(2C), 136.48, 154.77, 158.84, 159.89, 167.72, 178.86. MS m/z (%): 490.09 (M + 2, 25.22), 488.91 (M+, 26.64), 363.31 (100). Anal. calcd for C24H14BrN3O2S (488.36): C, 59.03; H, 2.89; N 8.60; found: C, 59.31; H, 3.02; N, 8.87.
Benzyl chloride (10 mmol, 1.26 g) was added to a well-stirred suspension of 1,3,4-oxadiazole-2-thiol 9 (10 mmol, 3.84 g) and anhydrous potassium carbonate (10 mmol, 1.38 g) in dry acetone (20 mL). The reaction mixture was heated to reflux for 6 h, and then filtered off. Compound 11 was produced by evaporating the excess solvent and crystallizing it from ethanol.
To a well-stirred suspension of 1,3,4-oxadiazole-2-thiol 9 (10 mmol, 3.84 g) and anhydrous potassium carbonate (10 mmol, 1.38 g) in dry acetone (20 mL), benzyl chloride (10 mmol, 1.26 g) was added. The reaction mixture was heated under reflux for 6 h, and then filtered off. Excess solvent was evaporated, and the obtained solid was dried and crystallized from ethanol to give compound 11.
Yellowish green powder, yield 70%, m.p. 142–144 °C. IR (KBr, cm−1): 3099 (CH aromatic), 2929 (CH aliphatic), 1587 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 4.62 (s, 2H, CH2), 7.24–7.36 (m, 3H, Ar-H), 7.49 (d, 2H, J = 7.2 Hz, Ar-H), 7.67–7.69 (m, 3H, Ar-H), 7.82 (t, 1H, Ar-H), 8.06–8.29 (m, 4H, Ar-H), 8.83 (d, 1H, J = 8.4 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 33.16, 117.36, 121.99, 124.39, 124.52, 125.82, 125.91, 126.21(2C), 126.59, 129.25, 129.63(2C), 129.81(2C), 130.57, 130.83, 131.63(2C), 142.34, 143.10, 148.17, 155.38, 166.40. MS m/z (%): 476.74 (M + 2, 38.62), 474.76 (M+, 37.00), 437.47 (100). Anal. calcd for C24H16BrN3OS (474.38): C, 60.77; H, 3.40; N 8.86; found: C, 60.90; H, 3.62; N, 9.04.
The alkylating agent (20 mmol) was added to a well-stirred suspension of 1,3,4-oxadiazole-2-thiol 9 (10 mmol, 3.84 g) and potassium hydroxide (20 mmol, 1.12 g) in a mixture of ethanol (20 mL) and water (10 mL). The reaction mixture was agitated at 50–60 °C for 4–6 h. Compounds 12a–d were produced by filtering out, drying and crystallizing the produced solid from ethanol.
White powder, yield 70%, m.p. 170–172 °C. IR (KBr, cm−1): 3057 (CH aromatic), 2931 (CH aliphatic), 1571 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 2.87 (3, 3H, CH3), 7.75–7.79 (m, 3H, Ar-H), 7.89 (t, 1H, Ar-H), 8.16 (d, 1H, J = 8.4 Hz, Ar-H), 8.26 (d, 2H, J = 8 Hz, Ar-H), 8.51 (s, 1H, Ar-H), 9.00 (d, 1H, J = 8.4 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 14.92, 119.84, 122.17, 123.22, 126.31, 127.66, 129.31(2C), 130.02, 130.40, 132.02(2C), 138.80, 144.30, 149.04, 153.79, 163.66, 165.00. MS m/z (%): 400.18 (M + 2, 21.71), 398.73 (M+, 22.46), 258.95(100). Anal. calcd for C18H12BrN3OS (398.28): C, 54.28; H, 3.04; N 10.55; found: C, 54.56; H, 3.21; N, 10.79.
White powder, yield 73%, m.p. 121–123 °C. IR (KBr, cm−1): 3055 (CH aromatic), 2962 (CH aliphatic), 1573 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 1.49 (t, 3H, CH3), 3.31–3.44 (q, 2H, CH2), 7.74–7.79 (m, 3H, Ar-H), 7.89 (t, 1H, Ar-H), 8.15 (d, 1H, J = 8 Hz, Ar-H), 8.24 (d, 2H, J = 8.4 Hz, Ar-H), 8.47 (s, 1H, Ar-H), 8.98 (d, 1H, J = 8 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 14.87, 27.80, 118.11, 120.80, 122.16, 125.94, 127.29, 129.70, 129.99(2C) 130.36, 131.34, 132.03(2C), 137.15, 148.66, 155.15, 163.64, 165.31 MS m/z (%): 414.16 (M + 2, 22.22), 412.18 (M+, 22.67), 190.72(100). Anal. calcd for C19H14BrN3OS (412.31): C, 55.35; H, 3.42; N 10.19; found: C, 55.17; H, 3.64; N, 10.45.
White powder, yield 69%, m.p. 117–119 °C. IR (KBr, cm−1): 3084 (CH aromatic), 2980 (CH aliphatic), 1598 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 4.10 (d, 2H, J = 6.8 Hz, SCH2), 5.24 (d, 1H, Jcis = 10 Hz, CHCH), 5.43 (d, 1H, Jtrans = 17.2 Hz, –CHCH), 6.00–6.13 (m, 1H, CHCH2), 7.74–7.78 (m, 3H, Ar-H), 7.88 (t, 1H, Ar-H), 8.13 (d, 1H, J = 8 Hz, Ar-H), 8.23 (d, 2H, J = 7.6 Hz, Ar-H), 8.44 (s, 1H, Ar-H), 8.97 (d, 1H, J = 8 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 34.90, 118.10, 119.84, 121.48, 121.84, 123.89, 125.57, 127.96, 129.03(2C), 129.68, 130.69, 132.41(2C), 133.07, 136.47, 148.00, 154.76, 163.65, 165.69. MS m/z (%): 426.79 (M + 2, 17.69), 424.77 (M+, 18.08), 222.87(100). Anal. calcd for C20H14BrN3OS (424.32): C, 56.61; H, 3.33; N 9.90; found: C, 56.86; H, 3.50; N, 10.14.
White powder, yield 72%, m.p. 176–178 °C. IR (KBr, cm−1): 3080 (CH aromatic), 2972 (CH aliphatic), 1587 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 4.28 (d, 2H, J = 6.4 Hz, SCH2), 6.50–6.54 (m, 1H, C–CH2), 6.80 (d, 1H, J = 15.6 Hz, CH-ph), 7.23–7.29 (m, 3H, Ar-H), 7.42 (d, 2H, J = 6.8 Hz, Ar-H), 7.71–7.77 (m, 3H, Ar-H), 7.85 (t, 1H, Ar-H), 8.12 (d, 1H, J = 8 Hz, Ar-H), 8.19 (d, 2H, J = 7.6 Hz, Ar-H), 8.43 (s, 1H, Ar-H), 8.96 (d, 1H, J = 8.4 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 35.35, 122.18, 122.64, 124.49, 124.93, 125.83, 126.27, 126.73, 129.23(2C), 129.71(2C), 129.93(2C), 130.35, 131.29(2C), 132.39, 134.46, 136.32, 137.17, 139.95, 148.80, 155.14, 162.89, 165.13. MS m/z (%): 502.37 (M + 2, 23.55), 500.27 (M+, 25.10), 434.76(100). Anal. calcd for C26H18BrN3OS (500.41): C, 62.41; H, 3.63; N 8.40; found: C, 62.32; H, 3.79; N, 8.67.
A mixture of 1,3,4-oxadiazole-2-thiol 9 (10 mmol, 3.84 g) and 2-chloroacetic acid (10 mmol, 0.95 g) in methylene chloride (20 mL) containing a few drops of TEA was heated under reflux for 10 h. After completion of the reaction, the excess solvent was evaporated under vacuum and the crude precipitate was crystallized from ethanol to give compound 13.
White powder, yield 69%, m.p. 258–260 °C. IR (KBr, cm−1): 3419 (OH), 3080 (CH aromatic), 2983 (CH aliphatic), 1716 (CO), 1598 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 4.19 (s, 2H, CH2), 5.72 (s, 1H, OH, D2O exchangeable), 7.76–7.80 (m, 3H, Ar-H), 7.90 (t, 1H, Ar-H), 8.17 (d, 1H, J = 8.4 Hz, Ar-H), 8.29 (d, 2H, J = 8.00 Hz, Ar-H), 8.54 (s, 1H, Ar-H), 9.00 (d, 1H, J = 8.0 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 36.62, 118.54, 122.91, 124.89, 125.91, 128.57, 128.93, 129.55(2C), 130.21, 131.28 132.24(2C), 136.62, 148.27, 155.29, 164.04, 166.34, 169.00. MS m/z (%): 444.62 (M + 2, 20.14), 442.56 (M+, 21.06), 326.68(100). Anal. calcd for C19H12BrN3O3S (442.29): C, 51.60; H, 2.73; N 9.50; found: C, 51.78; H, 2.90; N, 9.43.
An equimolar amount of 1,3,4-oxadiazole-2-thiol 9 (10 mmol, 3.84 g) and compound 4 (10 mmol, 2.11 g) in methylene chloride (20 mL) containing a few drops of TEA was heated under reflux for 6 h. The reaction mixture was filtered, and the resulting solid thus obtained was crystallized from ethanol to give compound 14.
White powder, yield 86%, m.p. 219–221 °C. IR (KBr, cm−1): 3446 (NH), 3061 (CH aromatic), 2927 (CH aliphatic), 1670 (CO), 1597 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 2.57 (s, 3H, CH3), 4.51 (s, 2H, CH2), 7.71–7.75 (m, 5H, Ar-H), 7.87–7.93 (m, 3H, Ar-H), 8.16 (d, 1H, J = 8.4 Hz, Ar-H), 8.21 (d, 2H, J = 8 Hz, Ar-H), 8.49 (s, 1H, Ar-H), 8.98 (d, 1H, J = 8.4 Hz, Ar-H), 10.90 (s, 1H, NH, D2O exchangeable). MS m/z (%): 561.96 (M + 2, 17.69), 559.09 (M+, 18.60), 404.96(100). Anal. calcd for C27H19BrN4O3S (559.44): C, 57.97; H, 3.42; N 10.01; found: C, 58.12; H, 3.61; N, 10.29.
The corresponding acetamide derivative 5a,b (10 mmol) was added to a well-stirred solution of 1,3,4-oxadiazole-2-thiol 9 (10 mmol, 3.84 g) and potassium hydroxide (20 mmol, 1.12 g) in a mixture of ethanol (20 mL) and water (10 mL). The reaction mixture was heated to reflux for 6–8 h. Compound 15a,b was produced by filtering out, drying and crystallizing the produced solid from ethanol.
White powder, yield 50% (2.4 g), m.p. 292–294 °C. IR (KBr, cm−1): 3429 (NH), 3061 (CH aromatic), 2968 (CH aliphatic), 1672 (CO), 1587 (CN). 1HNMR (400 MHz, DMSO-d6), δ ppm: 4.27 (s, 2H, CH2), 7.07 (t, 1H, Ar-H), 7.43 (d, 2H, J = 7.6 Hz, Ar-H), 7.57 (t, 1H, Ar-H), 7.70 (d, 2H, J = 7.7 Hz, Ar-H), 7.80 (t, 2H, Ar-H), 7.86 (t, 1H, Ar-H), 8.17 (d, 2H, J = 8.Hz, Ar-H), 8.26 (d, 1H, J = 8.4 Hz, Ar-H), 8.38 (s, 1H, Ar-H) 9.12 (d, 1H, J = 8.4 Hz, Ar-H), 10.96 (s, 1H, NH, D2O exchangeable). 13C NMR (100 MHz, DMSO-d6), δ ppm: 34.00, 117.20, 118.86, 119.91, 120.87(2C), 122.54, 123.89, 128.94(2C), 129.92(2C), 130.60, 131.59(2C), 133.23, 134.60, 135.56, 138.29, 141.97, 148.67, 149.94, 154.62, 163.65, 168.03. MS m/z (%): 519.48 (M + 2, 46.21), 517.38 (M+, 43.91), 195.66(100). Anal. calcd for C25H17BrN4O2S (517.40): C, 58.04; H, 3.31; N 10.83; found: C, 58.31; H, 3.47; N, 11.04.
Grayish white powder, yield 55%, m.p. 565–267 °C. IR (KBr, cm−1): 3446 (NH), 3078 (CH aromatic), 2956 (CH aliphatic), 1651 (CO), 1591 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 3.83 (s, 3H, OCH3), 4.24 (s, 2H, CH2), 7.10 (d, 2H, J = 6.4 Hz, Ar-H), 7.33 (d, 2H, J = 6 Hz, Ar-H), 7.61–7.78 (m, 4H, Ar-H), 8.18–8.36 (m, 4H, Ar-H), 9.13 (d, 1H, J = 7.2 Hz. Ar-H), 10.75 (s, 1H, NH, D2O exchangeable). 13C NMR (100 MHz, DMSO-d6), δ ppm: 33.63, 56.06, 114.54(2C), 116.52, 117.58, 119.90, 124.21(2C), 125.28, 129.24, 129.55, 129.91(2C), 130.60, 132.27(2C), 132.93, 137.30, 138.97, 148.64, 149.28, 153.35, 155.31, 160.05, 166.67. MS m/z (%): 549.92 (M + 2, 24.86), 547.22 (M+, 28.38), 398.33 (100). Anal. calcd for C26H19BrN4O3S (547.43): C, 57.05; H, 3.50; N 10.23; found: C, 57.29; H, 3.62; N, 10.51.
Ethyl chloroacetate (10 mmol, 1.22 g) was added to a well-stirred suspension of 1,3,4-oxadiazole-2-thiol 9 (10 mmol, 3.84 g) and anhydrous potassium carbonate (10 mmol, 1.38 g) in dry acetone (20 mL). The reaction mixture was heated to reflux for 5 h. Compound 6 was produced by filtering out, drying and crystallizing the produced precipitate from ethanol.
White powder, yield 87%, m.p. 310–312 °C. IR (KBr, cm−1): 3077 (CH aromatic), 2983 (CH aliphatic), 1739 (CO), 1588 (CN). 1H NMR (400 MHz, DMSO-d6), δ ppm: 1.20 (t, 3H, CH3), 4.17–4.22 (q, 2H, CH2), 4.41 (s, 2H, CH2), 7.79–7.83 (m, 3H, Ar-H), 7.93 (t, 1H, Ar-H), 8.21 (d, 1H, J = 8.4 Hz, Ar-H), 8.31 (d, 2H, J = 8.8 Hz, Ar-H), 8.59 (s, 1H, Ar-H), 9.00 (d, 1H, J = 8.4 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 14.14, 32.14, 62.29, 118.74, 122.04, 124.58, 125.55, 126.22, 129.01, 129.66(2C), 130.38, 130.68, 132.04(2C), 135.47, 141.22, 148.62, 155.22, 166.38, 167.66. MS m/z (%): 472.95 (M + 2, 23.61), 470.52 (M+, 25.19), 321.42(100). Anal. calcd for C21H16BrN3O3S (470.34): C, 53.63; H, 3.43; N 8.93; found: C, 53.88; H, 3.61; N, 9.12.
A suspension of 1,3,4-oxadiazole-2-thiol 9 (10 mmol, 3.84 g) and an appropriate secondary amine (10 mmol) was heated under reflux in ethanol (30 mL) with 36% formaldehyde (20 mmol) for 4–6 h. After cooling to room temperature, the resultant solid was crystallized from ethanol to get compound 17a–e.
Buff powder, yield 82%, m.p. 210–212 °C. IR (KBr, cm−1): 3059 (CH aromatic), 2974 (CH aliphatic), 1587 (CN), 1267 (CS). 1H NMR (400 MHz, DMSO-d6), δ ppm: 2.82–2.86 (m, 4H, CH2–N–CH2), 3.60–3.62 (m, 4H, CH2–O–CH2), 5.18 (s, 2H, CH2), 7.78–7.81 (m, 3H, Ar-H), 7.91 (t, 1H, Ar-H), 8.20 (d, 1H, J = 8.0 Hz. Ar-H), 8.28 (d, 2H, J = 8.8 Hz, Ar-H), 8.45 (s, 1H, Ar-H), 8.95 (d, 1H, J = 8.0 Hz, Ar-H), 13C NMR (100 MHz, DMSO-d6), δ ppm: 53.95(2C), 66.38(2C), 79.88, 122.55, 123.38, 124.87, 125.93, 126.61, 127.63, 128.61(2C), 129.93, 130.68, 131.75(2C), 137.82, 143.88, 155.15, 157.82, 179.34. MS m/z (%): 485.95 (M + 2, 17.04), 483.52 (M+, 14.99), 437.94 (100). Anal. calcd for C22H19BrN4O2S (483.38): C, 54.66; H, 3.96; N 11.59; found: C, 54.79; H, 4.12; N, 11.78.
Yellow powder, yield 77%, m.p. 192–194 °C, IR (KBr, cm−1): 3014 (CH aromatic), 2933 (CH aliphatic), 1587 (CN), 1246 (CS). 1H NMR (400 MHz, DMSO-d6), δ ppm: 1.35–1.64 (m, 6H, piperidine), 2.73–3.01 (m, 4H, piperidine), 5.17 (s, 2H, CH2), 7.76–7.88 (m, 3H, Ar-H), 7.87 (t, 1H, Ar-H), 8.16 (d, 1H, J = 8.0 Hz. Ar-H), 8.26 (d, 2H, J = 8.4 Hz, Ar-H), 8.36 (s, 1H, Ar-H), 9.15 (d, 1H, J = 8.0 Hz, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 22.23, 23.31(2C), 44.68(2C), 83.48, 116.21, 122.53, 124.69, 126.24, 126.60, 128.27, 129.25(2C), 130.93, 131.27, 132.25(2C), 137.60, 149.27, 155.00, 155.97, 181.05. MS m/z (%): 483.94 (M + 2, 36.44), 481.27 (M+, 36.79), 467.04 (100). Anal. calcd for C23H21BrN4OS (481.41): C, 57.38; H, 4.40; N 11.64; found: C, 57.45; H, 4.63; N, 11.90.
Yellow powder, yield 74%, m.p. 219–221 °C. IR (KBr, cm−1): 3423 (NH), 3064 (CH aromatic), 2931 (CH aliphatic), 1593 (CN), 1249 (CS). 1H NMR (400 MHz, DMSO-d6), δ ppm: 1.04 (s, 1H, NH, D2O exchangeable), 2.61 (s, 4H, piperazine), 2.91–3.07 (s, 4H, piperazine), 5.14 (s, 2H, CH2), 7.73–7.78 (m, 3H, Ar-H), 7.86 (t, 1H, Ar-H), 8.14 (d, 1H, J = 8.0 Hz, Ar-H), 8.26 (d, 2H, J = 7.6 Hz, Ar-H), 8.32 (s, 1H, Ar-H), 9.25 (d, 1H, J = 8.0 Hz, Ar-H). MS m/z (%): 484.08 (M + 2, 24.53), 481.98 (M+, 26.48), 284.45 (100). Anal. calcd for C22H20BrN5OS (482.40): C, 54.78; H, 4.18; N 14.52; found: C, 54.97; H, 4.40; N, 14.76.
Yellow powder, yield 80%, m.p. 156–158 °C. IR (KBr, cm−1): 3055 (CH aromatic), 2995 (CH aliphatic), 1589 (CN), 1265 (CS). 1H NMR (400 MHz, DMSO-d6), δ ppm: 2.83 (s, 3H, CH3), 3.18 (s, 4H, piperazine), 3.96 (s, 4H, piperazine), 5.23 (s, 2H, CH2), 7.65 (t, 1H, Ar-H), 7.71 (d, 2H, J = 8.4 Hz, Ar-H), 7.77 (t, 1H, Ar-H), 7.95 (d, 1H, J = 8.0 Hz, Ar-H), 8.07–8.11 (m, 3H, Ar-H), 8.85 (s, 1H, Ar-H). 13C NMR (100 MHz, DMSO-d6), δ ppm: 18.67, 53.39(2C), 56.43(2C), 69.34, 121.19, 121.87, 123.60, 125.24, 125.56, 128.30, 128.55, 129.42(2C), 130.23, 130.84, 132.18(2C), 137.21, 154.62, 154.72, 180.76. MS m/z (%): 498.15 (M + 2, 19.76), 496.72 (M+, 20.59), 323.33 (100). Anal. calcd for C23H22BrN5OS (496.43): C, 55.65; H, 4.47; N 14.11; found: C, 55.81; H, 4.63; N, 13.98.
Buff powder, yield 77%, m.p. 266–286 °C. IR (KBr, cm−1): 3055 (CH aromatic), 2997 (CH aliphatic), 1573 (CN), 1238 (CS). 1H NMR (400 MHz, DMSO-d6), δ ppm: 5.53 (s, 2H, CH2), 6.80 (t, 2H, Ar-H), 7.05 (d, 4H, J = 8 Hz, Ar-H), 7.21 (t, 4H, Ar-H), 7.71–7.76 (m, 3H, Ar-H), 7.87 (t, 1H, Ar-H), 8.13 (d, 1H, J = 8.4 Hz, Ar-H), 8.19 (d, 2H, J = 8.4 Hz, Ar-H), 8.33 (s, 1H, Ar-H), 8.74 (d, 1H, J = 8.4 Hz, Ar-H). MS m/z (%): 567.00 (M + 2, 15.43), 565.25 (M+, 17.63), 322.87 (100). Anal. calcd for C30H21BrN4OS (565.49): C, 63.72; H, 3.74; N, 9.91; found: C, 63.54; H, 3.89; N, 10.13.
Footnote |
† Electronic supplementary information (ESI) available. See DOI: https://doi.org/10.1039/d4ra06712f |
This journal is © The Royal Society of Chemistry 2024 |