Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.940
Filtrer
1.
Drug Dev Res ; 85(4): e22216, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38831547

RÉSUMÉ

A new series of quinoxaline-sulfonamide derivatives 3-12 were synthesized using fragment-based drug design by reaction of quinoxaline sulfonyl chloride (QSC) with different amines and hydrazines. The quinoxaline-sulfonamide derivatives were evaluated for antidiabetic and anti-Alzheimer's potential against α-glucosidase, α-amylase, and acetylcholinesterase enzymes. These derivatives showed good to moderate potency against α-amylase and α-glucosidase with inhibitory percentages between 24.34 ± 0.01%-63.09 ± 0.02% and 28.95 ± 0.04%-75.36 ± 0.01%, respectively. Surprisingly, bis-sulfonamide quinoxaline derivative 4 revealed the most potent activity with inhibitory percentages of 75.36 ± 0.01% and 63.09 ± 0.02% against α-glucosidase and α-amylase compared to acarbose (IP = 57.79 ± 0.01% and 67.33 ± 0.01%), respectively. Moreover, the quinoxaline derivative 3 exhibited potency as α-glucosidase and α-amylase inhibitory with a minute decline from compound 4 and acarbose with inhibitory percentages of 44.93 ± 0.01% and 38.95 ± 0.01%. Additionally, in vitro acetylcholinesterase inhibitory activity for designed derivatives exhibited weak to moderate activity. Still, sulfonamide-quinoxaline derivative 3 emerged as the most active member with inhibitory percentage of 41.92 ± 0.02% compared with donepezil (IP = 67.27 ± 0.60%). The DFT calculations, docking simulation, target prediction, and ADMET analysis were performed and discussed in detail.


Sujet(s)
Anticholinestérasiques , Inhibiteurs des glycoside hydrolases , Simulation de docking moléculaire , Quinoxalines , Sulfonamides , alpha-Amylases , alpha-Glucosidase , Quinoxalines/composition chimique , Quinoxalines/pharmacologie , Anticholinestérasiques/composition chimique , Anticholinestérasiques/pharmacologie , Inhibiteurs des glycoside hydrolases/pharmacologie , Inhibiteurs des glycoside hydrolases/composition chimique , alpha-Amylases/antagonistes et inhibiteurs , alpha-Amylases/métabolisme , alpha-Glucosidase/métabolisme , alpha-Glucosidase/composition chimique , Sulfonamides/composition chimique , Sulfonamides/pharmacologie , Humains , Hypoglycémiants/composition chimique , Hypoglycémiants/pharmacologie , Relation structure-activité , Acetylcholinesterase/métabolisme , Modèles moléculaires , Pharmacophore
2.
Pestic Biochem Physiol ; 202: 105943, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38879303

RÉSUMÉ

In this study, a new series of thiazolo[4,5-b]quinoxaline derivatives 3-8 were synthesized by treating 2,3-dichloroquinoxaline with thiosemicarbazone and thiourea derivatives under reflux conditions. The chemical structure of the newly designed derivatives was conducted using spectroscopic techniques. The insecticidal bioassay of the designed derivatives was evaluated against the 2nd and 4th larvae of S. litura after five days as toxicity agents via median lethal concentration (LC50) and the lethal time values (LT50). The results indicated that all the tested compounds had insecticidal effects against both instar larvae of S. litura with variable values. Among them, thiazolo[4,5-b]quinoxaline derivative 3 was the most toxic, with LC50 = 261.88 and 433.68 ppm against 2nd and 4th instar larvae, respectively. Moreover, the thiazolo[4,5-b]quinoxaline derivative 3 required the least time to kill the 50% population (LT50) of 2nd larvae were 20.88, 13.2, and 15.84 hs with 625, 1250, and 2500 ppm, respectively, while for the 4th larval instar were 2.75, 2.08, and 1.76 days with concentrations of 625, 1250, and 2500 ppm, respectively. Larvae's morphological and histological studies for the most active derivative 3 were investigated. According to SEM analysis, the exterior morphology of the cuticle and head capsule was affected. In addition, there were some histological alterations in the cuticle layers and the midgut tissues. Columnar cells began breaking down, and vacuolization occurred in the peritrophic membrane. Moreover, treating 4th S litura larvae hemolymph with compound 3 showed significant changes in biochemical analysis, such as total proteins, GPT, GOT, acetylcholinesterase (AChE), and alkaline phosphatase (AlP). Finally, the toxicity prediction of the most active derivative revealed non-corrosive, non-irritant to the eye, non-respiratory toxicity, non-sensitivity to the skin, non-hepatotoxic, and don't have toxicity on minnow toxicity and T. pyriformis indicating a good toxicity profile for human.


Sujet(s)
Insecticides , Larve , Quinoxalines , Spodoptera , Animaux , Insecticides/synthèse chimique , Insecticides/pharmacologie , Insecticides/toxicité , Insecticides/composition chimique , Quinoxalines/toxicité , Quinoxalines/pharmacologie , Quinoxalines/synthèse chimique , Quinoxalines/composition chimique , Larve/effets des médicaments et des substances chimiques , Spodoptera/effets des médicaments et des substances chimiques , Spodoptera/croissance et développement , Thiazoles/composition chimique
3.
Eur Rev Med Pharmacol Sci ; 28(10): 3548-3555, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38856130

RÉSUMÉ

OBJECTIVE: Extended-spectrum beta-lactamases (ESBLs) targeting beta-lactam antibiotics pose a major healthcare challenge. Carbapenems are known to be less impacted. However, the emergence of carbapenem-resistant strains can add further complexity to this existing challenge. With slow drug discovery and rapid resistance, repurposing existing drugs is crucial. This research study aims to provide insight into the antimicrobial effectiveness of 3-hydrazinoquinoxaline-2-thiol against diverse clinical ESBL-producing isolates. MATERIALS AND METHODS: The broth microdilution assay was conducted on a total of sixty-nine clinical ESBL-producing isolates to assess the minimum inhibitory concentrations (MICs) of 3-hydrazinoquinoxaline-2-thiol. The assay was conducted in triplicate, and the average MIC values were calculated. RESULTS: The most repeatedly observed MIC was 64 µg/ml (37.7%), followed by 256 µg/ml (23.2%) and 128 µg/ml (17.4%). Other MICs: 32 µg/ml (11.6%), 16 µg/ml (7.2%), 4-8 µg/ml (1.4%). CONCLUSIONS: This study demonstrated an effect of 3-hydrazinoquinoxaline-2-thiol on various ESBL-producing strains in vitro, indicating its promising therapeutic potential. To comprehensively understand the drug, rigorous testing, including pharmacokinetics, resistance assays, safety assessments, and exploration of potential synergies with other antibiotics against ESBL-producing organisms, is crucial.


Sujet(s)
Antibactériens , Tests de sensibilité microbienne , Quinoxalines , bêta-Lactamases , Quinoxalines/pharmacologie , bêta-Lactamases/métabolisme , Antibactériens/pharmacologie , Humains
4.
J Enzyme Inhib Med Chem ; 39(1): 2367128, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38913598

RÉSUMÉ

Inhibition of α-glucosidase and α-amylase are key tactics for managing blood glucose levels. Currently, stronger, and more accessible inhibitors are needed to treat diabetes. Indeno[1,2-b] quinoxalines-carrying thiazole hybrids 1-17 were created and described using NMR. All analogues were tested for hypoglycaemic effect against STZ-induced diabetes in mice. Compounds 4, 6, 8, and 16 were the most potent among the synthesised analogues. These hybrids were examined for their effects on plasma insulin, urea, creatinine, GSH, MDA, ALT, AST, and total cholesterol. Moreover, these compounds were tested against α-glucosidase and α-amylase enzymes in vitro. The four hybrids 4, 6, 8, and 16 represented moderate to potent activity with IC50 values 0.982 ± 0.04, to 10.19 ± 0.21 for α-glucosidase inhibition and 17.58 ± 0.74 to 121.6 ± 5.14 µM for α-amylase inhibition when compared to the standard medication acarbose with IC50=0.316 ± 0.02 µM for α-glucosidase inhibition and 31.56 ± 1.33 µM for α-amylase inhibition. Docking studies as well as in silico ADMT were done.


Sujet(s)
Relation dose-effet des médicaments , Inhibiteurs des glycoside hydrolases , Hypoglycémiants , Simulation de docking moléculaire , Quinoxalines , Thiazoles , alpha-Amylases , alpha-Glucosidase , Quinoxalines/pharmacologie , Quinoxalines/composition chimique , Quinoxalines/synthèse chimique , alpha-Amylases/antagonistes et inhibiteurs , alpha-Amylases/métabolisme , alpha-Glucosidase/métabolisme , Hypoglycémiants/pharmacologie , Hypoglycémiants/composition chimique , Hypoglycémiants/synthèse chimique , Animaux , Souris , Relation structure-activité , Inhibiteurs des glycoside hydrolases/pharmacologie , Inhibiteurs des glycoside hydrolases/synthèse chimique , Inhibiteurs des glycoside hydrolases/composition chimique , Structure moléculaire , Thiazoles/composition chimique , Thiazoles/pharmacologie , Thiazoles/synthèse chimique , Diabète expérimental/traitement médicamenteux , Streptozocine , Halogénation , Mâle , Antienzymes/pharmacologie , Antienzymes/composition chimique , Antienzymes/synthèse chimique
5.
Molecules ; 29(11)2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38893377

RÉSUMÉ

Plant pathogenic fungi pose a major threat to global food security, ecosystem services, and human livelihoods. Effective and broad-spectrum fungicides are needed to combat these pathogens. In this study, a novel antifungal 2-oxyacetate hydrazide quinoxaline scaffold as a simple analogue was designed and synthesized. Their antifungal activities were evaluated against Botrytis cinerea (B. cinerea), Altemaria solani (A. solani), Gibberella zeae (G. zeae), Rhizoctonia solani (R. solani), Colletotrichum orbiculare (C. orbiculare), and Alternaria alternata (A. alternata). These results demonstrated that most compounds exhibited remarkable inhibitory activities and possessed better efficacy than ridylbacterin, such as compound 15 (EC50 = 0.87 µg/mL against G. zeae, EC50 = 1.01 µg/mL against C. orbiculare) and compound 1 (EC50 = 1.54 µg/mL against A. alternata, EC50 = 0.20 µg/mL against R. solani). The 3D-QSAR analysis of quinoxaline-2-oxyacetate hydrazide derivatives has provided new insights into the design and optimization of novel antifungal drug molecules based on quinoxaline.


Sujet(s)
Antifongiques , Tests de sensibilité microbienne , Relation quantitative structure-activité , Quinoxalines , Antifongiques/pharmacologie , Antifongiques/synthèse chimique , Antifongiques/composition chimique , Quinoxalines/pharmacologie , Quinoxalines/composition chimique , Quinoxalines/synthèse chimique , Conception de médicament , Alternaria/effets des médicaments et des substances chimiques , Rhizoctonia/effets des médicaments et des substances chimiques , Botrytis/effets des médicaments et des substances chimiques , Structure moléculaire , Colletotrichum/effets des médicaments et des substances chimiques , Gibberella/effets des médicaments et des substances chimiques
6.
PLoS One ; 19(5): e0303373, 2024.
Article de Anglais | MEDLINE | ID: mdl-38728271

RÉSUMÉ

BACKGROUND: Candida represents a prevalent fungal infection, notable for its substantial implications on morbidity and mortality rates. In the landscape of prospective treatments, quinoxaline derivatives emerge as a category of compact compounds exhibiting notable potential in addressing infections. These derivatives showcase promising antimicrobial efficacy coupled with favorable pharmacokinetic and safety characteristics. AIMS: The central aim of this investigation was to examine the antifungal characteristics of 2-Chloro-3-hydrazinylquinoxaline against diverse strains of Candida and Aspergillus in vitro. Additionally, we endeavored to assess the in vivo efficacy of 2-Chloro-3-hydrazinylquinoxaline using a murine model for oral candidiasis induced by C. albicans cells ATCC 10231. RESULTS: 2-Chloro-3-hydrazinylquinoxaline demonstrated noteworthy effectiveness when tested against various reference strains of Candida species. It exhibited heightened efficacy, particularly against Candida krusei isolates. However, its performance against Candida albicans, Candida tropicalis, Candida glabrata, Candida parapsilosis, and Candida auris isolates exhibited variability. Notably, 2-Chloro-3-hydrazinylquinoxaline manifests variable efficacy against Aspergillus fumigatus, Aspergillus niger, Aspergillus terreus and Aspergillus flavus and no effect against Aspergillus brasiliensis. In a murine model, 2-Chloro-3-hydrazinylquinoxaline exhibited significant efficacy in combating the C. albicans cells ATCC 10231 strain, underscoring its potential as a viable treatment option. CONCLUSION: 2-Chloro-3-hydrazinylquinoxaline has demonstrated substantial potential in effectively addressing various Candida and Aspergillus species, showcasing dual attributes of antifungal and anti-inflammatory properties. However, to attain a more comprehensive understanding of its therapeutic capabilities, further investigations, incorporating additional tests and experiments, are imperative.


Sujet(s)
Antifongiques , Candida , Tests de sensibilité microbienne , Quinoxalines , Antifongiques/pharmacologie , Antifongiques/composition chimique , Quinoxalines/pharmacologie , Quinoxalines/composition chimique , Animaux , Candida/effets des médicaments et des substances chimiques , Souris , Modèles animaux de maladie humaine , Candidose/traitement médicamenteux , Candidose/microbiologie , Femelle
7.
Int J Biol Macromol ; 269(Pt 1): 132020, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38704061

RÉSUMÉ

A series of D-ring fused 16-substituted steroidal quinoxalin-2(1H)-one attached to an electron-releasing (ER) or electron-withdrawing (EW) groups via steroidal oxoacetate intermediate were synthesized to investigate their protein aggregation inhibition potential using human lysozyme (HLZ). The influence of the type of substituent at the C-6 positions of the quinoxalin-2(1H)-one ring on the protein aggregation inhibition potential was observed, showing that the EW moiety improved the protein aggregation inhibition potency. Of all the evaluated compounds, NO2-substituted quinoxalin-2(1H)-one derivative 13 was the most active compound and had a maximum protein aggregation inhibition effect. Significant stabilization effects strongly support the binding of the most biologically active steroidal quinoxalin-2(1H)-one with docking studies. The predicted physicochemical and ADME properties lie within a drug-like space which shows no violation of Lipinski's rule of five except compounds 12 and 13. Combined, our results suggest that D-ring fused 16-substituted steroidal quinoxalin-2(1H)-one has the potential to modulate the protein aggregation inhibition effect.


Sujet(s)
Simulation de docking moléculaire , Lysozyme , Agrégats de protéines , Quinoxalines , Quinoxalines/composition chimique , Quinoxalines/pharmacologie , Agrégats de protéines/effets des médicaments et des substances chimiques , Humains , Lysozyme/composition chimique , Lysozyme/métabolisme , Stéroïdes/composition chimique , Stéroïdes/pharmacologie , Pliage des protéines
8.
J Inorg Biochem ; 257: 112600, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38759261

RÉSUMÉ

Rhenium complexes show great promise as anticancer drug candidates. Specifically, compounds with a Re(CO)3(NN)(py)+ core in their architecture have shown cytotoxicity equal to or greater than that of well-established anticancer drugs based on platinum or organic molecules. This study aimed to evaluate how the strength of the interaction between rhenium(I) tricarbonyl complexes fac-[Re(CO)3(NN)(py)]+, NN = 1,10-phenanthroline (phen), dipyrido[3,2-f:2',3'-h]quinoxaline (dpq) or dipyrido[3,2-a:2'3'-c]phenazine (dppz) and biomolecules (protein, lipid and DNA) impacted the corresponding cytotoxic effect in cells. Results showed that fac-[Re(CO)3(dppz)(py)]+ has higher Log Po/w and binding constant (Kb) with biomolecules (protein, lipid and DNA) compared to complexes of fac-[Re(CO)3(phen)(py)]+ and fac-[Re(CO)3(dpq)(py)]+. As consequence, fac-[Re(CO)3(dppz)(py)]+ exhibited the highest cytotoxicity (IC50 = 8.5 µM for HeLa cells) for fac-[Re(CO)3(dppz)(py)]+ among the studied compounds (IC50 > 15 µM). This highest cytotoxicity of fac-[Re(CO)3(dppz)(py)]+ are probably related to its lipophilicity, higher permeation of the lipid bilayers of cells, and a more potent interaction of the dppz ligand with biomolecules (protein and DNA). Our findings open novel avenues for rational drug design and highlight the importance of considering the chemical structures of rhenium complexes that strongly interact with biomolecules (proteins, lipids, and DNA).


Sujet(s)
Antinéoplasiques , Complexes de coordination , ADN , Rhénium , Rhénium/composition chimique , Humains , Complexes de coordination/pharmacologie , Complexes de coordination/composition chimique , Complexes de coordination/synthèse chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , ADN/composition chimique , ADN/métabolisme , Phénanthrolines/composition chimique , Phénanthrolines/pharmacologie , Quinoxalines/composition chimique , Quinoxalines/pharmacologie , Phénazines/composition chimique , Phénazines/pharmacologie , Lignée cellulaire tumorale , Cellules HeLa
9.
Chem Biol Interact ; 395: 111036, 2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38705443

RÉSUMÉ

Gelsemium elegans Benth. (G. elegans) is a traditional medicinal herb that has anti-inflammatory, analgesic, sedative, and detumescence effects. However, it can also cause intestinal side effects such as abdominal pain and diarrhea. The toxicological mechanisms of gelsenicine are still unclear. The objective of this study was to assess enterotoxicity induced by gelsenicine in the nematodes Caenorhabditis elegans (C. elegans). The nematodes were treated with gelsenicine, and subsequently their growth, development, and locomotion behavior were evaluated. The targets of gelsenicine were predicted using PharmMapper. mRNA-seq was performed to verify the predicted targets. Intestinal permeability, ROS generation, and lipofuscin accumulation were measured. Additionally, the fluorescence intensities of GFP-labeled proteins involved in oxidative stress and unfolded protein response in endoplasmic reticulum (UPRER) were quantified. As a result, the treatment of gelsenicine resulted in the inhibition of nematode lifespan, as well as reductions in body length, width, and locomotion behavior. A total of 221 targets were predicted by PharmMapper, and 731 differentially expressed genes were screened out by mRNA-seq. GO and KEGG enrichment analysis revealed involvement in redox process and transmembrane transport. The permeability assay showed leakage of blue dye from the intestinal lumen into the body cavity. Abnormal mRNAs expression of gem-4, hmp-1, fil-2, and pho-1, which regulated intestinal development, absorption and catabolism, transmembrane transport, and apical junctions, was observed. Intestinal lipofuscin and ROS were increased, while sod-2 and isp-1 expressions were decreased. Multiple proteins in SKN-1/DAF-16 pathway were found to bind stably with gelsenicine in a predictive model. There was an up-regulation in the expression of SKN-1:GFP, while the nuclear translocation of DAF-16:GFP exhibited abnormality. The UPRER biomarker HSP-4:GFP was down-regulated. In conclusion, the treatment of gelsenicine resulted in the increase of nematode intestinal permeability. The toxicological mechanisms underlying this effect involved the disruption of intestinal barrier integrity, an imbalance between oxidative and antioxidant processes mediated by the SKN-1/DAF-16 pathway, and abnormal unfolded protein reaction.


Sujet(s)
Caenorhabditis elegans , Espèces réactives de l'oxygène , Animaux , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Caenorhabditis elegans/métabolisme , Espèces réactives de l'oxygène/métabolisme , Quinoxalines/pharmacologie , Protéines de Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/génétique , Stress oxydatif/effets des médicaments et des substances chimiques , Intestins/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Muqueuse intestinale/effets des médicaments et des substances chimiques , Gelsemium/composition chimique , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Perméabilité/effets des médicaments et des substances chimiques , Lipofuscine/métabolisme , Locomotion/effets des médicaments et des substances chimiques , Alcaloïdes indoliques
10.
Eur J Med Chem ; 274: 116536, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-38805936

RÉSUMÉ

G-quadruplexes (G4s) are commonly formed in the G-rich strand of telomeric DNA. Ligands targeting telomeric G4 induce DNA damage and telomere dysfunction, which makes them potential antitumor drugs. New telomeric G4 ligands with drug-likeness are still needed to be exploited, especially with their antitumor mechanisms thoroughly discussed. In this study, a novel series of quinoxaline analogs were rationally designed and synthesized. Among them, R1 was the most promising ligand for its cytotoxic effects on tumor cells and stabilizing ability with telomeric G4. Cellular assays illustrated that R1 stabilized G4 and induced R-loop accumulation in the telomeric regions, subsequently triggering DNA damage responses, cell cycle arrest in G2/M phase, apoptosis and antiproliferation. Moreover, R1 evoked immunogenic cell death (ICD) in tumor cells, which promoted the maturation of bone marrow derived dendritic cells (BMDCs). In breast cancer mouse model, R1 exhibited a significant decrease in tumor burden through the immunomodulatory effects, including the increase of CD4+ and CD8+ T cells in tumors and cytokine levels in sera. Our research provides a new idea that targeting telomeric G4 induces DNA damage responses, causing antitumor effects both in vitro and in vivo, partially due to the enhancement of immunomodulation.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , G-quadruplexes , Quinoxalines , Télomère , G-quadruplexes/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Quinoxalines/composition chimique , Quinoxalines/pharmacologie , Quinoxalines/synthèse chimique , Animaux , Humains , Télomère/effets des médicaments et des substances chimiques , Ligands , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Structure moléculaire , Tests de criblage d'agents antitumoraux , Relation structure-activité , Relation dose-effet des médicaments , Femelle , Immunomodulation/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris de lignée BALB C , Altération de l'ADN/effets des médicaments et des substances chimiques
11.
J Mater Chem B ; 12(17): 4197-4207, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38595311

RÉSUMÉ

Second near-infrared (NIR-II) fluorescence imaging shows huge application prospects in clinical disease diagnosis and surgical navigation, while it is still a big challenge to exploit high performance NIR-II dyes with long-wavelength absorption and high fluorescence quantum yield. Herein, based on planar π-conjugated donor-acceptor-donor systems, three NIR-II dyes (TP-DBBT, TP-TQ1, and TP-TQ2) were synthesized with bulk steric hindrance, and the influence of acceptor engineering on absorption/emission wavelengths, fluorescence efficiency and photothermal properties was systematically investigated. Compared with TP-DBBT and TP-TQ2, the TP-TQ1 based on 6,7-diphenyl-[1,2,5]thiadiazoloquinoxaline can well balance absorption/emission wavelengths, NIR-II fluorescence brightness and photothermal effects. And the TP-TQ1 nanoparticles (NPs) possess high absorption ability at a peak absorption of 877 nm, with a high relative quantum yield of 0.69% for large steric hindrance hampering the close π-π stacking interactions. Furthermore, the TP-TQ1 NPs show a desirable photothermal conversion efficiency of 48% and good compatibility. In vivo experiments demonstrate that the TP-TQ1 NPs can serve as a versatile theranostic agent for NIR-II fluorescence/photoacoustic imaging-guided tumor phototherapy. The molecular planarization strategy provides an approach for designing efficient NIR-II fluorophores with extending absorption/emission wavelength, high fluorescence brightness, and outstanding phototheranostic performance.


Sujet(s)
Colorants fluorescents , Rayons infrarouges , Quinoxalines , Thiadiazoles , Quinoxalines/composition chimique , Quinoxalines/synthèse chimique , Quinoxalines/pharmacologie , Colorants fluorescents/composition chimique , Colorants fluorescents/synthèse chimique , Animaux , Souris , Humains , Thiadiazoles/composition chimique , Nanomédecine théranostique , Structure moléculaire , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Imagerie optique , Souris de lignée BALB C , Femelle , Photothérapie/méthodes , Survie cellulaire/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Taille de particule
12.
Eur J Med Chem ; 271: 116360, 2024 May 05.
Article de Anglais | MEDLINE | ID: mdl-38614060

RÉSUMÉ

Cancer is a leading cause of death and a major health problem worldwide. While many effective anticancer agents are available, most drugs currently on the market are not specific, raising issues like the common side effects of chemotherapy. However, recent research hold promises for the development of more efficient and safer anticancer drugs. Quinoxaline and its derivatives are becoming recognized as a novel class of chemotherapeutic agents with activity against different tumors. The present review compiles and discusses studies concerning the therapeutic potential of the anticancer activity of quinoxaline derivatives, covering articles published between January 2018 and January 2023.


Sujet(s)
Antinéoplasiques , Tumeurs , Quinoxalines , Quinoxalines/composition chimique , Quinoxalines/pharmacologie , Quinoxalines/synthèse chimique , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Tumeurs/traitement médicamenteux , Animaux , Structure moléculaire , Développement de médicament , Prolifération cellulaire/effets des médicaments et des substances chimiques , Découverte de médicament , Tests de criblage d'agents antitumoraux , Relation structure-activité
13.
Eur J Med Chem ; 270: 116377, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38581731

RÉSUMÉ

Evading the cellular apoptosis mechanism by modulating multiple pathways poses a sturdy barrier to effective chemotherapy. Cancer cell adeptly resists the apoptosis signaling pathway by regulating anti and pro-apoptotic proteins to escape cell death. Nevertheless, bypassing the apoptotic pathway through necroptosis, an alternative programmed cell death process, maybe a potential therapeutic modality for apoptosis-resistant cells. However, synthetic mono-quinoxaline-based intercalator-induced cellular necroptosis as an anti-cancer perspective remains under-explored. To address this concern, we undertook the design and synthesis of quinoxaline-based small molecules (3a-3l). Our approach involved enhancing the π-surface of the mandatory benzyl moiety to augment its ability to induce DNA structural alteration via intercalation, thereby promoting cytotoxicity across various cancer cell lines (HCT116, HT-29, and HeLa). Notably, the potent compound 3a demonstrated the capacity to induce DNA damage in cancer cells, leading to the induction of ZBP1-mediated necroptosis in the RIP3-expressed cell line (HT-29), where Z-VAD effectively blocked apoptosis-mediated cell death. Interestingly, we observed that 3a induced RIP3-driven necroptosis in combination with DNA hypomethylating agents, even in the RIP3-silenced cell lines (HeLa and HCT116). Overall, our synthesized compound 3a emerged as a promising candidate against various cancers, particularly in apoptosis-compromised cells, through the induction of necroptosis.


Sujet(s)
Nécroptose , Tumeurs , Humains , Quinoxalines/pharmacologie , Apoptose , Cellules HT29 , ADN/pharmacologie , Nécrose/induit chimiquement , Protein kinases/métabolisme
14.
Chem Biol Drug Des ; 103(3): e14499, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38444047

RÉSUMÉ

In this paper, we report the synthesis of quinoxaline-isoxazole-piperazine conjugates. The anticancer activity was evaluated against three human cancer cell lines, including MCF-7 (breast), HepG-2 (liver), and HCT-116 (colorectal). The outcomes of the tested compounds 5d, 5e, and 5f have shown more potent activity when compared to the standard drug erlotinib. In a cell survivability test (MCF-10A), three potent compounds (5d, 5e, and 5f) were evaluated against the normal breast cell line, although neither of them displayed any significant cytotoxicity with IC50 values greater than 84 µM. Furthermore, the compounds 5d, 5e, and 5f were tested for tyrosine kinase EGFR inhibitory action using erlotinib as the reference drug and compound 5e was shown to be more potent in inhibiting the tyrosine kinase EGFR than sorafenib. In addition to this, molecular docking studies of compounds 5d, 5e, and 5f demonstrated that these compounds had more EGFR-binding interactions. The potent compounds 5d, 5e, and 5f were subjected to in silico pharmacokinetic assessment by SWISS, ADME, and pkCSM. While the compounds 5d, 5e, and 5f followed Lipinski, Veber, Egan, and Muegge rules without any deviation.


Sujet(s)
Antinéoplasiques , Quinoxalines , Humains , Simulation de docking moléculaire , Chlorhydrate d'erlotinib/pharmacologie , Quinoxalines/pharmacologie , Antinéoplasiques/pharmacologie , Isoxazoles , Pipérazine , Protein-tyrosine kinases , Récepteurs ErbB
15.
Chem Biol Drug Des ; 103(3): e14502, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38453260

RÉSUMÉ

We designed and synthesized thirty novel quinoxaline aryl ethers as anticancer agents, and the structures of final compounds were confirmed with various analytical techniques like Mass, 1 H NMR, 13 C NMR, FTIR, and elemental analyses. The compounds were tested against three cancer cell lines: colon cancer (HCT-116), breast cancer (MDA-MB-231), prostate cancer (DU-145), and one normal cell line: human embryonic kidney cell line (HEK-293). The obtained results indicate that two compounds, FQ and MQ, with IC50 values < 16 µM, were the most active compounds. Molecular docking studies revealed the binding of FQ and MQ molecules in the active site of the c-Met kinase (PDB ID: 3F66, 1.40 Å). Furthermore, QikProp ADME prediction and the MDS analysis preserved those critical docking data of both compounds, FQ and MQ. Western blotting was used to confirm the impact of the compounds FQ and MQ on the inhibition of the c-Met kinase receptor. The apoptosis assays were performed to investigate the mechanism of cell death for the most active compounds, FQ and MQ. The Annexin V/7-AAD assay indicated apoptosis in MDA-MB-231 cells treated with FQ and MQ, with FQ (21.4%) showing a higher efficacy in killing MDA-MB-231 cells than MQ (14.25%). The Caspase 3/7 7-AAD assay further supported these findings, revealing higher percentages of apoptotic cells for FQ-treated MDA-MB-231 cells (41.8%). The results obtained from the apoptosis assay conclude that FQ exhibits better anticancer activity against MDA-MB-231 cells than MQ.


Sujet(s)
Antinéoplasiques , Éthers , Humains , Lignée cellulaire tumorale , Simulation de docking moléculaire , Quinoxalines/pharmacologie , Cellules HEK293 , Prolifération cellulaire , Tests de criblage d'agents antitumoraux , Antinéoplasiques/composition chimique , Apoptose , Structure moléculaire , Relation structure-activité
16.
J Biochem Mol Toxicol ; 38(4): e23690, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38493304

RÉSUMÉ

The cytotoxic activity, EGFR/VEGFR2 target inhibition, apoptotic activity, RT-PCR gene expression, in vivo employing a solid-Ehrlich carcinoma model, and in silico investigations for highlighting the binding affinity of eight quinoxaline derivatives were tested for anticancer activities. The results showed that compound 8 (N-allyl quinoxaline) had potent cytotoxicity against A594 and MCF-7 cancer cells with IC50 values of 0.86 and 1.06 µM, respectively, with noncytotoxic activity against WISH and MCF-10A cells having IC50 values more than 100 µM. Furthermore, it strongly induced apoptotic cell death in A549 and MCF-7 cells by 43.13% and 34.07%, respectively, stopping the cell cycle at S and G1-phases. For the molecular target, the results showed that compound 8 had a promising EGFR inhibition activity with an IC50 value of 0.088 µM compared to Sorafenib (IC50 = 0.056 µM), and it had a promising VEGFR2 inhibition activity with an IC50 value of 0.108 µM compared to Sorafenib (IC50 = 0.049 µM). Treatment with compound 8 ameliorated biochemical and histochemical parameters near normal in the in vivo investigation, with a tumor inhibition ratio of 68.19% compared to 64.8% for 5-FU treatment. Finally, the molecular docking study demonstrated the binding affinity through binding energy and interactive binding mode inside the EGFR/VEGFR2 proteins. Potent EGFR and VEGFR2 inhibition of compound 8 suggests its potential for development as a selective anticancer drug.


Sujet(s)
Antinéoplasiques , Quinoxalines , Humains , Relation structure-activité , Sorafénib/pharmacologie , Simulation de docking moléculaire , Quinoxalines/pharmacologie , Apoptose , Antinéoplasiques/composition chimique , Récepteurs ErbB/métabolisme , Prolifération cellulaire , Structure moléculaire , Tests de criblage d'agents antitumoraux , Inhibiteurs de protéines kinases/pharmacologie
17.
Cancer Immunol Res ; 12(5): 631-643, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38407902

RÉSUMÉ

Chimeric antigen receptor (CAR) T cells are emerging as an effective antitumoral therapy. However, their therapeutic effects on solid tumors are limited because of their short survival time and the immunosuppressive tumor microenvironment. Memory T cells respond more vigorously and persist longer than their naïve/effector counterparts. Therefore, promoting CAR T-cell development into memory T cells could further enhance their antitumoral effects. HI-TOPK-032 is a T-LAK cell-originated protein kinase (TOPK)-specific inhibitor that moderately represses some types of tumors. However, it is unknown whether HI-TOPK-032 works on hepatocellular carcinoma (HCC) and whether it impacts antitumoral immunity. Using both subcutaneous and orthotopic xenograft tumor models of two human HCC cell lines, Huh-7 and HepG2, we found that HI-TOPK-032 significantly improved proliferation/persistence of CD8+ CAR T cells, as evidenced by an increase in CAR T-cell counts or frequency of Ki-67+CD8+ cells and a decrease in PD-1+LAG-3+TIM-3+CD8+ CAR T cells in vivo. Although HI-TOPK-032 did not significantly suppress HCC growth, it enhanced the capacity of CAR T cells to inhibit tumor growth. Moreover, HI-TOPK-032 augmented central memory CD8+ T cell (TCM) frequency while increasing eomesodermin expression in CD8+ CAR T cells in tumor-bearing mice. Moreover, it augmented CD8+ CAR TCM cells in vitro and reduced their expression of immune checkpoint molecules. Finally, HI-TOPK-032 inhibited mTOR activation in CAR T cells in vitro and in tumors, whereas overactivation of mTOR reversed the effects of HI-TOPK-032 on CD8+ TCM cells and tumor growth. Thus, our studies have revealed mechanisms underlying the antitumoral effects of HI-TOPK-032 while advancing CAR T-cell immunotherapy.


Sujet(s)
Carcinome hépatocellulaire , Immunothérapie adoptive , Indolizine , Tumeurs du foie , Cellules T mémoire , Quinoxalines , Animaux , Humains , Souris , Carcinome hépatocellulaire/thérapie , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/anatomopathologie , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Immunothérapie adoptive/méthodes , Tumeurs du foie/immunologie , Tumeurs du foie/thérapie , Tumeurs du foie/anatomopathologie , Cellules T mémoire/effets des médicaments et des substances chimiques , Cellules T mémoire/immunologie , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Récepteurs chimériques pour l'antigène/immunologie , Microenvironnement tumoral/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Indolizine/pharmacologie , Indolizine/usage thérapeutique , Quinoxalines/pharmacologie , Quinoxalines/usage thérapeutique
18.
Int J Biol Macromol ; 263(Pt 1): 130175, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38360242

RÉSUMÉ

Diabetes mellitus is a multifactorial disease and its effective therapy often demands several drugs with different modes of action. Herein, we report a rational design and synthesis of multi-targeting novel molecular hybrids comprised of EGCG and quinoxaline derivatives that can effectively inhibit α-glucosidase, α-amylase as well as control oxidative stress by scavenging ROS. The hybrids showed superior inhibition of α-glucosidase along with similar α-amylase inhibition as compared to standard drug, acarbose. Most potent compound, 15c showed an IC50 of 0.50 µM (IC50 of acarbose 190 µM) against α-glucosidase. Kinetics studies with 15c revealed a competitive inhibition against α-glucosidase. Binding affinity of 15c (-9.5 kcal/mol) towards α-glucosidase was significantly higher than acarbose (-7.7 kcal/mol). 15c exhibited remarkably high antioxidant activity (IC50 = 18.84 µM), much better than vitamin C (IC50 = 33.04 µM). Of note, acarbose shows no antioxidant activity. Furthermore, α-amylase activity was effectively inhibited by 15c with an IC50 value of 16.35 µM. No cytotoxicity was observed for 15c (up to 40 µM) in MCF-7 cells. Taken together, we report a series of multi-targeting molecular hybrids capable of inhibiting carbohydrate hydrolysing enzymes as well as reducing oxidative stress, thus representing an advancement towards effective and novel therapeutic approaches for diabetes.


Sujet(s)
Diabète , Hypoglycémiants , Humains , Hypoglycémiants/pharmacologie , Hypoglycémiants/composition chimique , Acarbose/pharmacologie , Acarbose/composition chimique , alpha-Glucosidase/métabolisme , alpha-Amylases/composition chimique , Quinoxalines/pharmacologie , Antioxydants/composition chimique , Stress oxydatif , Simulation de docking moléculaire , Inhibiteurs des glycoside hydrolases/composition chimique
19.
J Med Chem ; 67(5): 3571-3589, 2024 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-38385264

RÉSUMÉ

PAR4 is a promising antithrombotic target with potential for separation of efficacy from bleeding risk relative to current antiplatelet therapies. In an effort to discover a novel PAR4 antagonist chemotype, a quinoxaline-based HTS hit 3 with low µM potency was identified. Optimization of the HTS hit through the use of positional SAR scanning and the design of conformationally constrained cores led to the discovery of a quinoxaline-benzothiazole series as potent and selective PAR4 antagonists. The lead compound 48, possessing a 2 nM IC50 against PAR4 activation by γ-thrombin in platelet-rich plasma (PRP) and greater than 2500-fold selectivity versus PAR1, demonstrated robust antithrombotic efficacy and minimal bleeding in the cynomolgus monkey models.


Sujet(s)
Fibrinolytiques , Thrombose , Animaux , Fibrinolytiques/pharmacologie , Fibrinolytiques/usage thérapeutique , Macaca fascicularis , Quinoxalines/pharmacologie , Quinoxalines/usage thérapeutique , Récepteurs à la thrombine , Thrombine , Hémorragie , Thrombose/traitement médicamenteux , Thrombose/prévention et contrôle , Récepteur de type PAR-1 , Plaquettes , Agrégation plaquettaire
20.
Molecules ; 29(2)2024 Jan 17.
Article de Anglais | MEDLINE | ID: mdl-38257377

RÉSUMÉ

A facile methodology for the construction of a complex heterocycle indazolo-fused quinoxalinone has been developed via an Ugi four-component reaction (U-4CR) followed by an intramolecular Ullmann reaction. The expeditious process features an operationally simple approach, time efficiency, and a broad substrate scope. Biological activity was evaluated and demonstrated that compound 6e inhibits human colon cancer cell HCT116 proliferation with an IC50 of 2.1 µM, suggesting potential applications for developing a drug lead in medicinal chemistry.


Sujet(s)
Tumeurs du côlon , Quinoxalines , Humains , Quinoxalines/pharmacologie , Prolifération cellulaire , Chimie pharmaceutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...