Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.003
Filtrar
1.
Eur J Med Chem ; 270: 116377, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38581731

RESUMO

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.


Assuntos
Necroptose , Neoplasias , Humanos , Quinoxalinas/farmacologia , Apoptose , Células HT29 , DNA/farmacologia , Necrose/induzido quimicamente , Proteínas Quinases/metabolismo
2.
JCO Precis Oncol ; 8: e2300406, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38603651

RESUMO

PURPOSE: Despite fibroblast growth factor receptor (FGFR) inhibitors being approved in tumor types with select FGFR rearrangements or gene mutations, amplifications of FGFR represent the most common FGFR alteration across malignancies. Subprotocol K1 (EAY131-K1) of the National Cancer Institute-MATCH platform trial was designed to evaluate the antitumor efficacy of the oral FGFR1-4 inhibitor, erdafitinib, in patients with tumors harboring FGFR1-4 amplification. METHODS: EAY131-K1 was an open-label, single-arm, phase II study with central confirmation of presence of FGFR1-4 amplification in tumors. Patients with urothelial carcinoma were excluded. Enrolled patients received oral erdafitinib at a starting dose of 8 mg once daily continuously with escalation to 9 mg once daily continuously, on the basis of predefined time point assessments of phosphate levels, until disease progression or intolerable toxicity. The primary end point was centrally assessed objective response rate (ORR), with key secondary end points being 6-month progression-free survival (PFS6), PFS, overall survival (OS), and safety. RESULTS: Thirty-five patients were enrolled into this study with 18 included in the prespecified primary efficacy analysis. The median age of the 18 patients was 60 years, and 78% had received ≥3 previous lines of therapy. There were no confirmed responses to erdafitinib; however, five patients experienced stable disease (SD) as best response. One patient with an FGFR1-amplified breast cancer had a prolonged PFS >168 days (5.5 months). The median PFS was 1.7 months (90% CI, 1.1 to 1.8 months) and the median OS was 4.2 months (90% CI, 2.3 to 9.3 months). The estimated PFS6 rate was 13.8% (90% CI, 3.3 to 31.6). The majority of toxicities were grade 1 to 2 in nature, although there was one grade 5 treatment-related adverse event. CONCLUSION: Erdafitinib did not meet its primary end point of efficacy as determined by ORR in treatment-refractory solid tumors harboring FGFR1-4 amplifications. Our findings support that rearrangements and gene mutations, but not amplifications, of FGFR remain the established FGFR alterations with approved indications for FGFR inhibition.


Assuntos
Carcinoma de Células de Transição , Quinoxalinas , Neoplasias da Bexiga Urinária , Estados Unidos , Humanos , Pessoa de Meia-Idade , National Cancer Institute (U.S.) , Neoplasias da Bexiga Urinária/tratamento farmacológico , Pirazóis/uso terapêutico
3.
JCO Precis Oncol ; 8: e2300407, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38603650

RESUMO

PURPOSE: Subprotocol K2 (EAY131-K2) of the NCI-MATCH platform trial was an open-label, single-arm, phase II study designed to evaluate the antitumor efficacy of the oral FGFR1-4 inhibitor, erdafitinib, in patients with tumors harboring FGFR1-4 mutations or fusions. METHODS: Central confirmation of tumor FGFR1-4 mutations or fusions was required for outcome analysis. Patients with urothelial carcinoma were excluded. Enrolled subjects received oral erdafitinib at a starting dose of 8 mg daily continuously until intolerable toxicity or disease progression. The primary end point was objective response rate (ORR) with key secondary end points of safety, progression-free survival (PFS), and overall survival (OS). RESULTS: Thirty-five patients were enrolled, and 25 patients were included in the primary efficacy analysis as prespecified in the protocol. The median age was 61 years, and 52% of subjects had received ≥3 previous lines of therapy. The confirmed ORR was 16% (4 of 25 [90% CI, 5.7 to 33.0], P = .034 against the null rate of 5%). An additional seven patients experienced stable disease as best-confirmed response. Four patients had a prolonged PFS including two with recurrent WHO grade IV, IDH1-/2-wildtype glioblastoma. The median PFS and OS were 3.6 months and 11.0 months, respectively. Erdafitinib was manageable with no new safety signals. CONCLUSION: This study met its primary end point in patients with several pretreated solid tumor types harboring FGFR1-3 mutations or fusions. These findings support advancement of erdafitinib for patients with fibroblast growth factor receptor-altered tumors outside of currently approved indications in a potentially tumor-agnostic manner.


Assuntos
Carcinoma de Células de Transição , Quinoxalinas , Neoplasias da Bexiga Urinária , Humanos , Pessoa de Meia-Idade , Neoplasias da Bexiga Urinária/tratamento farmacológico , Pirazóis/uso terapêutico , Pirazóis/efeitos adversos , Mutação
4.
J Biochem Mol Toxicol ; 38(4): e23690, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38493304

RESUMO

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.


Assuntos
Antineoplásicos , Quinoxalinas , Humanos , Relação Estrutura-Atividade , Sorafenibe/farmacologia , Simulação de Acoplamento Molecular , Quinoxalinas/farmacologia , Apoptose , Antineoplásicos/química , Receptores ErbB/metabolismo , Proliferação de Células , Estrutura Molecular , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores de Proteínas Quinases/farmacologia
5.
J Hazard Mater ; 469: 134068, 2024 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-38521040

RESUMO

The abuse of olaquindox (OLA) as both an antimicrobial agent and a growth promoter poses significant threats to the environment and human health. While nanoreactors have proven effective in hazard detection, their widespread adoption has been hindered by tedious chemical processes and limited functionality. In this study, we introduce a novel green self-assembly strategy utilizing invertase, horseradish peroxidase, antibodies, and gold nanoclusters to form an aggregation-induced emission-type zeolitic imidazolate framework-8 nanoreactor. The results demonstrate that the lateral flow immunoassay not only allows for qualitative naked eye detection but also enables optical analysis through the fluorescence generated by aggregated gold nanoclusters and enzyme-catalyzed enhancement of visible colorimetric signals. To accommodate more detection scenarios, the photothermal effects and redox reactions of the nanoreactor can fulfill the requirements of thermal sensing and electrochemical analysis for smartphone applications. Remarkably, the proposed approach achieves a detection limit 17 times lower than conventional methods. Besides, the maximum linear range spans from 0.25 to 5 µg/L with high specificity, and the recovery is 85.2-112.9% in environmental water and swine urine. The application of this high-performance nanoreactor opens up avenues for the construction of multifunctional biosensors with great potential in monitoring hazardous materials.


Assuntos
Quinoxalinas , Smartphone , Zeolitas , Animais , Biônica , Ouro , Nanotecnologia , Suínos
6.
Chem Res Toxicol ; 37(4): 528-539, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38507288

RESUMO

Quinoxaline 1,4-di-N-Oxides (QdNOs) have been used as synthetic antimicrobial agents in animal husbandry and aquaculture. The metabolism and potential toxicity have been also concerns in recently years. The metabolism investigations showed that there were 8 metabolites of Carbadox (CBX), 34 metabolites of Cyadox (CYA), 33 metabolites of Mequindox (MEQ), 35 metabolites of Olaquindox (OLA), and 56 metabolites of Quinocetone (QCT) in different animals. Among them, Cb3 and Cb8, M6, and O9 are metabolic residual markers of CBX, MEQ and OLA, which are associated with N → O reduction. Toxicity studies revealed that QdNOs exhibited severe tumorigenicity, cytotoxicity, and adrenal toxicity. Metabolic toxicology showed that toxicity of QdNOs metabolites might be related to the N → O group reduction, and some metabolites exhibited higher toxic effects than the precursor, which could provide guidance for further research on the metabolic toxicology of QdNOs and provide a wealth of information for food safety evaluation.


Assuntos
Óxidos , Quinoxalinas , Animais , Quinoxalinas/toxicidade , Quinoxalinas/metabolismo , Carbadox , Estresse Oxidativo
7.
Chem Biol Drug Des ; 103(3): e14499, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38444047

RESUMO

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.


Assuntos
Antineoplásicos , Quinoxalinas , Humanos , Simulação de Acoplamento Molecular , Cloridrato de Erlotinib/farmacologia , Quinoxalinas/farmacologia , Antineoplásicos/farmacologia , Isoxazóis , Piperazina , Proteínas Tirosina Quinases , Receptores ErbB
8.
Chem Biol Drug Des ; 103(3): e14502, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38453260

RESUMO

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.


Assuntos
Antineoplásicos , Éteres , Humanos , Linhagem Celular Tumoral , Simulação de Acoplamento Molecular , Quinoxalinas/farmacologia , Células HEK293 , Proliferação de Células , Ensaios de Seleção de Medicamentos Antitumorais , Antineoplásicos/química , Apoptose , Estrutura Molecular , Relação Estrutura-Atividade
9.
Korean J Gastroenterol ; 83(3): 111-118, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38522854

RESUMO

Background/Aims: This study compared the effectiveness and safety of glecaprevir/pibrentasvir (GLE/PIB) and sofosbuvir/ledipasvir (SOF/LDV) in real-life clinical practice. Methods: The data from genotype 1 or 2 chronic hepatitis C patients treated with GLE/PIB or sofosbuvir + ribavirin or SOF/LDV in South Korea were collected retrospectively. The analysis included the treatment completion rate, sustained virologic response at 12 weeks (SVR12) test rate, treatment effectiveness, and adverse events. Results: Seven hundred and eighty-two patients with genotype 1 or 2 chronic hepatitis C who were treated with GLE/PIB (n=575) or SOF/LDV (n=207) were included in this retrospective study. The baseline demographic and clinical characteristics revealed significant statistical differences in age, genotype, ascites, liver cirrhosis, and hepatocellular carcinoma between the GLE/PIB and SOF/LDV groups. Twenty-two patients did not complete the treatment protocol. The treatment completion rate was high for both regimens without statistical significance (97.7% vs. 95.7%, p=0.08). The overall SVR12 of intention-to-treat analysis was 81.2% vs. 80.7% without statistical significance (p=0.87). The overall SVR12 of per protocol analysis was 98.7% vs. 100% without statistical significance (p=0.14). Six patients treated with GLE/PIB experienced treatment failure. They were all male, genotype 2, and showed a negative hepatitis C virus RNA level at the end of treatment. Two patients treated with GLE/PIB stopped medication because of fever and abdominal discomfort. Conclusions: Both regimens had similar treatment completion rates, effectiveness, and safety profiles. Therefore, the SOF/LDV regimen can also be considered a viable DAA for the treatment of patients with genotype 1 or 2 chronic hepatitis C.


Assuntos
Ácidos Aminoisobutíricos , Benzimidazóis , Ciclopropanos , Fluorenos , Hepatite C Crônica , Lactamas Macrocíclicas , Leucina/análogos & derivados , Neoplasias Hepáticas , Prolina/análogos & derivados , Pirrolidinas , Quinoxalinas , Sulfonamidas , Humanos , Masculino , Sofosbuvir/uso terapêutico , Antivirais/uso terapêutico , Hepatite C Crônica/tratamento farmacológico , Hepacivirus/genética , Estudos Retrospectivos , Resultado do Tratamento , Neoplasias Hepáticas/tratamento farmacológico , Genótipo , Quimioterapia Combinada
11.
J Hazard Mater ; 469: 133967, 2024 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-38457978

RESUMO

Diclofop-methyl, an aryloxyphenoxypropionate (AOPP) herbicide, is a chiral compound with two enantiomers. Microbial detoxification and degradation of various enantiomers is garnering immense research attention. However, enantioselective catabolism of diclofop-methyl has been rarely explored, especially at the molecular level. This study cloned two novel hydrolase genes (dcmA and dcmH) in Sphingopyxis sp. DBS4, and characterized them for diclofop-methyl degradation. DcmA, a member of the amidase superfamily, exhibits 26.1-45.9% identity with functional amidases. Conversely, DcmH corresponded to the DUF3089 domain-containing protein family (a family with unknown function), sharing no significant similarity with other biochemically characterized proteins. DcmA exhibited a broad spectrum of substrates, with preferential hydrolyzation of (R)-(+)-diclofop-methyl, (R)-(+)-quizalofop-ethyl, and (R)-(+)-haloxyfop-methyl. DcmH also preferred (R)-(+)-quizalofop-ethyl and (R)-(+)-haloxyfop-methyl degradation while displaying no apparent enantioselective activity towards diclofop-methyl. Using site-directed mutagenesis and molecular docking, it was determined that Ser175 was the fundamental residue influencing DcmA's activity against the two enantiomers of diclofop-methyl. For the degradation of AOPP herbicides, DcmA is an enantioselective amidase that has never been reported in research. This study provided novel hydrolyzing enzyme resources for the remediation of diclofop-methyl in the environment and deepened the understanding of enantioselective degradation of chiral AOPP herbicides mediated by microbes.


Assuntos
Éteres Difenil Halogenados , Herbicidas , Maleatos , Propionatos , Quinoxalinas , Herbicidas/metabolismo , Hidrolases , Simulação de Acoplamento Molecular , Estereoisomerismo , Produtos da Oxidação Avançada de Proteínas
12.
Basic Clin Pharmacol Toxicol ; 134(5): 629-642, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38501576

RESUMO

The effectiveness of natural killer (NK) cells transferred adoptively in combating solid tumours is limited by challenges such as their difficulty in penetrating tumours from the bloodstream and maintaining viability without the support of interleukin-2 (IL-2). Genetically modified NK-92MI cells, which can release IL-2 to sustain their viability, have been identified as a promising alternative. This adaptation addresses the negative consequences of systemic IL-2 administration. The role of PSD-95/discs large/ZO-1 (PDZ)-binding kinase (PBK) in cancer development is recognized, but its effects on immunity are not fully understood. This study explores how PBK expression influences the ability of NK-92MI cells to infiltrate ovarian tumours. Elevated levels of PBK expression have been found in various cancers, including ovarian cancer (OV), with analyses showing higher PBK mRNA levels in tumour tissues compared to normal ones. Immunohistochemistry has confirmed increased PBK expression in OV tissues. Investigations into PBK's role in immune regulation reveal its association with immune cell infiltration, indicating a potentially compromised immune environment in OV with high PBK expression. The small-molecule inhibitor HI-TOPK-032, which inhibits PBK, enhances the cytotoxicity of NK-92MI cells toward OV cells. It increases the production of interferon-γ and tumour necrosis factor-α, reduces apoptosis and encourages cell proliferation. Mechanistic studies showed that contact with OV cells treated with HI-TOPK-032 upregulates CD107a on NK-92 cells. In vivo studies demonstrated that HI-TOPK-032 improves the antitumour effects of NK-92MI cells in OVCAR3Luc xenografts, extending survival without significant side effects. Safety assessments in mice confirm HI-TOPK-032's favourable safety profile, highlighting its potential as a viable antitumour therapy. These results suggest that combining NK-92MI cells with HI-TOPK-032 enhances antitumour effectiveness against OV, indicating a promising, safe and effective treatment strategy that warrants further clinical investigation.


Assuntos
Indolizinas , Interleucina-2 , Neoplasias Ovarianas , Quinoxalinas , Humanos , Camundongos , Animais , Feminino , Apoptose , Linhagem Celular Tumoral , Neoplasias Ovarianas/tratamento farmacológico , MAP Quinases Reguladas por Sinal Extracelular , Células Matadoras Naturais
13.
Mikrochim Acta ; 191(4): 221, 2024 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-38536529

RESUMO

The successful fabrication is reported of highly crystalline Co nanoparticles interconnected with zeolitic imidazolate framework (ZIF-12) -based amorphous porous carbon using the molten-salt-assisted approach utilizing NaCl. Single crystal diffractometers (XRD), and X-ray photoelectron spectroscopy (XPS) analyses confirm the codoped amorphous carbon structure. Crystallite size was calculated by Scherrer (34 nm) and Williamson-Hall models (42 nm). The magnetic properties of NPCS (N-doped porous carbon sheet) were studied using a vibrating sample magnetometer (VSM). The NPCS has a magnetic saturation (Ms) value of 1.85 emu/g. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) analyses show that Co/Co3O4 nanoparticles are homogeneously distributed in the carbon matrix. While a low melting point eutectic salt acts as an ionic liquid solvent, ZIF-12, at high temperature, leading cobalt nanoparticles with a trace amount of Co3O4 interconnected by conductive amorphous carbon. In addition, the surface area (89.04 m2/g) and pore architectures of amorphous carbon embedded with Co nanoparticles are created using the molten salt approach. Thanks to this inexpensive and effective method, the optimal composite porous carbon structures were obtained with the strategy using NaCl salt and showed distinct electrochemical performance on electrochemical methodology revealing the analytical profile of Erdatifinib (ERD) as a sensor modifier. The linear response spanned from 0.01 to 7.38 µM, featuring a limit of detection (LOD) of 3.36 nM and a limit of quantification (LOQ) of 11.2 nM. The developed sensor was examined in terms of selectivity, repeatability, and reproducibility. The fabricated electrode was utilized for the quantification of Erdafitinib in urine samples and pharmaceutical dosage forms. This research provides a fresh outlook on the advancements in electrochemical sensor technology concerning the development and detection of anticancer drugs within the realms of medicine and pharmacology.


Assuntos
Antineoplásicos , Cobalto , Óxidos , Pirazóis , Quinoxalinas , Cloreto de Sódio , Reprodutibilidade dos Testes , Carbono/química
14.
Clin Pharmacol Ther ; 115(4): 825-838, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38339803

RESUMO

The integration of pharmacokinetic-pharmacodynamic (PK-PD) modeling and simulations with artificial intelligence/machine learning algorithms is one of the most attractive areas of the pharmacometric research. These hybrid techniques are currently under investigation to perform several tasks, among which precision dosing. In this scenario, this paper presents and evaluates a new framework embedding PK-PD models into a reinforcement learning (RL) algorithm, Q-learning (QL), to personalize pharmacological treatment. Each patient is represented with a set of PK-PD parameters and has a personal QL agent which optimizes the individual treatment. In the training phase, leveraging PK-PD simulations, the QL agent assesses different actions, defined consistently with the clinical knowledge to consider only plausible dose-adjustments, in order to find the optimal rules. The proposed framework was evaluated to optimize the erdafitinib treatment in patients with metastatic urothelial carcinoma. This drug was approved by the US Food and Drug Administration (FDA) with a dose-adaptive protocol based on monitoring the levels of serum phosphate, which represent a biomarker of both treatment efficacy and toxicity. To evaluate the flexibility of the methodology, a heterogeneous virtual population of 141 patients was generated using an erdafitinib population PK (PopPK)-PD literature model. For each patient, treatment response was simulated by using both QL-optimized protocol and the clinical one. QL agents outperform the approved dose-adaptive rules, increasing more than 10% the efficacy and the safety of treatment at each end point. Results confirm the great potentialities of the integration of PopPK-PD models and RL algorithms to optimize precision dosing tasks.


Assuntos
Carcinoma de Células de Transição , Pirazóis , Quinoxalinas , Neoplasias da Bexiga Urinária , Estados Unidos , Humanos , Inteligência Artificial
15.
J Med Chem ; 67(5): 3571-3589, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38385264

RESUMO

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.


Assuntos
Fibrinolíticos , Trombose , Animais , Fibrinolíticos/farmacologia , Fibrinolíticos/uso terapêutico , Macaca fascicularis , Quinoxalinas/farmacologia , Quinoxalinas/uso terapêutico , Receptores de Trombina , Trombina , Hemorragia , Trombose/tratamento farmacológico , Trombose/prevenção & controle , Receptor PAR-1 , Plaquetas , Agregação Plaquetária
16.
Int J Biol Macromol ; 263(Pt 1): 130175, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38360242

RESUMO

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.


Assuntos
Diabetes Mellitus , Hipoglicemiantes , Humanos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/química , Acarbose/farmacologia , Acarbose/química , alfa-Glucosidases/metabolismo , alfa-Amilases/química , Quinoxalinas/farmacologia , Antioxidantes/química , Estresse Oxidativo , Simulação de Acoplamento Molecular , Inibidores de Glicosídeo Hidrolases/química
17.
Food Chem ; 443: 138539, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38320375

RESUMO

Quinoxalines are a class of veterinary drugs with antibacterial and growth-promoting functions. They are often widely used to treat and prevent animal diseases and are illegally used as animal growth promoters to increase economic benefits. Quinoxalines could be easily metabolized in animals to various residue markers and remain in animal-derived foods, which would pose a serious threat to human health. Consequently, it is necessary to detect the residues of quinoxalines and their metabolites. This article reviewed and evaluated immunoassays for quinoxalines and their metabolites in animal-derived foods, mainly including enzyme-linked immunosorbent assays, fluorescence immunosorbent assays, immunochromatography, and surface plasmon resonance biosensors. In addition, we deeply explored the design of haptens for quinoxalines and their metabolites and analyzed the effect of haptens on antibody performance. This paper aims to provide guidance and references for their accurate and sensitive detection, thereby ensuring food safety and human public health.


Assuntos
Anticorpos , Quinoxalinas , Animais , Humanos , Quinoxalinas/análise , Ensaio de Imunoadsorção Enzimática/métodos , Imunoensaio , Haptenos/química
18.
Adv Ther ; 41(2): 744-758, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38169058

RESUMO

INTRODUCTION: In 2019, an 8-week regimen of glecaprevir/ pibrentasvir (GLE/PIB) was FDA-approved for treatment of chronic hepatitis C (HCV) in patients with cirrhosis. We used data from the Chronic Hepatitis Cohort Study (CHeCS) to evaluate treatment response and adverse events among patients with HCV and cirrhosis under routine clinical care. METHODS: Using an intention-to-treat (ITT)/modified ITT (mITT) approach, endpoints were (1) sustained virological response (SVR) at 12 weeks (SVR12) post-treatment; and (2) adverse events (AEs)/serious AEs during treatment. Patients with cirrhosis from two CHeCS sites were included if they were prescribed GLE/PIB from August 2017 to June 2020. Detailed treatment and clinical data were collected. Patient baseline characteristics were described with mean/standard deviation (std) for continuous variables, and proportions for categorical variables. Analyses were propensity score adjusted. The final model retained variables that were significant with p value < 0.05. RESULTS: The ITT sample included 166 patients, with 43, 116, and 7 patients in the 8-week, 12-week, and > 12-week planned treatment groups. Among them, 159 had confirmed SVR (95.8%, LCL 93.2%). The mITT analysis included 160 patients after excluding 6 with unknown HCV RNA results; 159 achieved SVR (99.4%, LCL 98.3%). There were no significant differences in rates of SVR between the 8-week and 12-week regimens in either analysis, nor any association with patient characteristics. SAEs were experienced by 1 patient (2%) in the 8-week group, 7 (5%) in the 12-week group (including one death), and 2 (29%) in the > 12-week group; 4 patients (from the 12-week group) experienced serious AEs or hepatic events that were "likely attributable" to GLE/PIB treatment. CONCLUSION: An 8-week regimen of GLE/PIB is well tolerated and highly effective among US patients with HCV and cirrhosis receiving routine clinical care.


Assuntos
Ácidos Aminoisobutíricos , Benzimidazóis , Ciclopropanos , Hepatite C Crônica , Lactamas Macrocíclicas , Leucina/análogos & derivados , Cirrose Hepática , Prolina/análogos & derivados , Sulfonamidas , Humanos , Estudos de Coortes , Resultado do Tratamento , Cirrose Hepática/complicações , Cirrose Hepática/tratamento farmacológico , Quinoxalinas/efeitos adversos , Pirrolidinas/uso terapêutico , Hepatite C Crônica/complicações , Hepatite C Crônica/tratamento farmacológico , Hepatite C Crônica/genética , Hepacivirus/genética , Antivirais/efeitos adversos , Genótipo
19.
Adv Sci (Weinh) ; 11(12): e2307870, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38233204

RESUMO

For tumor treatment, the ultimate goal in tumor therapy is to eliminate the primary tumor, manage potential metastases, and trigger an antitumor immune response, resulting in the complete clearance of all malignant cells. Tumor microenvironment (TME) refers to the local biological environment of solid tumors and has increasingly become an attractive target for cancer therapy. Neutrophils within TME of gastric cancer (GC) spontaneously undergo ferroptosis, and this process releases oxidized lipids that limit T cell activity. Enhanced photodynamic therapy (PDT) mediated by di-iodinated IR780 (Icy7) significantly increases the production of reactive oxygen species (ROS). Meanwhile, neutrophil ferroptosis can be triggered by increased ROS generation in the TME. In this study, a liposome encapsulating both ferroptosis inhibitor Liproxstatin-1 and modified photosensitizer Icy7, denoted LLI, significantly inhibits tumor growth of GC. LLI internalizes into MFC cells to generate ROS causing immunogenic cell death (ICD). Simultaneously, liposome-deliver Liproxstatin-1 effectively inhibits the ferroptosis of tumor neutrophils. LLI-based immunogenic PDT and neutrophil-targeting immunotherapy synergistically boost the anti-PD-1 treatment to elicit potent TME and systemic antitumor immune response with abscopal effects. In conclusion, LLI holds great potential for GC immunotherapy.


Assuntos
Ferroptose , Fotoquimioterapia , Quinoxalinas , Compostos de Espiro , Neoplasias Gástricas , Humanos , Neutrófilos , Lipossomos , Espécies Reativas de Oxigênio , Microambiente Tumoral
20.
Molecules ; 29(2)2024 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-38257377

RESUMO

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.


Assuntos
Neoplasias do Colo , Quinoxalinas , Humanos , Quinoxalinas/farmacologia , Proliferação de Células , Química Farmacêutica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...