Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Int Immunopharmacol ; 96: 107645, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33894488

RESUMO

Immunosuppression is one of the main mechanisms facilitating tumor expansion. It may be driven by immune checkpoint protein expression, anti-inflammatory cytokine secretion or enhanced metabolic enzyme production, leading to the subsequent build-up of metabolites such as adenosine. Under physiological conditions, adenosine prevents the development of tissue damage resulting from a prolonged immune response; the same mechanism might be employed by tumor tissue to promote immunosuppression. Immune cells expressing A2A and A2B adenosine receptors present in an adenosine-rich environment have suppressed effector functions, such as cytotoxicity, proinflammatory cytokine release, antigen presentation and others, making them inert to cancer cells. This study was designed to investigate the dual antagonist potential of SEL330-639 to abolish adenosine-driven immunosuppression. SEL330-639 has slow dissociation kinetics. It inhibits cAMP production in human CD4+ cells, CD8+ cells and moDCs, which leads to diminished CREB phosphorylation and restoration of antitumor cytokine production (IL-2, TNFα, IL-12) in multiple primary human immune cells. The aforementioned results were additionally validated by gene expression analysis and functional assays in which NK cell line cytotoxicity was recovered by SEL330-639. Adenosine-driven immunosuppression is believed to preclude the effectiveness of immune checkpoint inhibitor therapies. Hence, there is an urgent need to develop new immuno-oncological strategies. Here, we comprehensively characterize SEL330-639, a novel dual A2A/A2B receptor antagonist effective in both lymphoid and myeloid cell populations with nanomolar potency. Due to its tight binding to the A2A and A2B receptors, this binding is sustained even at high adenosine concentrations mimicking the upper limit of the range of adenosine levels observed in the tumor microenvironment.


Assuntos
Antagonistas do Receptor A2 de Adenosina/farmacologia , Adenosina/imunologia , Terapia de Imunossupressão/métodos , Animais , Linhagem Celular , AMP Cíclico/antagonistas & inibidores , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Citocinas/metabolismo , Células Dendríticas/metabolismo , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Cinética , Fosforilação/efeitos dos fármacos , Ratos , Receptor A2A de Adenosina/efeitos dos fármacos , Receptor A2A de Adenosina/genética , Receptor A2A de Adenosina/imunologia , Receptor A2B de Adenosina/efeitos dos fármacos , Receptor A2B de Adenosina/genética , Receptor A2B de Adenosina/imunologia , Linfócitos T/metabolismo
2.
Bioorg Med Chem Lett ; 29(4): 646-653, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30626557

RESUMO

In oncology, the "Warburg effect" describes the elevated production of energy by glycolysis in cancer cells. The ubiquitous and hypoxia-induced 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) plays a noteworthy role in the regulation of glycolysis by producing fructose-2,6-biphosphate (F-2,6-BP), a potent activator of the glycolysis rate-limiting phosphofructokinase PFK-1. Series of amides and sulfonamides derivatives based on a N-aryl 6-aminoquinoxaline scaffold were synthesized and tested for their inhibition of PFKFB3 in vitro in a biochemical assay as well as in HCT116 cells. The carboxamide series displayed satisfactory kinetic solubility and metabolic stability, and within this class, potent lead compounds with low nanomolar activity have been identified with a suitable profile for further in vivo evaluation.


Assuntos
Amidas/química , Fosfofrutoquinase-2/antagonistas & inibidores , Quinoxalinas/química , Quinoxalinas/farmacologia , Sulfonamidas/química , Células HCT116 , Humanos , Cinética , Solubilidade
3.
Bioorg Med Chem Lett ; 29(4): 607-613, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30626559

RESUMO

Maternal embryonic leucine zipper kinase (MELK) is involved in several key cellular processes and displays increased levels of expression in numerous cancer classes (colon, breast, brain, ovary, prostate and lung). Although no selective MELK inhibitors have yet been approved, increasing evidence suggest that inhibition of MELK would constitute a promising approach for cancer therapy. A weak high-throughput screening hit (17, IC50 ≈ 5 µM) with lead-like properties was optimized for MELK inhibition. The early identification of a plausible binding mode by molecular modeling offered guidance in the choice of modifications towards compound 52 which displayed a 98 nM IC50. A good selectivity profile was achieved for a representative member of the series (29) in a 486 protein kinase panel. Future elaboration of 52 has the potential to deliver compounds for further development with chemotherapeutic aims.


Assuntos
Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Tiofenos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Ensaios de Triagem em Larga Escala , Humanos , Concentração Inibidora 50
4.
ChemMedChem ; 14(1): 169-181, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30378281

RESUMO

Energy and biomass production in cancer cells are largely supported by aerobic glycolysis in what is called the Warburg effect. The process is regulated by key enzymes, among which phosphofructokinase PFK-2 plays a significant role by producing fructose-2,6-biphosphate; the most potent activator of the glycolysis rate-limiting step performed by phosphofructokinase PFK-1. Herein, the synthesis, biological evaluation and structure-activity relationship of novel inhibitors of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), which is the ubiquitous and hypoxia-induced isoform of PFK-2, are reported. X-ray crystallography and docking were instrumental in the design and optimisation of a series of N-aryl 6-aminoquinoxalines. The most potent representative, N-(4-methanesulfonylpyridin-3-yl)-8-(3-methyl-1-benzothiophen-5-yl)quinoxalin-6-amine, displayed an IC50 of 14 nm for the target and an IC50 of 0.49 µm for fructose-2,6-biphosphate production in human colon carcinoma HCT116 cells. This work provides a new entry in the field of PFKFB3 inhibitors with potential for development in oncology.


Assuntos
Descoberta de Drogas , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Fosfofrutoquinase-2/antagonistas & inibidores , Quinoxalinas/química , Quinoxalinas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Células HCT116 , Humanos , Ácido Láctico/antagonistas & inibidores , Ácido Láctico/biossíntese , Modelos Moleculares , Estrutura Molecular , Fosfofrutoquinase-2/metabolismo , Quinoxalinas/síntese química , Relação Estrutura-Atividade
5.
Oncotarget ; 8(20): 33779-33795, 2017 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-28422713

RESUMO

Inhibition of oncogenic transcriptional programs is a promising therapeutic strategy. A substituted tricyclic benzimidazole, SEL120-34A, is a novel inhibitor of Cyclin-dependent kinase 8 (CDK8), which regulates transcription by associating with the Mediator complex. X-ray crystallography has shown SEL120-34A to be a type I inhibitor forming halogen bonds with the protein's hinge region and hydrophobic complementarities within its front pocket. SEL120-34A inhibits phosphorylation of STAT1 S727 and STAT5 S726 in cancer cells in vitro. Consistently, regulation of STATs- and NUP98-HOXA9- dependent transcription has been observed as a dominant mechanism of action in vivo. Treatment with the compound resulted in a differential efficacy on AML cells with elevated STAT5 S726 levels and stem cell characteristics. In contrast, resistant cells were negative for activated STAT5 and revealed lineage commitment. In vivo efficacy in xenotransplanted AML models correlated with significant repression of STAT5 S726. Favorable pharmacokinetics, confirmed safety and in vivo efficacy provide a rationale for the further clinical development of SEL120-34A as a personalized therapeutic approach in AML.


Assuntos
Antineoplásicos/farmacologia , Quinase 8 Dependente de Ciclina/antagonistas & inibidores , Leucemia Mieloide Aguda/metabolismo , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Quinase 8 Dependente de Ciclina/química , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/genética , Camundongos , Modelos Moleculares , Conformação Molecular , Fosforilação/efeitos dos fármacos , Ligação Proteica , Inibidores de Proteínas Quinases/química , Fator de Transcrição STAT1/química , Fator de Transcrição STAT5/química , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Biochim Biophys Acta ; 1854(10 Pt B): 1617-29, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26006748

RESUMO

Cyclin-dependent kinase (CDK) inhibitors have been developed as potential anticancer therapeutics and several nonselective compounds are currently in advanced clinical trials. This review is focused on the key biological roles of CDK8 kinase, which provide a proof-of-principle for continued efforts toward effective cancer treatment, targeting activity of this CDK family member. Among currently identified kinase inhibitors, several displayed significant selectivity for CDK8 and notably the effectiveness in targeting cancer specific gene expression programs. Structural features of CDK8 and available ligands were discussed from a perspective of the rational drug design process. Current state of the art confirms that further development of CDK8 inhibitors will translate into targeted therapies in oncology. This article is part of a Special Issue entitled:Inhibitors of Protein Kinases.


Assuntos
Quinase 8 Dependente de Ciclina/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Transcrição Gênica/efeitos dos fármacos , Quinase 8 Dependente de Ciclina/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Estrutura Molecular , Neoplasias/genética , Niacinamida/análogos & derivados , Niacinamida/química , Niacinamida/uso terapêutico , Compostos de Fenilureia/química , Compostos de Fenilureia/uso terapêutico , Conformação Proteica , Inibidores de Proteínas Quinases/química , Transdução de Sinais/efeitos dos fármacos , Sorafenibe
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA