Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Tissue Eng Part A ; 30(1-2): 75-83, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37772690

RESUMO

Abdominal aortic aneurysm (AAA) is a critical, multifactorial cardiovascular disorder marked by localized dilatation of the abdominal aorta. A major challenge to countering the pathophysiology of AAAs lies in the naturally irreversible breakdown of elastic fibers in the aorta wall, which is linked to the poor elastogenicity of adult and diseased vascular smooth muscle cells (SMCs) and their impaired ability to assemble mature elastic fibers in a chronic proteolytic tissue milieu. We have previously shown that these are downstream effects of neutrophil elastase-induced activation of the epidermal growth factor receptor (EGFR) activity in aneurysmal SMCs. The novelty of this study lies in investigating the benefits of an EGFR inhibitor drug, afatinib (used to treat nonsmall cell lung cancer), for proelastogenic and antiproteolytic stimulation of aneurysmal SMCs. In in vitro cell cultures, we have shown that safe doses of 0.5 and 1 nM afatinib inhibit EGFR and p-extracellular signal-regulated kinases 1/2 protein expression by 50-70% and downstream elastolytic matrix metalloprotease 2 (MMP2) versus untreated control cultures. In addition, elastin production on a per cell basis was significantly upregulated by afatinib doses within the 0.1-1 nM dose range, which was further validated through transmission electron microscopy showing significantly increased presence of tropoelastin coacervates and maturing elastic fibers upon afatinib treatment at the above doses. Therefore, our studies for the first time demonstrate the therapeutic benefits of afatinib toward use for elastic matrix repair in small AAAs.


Assuntos
Aneurisma Aórtico , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Ratos , Animais , Humanos , Afatinib/farmacologia , Afatinib/metabolismo , Ratos Sprague-Dawley , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Aneurisma Aórtico/metabolismo , Elastina/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/farmacologia , Miócitos de Músculo Liso
2.
J Mol Model ; 27(10): 309, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34599372

RESUMO

Chronic myeloid leukemia (CML) is a pathological condition associated with the uncontrolled proliferation of white blood cells and respective loss of function. Imatinib was the first drug that could effectively treat this condition, but its use is hindered by the development of mutations of the BCR-ABL protein, which are the cause of resistance. Therefore, dasatinib and afatinib present similarities that can be explored to discover new molecules capable of overcoming the effects of imatinib. Afatinib exhibited electronic and docking behavior, indicating that a replacement with some minor modifications could design a new potential inhibitor. The amide group in each candidate is clearly of pharmacophoric importance, and it needs to concentrate a negative region. Sulfur group presents a good pharmacophoric profile, which was shown by dasatinib results, adding to the influence of the Met318 residue in the target protein active site configuration. This behavior suggests that the sulfur atom and other fragments that have an affinity for the methionine sidechain may provide a significant positive effect when present in TKI molecules such as afatinib or dasatinib.


Assuntos
Afatinib/química , Dasatinibe/química , Proteínas de Fusão bcr-abl/química , Afatinib/metabolismo , Afatinib/farmacologia , Antineoplásicos/química , Antineoplásicos/metabolismo , Domínio Catalítico , Dasatinibe/metabolismo , Dasatinibe/farmacologia , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Humanos , Mesilato de Imatinib/química , Mesilato de Imatinib/metabolismo , Mesilato de Imatinib/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Metionina/química , Simulação de Acoplamento Molecular , Mutação , Teoria Quântica , Enxofre/química
3.
ACS Chem Biol ; 16(9): 1644-1653, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34397208

RESUMO

Covalent inhibition is a powerful strategy to develop potent and selective small molecule kinase inhibitors. Targeting the conserved catalytic lysine is an attractive method for selective kinase inactivation. We have developed novel, selective inhibitors of phosphoinositide 3-kinase δ (PI3Kδ) which acylate the catalytic lysine, Lys779, using activated esters as the reactive electrophiles. The acylating agents were prepared by adding the activated ester motif to a known selective dihydroisobenzofuran PI3Kδ inhibitor. Three esters were designed, including an acetate ester which was the smallest lysine modification evaluated in this work. Covalent binding to the enzyme was characterized by intact protein mass spectrometry of the PI3Kδ-ester adducts. An enzymatic digest coupled with tandem mass spectrometry identified Lys779 as the covalent binding site, and a biochemical activity assay confirmed that PI3Kδ inhibition was a direct result of covalent lysine acylation. These results indicate that a simple chemical modification such as lysine acetylation is sufficient to inhibit kinase activity. The selectivity of the compounds was evaluated against lipid kinases in cell lysates using a chemoproteomic binding assay. Due to the conserved nature of the catalytic lysine across the kinome, we believe the covalent inhibition strategy presented here could be applicable to a broad range of clinically relevant targets.


Assuntos
Acrilamidas/química , Adenina/análogos & derivados , Afatinib/química , Compostos de Anilina/química , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Lisina/química , Inibidores de Fosfoinositídeo-3 Quinase/química , Piperidinas/química , Acetilação , Acrilamidas/metabolismo , Adenina/química , Adenina/metabolismo , Afatinib/metabolismo , Sequência de Aminoácidos , Compostos de Anilina/metabolismo , Catálise , Domínio Catalítico , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Humanos , Espectrometria de Massas , Simulação de Acoplamento Molecular , Inibidores de Fosfoinositídeo-3 Quinase/metabolismo , Piperidinas/metabolismo , Ligação Proteica , Conformação Proteica , Especificidade por Substrato
4.
J Med Chem ; 64(10): 6569-6580, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-33719426

RESUMO

KRAS, the most common oncogenic driver in human cancers, is controlled and signals primarily through protein-protein interactions (PPIs). The interaction between KRAS and SOS1, crucial for the activation of KRAS, is a typical, challenging PPI with a large contact surface area and high affinity. Here, we report that the addition of only one atom placed between Y884SOS1 and A73KRAS is sufficient to convert SOS1 activators into SOS1 inhibitors. We also disclose the discovery of BI-3406. Combination with the upstream EGFR inhibitor afatinib shows in vivo efficacy against KRASG13D mutant colorectal tumor cells, demonstrating the utility of BI-3406 to probe SOS1 biology. These findings challenge the dogma that large molecules are required to disrupt challenging PPIs. Instead, a "foot in the door" approach, whereby single atoms or small functional groups placed between key PPI interactions, can lead to potent inhibitors even for challenging PPIs such as SOS1-KRAS.


Assuntos
Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteína SOS1/metabolismo , Afatinib/química , Afatinib/metabolismo , Afatinib/uso terapêutico , Regulação Alostérica/efeitos dos fármacos , Sítios de Ligação , Domínio Catalítico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Humanos , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Mapas de Interação de Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/genética , Quinazolinas/química , Quinazolinas/metabolismo , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico , Proteína SOS1/agonistas , Proteína SOS1/antagonistas & inibidores , Proteína SOS1/genética
5.
J Mater Chem B ; 8(35): 7931-7940, 2020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32779670

RESUMO

Molecular targeted-photodynamic combinational therapy is a promising strategy to enhance antitumor effects; meanwhile, current nanocarriers face challenges of limited selective delivery and release of therapeutic agents to specific tumor sites, which significantly compromises their therapeutic efficacy. Herein, we report active-targeting, enzyme- and ROS-dual responsive nanoparticles (HPGBCA) consisting of CD44-targeting hyaluronic acid (HA) shells and afatinib (AFT)-loaded, ROS-sensitive poly(l-lysine)-conjugated chlorin e6 (Ce6) derivative nanoparticle cores (PGBCA). HPGBCA can actively carry AFT and Ce6 specifically to tumor cells due to the negatively charged HA and CD44-mediated active targeting. Subsequently, hyaluronidase in the endosome will further spur the degradation of the HA shell to prompt exposure of the positively charged PGBCA core for rapid endosomal escape and intracellular delivery of AFT and Ce6. Furthermore, the generation of ROS produced by Ce6 under NIR irradiation can trigger the rapid oxidation of the thioether linker to facilitate the release of AFT into the cytoplasm. In vitro and in vivo studies demonstrated that the released AFT and excessive ROS at the local site can synergistically induce cell apoptosis to enhance the therapeutic efficacy without side effects. Our developed intelligent nanoparticle provides new avenues to achieve on-demand, specific intracellular drug release for improved molecular targeted-photodynamic combination therapeutic efficacy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Liberação Controlada de Fármacos , Receptores ErbB/antagonistas & inibidores , Espaço Intracelular/metabolismo , Neoplasias Pulmonares/patologia , Nanopartículas/química , Inibidores de Proteínas Quinases/metabolismo , Afatinib/química , Afatinib/metabolismo , Afatinib/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Clorofilídeos , Portadores de Fármacos/química , Humanos , Ácido Hialurônico/química , Luz , Neoplasias Pulmonares/tratamento farmacológico , Terapia de Alvo Molecular , Fotoquimioterapia , Porfirinas/química , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Espécies Reativas de Oxigênio/metabolismo
6.
J Chem Inf Model ; 59(1): 309-315, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30481018

RESUMO

The anticancer drug afatinib has been found to be more effective at inhibiting the oncogenic EGFR mutant exon 19 deletion (19del) over the oncogenic EGFR mutant L858R. The underlying mechanism has been hypothesized to result from differences in structural constraints introduced by the mutations and stabilizing interactions afforded by a buried water molecule in 19del (Kannan, S.; et al. Sci. Rep. 2017, 7, 1540). The COSMIC cancer database is mined for EGFR sequences to discover that several mutations in the form of Single nucleotide polymorphisms (SNPs) line this hydration cavity. In this work, the effects of these SNPs on the affinity of afatinib for EGFRWT and oncogenic mutants EGFRL858R and EGFR19del were studied using free energy perturbation and thermodynamic Integration calculations. The simulations reveal that several SNPs have significant effects on the affinity of afatinib for the mutant EGFRs carrying the SNPs and may thus have clinical implications relating to emergence of resistance to afatinib, thus potentially impacting the choice of EGFR inhibitors in the clinic.


Assuntos
Afatinib/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Simulação de Dinâmica Molecular , Polimorfismo de Nucleotídeo Único , Afatinib/farmacologia , Mineração de Dados , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/química , Humanos , Mutação , Ligação Proteica/genética , Conformação Proteica , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...