Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Nature ; 557(7704): 228-232, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29686415

RESUMO

Most drugs are developed through iterative rounds of chemical synthesis and biochemical testing to optimize the affinity of a particular compound for a protein target of therapeutic interest. This process is challenging because candidate molecules must be selected from a chemical space of more than 1060 drug-like possibilities 1 , and a single reaction used to synthesize each molecule has more than 107 plausible permutations of catalysts, ligands, additives and other parameters 2 . The merger of a method for high-throughput chemical synthesis with a biochemical assay would facilitate the exploration of this enormous search space and streamline the hunt for new drugs and chemical probes. Miniaturized high-throughput chemical synthesis3-7 has enabled rapid evaluation of reaction space, but so far the merger of such syntheses with bioassays has been achieved with only low-density reaction arrays, which analyse only a handful of analogues prepared under a single reaction condition8-13. High-density chemical synthesis approaches that have been coupled to bioassays, including on-bead 14 , on-surface 15 , on-DNA 16 and mass-encoding technologies 17 , greatly reduce material requirements, but they require the covalent linkage of substrates to a potentially reactive support, must be performed under high dilution and must operate in a mixture format. These reaction attributes limit the application of transition-metal catalysts, which are easily poisoned by the many functional groups present in a complex mixture, and of transformations for which the kinetics require a high concentration of reactant. Here we couple high-throughput nanomole-scale synthesis with a label-free affinity-selection mass spectrometry bioassay. Each reaction is performed at a 0.1-molar concentration in a discrete well to enable transition-metal catalysis while consuming less than 0.05 milligrams of substrate per reaction. The affinity-selection mass spectrometry bioassay is then used to rank the affinity of the reaction products to target proteins, removing the need for time-intensive reaction purification. This method enables the primary synthesis and testing steps that are critical to the invention of protein inhibitors to be performed rapidly and with minimal consumption of starting materials.


Assuntos
Nanotecnologia/métodos , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/síntese química , Proteínas/química , Bioensaio , Catálise , Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Quinase 1 do Ponto de Checagem/química , Avaliação Pré-Clínica de Medicamentos , Cinética , Ligantes , Espectrometria de Massas , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/química , Proteínas/antagonistas & inibidores , Especificidade por Substrato
2.
ACS Chem Biol ; 17(9): 2595-2604, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-36044633

RESUMO

Although current antiretroviral therapy can control HIV-1 replication and prevent disease progression, it is not curative. Identifying mechanisms that can lead to eradication of persistent viral reservoirs in people living with HIV-1 (PLWH) remains an outstanding challenge to achieving cure. Utilizing a phenotypic screen, we identified a novel chemical class capable of killing HIV-1 infected peripheral blood mononuclear cells. Tool compounds ICeD-1 and ICeD-2 ("inducer of cell death-1 and 2"), optimized for potency and selectivity from screening hits, were used to deconvolute the mechanism of action using a combination of chemoproteomic, biochemical, pharmacological, and genetic approaches. We determined that these compounds function by modulating dipeptidyl peptidase 9 (DPP9) and activating the caspase recruitment domain family member 8 (CARD8) inflammasome. Efficacy of ICeD-1 and ICeD-2 was dependent on HIV-1 protease activity and synergistic with efavirenz, which promotes premature activation of HIV-1 protease at high concentrations in infected cells. This in vitro synergy lowers the efficacious cell kill concentration of efavirenz to a clinically relevant dose at concentrations of ICeD-1 or ICeD-2 that do not result in complete DPP9 inhibition. These results suggest engagement of the pyroptotic pathway as a potential approach to eliminate HIV-1 infected cells.


Assuntos
Infecções por HIV , HIV-1 , Alcinos , Benzoxazinas , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Ciclopropanos , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Infecções por HIV/tratamento farmacológico , HIV-1/metabolismo , Humanos , Inflamassomos/metabolismo , Leucócitos Mononucleares , Proteínas de Neoplasias/metabolismo
3.
Mol Cancer Ther ; 19(6): 1298-1307, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32229606

RESUMO

The programmed cell death 1 (PD-1) pathway represents a major immune checkpoint, which may be engaged by cells in the tumor microenvironment to overcome active T-cell immune surveillance. Pembrolizumab (Keytruda®, MK-3475) is a potent and highly selective humanized mAb of the IgG4/kappa isotype designed to directly block the interaction between PD-1 and its ligands, PD-L1 and PD-L2. This blockade enhances the functional activity of T cells to facilitate tumor regression and ultimately immune rejection. Pembrolizumab binds to human and cynomolgus monkey PD-1 with picomolar affinity and blocks the binding of human and cynomolgus monkey PD-1 to PD-L1 and PD-L2 with comparable potency. Pembrolizumab binds both the C'D and FG loops of PD-1. Pembrolizumab overcomes human and cynomolgus monkey PD-L1-mediated immune suppression in T-cell cultures by enhancing IL2 production following staphylococcal enterotoxin B stimulation of healthy donor and cancer patient cells, and IFNγ production in human primary tumor histoculture. Ex vivo and in vitro studies with human and primate T cells show that pembrolizumab enhances antigen-specific T-cell IFNγ and IL2 production. Pembrolizumab does not mediate FcR or complement-driven effector function against PD-1-expressing cells. Pembrolizumab displays dose-dependent clearance and half-life in cynomolgus monkey pharmacokinetic and toxicokinetic studies typical for human IgG4 antibodies. In nonhuman primate toxicology studies, no findings of toxicologic significance were observed. The preclinical data for pembrolizumab are consistent with the clinical anticancer activity and safety that has been demonstrated in human clinical trials.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/farmacocinética , Leucócitos Mononucleares/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Linfócitos T/efeitos dos fármacos , Animais , Antineoplásicos Imunológicos/farmacocinética , Antineoplásicos Imunológicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Antígeno B7-H1/metabolismo , Feminino , Humanos , Inibidores de Checkpoint Imunológico/farmacocinética , Inibidores de Checkpoint Imunológico/farmacologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/patologia , Macaca fascicularis , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/imunologia , Neoplasias/patologia , Proteína 2 Ligante de Morte Celular Programada 1/antagonistas & inibidores , Proteína 2 Ligante de Morte Celular Programada 1/imunologia , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia , Distribuição Tecidual , Testes de Toxicidade
4.
J Mass Spectrom ; 41(1): 43-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16317713

RESUMO

In the work reported here, a novel interface, the nanosplitter, is incorporated into the drug metabolism laboratory in order to enhance the analytical capabilities of detecting and identifying drug-related metabolites to support drug metabolism studies during the drug development process. When an existing LC-MS-radiometric detector (RD) system is coupled with this nanosplitter, the system becomes capable of performing dynamic microspray under a typical analytical LC method. With the superior MS sensitivity offered by this system, most of the analytical LC methods developed for metabolite profiling can then be easily adopted for metabolite identification work. The improvement of these analytical capabilities can streamline the entire process of the drug metabolism study. In the experiments presented here, the nanosplitter interface coupled with analytical HPLC systems (e.g. 4.6 x 250 mm column @ 1 ml/min) demonstrated significant increases in MS signal (2x to 40x peak area) when compared to the standard LC-MS interface for both in vitro and in vivo metabolism studies. Furthermore, this signal gain facilitated the MS detection of additional metabolites (observed in the radiometric trace) that were below the MS level of detection when using the standard interface.


Assuntos
Cromatografia Líquida de Alta Pressão/instrumentação , Espectrometria de Massas/instrumentação , Preparações Farmacêuticas/metabolismo , Animais , Quinases da Glicogênio Sintase/química , Quinases da Glicogênio Sintase/metabolismo , Haplorrinos , Hepatócitos/química , Nanotecnologia , Preparações Farmacêuticas/química , Preparações Farmacêuticas/urina , Ratos , Padrões de Referência
5.
J Biomol Screen ; 21(6): 608-19, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26969322

RESUMO

The primary objective of early drug discovery is to associate druggable target space with a desired phenotype. The inability to efficiently associate these often leads to failure early in the drug discovery process. In this proof-of-concept study, the most tractable starting points for drug discovery within the NF-κB pathway model system were identified by integrating affinity selection-mass spectrometry (AS-MS) with functional cellular assays. The AS-MS platform Automated Ligand Identification System (ALIS) was used to rapidly screen 15 NF-κB proteins in parallel against large-compound libraries. ALIS identified 382 target-selective compounds binding to 14 of the 15 proteins. Without any chemical optimization, 22 of the 382 target-selective compounds exhibited a cellular phenotype consistent with the respective target associated in ALIS. Further studies on structurally related compounds distinguished two chemical series that exhibited a preliminary structure-activity relationship and confirmed target-driven cellular activity to NF-κB1/p105 and TRAF5, respectively. These two series represent new drug discovery opportunities for chemical optimization. The results described herein demonstrate the power of combining ALIS with cell functional assays in a high-throughput, target-based approach to determine the most tractable drug discovery opportunities within a pathway.


Assuntos
Descoberta de Drogas , Ensaios de Triagem em Larga Escala/métodos , NF-kappa B/antagonistas & inibidores , Relação Estrutura-Atividade , Ligantes , Espectrometria de Massas/métodos , NF-kappa B/química , Ligação Proteica , Transdução de Sinais/efeitos dos fármacos , Fator 5 Associado a Receptor de TNF/antagonistas & inibidores , Fator 5 Associado a Receptor de TNF/química , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/química
6.
J Chromatogr A ; 1053(1-2): 151-9, 2004 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-15543981

RESUMO

Several pairs of analytes in plasma were investigated to demonstrate the successful utility of a novel interface in quantitative bioanalytical LC-MS and LC-MS/MS. Recently in our laboratory, an interface (the nanosplitter) was developed that allows the coupling of normal-bore liquid chromatography with microelectrospray mass spectrometry. The post-column concentric split minimizes turbulence and is shown to produce significant gains in the mass spectrometric signal. This configuration of the splitter allows sampling of the center portion of the parabolic HPLC plug, which maintains chromatographic integrity while producing high split ratios and effectively conserving nearly 99.9% of the sample. When utilizing a Finnigan mass spectrometer (with a heated capillary interface design), the signal gain with the nanosplitter ranged from 5 to 16 times the peak area obtained using the conventional interface without splitting. The linearity of the nanosplitter and conventional interface are shown to be comparable for all analytes tested. The nanosplitter was also fitted to a Sciex mass spectrometer and the results were compared to those from turbo ionspray. While in this case no significant signal improvement was observed, when normalized to the actual analyte mass introduced into the MS, the mass sensitivity was still increased 270-fold. The variations in signal gain utilizing the nanosplitter on instruments from different manufacturers reflect the inherent differences in the source designs while confirming the benefits of coupling high flow LC separations with low flow mass spectrometric detection.


Assuntos
Cromatografia Líquida/métodos , Espectrometria de Massas por Ionização por Electrospray/métodos , Quinases da Glicogênio Sintase/isolamento & purificação , Preparações Farmacêuticas/isolamento & purificação
7.
J Proteome Res ; 6(6): 2269-76, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17472361

RESUMO

Advances in analytical techniques, specifically in mass spectrometry, have allowed for both facile protein identification and routine sequencing of proteins at increased sensitivity levels. Protein modifications present additional challenges because they occur at low stoichiometries and often change the analytical behavior of the molecule. For example, characterization of protein phosphorylation provides crucial information to signaling processes that are often associated with disease. Research into protein phosphorylation requires inter-disciplinary co-operation involving multiple investigators with expertise in diverse scientific fields. As such, techniques must be simple, effective, and incorporate multiple checkpoints that confirm the sample contains a phosphorylated protein in order to ensure resources are conserved. In this study, tumor progression locus 2 (Tpl2), which has been implicated in cell cycle regulation and has been shown to play a significant role in critical signal transduction pathways, was transfected into 293T cells, overexpressed and isolated from the cell lysate. Isolated proteins were separated via 1D gel electrophoresis, and their phosphorylation was confirmed using phosphospecific staining. The bands were excised and subjected to tryptic digestion and immobilized metal affinity chromatography (IMAC) prior to analysis by capillary-LC-MS/MS. Three phosphorylation sites were detected on Tpl2. One site had previously been reported in the literature but had not been characterized by mass spectrometric methods until this time; two additional novel sites of phosphorylation were detected.


Assuntos
Cromatografia Líquida/métodos , Imidazóis/química , MAP Quinase Quinase Quinases/química , Espectrometria de Massas/métodos , Fosfopeptídeos/análise , Proteínas Proto-Oncogênicas/química , Sequência de Aminoácidos , Humanos , Dados de Sequência Molecular , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA