Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Med Chem Lett ; 11(11): 2244-2252, 2020 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-33214836

RESUMO

The successful application of immunotherapy in the treatment of cancer relies on effective engagement of immune cells in the tumor microenvironment. Phosphoinositide 3-kinase γ (PI3Kγ) is highly expressed in tumor-associated macrophages, and its expression levels are associated with tumor immunosuppression and growth. Selective inhibition of PI3Kγ offers a promising strategy in immuno-oncology, which has led to the development of numerous potent PI3Kγ inhibitors with variable selectivity profiles. To facilitate further investigation of the therapeutic potential of PI3Kγ inhibition, we required a potent and PI3Kγ-selective tool compound with sufficient metabolic stability for use in future in vivo studies. Herein, we describe some of our efforts to realize this goal through the systematic study of SARs within a series of 7-azaindole-based PI3Kγ inhibitors. The large volume of data generated from this study helped guide our subsequent lead optimization efforts and will inform further development of PI3Kγ-selective inhibitors for use in immunomodulation.

2.
J Med Chem ; 63(19): 11235-11257, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-32865410

RESUMO

The selective inhibition of the lipid signaling enzyme PI3Kγ constitutes an opportunity to mediate immunosuppression and inflammation within the tumor microenvironment but is difficult to achieve due to the high sequence homology across the class I PI3K isoforms. Here, we describe the design of a novel series of potent PI3Kγ inhibitors that attain high isoform selectivity through the divergent projection of substituents into both the "selectivity" and "alkyl-induced" pockets within the adenosine triphosphate (ATP) binding site of PI3Kγ. These efforts have culminated in the discovery of 5-[2-amino-3-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-5-yl]-2-[(1S)-1-cyclopropylethyl]-7-(trifluoromethyl)-2,3-dihydro-1H-isoindol-1-one (4, IC50 = 0.064 µM, THP-1 cells), which displays >600-fold selectivity for PI3Kγ over the other class I isoforms and is a promising step toward the identification of a clinical development candidate. The structure-activity relationships identified throughout this campaign demonstrate that greater γ-selectivity can be achieved by inhibitors that occupy an "alkyl-induced" pocket and possess bicyclic hinge-binding motifs capable of forming more than one hydrogen bond to the hinge region of PI3Kγ.


Assuntos
Classe Ib de Fosfatidilinositol 3-Quinase/efeitos dos fármacos , Desenho de Fármacos , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Animais , Cristalografia por Raios X , Humanos , Simulação de Acoplamento Molecular , Inibidores de Fosfoinositídeo-3 Quinase/química , Inibidores de Fosfoinositídeo-3 Quinase/farmacocinética , Ratos , Relação Estrutura-Atividade
3.
J Med Chem ; 63(8): 3935-3955, 2020 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-32212732

RESUMO

CD73 is an extracellular mediator of purinergic signaling. When upregulated in the tumor microenvironment, CD73 has been implicated in the inhibition of immune function through overproduction of adenosine. Traditional efforts to inhibit CD73 have involved antibody therapy or the development of small molecules, the most potent of which mimic the acidic and ionizable structure of the enzyme's natural substrate, adenosine 5'-monophosphate (AMP). Here, we report the systematic discovery of a novel class of non-nucleotide CD73 inhibitors that are more potent than all other nonphosphonate inhibitor classes reported to date. These efforts have culminated in the discovery of 4-({5-[4-fluoro-1-(2H-indazol-6-yl)-1H-1,2,3-benzotriazol-6-yl]-1H-pyrazol-1-yl}methyl)benzonitrile (73, IC50 = 12 nM) and 4-({5-[4-chloro-1-(2H-indazol-6-yl)-1H-1,2,3-benzotriazol-6-yl]-1H-pyrazol-1-yl}methyl)benzonitrile (74, IC50 = 19 nM). Cocrystallization of 74 with human CD73 demonstrates a competitive binding mode. These compounds show promise for the improvement of drug-like character via the attenuation of the acidity and low membrane permeability inherent to known nucleoside inhibitors of CD73.


Assuntos
5'-Nucleotidase/antagonistas & inibidores , Descoberta de Drogas/métodos , Triazóis/química , Triazóis/farmacologia , 5'-Nucleotidase/metabolismo , Animais , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Cristalografia por Raios X/métodos , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos
4.
Sci Adv ; 5(6): eaax2650, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31223657

RESUMO

Dinoflagelates and cyanobacteria produce saxitoxin (STX), a lethal bis-guanidinium neurotoxin causing paralytic shellfish poisoning. A number of metazoans have soluble STX-binding proteins that may prevent STX intoxication. However, their STX molecular recognition mechanisms remain unknown. Here, we present structures of saxiphilin (Sxph), a bullfrog high-affinity STX-binding protein, alone and bound to STX. The structures reveal a novel high-affinity STX-binding site built from a "proto-pocket" on a transferrin scaffold that also bears thyroglobulin domain protease inhibitor repeats. Comparison of Sxph and voltage-gated sodium channel STX-binding sites reveals a convergent toxin recognition strategy comprising a largely rigid binding site where acidic side chains and a cation-π interaction engage STX. These studies reveal molecular rules for STX recognition, outline how a toxin-binding site can be built on a naïve scaffold, and open a path to developing protein sensors for environmental STX monitoring and new biologics for STX intoxication mitigation.


Assuntos
Proteínas de Transporte/metabolismo , Saxitoxina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Linhagem Celular , Cianobactérias/metabolismo , Humanos , Peptídeo Hidrolases/metabolismo , Inibidores de Proteases/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Rana catesbeiana , Células Sf9 , Canais de Sódio/metabolismo , Tireoglobulina/metabolismo , Transferrina/metabolismo
5.
Angew Chem Int Ed Engl ; 58(6): 1689-1693, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30488599

RESUMO

The bis-guanidinium toxins are a collection of natural products that display nanomolar potency against select isoforms of eukaryotic voltage-gated Na+ ion channels. We describe a synthetic strategy that enables access to four of these poisons, namely 11-saxitoxinethanoic acid, C13-acetoxy saxitoxin, decarbamoyl saxitoxin, and saxitoxin. Highlights of this work include an unusual Mislow-Evans rearrangement and a late-stage Stille ketene acetal coupling. The IC50 value of 11-saxitoxinethanoic acid was measured against rat NaV 1.4, and found to be 17.0 nm, similar to those of the sulfated toxins gonyautoxin II and III.


Assuntos
Produtos Biológicos/síntese química , Saxitoxina/análogos & derivados , Saxitoxina/síntese química , Produtos Biológicos/química , Estrutura Molecular , Saxitoxina/química , Estereoisomerismo
6.
Proc Natl Acad Sci U S A ; 115(21): E4900-E4909, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29669921

RESUMO

CLC proteins are a ubiquitously expressed family of chloride-selective ion channels and transporters. A dearth of pharmacological tools for modulating CLC gating and ion conduction limits investigations aimed at understanding CLC structure/function and physiology. Herein, we describe the design, synthesis, and evaluation of a collection of N-arylated benzimidazole derivatives (BIMs), one of which (BIM1) shows unparalleled (>20-fold) selectivity for CLC-Ka over CLC-Kb, the two most closely related human CLC homologs. Computational docking to a CLC-Ka homology model has identified a BIM1 binding site on the extracellular face of the protein near the chloride permeation pathway in a region previously identified as a binding site for other less selective inhibitors. Results from site-directed mutagenesis experiments are consistent with predictions of this docking model. The residue at position 68 is 1 of only ∼20 extracellular residues that differ between CLC-Ka and CLC-Kb. Mutation of this residue in CLC-Ka and CLC-Kb (N68D and D68N, respectively) reverses the preference of BIM1 for CLC-Ka over CLC-Kb, thus showing the critical role of residue 68 in establishing BIM1 selectivity. Molecular docking studies together with results from structure-activity relationship studies with 19 BIM derivatives give insight into the increased selectivity of BIM1 compared with other inhibitors and identify strategies for further developing this class of compounds.


Assuntos
Canais de Cloreto/antagonistas & inibidores , Cloretos/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Sítios de Ligação , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Eletrofisiologia , Humanos , Simulação de Acoplamento Molecular , Mutagênese Sítio-Dirigida , Mutação , Conformação Proteica , Relação Estrutura-Atividade , Xenopus laevis
7.
Science ; 354(6314): 865-869, 2016 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-27856903

RESUMO

The steroidal neurotoxin (-)-batrachotoxin functions as a potent agonist of voltage-gated sodium ion channels (NaVs). Here we report concise asymmetric syntheses of the natural (-) and non-natural (+) antipodes of batrachotoxin, as well both enantiomers of a C-20 benzoate-modified derivative. Electrophysiological characterization of these molecules against NaV subtypes establishes the non-natural toxin enantiomer as a reversible antagonist of channel function, markedly different in activity from (-)-batrachotoxin. Protein mutagenesis experiments implicate a shared binding side for the enantiomers in the inner pore cavity of NaV These findings motivate and enable subsequent studies aimed at revealing how small molecules that target the channel inner pore modulate NaV dynamics.


Assuntos
Batraquiotoxinas/síntese química , Batraquiotoxinas/farmacologia , Proteínas Musculares/antagonistas & inibidores , Bloqueadores do Canal de Sódio Disparado por Voltagem/síntese química , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Sítios de Ligação , Proteínas Musculares/química , Proteínas Musculares/genética , Mutação Puntual , Estrutura Secundária de Proteína , Ratos , Canais de Sódio/química , Canais de Sódio/genética , Bloqueadores do Canal de Sódio Disparado por Voltagem/química
8.
Proc Natl Acad Sci U S A ; 113(21): 5856-61, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27162340

RESUMO

Improper function of voltage-gated sodium channels (NaVs), obligatory membrane proteins for bioelectrical signaling, has been linked to a number of human pathologies. Small-molecule agents that target NaVs hold considerable promise for treatment of chronic disease. Absent a comprehensive understanding of channel structure, the challenge of designing selective agents to modulate the activity of NaV subtypes is formidable. We have endeavored to gain insight into the 3D architecture of the outer vestibule of NaV through a systematic structure-activity relationship (SAR) study involving the bis-guanidinium toxin saxitoxin (STX), modified saxitoxins, and protein mutagenesis. Mutant cycle analysis has led to the identification of an acetylated variant of STX with unprecedented, low-nanomolar affinity for human NaV1.7 (hNaV1.7), a channel subtype that has been implicated in pain perception. A revised toxin-receptor binding model is presented, which is consistent with the large body of SAR data that we have obtained. This new model is expected to facilitate subsequent efforts to design isoform-selective NaV inhibitors.


Assuntos
Proteínas Musculares/química , Canal de Sódio Disparado por Voltagem NAV1.2/química , Canal de Sódio Disparado por Voltagem NAV1.5/química , Canal de Sódio Disparado por Voltagem NAV1.7/química , Proteínas Recombinantes/química , Saxitoxina/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/química , Animais , Sítios de Ligação , Células CHO , Cricetulus , Desenho de Fármacos , Expressão Gênica , Células HEK293 , Humanos , Cinética , Simulação de Acoplamento Molecular , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutação , Canal de Sódio Disparado por Voltagem NAV1.2/genética , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Técnicas de Patch-Clamp , Ligação Proteica , Conformação Proteica , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saxitoxina/química , Bloqueadores dos Canais de Sódio/química , Canais de Sódio/genética , Canais de Sódio/metabolismo , Relação Estrutura-Atividade , Tetrodotoxina/química , Tetrodotoxina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...