Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 65(7): 5300-5316, 2022 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-35302767

RESUMO

Bruton's tyrosine kinase (BTK), a Tec family tyrosine kinase, is critical in immune pathways as an essential intracellular signaling element, participating in both adaptive and immune responses. Currently approved BTK inhibitors are irreversible covalent inhibitors and limited to oncology indications. Herein, we describe the design of covalent reversible BTK inhibitors and the discoveries of PRN473 (11) and rilzabrutinib (PRN1008, 12). These compounds have exhibited potent and durable inhibition of BTK, in vivo efficacy in rodent arthritis models, and clinical efficacy in canine pemphigus foliaceus. Compound 11 has completed phase 1 trials as a topical agent, and 12 is in phase 3 trials for pemphigus vulgaris and immune thrombocytopenia.


Assuntos
Inibidores de Proteínas Quinases , Transdução de Sinais , Tirosina Quinase da Agamaglobulinemia , Animais , Cães , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
2.
Mol Cancer Ther ; 16(12): 2668-2676, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28978721

RESUMO

An increasing number of cancers are known to harbor mutations, translocations, or amplifications in the fibroblast growth factor receptor (FGFR) family of kinases. The FGFR inhibitors evaluated in clinical trials to date have shown promise at treating these cancers. Here, we describe PRN1371, an irreversible covalent inhibitor of FGFR1-4 targeting a cysteine within the kinase active site. PRN1371 demonstrated strong FGFR potency and excellent kinome-wide selectivity in a number of biochemical and cellular assays, including in various cancer cell lines exhibiting FGFR alterations. Furthermore, PRN1371 maintained FGFR inhibition in vivo, not only when circulating drug levels were high but also after the drug had been cleared from circulation, indicating the possibility of sustained FGFR inhibition in the clinic without the need for continuous drug exposure. Durable tumor regression was also obtained in multiple tumor xenografts and patient-derived tumor xenograft models and was sustained even using an intermittent dosing strategy that provided drug holidays. PRN1371 is currently under clinical investigation for treatment of patients with solid tumors. Mol Cancer Ther; 16(12); 2668-76. ©2017 AACR.


Assuntos
Piridonas/uso terapêutico , Pirimidinas/uso terapêutico , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Camundongos , Piridonas/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Med Chem ; 60(15): 6516-6527, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28665128

RESUMO

Aberrant signaling of the FGF/FGFR pathway occurs frequently in cancers and is an oncogenic driver in many solid tumors. Clinical validation of FGFR as a therapeutic target has been demonstrated in bladder, liver, lung, breast, and gastric cancers. Our goal was to develop an irreversible covalent inhibitor of FGFR1-4 for use in oncology indications. An irreversible covalent binding mechanism imparts many desirable pharmacological benefits including high potency, selectivity, and prolonged target inhibition. Herein we report the structure-based design, medicinal chemistry optimization, and unique ADME assays of our irreversible covalent drug discovery program which culminated in the discovery of compound 34 (PRN1371), a highly selective and potent FGFR1-4 inhibitor.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Piridonas/farmacologia , Pirimidinas/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/síntese química , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Cães , Desenho de Fármacos , Estabilidade de Medicamentos , Feminino , Humanos , Absorção Intestinal , Macaca fascicularis , Masculino , Piridonas/administração & dosagem , Piridonas/síntese química , Piridonas/farmacocinética , Pirimidinas/administração & dosagem , Pirimidinas/síntese química , Pirimidinas/farmacocinética , Ratos Sprague-Dawley , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Solubilidade , Relação Estrutura-Atividade
4.
Bioorg Med Chem Lett ; 27(3): 632-635, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28025004

RESUMO

Reactive metabolites have been putatively linked to many adverse drug reactions including idiosyncratic toxicities for a number of drugs with black box warnings or withdrawn from the market. Therefore, it is desirable to minimize the risk of reactive metabolite formation for lead molecules in optimization, in particular for non-life threatening chronic disease, to maximize benefit to risk ratio. This article describes our effort in addressing reactive metabolite issues for a series of 3-amino-2-pyridone inhibitors of BTK, e.g. compound 1 has a value of 459pmol/mg protein in the microsomal covalent binding assay. Parallel approaches were taken to successfully resolve the issues: establishment of a predictive screening assay with correlation association of covalent binding assay, identification of the origin of reactive metabolite formation using MS/MS analysis of HLM as well as isolation and characterization of GSH adducts. This ultimately led to the discovery of compound 7 (RN941) with significantly reduced covalent binding of 26pmol/mg protein.


Assuntos
Inibidores de Proteínas Quinases/química , Proteínas Tirosina Quinases/antagonistas & inibidores , Piridonas/química , Tirosina Quinase da Agamaglobulinemia , Glutationa/química , Espectroscopia de Ressonância Magnética , Microssomos/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Piridonas/metabolismo , Espectrometria de Massas em Tandem
5.
Nat Chem Biol ; 11(7): 525-31, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26006010

RESUMO

Drugs with prolonged on-target residence times often show superior efficacy, yet general strategies for optimizing drug-target residence time are lacking. Here we made progress toward this elusive goal by targeting a noncatalytic cysteine in Bruton's tyrosine kinase (BTK) with reversible covalent inhibitors. Using an inverted orientation of the cysteine-reactive cyanoacrylamide electrophile, we identified potent and selective BTK inhibitors that demonstrated biochemical residence times spanning from minutes to 7 d. An inverted cyanoacrylamide with prolonged residence time in vivo remained bound to BTK for more than 18 h after clearance from the circulation. The inverted cyanoacrylamide strategy was further used to discover fibroblast growth factor receptor (FGFR) kinase inhibitors with residence times of several days, demonstrating the generalizability of the approach. Targeting of noncatalytic cysteines with inverted cyanoacrylamides may serve as a broadly applicable platform that facilitates 'residence time by design', the ability to modulate and improve the duration of target engagement in vivo.


Assuntos
Acrilamidas/farmacocinética , Linfócitos B/efeitos dos fármacos , Cianoacrilatos/farmacocinética , Inibidores de Proteínas Quinases/farmacocinética , Proteínas Tirosina Quinases/antagonistas & inibidores , Acrilamidas/síntese química , Tirosina Quinase da Agamaglobulinemia , Animais , Linfócitos B/enzimologia , Linfócitos B/patologia , Linhagem Celular Tumoral , Cristalografia por Raios X , Cianoacrilatos/síntese química , Dasatinibe , Feminino , Expressão Gênica , Humanos , Ligantes , Simulação de Acoplamento Molecular , Inibidores de Proteínas Quinases/síntese química , Estrutura Terciária de Proteína , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/genética , Pirimidinas/farmacocinética , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Células Sf9 , Spodoptera , Relação Estrutura-Atividade , Especificidade por Substrato , Tiazóis/farmacocinética , Fatores de Tempo
6.
J Med Chem ; 57(6): 2683-91, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24520947

RESUMO

Inhibition of spleen tyrosine kinase has attracted much attention as a mechanism for the treatment of cancers and autoimmune diseases such as asthma, rheumatoid arthritis, and systemic lupus erythematous. We report the structure-guided optimization of pyridazine amide spleen tyrosine kinase inhibitors. Early representatives of this scaffold were highly potent and selective but mutagenic in an Ames assay. An approach that led to the successful identification of nonmutagenic examples, as well as further optimization to compounds with reduced cardiovascular liabilities is described. Select pharmacokinetic and in vivo efficacy data are presented.


Assuntos
Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Piridazinas/síntese química , Piridazinas/farmacologia , Baço/enzimologia , Amidas/síntese química , Amidas/farmacologia , Animais , Biologia Computacional , Simulação por Computador , Desenho de Fármacos , Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Humanos , Técnicas In Vitro , Camundongos , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Conformação Molecular , Mutagênese/efeitos dos fármacos , Testes de Mutagenicidade , Inibidores de Proteínas Quinases/farmacocinética , Piridazinas/farmacocinética , Ratos , Baço/efeitos dos fármacos , Relação Estrutura-Atividade , Difração de Raios X
7.
J Med Chem ; 55(17): 7686-95, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-22877085

RESUMO

Inhibition of PI3Kδ is considered to be an attractive mechanism for the treatment of inflammatory diseases and leukocyte malignancies. Using a structure-based design approach, we have identified a series of potent and selective benzimidazole-based inhibitors of PI3Kδ. These inhibitors do not occupy the selectivity pocket between Trp760 and Met752 that is induced by other families of PI3Kδ inhibitors. Instead, the selectivity of the compounds for inhibition of PI3Kδ relative to other PI3K isoforms appears to be due primarily to the strong interactions these inhibitors are able to make with Trp760 in the PI3Kδ binding pocket. The pharmacokinetic properties and the ability of compound 5 to inhibit the function of B-cells in vivo are described.


Assuntos
Benzimidazóis/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Benzimidazóis/química , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Inibidores de Proteínas Quinases/química , Espectrometria de Massas por Ionização por Electrospray , Espectrofotometria Ultravioleta
9.
Toxicol Sci ; 118(1): 266-75, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20810542

RESUMO

The cellular function of kinases combined with the difficulty of designing selective small molecule kinase inhibitors (SMKIs) poses a challenge for drug development. The late-stage attrition of SMKIs could be lessened by integrating safety information of kinases into the lead optimization stage of drug development. Herein, a mathematical model to predict bone marrow toxicity (BMT) is presented which enables the rational design of SMKIs away from this safety liability. A specific example highlights how this model identifies critical structural modifications to avoid BMT. The model was built using a novel algorithm, which selects 19 representative kinases from a panel of 277 based upon their ATP-binding pocket sequences and ability to predict BMT in vivo for 48 SMKIs. A support vector machine classifier was trained on the selected kinases and accurately predicts BMT with 74% accuracy. The model provides an efficient method for understanding SMKI-induced in vivo BMT earlier in drug discovery.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Desenho de Fármacos , Inibidores de Proteínas Quinases/toxicidade , Proteômica/métodos , Trifosfato de Adenosina/metabolismo , Algoritmos , Animais , Inteligência Artificial , Células da Medula Óssea/enzimologia , Biologia Computacional , Simulação por Computador , Humanos , Modelos Biológicos , Estrutura Molecular , Peso Molecular , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/metabolismo , Proteínas Quinases/química , Proteínas Quinases/metabolismo , Curva ROC
10.
J Pharmacol Exp Ther ; 327(3): 610-9, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18776065

RESUMO

P38alpha is a protein kinase that regulates the expression of inflammatory cytokines, suggesting a role in the pathogenesis of diseases such as rheumatoid arthritis (RA) or systemic lupus erythematosus. Here, we describe the preclinical pharmacology of pamapimod, a novel p38 mitogen-activated protein kinase inhibitor. Pamapimod inhibited p38alpha and p38beta enzymatic activity, with IC(50) values of 0.014 +/- 0.002 and 0.48 +/- 0.04 microM, respectively. There was no activity against p38delta or p38gamma isoforms. When profiled across 350 kinases, pamapimod bound only to four kinases in addition to p38. Cellular potency was assessed using phosphorylation of heat shock protein-27 and c-Jun as selective readouts for p38 and c-Jun NH(2)-terminal kinase (JNK), respectively. Pamapimod inhibited p38 (IC(50), 0.06 microM), but inhibition of JNK was not detected. Pamapimod also inhibited lipopolysaccharide (LPS)-stimulated tumor necrosis factor (TNF) alpha production by monocytes, interleukin (IL)-1beta production in human whole blood, and spontaneous TNFalpha production by synovial explants from RA patients. LPS- and TNFalpha-stimulated production of TNFalpha and IL-6 in rodents also was inhibited by pamapimod. In murine collagen-induced arthritis, pamapimod reduced clinical signs of inflammation and bone loss at 50 mg/kg or greater. In a rat model of hyperalgesia, pamapimod increased tolerance to pressure in a dose-dependent manner, suggesting an important role of p38 in pain associated with inflammation. Finally, an analog of pamapimod that has equivalent potency and selectivity inhibited renal disease in lupus-prone MRL/lpr mice. Our study demonstrates that pamapimod is a potent, selective inhibitor of p38alpha with the ability to inhibit the signs and symptoms of RA and other autoimmune diseases.


Assuntos
Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Piridonas/farmacologia , Pirimidinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Animais , Artrite Reumatoide/tratamento farmacológico , Avaliação Pré-Clínica de Medicamentos , Humanos , Inflamação/tratamento farmacológico , Concentração Inibidora 50 , Interleucina-1beta/antagonistas & inibidores , Interleucina-6/antagonistas & inibidores , Nefropatias/prevenção & controle , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Osteoporose/prevenção & controle , Isoformas de Proteínas , Piridonas/uso terapêutico , Pirimidinas/uso terapêutico , Líquido Sinovial/imunologia , Líquido Sinovial/metabolismo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/antagonistas & inibidores
11.
J Med Chem ; 49(5): 1562-75, 2006 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-16509574

RESUMO

A novel class of highly selective inhibitors of p38 MAP kinase was discovered from high throughput screening. The synthesis and optimization of a series of 5-amino-N-phenyl-1H-pyrazol-4-yl-3-phenylmethanones is described. An X-ray crystal structure of this series bound in the ATP binding pocket of unphosphorylated p38alpha established the presence of a unique hydrogen bond between the exocyclic amine of the inhibitor and threonine 106 which likely contributes to the selectivity for p38. The crystallographic information was used to optimize the potency and physicochemical properties of the series. The incorporation of the 2,3-dihydroxypropoxy moiety on the pyrazole scaffold resulted in a compound with excellent drug-like properties including high oral bioavailability. These efforts identified 63 (RO3201195) as an orally bioavailable and highly selective inhibitor of p38 which was selected for advancement into Phase I clinical trials.


Assuntos
Anti-Inflamatórios/síntese química , Pirazóis/síntese química , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Trifosfato de Adenosina/química , Administração Oral , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Artrite Experimental/tratamento farmacológico , Sítios de Ligação , Disponibilidade Biológica , Linhagem Celular , Cristalografia por Raios X , Cães , Feminino , Haplorrinos , Humanos , Interleucina-1/antagonistas & inibidores , Interleucina-1/biossíntese , Interleucina-6/antagonistas & inibidores , Interleucina-6/biossíntese , Modelos Moleculares , Pirazóis/química , Pirazóis/farmacologia , Ratos , Ratos Endogâmicos Lew , Estereoisomerismo , Relação Estrutura-Atividade , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/química
12.
J Med Chem ; 46(22): 4702-13, 2003 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-14561090

RESUMO

Inhibition of the biosynthesis of proinflammatory cytokines such as tumor necrosis factor and interleukin-1 via p38 has been an approach toward the development of a disease modifying agent for the treatment of chronic inflammation and autoimmune diseases. The development of a new core structure of p38 inhibitors, 3-(4-fluorophenyl)-2-(pyridin-4-yl)-1H-pyrrolo[3,2-b] pyridine, is described. X-ray crystallographic data of the lead bound to the active site of p38 was used to guide the optimization of the series. Specific focus was placed on modulating the physical properties of the core while maintaining potent inhibition of p38. These efforts identified 42c as a potent inhibitor of p38, which also possessed the required physical properties worthy of advanced studies.


Assuntos
Inibidores Enzimáticos/síntese química , Indóis/síntese química , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Piridinas/síntese química , Administração Oral , Animais , Disponibilidade Biológica , Linhagem Celular , Cristalografia por Raios X , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Ligação de Hidrogênio , Técnicas In Vitro , Indóis/química , Injeções Intravenosas , Proteína Quinase 14 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/química , Modelos Moleculares , Ligação Proteica , Isoformas de Proteínas , Piridinas/química , Piridinas/farmacologia , Ratos , Relação Estrutura-Atividade , Fator de Necrose Tumoral alfa/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA