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.
Toxicol Appl Pharmacol ; 438: 115905, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35122773

RESUMO

Systemic therapies targeting transforming growth factor beta (TGFß) or TGFßR1 kinase (ALK5) have been plagued by toxicities including cardiac valvulopathy and bone physeal dysplasia in animals, posing a significant challenge for clinical development in pulmonary indications. The current work aims to demonstrate that systemic ALK5-associated toxicities can be mitigated through localized lung delivery. Lung-selective (THRX-144644) and systemically bioavailable (galunisertib) ALK5 inhibitors were compared to determine whether lung selectivity is sufficient to maintain local tissue concentrations while mitigating systemic exposure and consequent pathway-related findings. Both molecules demonstrated potent ALK5 activity in rat precision cut lung slices (PCLS; p-SMAD3 half-maximal inhibitory concentration [IC50], 141 nM and 1070 nM for THRX-144644 and galunisertib, respectively). In 14-day repeat-dose studies in rats, dose-related cardiac valvulopathy was recapitulated with oral galunisertib at doses ≥150 mg/kg/day. In contrast, inhaled nebulized THRX-144644 did not cause similar systemic findings up to the maximally tolerated doses in rats or dogs (10 and 1.5 mg/kg/day, respectively). THRX-144644 lung-to-plasma ratios ranged from 100- to 1200-fold in rats and dogs across dose levels. THRX-144644 lung trough (24 h) concentrations in rats and dogs ranged from 3- to 17-fold above the PCLS IC50 across tolerated doses. At a dose level exceeding tolerability (60 mg/kg/day; 76-fold above PCLS IC50) minimal heart and bone changes were observed when systemic drug concentrations reached pharmacologic levels. In conclusion, the current preclinical work demonstrates that localized pulmonary delivery of an ALK5 inhibitor leads to favorable TGFß pathway pharmacodynamic inhibition in lung while minimizing key systemic toxicities.


Assuntos
Pulmão/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Administração Oral , Animais , Cães , Feminino , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pirazóis/toxicidade , Quinolinas/toxicidade , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo
2.
Bioorg Med Chem ; 28(2): 115227, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31862310

RESUMO

Aberrant hedgehog (Hh) pathway signaling is implicated in multiple cancer types and targeting the Smoothened (SMO) receptor, a key protein of the Hh pathway, has proven effective in treating metastasized basal cell carcinoma. Our lead optimization effort focused on a series of heteroarylamides. We observed that a methyl substitution ortho to the heteroaryl groups on an aniline core significantly improved the potency of this series of compounds. These findings predated the availability of SMO crystal structure in 2013. Here we retrospectively applied quantum mechanics calculations to demonstrate the o-Me substitution favors the bioactive conformation by inducing a dihedral twist between the heteroaryl rings and the core aniline. The o-Me also makes favorable hydrophobic interactions with key residue side chains in the binding pocket. From this effort, two compounds (AZD8542 and AZD7254) showed excellent pharmacokinetics across multiple preclinical species and demonstrated in vivo activity in abrogating the Hh paracrine pathway as well as anti- tumor effects.


Assuntos
Antineoplásicos/farmacologia , Benzamidas/farmacologia , Descoberta de Drogas , Imidazóis/farmacologia , Receptor Smoothened/antagonistas & inibidores , Proteína GLI1 em Dedos de Zinco/antagonistas & inibidores , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Benzamidas/síntese química , Benzamidas/química , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Imidazóis/síntese química , Imidazóis/química , Camundongos , Estrutura Molecular , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , Receptor Smoothened/metabolismo , Relação Estrutura-Atividade , Proteína GLI1 em Dedos de Zinco/metabolismo
3.
J Med Chem ; 61(3): 1061-1073, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29301085

RESUMO

Checkpoint kinase 1 (CHK1) inhibitors are potential cancer therapeutics that can be utilized for enhancing the efficacy of DNA damaging agents. Multiple small molecule CHK1 inhibitors from different chemical scaffolds have been developed and evaluated in clinical trials in combination with chemotherapeutics and radiation treatment. Scaffold morphing of thiophene carboxamide ureas (TCUs), such as AZD7762 (1) and a related series of triazoloquinolines (TZQs), led to the identification of fused-ring bicyclic CHK1 inhibitors, 7-carboxamide thienopyridines (7-CTPs), and 7-carboxamide indoles. X-ray crystal structures reveal a key intramolecular noncovalent sulfur-oxygen interaction in aligning the hinge-binding carboxamide group to the thienopyridine core in a coplanar fashion. An intramolecular hydrogen bond to an indole NH was also effective in locking the carboxamide in the preferred bound conformation to CHK1. Optimization on the 7-CTP series resulted in the identification of lead compound 44, which displayed respectable drug-like properties and good in vitro and in vivo potency.


Assuntos
Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Descoberta de Drogas , Compostos Heterocíclicos/química , Compostos Heterocíclicos/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinase 1 do Ponto de Checagem/química , Dano ao DNA , Humanos , Indóis/química , Modelos Moleculares , Domínios Proteicos , Piridinas/química
4.
J Chromatogr B Analyt Technol Biomed Life Sci ; 942-943: 107-12, 2013 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-24239935

RESUMO

Tropomyosin-related kinases (Trk) are tyrosine kinase receptors implicated in tumor proliferation, invasion, and survival signaling across a number of tumors, making them potentially attractive targets for the treatment of cancer. AZD7451 is a potent and selective inhibitor of Trk kinases currently undergoing a Phase I dose escalation in glioblastoma multiforme at the National Cancer Institute. A key part of early clinical testing for AZD7451 involves demonstrating that pharmacokinetic half-life and clinical exposures of AZD7451 are sufficient to inhibit Trk receptors in preclinical models. To address this need, an ultra sensitive analytical method was developed to measure the AZD7451 profile in human plasma. A liquid-liquid extraction recovered >80% of AZD7451 before quantitative analysis by ultra HPLC-MS/MS. A Varian Polaris(®) C18-A column and a mass transition of m/z 383.5→340.5 (m/z 389.6→342.0 for the internal standard [(2)H6]-AZD7451) was used, and a dynamic calibration range of 0.5-1000ng/mL was established, which provided a sensitive (<8.5% deviation), and precise (<6%) quantitative assay for AZD7451. AZD7451 demonstrated stability in human plasma at room temperature for 24h (<7% change) and after extraction at 4°C for 24h (<8% change), and was stable through 4 freeze/thaw cycles (<8% change). This method was used to measure AZD7451 plasma levels in clinical samples to confirm the sensitivity at several time points following AZD7451 treatment in subjects with glioblastoma.


Assuntos
2-Aminopurina/sangue , Antineoplásicos/sangue , Cromatografia Líquida de Alta Pressão/métodos , Inibidores de Proteínas Quinases/sangue , Pirazóis/sangue , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Espectrometria de Massas em Tandem/métodos , 2-Aminopurina/química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Estabilidade de Medicamentos , Humanos , Modelos Lineares , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Pirazóis/química , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
5.
J Med Chem ; 55(15): 6916-33, 2012 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-22779424

RESUMO

Novel non-fluoroquinolone inhibitors of bacterial type II topoisomerases (DNA gyrase and topoisomerase IV) are of interest for the development of new antibacterial agents that are not impacted by target-mediated cross-resistance with fluoroquinolones. N-Linked amino piperidines, such as 7a, generally show potent antibacterial activity, including against quinolone-resistant isolates, but suffer from hERG inhibition (IC(50) = 44 µM for 7a) and QT prolongation in vivo. We now disclose the finding that new analogues of 7a with reduced pK(a) due to substitution with an electron-withdrawing substituent in the piperidine moiety, such as R,S-7c, retained the Gram-positive activity of 7a but showed significantly less hERG inhibition (IC(50) = 233 µM for R,S-7c). This compound exhibited moderate clearance in dog, promising efficacy against a MRSA strain in a mouse infection model, and an improved in vivo QT profile as measured in a guinea pig in vivo model. As a result of its promising activity, R,S-7c was advanced into phase I clinical studies.


Assuntos
Antibacterianos/síntese química , Dioxanos/síntese química , Piperidinas/síntese química , Quinolonas/síntese química , Inibidores da Topoisomerase II/síntese química , Administração Oral , Animais , Antibacterianos/farmacologia , Antibacterianos/toxicidade , Disponibilidade Biológica , DNA Topoisomerase IV/antagonistas & inibidores , Dioxanos/farmacologia , Dioxanos/toxicidade , Cães , Farmacorresistência Bacteriana , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Cobaias , Staphylococcus aureus Resistente à Meticilina , Camundongos , Testes de Sensibilidade Microbiana , Piperidinas/farmacologia , Piperidinas/toxicidade , Quinolonas/farmacologia , Quinolonas/toxicidade , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Estereoisomerismo , Relação Estrutura-Atividade , Inibidores da Topoisomerase II/farmacologia , Inibidores da Topoisomerase II/toxicidade
6.
J Med Chem ; 55(11): 5130-42, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22551018

RESUMO

Checkpoint kinases CHK1 and CHK2 are activated in response to DNA damage that results in cell cycle arrest, allowing sufficient time for DNA repair. Agents that lead to abrogation of such checkpoints have potential to increase the efficacy of such compounds as chemo- and radiotherapies. Thiophenecarboxamide ureas (TCUs) were identified as inhibitors of CHK1 by high throughput screening. A structure-based approach is described using crystal structures of JNK1 and CHK1 in complex with 1 and 2 and of the CHK1-3b complex. The ribose binding pocket of CHK1 was targeted to generate inhibitors with excellent cellular potency and selectivity over CDK1and IKKß, key features lacking from the initial compounds. Optimization of 3b resulted in the identification of a regioisomeric 3-TCU lead 12a. Optimization of 12a led to the discovery of the clinical candidate 4 (AZD7762), which strongly potentiates the efficacy of a variety of DNA-damaging agents in preclinical models.


Assuntos
Antineoplásicos/síntese química , Inibidores de Proteínas Quinases/síntese química , Proteínas Quinases/metabolismo , Tiofenos/síntese química , Ureia/análogos & derivados , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Sítios de Ligação , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Quinase 1 do Ponto de Checagem , Cristalografia por Raios X , Dano ao DNA , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Desenho de Fármacos , Sinergismo Farmacológico , Ensaios de Triagem em Larga Escala , Irinotecano , Camundongos , Modelos Moleculares , Estrutura Molecular , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/química , Ratos , Estereoisomerismo , Relação Estrutura-Atividade , Tiofenos/química , Tiofenos/farmacologia , Ureia/síntese química , Ureia/química , Ureia/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
7.
Bioorg Med Chem Lett ; 22(6): 2330-7, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22342147

RESUMO

Checkpoint kinase 1 (Chk1, CHEK1) is a Ser/Thr protein kinase that plays a key role in mediating the cellular response to DNA-damage. Synthesis and evaluation of a previously described class of Chk1 inhibitors, triazoloquinolones/triazolones (TZs) is further described herein. Our investigation of structure-activity relationships led to the identification of potent inhibitors 14c, 14h and 16e. Key challenges included modulation of physicochemical properties and pharmacokinetic (PK) parameters to enable compound testing in a Chk1 specific hollow fiber pharmacodynamic model. In this model, 16e was shown to abrogate topotecan-induced cell cycle arrest in a dose dependent manner. The demonstrated activity of TZs in this model in combination with a chemotherapeutic agent as well as radiotherapy validates this series of Chk1 inhibitors. X-ray crystal structures (PDB code: 2YEX and 2YER) for an initial lead and an optimized analog are also presented.


Assuntos
Antineoplásicos/síntese química , Neoplasias do Colo/terapia , Inibidores de Proteínas Quinases/síntese química , Proteínas Quinases/metabolismo , Triazóis/síntese química , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Neoplasias do Colo/enzimologia , Terapia Combinada , Cristalografia por Raios X , Dano ao DNA , Relação Dose-Resposta a Droga , Humanos , Camundongos , Camundongos Nus , Modelos Moleculares , Conformação Proteica , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/uso terapêutico , Relação Estrutura-Atividade , Topotecan/farmacologia , Triazóis/farmacocinética , Triazóis/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Med Chem ; 54(22): 7834-47, 2011 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21999508

RESUMO

Novel non-fluoroquinolone inhibitors of bacterial type II topoisomerases (DNA gyrase and topoisomerase IV) are of interest for the development of new antibacterial agents that are not impacted by target-mediated cross-resistance with fluoroquinolones. Aminopiperidines that have a bicyclic aromatic moiety linked through a carbon to an ethyl bridge, such as 1, generally show potent broad-spectrum antibacterial activity, including quinolone-resistant isolates, but suffer from potent hERG inhibition (IC(50)= 3 µM for 1). We now disclose the finding that new analogues of 1 with an N-linked cyclic amide moiety attached to the ethyl bridge, such as 24m, retain the broad-spectrum antibacterial activity of 1 but show significantly less hERG inhibition (IC(50)= 31 µM for 24m) and higher free fraction than 1. One optimized analogue, compound 24l, showed moderate clearance in the dog and promising efficacy against Staphylococcus aureus in a mouse thigh infection model.


Assuntos
Antibacterianos/síntese química , Proteínas de Bactérias/antagonistas & inibidores , DNA Topoisomerases Tipo II/metabolismo , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Piperidinas/síntese química , Inibidores da Topoisomerase/síntese química , Animais , Antibacterianos/farmacocinética , Antibacterianos/farmacologia , Cães , Farmacorresistência Bacteriana , Canal de Potássio ERG1 , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Camundongos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Piperidinas/farmacocinética , Piperidinas/farmacologia , Conformação Proteica , Ratos , Infecções Estafilocócicas/tratamento farmacológico , Relação Estrutura-Atividade , Inibidores da Topoisomerase/farmacocinética , Inibidores da Topoisomerase/farmacologia
9.
J Med Chem ; 54(19): 6734-50, 2011 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-21899292

RESUMO

Structure-activity relationship analysis identified (+)-N-(3-aminopropyl)-N-[1-(5-benzyl-3-methyl-4-oxo-[1,2]thiazolo[5,4-d]pyrimidin-6-yl)-2-methylpropyl]-4-methylbenzamide (AZD4877), from a series of novel kinesin spindle protein (KSP) inhibitors, as exhibiting both excellent biochemical potency and pharmaceutical properties suitable for clinical development. The selected compound arrested cells in mitosis leading to the formation of the monopolar spindle phenotype characteristic of KSP inhibition and induction of cellular death. A favorable pharmacokinetic profile and notable in vivo efficacy supported the selection of this compound as a clinical candidate for the treatment of cancer.


Assuntos
Antineoplásicos/síntese química , Benzamidas/síntese química , Cinesinas/antagonistas & inibidores , Pirimidinonas/síntese química , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Benzamidas/farmacocinética , Benzamidas/farmacologia , Proteínas Sanguíneas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Hepatócitos/metabolismo , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Camundongos , Camundongos Nus , Microssomos Hepáticos/metabolismo , Ligação Proteica , Pirimidinonas/farmacocinética , Pirimidinonas/farmacologia , Ratos , Ratos Wistar , Solubilidade , Estereoisomerismo , Relação Estrutura-Atividade
10.
Cancer Chemother Pharmacol ; 66(2): 245-54, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19841922

RESUMO

PURPOSE: Pharmacokinetic/pharmacodynamic (PK/PD) models have been shown to be useful in predicting tumor growth rates in mouse xenografts. We applied novel PK/PD models to the published anticancer combination therapies of tumor growth inhibition to simulate synergistic changes in tumor growth rates. The parameters from the PK/PD model were further used to estimate clinical doses of the combination. METHODS: A PK/PD model was built that linked the dosing regimen of a compound to the inhibition of tumor growth in mouse xenograft models. Two subsequent PK/PD models were developed to simulate the published tumor growth profiles of combination treatments. Model I predicts the tumor growth curve assuming that the effect of two anticancer drugs, AZD7762 and irinotecan, is synergistic when given in combination. Model II predicts the tumor growth curve assuming that the effect of co-administering flavopiridol and irinotecan is maximally synergistic when dosed at an optimal interval. RESULTS: Model I was able to account for the synergistic effects of AZD7762 following the administration of irinotecan. When Model II was applied to the antitumor activity of irinotecan and flavopiridol combination therapy, the modeling was able to reproduce the optimal dosing interval between administrations of the compounds. Furthermore, Model II was able to estimate the biologically active dose of flavopiridol recommended for phase II studies. CONCLUSIONS: The timing of clinical combination therapy doses is often selected empirically. PK/PD models provide a theoretical structure useful in the design of the optimal clinical dose, frequency of administration and the optimal timing of administration between anticancer agents to maximize tumor suppression.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Avaliação Pré-Clínica de Medicamentos/estatística & dados numéricos , Algoritmos , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Proliferação de Células , Avaliação Pré-Clínica de Medicamentos/métodos , Sinergismo Farmacológico , Flavonoides/administração & dosagem , Genes cdc/efeitos dos fármacos , Inibidores do Crescimento/administração & dosagem , Humanos , Irinotecano , Camundongos , Camundongos Nus , Modelos Estatísticos , Transplante de Neoplasias , Piperidinas/administração & dosagem , Valor Preditivo dos Testes , Tiofenos/administração & dosagem , Ureia/administração & dosagem , Ureia/análogos & derivados , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Med Chem ; 48(18): 5639-43, 2005 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-16134929
12.
J Biol Chem ; 278(3): 1450-6, 2003 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-12403772

RESUMO

The signal-inducible phosphorylation of serines 32 and 36 of I kappa B alpha is critical in regulating the subsequent ubiquitination and proteolysis of I kappa B alpha, which then releases NF-kappa B to promote gene transcription. The multisubunit I kappa B kinase responsible for this phosphorylation contains two catalytic subunits, termed I kappa B kinase (IKK)-1 and IKK-2. BMS-345541 (4(2'-aminoethyl)amino-1,8-dimethylimidazo(1,2-a)quinoxaline) was identified as a selective inhibitor of the catalytic subunits of IKK (IKK-2 IC(50) = 0.3 microm, IKK-1 IC(50) = 4 microm). The compound failed to inhibit a panel of 15 other kinases and selectively inhibited the stimulated phosphorylation of I kappa B alpha in cells (IC(50) = 4 microm) while failing to affect c-Jun and STAT3 phosphorylation, as well as mitogen-activated protein kinase-activated protein kinase 2 activation in cells. Consistent with the role of IKK/NF-kappa B in the regulation of cytokine transcription, BMS-345541 inhibited lipopolysaccharide-stimulated tumor necrosis factor alpha, interleukin-1 beta, interleukin-8, and interleukin-6 in THP-1 cells with IC(50) values in the 1- to 5-microm range. Although a Dixon plot of the inhibition of IKK-2 by BMS-345541 showed a non-linear relationship indicating non-Michaelis-Menten kinetic binding, the use of multiple inhibition analyses indicated that BMS-345541 binds in a mutually exclusive manner with respect to a peptide inhibitor corresponding to amino acids 26-42 of I kappa B alpha with Ser-32 and Ser-36 changed to aspartates and in a non-mutually exclusive manner with respect to ADP. The opposite results were obtained when studying the binding to IKK-1. A binding model is proposed in which BMS-345541 binds to similar allosteric sites on IKK-1 and IKK-2, which then affects the active sites of the subunits differently. BMS-345541 was also shown to have excellent pharmacokinetics in mice, and peroral administration showed the compound to dose-dependently inhibit the production of serum tumor necrosis factor alpha following intraperitoneal challenge with lipopolysaccharide. Thus, the compound is effective against NF-kappa B activation in mice and represents an important tool for investigating the role of IKK in disease models.


Assuntos
Inibidores Enzimáticos/farmacologia , Imidazóis/farmacologia , NF-kappa B/fisiologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Quinoxalinas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Sítio Alostérico , Animais , Domínio Catalítico , Inibidores Enzimáticos/metabolismo , Feminino , Quinase I-kappa B , Imidazóis/metabolismo , Cinética , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Quinoxalinas/metabolismo , Transcrição Gênica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA