Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 20(13): 3521-30, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24780296

RESUMO

PURPOSE: Irinotecan (CPT-11) induced diarrhea occurs frequently in patients with cancer and limits its usage. Bacteria ß-glucuronidase (GUS) enzymes in intestines convert the nontoxic metabolite of CPT-11, SN-38G, to toxic SN-38, and finally lead to damage of intestinal epithelial cells and diarrhea. We previously reported amoxapine as a potent GUS inhibitor in vitro. To further understand the molecular mechanism of amoxapine and its potential for treatment of CPT-11-induced diarrhea, we studied the binding modes of amoxapine and its metabolites by docking and molecular dynamics simulation, and tested the in vivo efficacy on mice in combination with CPT-11. EXPERIMENTAL DESIGN: The binding of amoxapine, its metabolites, 7-hydroxyamoxapine and 8-hydroxyamoxapine, and a control drug loxapine with GUS was explored by computational protocols. The in vitro potencies of metabolites were measured by Escherichia coli GUS enzyme and cell-based assay. Low-dosage daily oral administration was designed to use along with CPT-11 to treat tumor-bearing mice. RESULTS: Computational modeling results indicated that amoxapine and its metabolites bound in the active site of GUS and satisfied critical pharmacophore features: aromatic features near bacterial loop residue F365' and hydrogen bond toward E413. Amoxapine and its metabolites were demonstrated as potent in vitro. Administration of low dosages of amoxapine with CPT-11 in mice achieved significant suppression of diarrhea and reduced tumor growth. CONCLUSIONS: Amoxapine has great clinical potential to be rapidly translated to human subjects for irinotecan-induced diarrhea.


Assuntos
Amoxapina/farmacologia , Antineoplásicos/toxicidade , Glicoproteínas/farmacologia , Substâncias Protetoras/farmacologia , Amoxapina/análogos & derivados , Amoxapina/química , Animais , Antineoplásicos Fitogênicos/toxicidade , Camptotecina/análogos & derivados , Camptotecina/toxicidade , Linhagem Celular Tumoral , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Feminino , Glicoproteínas/química , Irinotecano , Camundongos , Conformação Molecular , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Neoplasias/tratamento farmacológico , Neoplasias/mortalidade , Neoplasias/patologia , Substâncias Protetoras/química , Ligação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Biomol Screen ; 17(7): 957-65, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22535688

RESUMO

The active metabolite of the chemotherapeutic irinotecan, SN-38, is detoxified through glucuronidation and then excreted into the gastrointestinal tract. Intestinal bacteria convert the glucuronidated metabolite back to the toxic SN-38 using ß-glucuronidase (GUS), resulting in debilitating diarrhea. Inhibiting GUS activity may relieve this side effect of irinotecan. In this study, we sought to determine whether any known drugs have GUS inhibitory activity. We screened a library of Food and Drug Administration-approved drugs with a cell-free biochemical enzyme assay using purified bacterial GUS. After triage, five drugs were confirmed to inhibit purified bacterial GUS. Three of these were the monoamine oxidase inhibitors nialamide, isocarboxazid, and phenelzine with average IC(50) values for inhibiting GUS of 71, 128, and 2300 nM, respectively. The tricyclic antidepressant amoxapine (IC(50) = 388 nM) and the antimalarial mefloquine (IC(50) = 1.2 µM) also had activity. Nialamide, isocarboxazid, and amoxapine had no significant activity against purified mammalian GUS but showed potent activity for inhibiting endogenous GUS activity in a cell-based assay using living intact Escherichia coli with average IC(50) values of 17, 336, and 119 nM, respectively. Thus, nialamide, isocarboxazid, and amoxapine have potential to be repurposed as therapeutics to reduce diarrhea associated with irinotecan chemotherapy and warrant further investigation for this use.


Assuntos
Camptotecina/análogos & derivados , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Escherichia coli/enzimologia , Glucuronidase/antagonistas & inibidores , Amoxapina/farmacologia , Antineoplásicos Fitogênicos/metabolismo , Camptotecina/metabolismo , Descoberta de Drogas , Escherichia coli/metabolismo , Proteínas de Escherichia coli/antagonistas & inibidores , Irinotecano , Isocarboxazida/farmacologia , Mefloquina/farmacologia , Inibidores da Monoaminoxidase/farmacologia , Nialamida/farmacologia , Fenelzina/farmacologia
3.
Curr Chem Genomics ; 5: 13-20, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21643506

RESUMO

CPT-11 is a widely-used anti-cancer drug that is converted in vivo to its active metabolite, SN-38. In the liver, enzymes detoxify SN-38 by coupling it to a glucuronidate moiety and this inactive compound (SN-38G) is excreted into the gastrointestinal tract. In the intestine, commensal bacteria convert the SN-38G back to the active and toxic SN-38 using bacterial ß-glucuronidase enzyme (GUS). This intestinal SN-38 causes debilitating diarrhea that prevents dose-intensification and efficacy in a significant fraction of patients undergoing CPT-11 treatment for cancer. This CPT-11 metabolic pathway suggests that small molecule inhibitors of GUS may have utility as novel therapeutics for prevention of dose-limiting diarrhea resulting from CPT-11 therapy. To identify chemical inhibitors of GUS activity, we employed and validated a high throughput, fluorescence-based biochemical assay and used this assay to screen a compound library. Novel inhibitors of GUS were identified with IC(50) values ranging from 50 nM to 4.8 µM. These compounds may be useful as chemical probes for use in proof-of-concept experiments designed to determine the efficacy of GUS inhibitors in altering the intestinal metabolism of drugs. Our results demonstrate that this high throughput assay can be used to identify small molecule inhibitors of GUS.

4.
Science ; 330(6005): 831-5, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-21051639

RESUMO

The dose-limiting side effect of the common colon cancer chemotherapeutic CPT-11 is severe diarrhea caused by symbiotic bacterial ß-glucuronidases that reactivate the drug in the gut. We sought to target these enzymes without killing the commensal bacteria essential for human health. Potent bacterial ß-glucuronidase inhibitors were identified by high-throughput screening and shown to have no effect on the orthologous mammalian enzyme. Crystal structures established that selectivity was based on a loop unique to bacterial ß-glucuronidases. Inhibitors were highly effective against the enzyme target in living aerobic and anaerobic bacteria, but did not kill the bacteria or harm mammalian cells. Finally, oral administration of an inhibitor protected mice from CPT-11-induced toxicity. Thus, drugs may be designed to inhibit undesirable enzyme activities in essential microbial symbiotes to enhance chemotherapeutic efficacy.


Assuntos
Antineoplásicos Fitogênicos/toxicidade , Camptotecina/análogos & derivados , Inibidores Enzimáticos/farmacologia , Glucuronidase/antagonistas & inibidores , Glucuronidase/farmacologia , Animais , Antineoplásicos Fitogênicos/metabolismo , Bactérias Anaeróbias/efeitos dos fármacos , Camptotecina/metabolismo , Camptotecina/toxicidade , Linhagem Celular Tumoral , Colo/efeitos dos fármacos , Colo/microbiologia , Colo/patologia , Cristalografia por Raios X , Diarreia/prevenção & controle , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Escherichia coli/enzimologia , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/isolamento & purificação , Proteínas de Escherichia coli/metabolismo , Feminino , Glucuronidase/química , Glucuronidase/isolamento & purificação , Glucuronidase/metabolismo , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Irinotecano , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Pró-Fármacos/metabolismo , Pró-Fármacos/toxicidade , Conformação Proteica
5.
J Biomol Screen ; 14(9): 1092-101, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19675313

RESUMO

The phenomenon of antibiotic resistance has created a need for the development of novel antibiotic classes with nonclassical cellular targets. Unfortunately, target-based drug discovery against proteins considered essential for in vitro bacterial viability has yielded few new therapeutic classes of antibiotics. Targeting the large proportion of genes considered nonessential that have yet to be explored by high-throughput screening, for example, RecA, can complement these efforts. Recent evidence suggests that RecA-controlled processes are responsible for tolerance to antibiotic chemotherapy and are involved in pathways that ultimately lead to full-fledged antibiotic resistance. Therefore inhibitors of RecA may serve as therapeutic adjuvants in combination chemotherapy of bacterial infectious diseases. Toward the goal of validating RecA as a novel target in the chemotherapy of bacterial infections, the authors have screened 35,780 small molecules against RecA. In total, 80 small molecules were identified as primary hits and could be clustered in 6 distinct chemotype clades. The most potent class of hits was further examined, and 1 member compound was found to inhibit RecA-mediated strand exchange and prevent ciprofloxacin-induced SOS expression in Escherichia coli. This compound represents the first small molecule demonstrating an ability to inhibit the bacterial SOS response in live bacterial cell cultures.


Assuntos
Inibidores Enzimáticos/farmacologia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Recombinases Rec A/antagonistas & inibidores , Resposta SOS em Genética/efeitos dos fármacos , Adenosina Trifosfatases/metabolismo , Anti-Infecciosos/farmacologia , Permeabilidade da Membrana Celular , Ciprofloxacina/farmacologia , Farmacorresistência Bacteriana , Inibidores Enzimáticos/química , Escherichia coli/metabolismo , Estrutura Molecular , Recombinases Rec A/metabolismo , Reprodutibilidade dos Testes , Bibliotecas de Moléculas Pequenas
6.
PLoS One ; 4(4): e5274, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19384407

RESUMO

BACKGROUND: Hypocatabolism of the amyloid beta-protein (Abeta) by insulin-degrading enzyme (IDE) is implicated in the pathogenesis of Alzheimer disease (AD), making pharmacological activation of IDE an attractive therapeutic strategy. However, it has not been established whether the proteolytic activity of IDE can be enhanced by drug-like compounds. METHODOLOGY/PRINCIPAL FINDINGS: Based on the finding that ATP and other nucleotide polyphosphates modulate IDE activity at physiological concentrations, we conducted parallel high-throughput screening campaigns in the absence or presence of ATP and identified two compounds--designated Ia1 and Ia2--that significantly stimulate IDE proteolytic activity. Both compounds were found to interfere with the crosslinking of a photoaffinity ATP analogue to IDE, suggesting that they interact with a bona fide ATP-binding domain within IDE. Unexpectedly, we observed highly synergistic activation effects when the activity of Ia1 or Ia2 was tested in the presence of ATP, a finding that has implications for the mechanisms underlying ATP-mediated activation of IDE. Notably, Ia1 and Ia2 activated the degradation of Abeta by approximately 700% and approximately 400%, respectively, albeit only when Abeta was presented in a mixture also containing shorter substrates. CONCLUSIONS/SIGNIFICANCE: This study describes the first examples of synthetic small-molecule activators of IDE, showing that pharmacological activation of this important protease with drug-like compounds is achievable. These novel activators help to establish the putative ATP-binding domain as a key modulator of IDE proteolytic activity and offer new insights into the modulatory action of ATP. Several larger lessons abstracted from this screen will help inform the design of future screening campaigns and facilitate the eventual development of IDE activators with therapeutic utility.


Assuntos
Química Farmacêutica , Ativadores de Enzimas/farmacologia , Insulina/metabolismo , Insulisina/metabolismo , Peptídeos beta-Amiloides/metabolismo , Hidrólise , Marcadores de Fotoafinidade
7.
J Biomol Screen ; 13(5): 396-405, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18480472

RESUMO

Twenty human proteins encode Phox/Bem1p (PB1) domains, which are involved in forming protein heterodimers. MEKK2, MEKK3, and MEK5 are 3 serine-threonine protein kinases that have PB1 domains. MEKK2, MEKK3, and MEK5 are the MAP3Ks and the MAP2K in the ERK5 mitogen-activated protein kinase (MAPK) signaling module. ERK5 is a critical MAPK for both development of the vasculature and vascular homeostasis in the adult, but no other MAPK has been shown to be critical in vascular maintenance in the adult animal. MEKK2 and MEKK3 are the only MAP3Ks shown to physically interact with and activate the MEK5-ERK5 signaling module. Interaction of MEKK2 or MEKK3 with MEK5 is mediated by heterodimerization of the MEKK2 (or MEKK3) PB1 and MEK5 PB1 domains. The authors have developed a homogeneous, time-resolved fluorescence resonance energy transfer (TR-FRET) assay to monitor PB1-PB1 domain heterodimerization. The assay uses a europium-chelate conjugated GST-MEK5 PB1 domain chimera, biotinylated MEKK2 PB1 domain, and streptavidin-Cy5. Interaction of the MEKK2 and MEK5 PB1 domains gives a robust FRET signal (Z' factor = 0.93), which is completely abrogated by mutation of 2 acidic residues (64D65E-->AA) within the MEK5 PB1 domain that causes loss of stable PB1-PB1 domain interaction. This assay can be used to study the specificity of PB1-PB1 domain interactions and to screen for molecules that can regulate MEKK2/MEKK3-MEK5 interactions. Disruption of PB1 domain interactions represents a novel approach for selectively regulating the ERK5 signaling pathway independent of kinase active site-directed adenosine triphosphate competitive inhibitors.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas/química , Técnicas de Química Combinatória , Dimerização , Glutationa Transferase/química , Humanos , MAP Quinase Quinase 5/química , Estrutura Terciária de Proteína
8.
J Biomol Screen ; 11(7): 729-35, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16928982

RESUMO

Familial amyotrophic lateral sclerosis (ALS) accounts for 10% of all ALS cases; approximately 25% of these cases are due to mutations in the Cu/Zn superoxide dismutase gene (SOD1). To date, 105 different mutations spanning all 5 exons have been identified in the SOD1 gene. Mutant SOD1-associated ALS is caused by a toxic gain of function of the mutated protein. Therefore, regardless of the specific mechanism whereby mutant SOD1 initiates motor neuron death, the authors hypothesize that measures that decrease levels of mutant SOD1 protein should ameliorate the phenotype in transgenic mice and potentially in patients with SOD1-mediated disease. They have designed 2 cell-based screening assays to identify small, brain-permeant molecules that inactivate expression of the SOD1 gene or increase the degradation of the SOD1 protein. Here they describe the development and optimization of these assays and the results of high-throughput screening using a variety of compound libraries, including a total of more than 116,000 compounds. The majority of the hit compounds identified that down-regulated SOD1 were shown to be toxic in a cell-based viability assay or were nonselective transcription inhibitors, but work is continuing on a number of nonspecific inhibitors of SOD1 expression. Ultimately, the authors believe that these 2 cell-based assays will provide powerful strategies to identify novel therapies for the treatment of inherited SOD1-associated forms of ALS.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/enzimologia , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/farmacologia , Superóxido Dismutase/metabolismo , Animais , Linhagem Celular , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/química , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Proteínas Mutantes/metabolismo , Células PC12 , Regiões Promotoras Genéticas/genética , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Superóxido Dismutase-1
9.
Chem Biol ; 12(7): 811-23, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16039528

RESUMO

Cyclin-dependent kinase 5 (Cdk5) is widely viewed as a possible target for a wide variety of neurological disorders. One pathological role attributed to Cdk5 is the abnormal phosphorylation of tau that may lead to the neuronal inclusions known as neurofibrillary tangles. A high through-put screen for inhibitors of Cdk5-mediated phosphorylation of tau resulted in three compounds with distinct mechanisms of action. One compound is competitive with ATP and has a high affinity for the Cdk5 ATP binding pocket. The second compound also competes with ATP, is noncompetitive with tau, and (uniquely among this class of inhibitors) displaces adjacent amino acid residues to make room for the nitrophenyl group. A third compound did not compete with ATP, but did compete with tau at low concentrations of tau. The SAR and charge optimization derived from cocrystals of the two ATP competitors along with cocrystals of three other ATP competitors map out the importance of filling and properly charging different regions of the ATP binding pocket. Taken together, this analysis shows how the structure of Cdk5 constrains the space of potential inhibitors and reveals a pocket unfilled in all of the structures. These leads could be a starting point for structure-based drug design of more potent and selective inhibitors.


Assuntos
Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Neurônios/efeitos dos fármacos , Proteínas tau/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Ligação Competitiva , Quinase 5 Dependente de Ciclina , Quinases Ciclina-Dependentes/química , Quinases Ciclina-Dependentes/metabolismo , Ativação Enzimática , Ligantes , Modelos Químicos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neurônios/enzimologia , Fosforilação/efeitos dos fármacos , Relação Estrutura-Atividade , Proteínas tau/química
10.
Bioorg Med Chem Lett ; 15(4): 1193-6, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15686940

RESUMO

A verbenachalcone derivative was synthesized and shown to protect N2a cells from caspase induction caused by serum starvation and to enhance the effect of NGF on neurite outgrowth in PC12 cells. As an initial investigation of the compound's mechanism(s) of action, we performed differential gene expression profiling in PC12 cells using oligonucleotide ( approximately 10,000 gene probes) microarrays. Gene expression patterns were compared in the presence of NGF (2 and 50 ng/mL) and NGF (2 ng/mL) plus the verbenachalcone derivative. Ten genes were significantly (2-fold; p0.05) up-regulated and seven genes were significantly down-regulated in the presence of the compound. These results were independently validated by quantitative real-time PCR for a subset of genes (cathepsin L, sigma-1 receptor and protein tyrosine phosphatase receptor type R). These genes or their protein products may represent useful therapeutic targets for treating neurodegeneration, such as Alzheimer's disease.


Assuntos
Inibidores de Caspase , Chalcona/análogos & derivados , Chalcona/síntese química , Regulação da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Animais , Caspases/genética , Linhagem Celular , Chalcona/farmacologia , Chalconas , Sinergismo Farmacológico , Indução Enzimática/efeitos dos fármacos , Perfilação da Expressão Gênica , Humanos , Células PC12 , Ratos , Relação Estrutura-Atividade
11.
Front Biosci ; 9: 2788-95, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15353314

RESUMO

Collagenases are a highly specific class of enzymes. In their native states, collagenases cleave only native triple helical collagen molecules at a single peptide bond between Gly775-Leu776 for Type I collagen and Gly775-Ile776 for Type II collagen. The linear sequence of collagen is about 1050 amino acids in length, where three linear peptide sequences are required to form a triple helical collagen molecule. At present, there exist no crystallographic structures of collagenase bound to native triple helical collagen; nor has it been shown that collagenase recognizes the triple helical conformation of collagen. In our study, we have used an inhibitor design structure-activity based approach to show that collagenase recognizes and cleaves triple helical collagen conformations in preference to non-triple helical collagen conformations.


Assuntos
Colágeno/química , Inibidores Enzimáticos/farmacologia , Metaloproteinase 1 da Matriz/química , Inibidores de Metaloproteinases de Matriz , Animais , Artrite/metabolismo , Catálise , Colagenases/química , Biologia Computacional/métodos , Cristalografia por Raios X , Bases de Dados de Proteínas , Desenho de Fármacos , Modelos Moleculares , Conformação Molecular , Peptídeos/química , Conformação Proteica , Software , Suínos
12.
J Biomol Screen ; 9(2): 122-31, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15006135

RESUMO

A high-throughput assay for tau phosphorylation by cdk5/p25 is described. Full-length recombinant tau was used as a substrate in the presence of saturating adenosine triphosphate (ATP). Using PHF-1, an antibody directed specifically against 2 tau phosphorylation epitopes (serine 396 and serine 404), an enzyme-linked immunosorbent assay (ELISA)-based colorimetric assay was formatted in 384-well plates. The assay was validated by measuring kinetic parameters for cdk5/p25 catalysis and known inhibitors. Rate constants for the site-specific phosphorylations at the PHF-1 epitopes were determined and suggested preferential phosphorylation at these sites. The performance of this assay in a high-throughput format was demonstrated and used to identify inhibitors of tau phosphorylation at specific epitopes phosphorylated by cdk5/p25.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Proteínas tau/metabolismo , Trifosfato de Adenosina/análise , Trifosfato de Adenosina/metabolismo , Anticorpos Monoclonais/metabolismo , Sítios de Ligação , Biotinilação , Células Cultivadas , Quinase 5 Dependente de Ciclina , Humanos , Cinética , Cinetina , Miniaturização , Fosforilação , Ligação Proteica , Purinas/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Fatores de Tempo
13.
Chem Biol ; 10(9): 837-46, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14522054

RESUMO

Neuronal ubiquitin C-terminal hydrolase (UCH-L1) has been linked to Parkinson's disease (PD), the progression of certain nonneuronal tumors, and neuropathic pain. Certain lung tumor-derived cell lines express UCH-L1 but it is not expressed in normal lung tissue, suggesting that this enzyme plays a role in tumor progression, either as a trigger or as a response. Small-molecule inhibitors of UCH-L1 would be helpful in distinguishing between these scenarios. By utilizing high-throughput screening (HTS) to find inhibitors and traditional medicinal chemistry to optimize their affinity and specificity, we have identified a class of isatin O-acyl oximes that selectively inhibit UCH-L1 as compared to its systemic isoform, UCH-L3. Three representatives of this class (30, 50, 51) have IC(50) values of 0.80-0.94 micro M for UCH-L1 and 17-25 micro M for UCH-L3. The K(i) of 30 toward UCH-L1 is 0.40 micro M and inhibition is reversible, competitive, and active site directed. Two isatin oxime inhibitors increased proliferation of the H1299 lung tumor cell line but had no effect on a lung tumor line that does not express UCH-L1. Inhibition of UCH-L1 expression in the H1299 cell line using RNAi had a similar proproliferative effect, suggesting that the UCH-L1 enzymatic activity is antiproliferative and that UCH-L1 expression may be a response to tumor growth. The molecular mechanism of this response remains to be determined.


Assuntos
Antineoplásicos/farmacologia , Isatina/farmacologia , Neoplasias Pulmonares/patologia , Oximas/farmacologia , Ubiquitina Tiolesterase/antagonistas & inibidores , Antineoplásicos/química , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Concentração Inibidora 50 , Isatina/química , Isoenzimas/antagonistas & inibidores , Oximas/química , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA