Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
BMC Bioinformatics ; 15: 261, 2014 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-25089037

RESUMO

BACKGROUND: Human epidermal growth factor receptor 2 (HER2) has an important role in cancer aggressiveness and poor prognosis. HER2 has been used as a drug target for cancers. In particular, to effectively treat HER2-positive cancer, small molecule inhibitors were developed to target HER2 kinase. Knowing that curcumin has been used as food to inhibit cancer activity, this study evaluated the efficacy of natural curcumins and curcumin analogs as HER2 inhibitors using in vitro and in silico studies. The curcumin analogs considered in this study composed of 4 groups classified by their core structure, ß-diketone, monoketone, pyrazole, and isoxazole. RESULTS: In the present study, both computational and experimental studies were performed. The specificity of curcumin analogs selected from the docked results was examined against human breast cancer cell lines. The screened curcumin compounds were then subjected to molecular dynamics simulation study. By modifying curcumin analogs, we found that protein-ligand affinity increases. The benzene ring with a hydroxyl group could enhance affinity by forming hydrophobic interactions and the hydrogen bond with the hydrophobic pocket. Hydroxyl, carbonyl or methoxy group also formed hydrogen bonds with residues in the adenine pocket and sugar pocket of HER2-TK. These modifications could suggest the new drug design for potentially effective HER2-TK inhibitors. Two outstanding compounds, bisdemethylcurcumin (AS-KTC006) and 3,5-bis((E)-3,4-dimethoxystyryl)isoxazole (AS-KTC021 ),were well oriented in the binding pocket almost in the simulation time, 30 ns. This evidence confirmed the results of cell-based assays and the docking studies. They possessed more distinguished interactions than known HER2-TK inhibitors, considering them as a promising drug in the near future. CONCLUSIONS: The series of curcumin compounds were screened using a computational molecular docking and followed by human breast cancer cell lines assay. Both AS-KTC006 and AS-KTC021 could inhibit breast cancer cell lines though inhibiting of HER2-TK. The intermolecular interactions were confirmed by molecular dynamics simulation studies. This information would explore more understanding of curcuminoid structures and HER2-TK.


Assuntos
Curcumina/análogos & derivados , Curcumina/farmacologia , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Desenho de Fármacos , Feminino , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Estrutura Terciária de Proteína
2.
ChemMedChem ; 9(1): 217-32, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24273104

RESUMO

As part of our research projects to identify new chemical entities of biological interest, we developed a synthetic approach and the biological evaluation of (7-aryl-1,5-naphthyridin-4-yl)ureas as a novel class of Aurora kinase inhibitors for the treatment of malignant diseases based on pathological cell proliferation. 1,5-Naphthyridine derivatives showed excellent inhibitory activities toward Aurora kinases A and B, and the most active compound, 1-cyclopropyl-3-[7-(1-methyl-1H-pyrazol-4-yl)-1,5-naphthyridin-4-yl]urea (49), displayed IC50 values of 13 and 107 nM against Aurora kinases A and B, respectively. In addition, the selectivity toward a panel of seven cancer-related protein kinases was highlighted. In vitro ADME properties were also determined in order to rationalize the difficulties in correlating antiproliferative activity with Aurora kinase inhibition. Finally, the good safety profile of these compounds imparts promising potential for their further development as anticancer agents.


Assuntos
Aurora Quinase A/antagonistas & inibidores , Aurora Quinase B/antagonistas & inibidores , Inibidores de Proteínas Quinases/análogos & derivados , Ureia/análogos & derivados , Animais , Aurora Quinase A/genética , Aurora Quinase A/metabolismo , Aurora Quinase B/genética , Aurora Quinase B/metabolismo , Células CACO-2 , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Células HCT116 , Meia-Vida , Humanos , Camundongos , Microssomos Hepáticos/metabolismo , Naftiridinas/química , Ligação Proteica , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Relação Estrutura-Atividade , Ureia/farmacocinética , Ureia/farmacologia
3.
J Med Chem ; 56(9): 3456-66, 2013 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-23550937

RESUMO

Curcumin is a biologically active component of curry powder. A structurally related class of mimetics possesses similar anti-inflammatory and anticancer properties. Mechanism has been examined by exploring kinase inhibition trends. In a screen of 50 kinases relevant to many forms of cancer, one member of the series (4, EF31) showed ≥85% inhibition for 10 of the enzymes at 5 µM, while 22 of the proteins were blocked at ≥40%. IC50 values for an expanded set of curcumin analogues established a rank order of potencies, and analyses of IKKß and AKT2 enzyme kinetics for 4 revealed a mixed inhibition model, ATP competition dominating. Our curcumin mimetics are generally selective for Ser/Thr kinases. Both selectivity and potency trends are compatible with protein sequence comparisons, while modeled kinase binding site geometries deliver a reasonable correlation with mixed inhibition. Overall, these analogues are shown to be pleiotropic inhibitors that operate at multiple points along cell signaling pathways.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Curcumina/análogos & derivados , Curcumina/farmacologia , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Curcumina/química , Humanos , Concentração Inibidora 50 , Cinética , Camundongos , Modelos Moleculares , Fosforilação/efeitos dos fármacos , Conformação Proteica , Inibidores de Proteínas Quinases/química , Proteínas Quinases/química , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
4.
Eur J Med Chem ; 55: 220-7, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22871485

RESUMO

Previously, we demonstrated that the multiple kinase inhibitor sorafenib mediates the repression of phospho-STAT3 in hepatocellular carcinoma cells. In this study, we used this kinase-independent mechanism as a molecular basis to use sorafenib as scaffold to develop a novel class of SHP-1-activating agents. The proof of principle of this premise was provided by SC-1, which on replacement of N-methylpicolinamide by a phenylcyano group showed abolished kinase activity while retaining phospho-STAT3 repressive activity. Structural optimization of SC-1 led to compound 6, which repressed phospho-STAT3 through SHP-1 activation and inhibited PLC5 cell proliferation at sub-micromolar potency. In light of the pivotal role of phospho-STAT3 in promoting tumorigenesis and drug resistance, this novel SHP-1-activating agent may have therapeutic relevance in cancer therapy.


Assuntos
Benzenossulfonatos/química , Benzenossulfonatos/farmacologia , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Piridinas/química , Piridinas/farmacologia , Fator de Transcrição STAT3/metabolismo , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Nus , Niacinamida/análogos & derivados , Compostos de Fenilureia , Fosfoproteínas/metabolismo , Proteínas Quinases/metabolismo , Sorafenibe , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Eur J Med Chem ; 54: 264-71, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22647219

RESUMO

p38 mitogen-activated protein kinase (p38 MAPK) is an important signal transducing enzyme involved in many cellular regulations, including signaling pathways, pain and inflammation. Several p38 MAPK inhibitors have been developed as drug candidates to treatment of autoimmune disorders, such as rheumatoid arthritis. In this paper we reported the docking, synthesis and pharmacological activity of novel urea-derivatives (4a-e) designed as p38 MAPK inhibitors. These derivatives presented good theoretical affinity to the target p38 MAPK, standing out compound 4e (LASSBio-998), which showed a better score value compared to the prototype GK-00687. This compound was able to reduce in vitro TNF-α production and was orally active in a hypernociceptive murine model sensible to p38 MAPK inhibitors. Otherwise, compound 4e presented a dose-dependent analgesic effect in a model of antigen (mBSA)-induced arthritis and anti-inflammatory profile in carrageenan induced paw edema, indicating its potential as a new antiarthritis prototype.


Assuntos
Desenho de Fármacos , Simulação de Acoplamento Molecular , Ureia/síntese química , Ureia/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Carragenina/farmacologia , Relação Dose-Resposta a Droga , Edema/induzido quimicamente , Edema/tratamento farmacológico , Feminino , Masculino , Camundongos , Nociceptividade/efeitos dos fármacos , Conformação Proteica , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/biossíntese , Ureia/análogos & derivados , Ureia/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/química
6.
Mini Rev Med Chem ; 12(7): 650-64, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22512549

RESUMO

Marine natural products offer an abundant source of pharmacologically active agents with great diversity and complexity, and the potential to produce valuable therapeutic entities. Indole alkaloids is one of the important class of marine-derived secondary metabolites, with wide occurrence amongst variety of marine sources such as sponges, tunicates, algae, worms and microorganisms and have been extensively studied for their biological activities. Among this chemical family, a sponge-derived bis-indole alkaloid fascaplysin (1) exhibited broad range of bioactivities including antibacterial, antifungal, antiviral, anti-HIV-1-RTase, p56 tyrosine kinase inhibition, antimalarial, anti-angiogenic, antiproliferative activity against numerous cancer cell lines, specific inhibition of cyclin-dependent kinase-4 (IC(50) 350 nM) and action as a DNA intercalator. In the present review, the chemical diversity of natural as well as synthetic analogues of fascaplysin has been reviewed with a detailed account on synthetic reports and pharmacological studies. Our analysis of the structure-activity relationships of this family of compounds highlights the existence of various potential leads for the development of novel anticancer agents.


Assuntos
Organismos Aquáticos/química , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Indóis/química , Indóis/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Produtos Biológicos/química , Quinase 4 Dependente de Ciclina/metabolismo , Humanos , Indóis/síntese química , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/síntese química , Especificidade por Substrato
7.
Mini Rev Med Chem ; 12(7): 618-31, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22512550

RESUMO

Marine invertebrates are a rich source of novel, bioactive secondary metabolites and have attracted a great deal of attention from scientists in the fields of chemistry, pharmacology, ecology, and molecular biology. This profilic natural source has produced several antitumor secondary metabolites and amongst these, indole alkaloids are of wide occurrence. Meridianins A-G (1-7) are indole alkaloids isolated from tunicate Aplidium meridianum and are known to inhibit variety of protein kinases associated with cancer and neurodegenerative diseases. These compounds also exhibited promising antiproliferative activity in several cancer cell lines. Amongst natural meridianins, meridianin E (5) showed potent and selective inhibition of CDK-1 and CDK-5. Several synthetic meridianin analogs exhibited potent and selective inhibition of glycogen synthase-3 (GSK-3) and dual-specificity tyrosine-phosphorylation regulated kinase 1A (Dyrk-1A) which are known to be implicated in progression of Alzheimer's disease. The present review provides the critical account of isolation, medicinal chemistry and pharmacology of meridianins. Our analysis of the structure-activity relationships of this family of compounds highlights the existence of various potential leads for the development of novel anticancer and anti-Alzheimer's agents.


Assuntos
Organismos Aquáticos/química , Alcaloides Indólicos/química , Alcaloides Indólicos/farmacologia , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Animais , Produtos Biológicos/química , Humanos , Alcaloides Indólicos/síntese química , Alcaloides Indólicos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/metabolismo , Proteínas Quinases/química , Relação Estrutura-Atividade
8.
Angiogenesis ; 14(3): 281-91, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21487803

RESUMO

Small molecular inhibitors of Cyclin dependent kinases (Cdks) are currently being developed as anticancer therapeutics due to their antiproliferative properties. The purine Cdk specific inhibitor (R)-roscovitine (seliciclib, CYC202) represents one of the most promising of these compounds. It is currently evaluated in clinical trials concerning cancer therapy. Recently, we have shown that roscovitine exerts potent antiangiogenic effects and elucidated Cdk5 as a new player in angiogenesis. These findings introduce Cdk5 as novel target for antiangiogenic therapy, and Cdk5 inhibitors as an attractive therapeutic approach. Here, we present the antiangiogenic profile of 15 derivatives of roscovitine in vitro and in vivo and provide structure activity relationships of the roscovitine analogs. The (S)-isomer LGR561 and the respective (R)- and (S)-isomers LGR848 and LGR849 strongly inhibited proliferation and cell cycle progression, induced cell death, and reduced migration of endothelial cells in vitro. In comparison to roscovitine, these compounds showed an increased potency to inhibit Cdk2, Cdk5, Cdk7, and Cdk9. By analyzing the effects of LGR561, LGR848, and LGR849 on endothelial cell tube formation, mouse aortic ring sprouting, angiogenesis in the chick chorioallantoic membrane, and neovessel formation in the mouse cornea, we elucidate the two (S)-isomers LGR561 and LGR849 as highly potent inhibitors of angiogenesis. This study provides first information on how to modify roscovitine to develop Cdk inhibitors with increased antiangiogenic activity and suggests the application of existing and the development of new Cdk inhibitors to inhibit both, cancer cell proliferation and angiogenesis.


Assuntos
Inibidores da Angiogênese/farmacologia , Aorta/metabolismo , Ciclo Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/antagonistas & inibidores , Células Endoteliais/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Inibidores de Proteínas Quinases , Purinas/farmacologia , Animais , Aorta/patologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Neovascularização da Córnea/tratamento farmacológico , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/patologia , Quinases Ciclina-Dependentes/metabolismo , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/patologia , Humanos , Camundongos , Técnicas de Cultura de Órgãos , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Roscovitina , Estereoisomerismo
9.
Curr Top Med Chem ; 11(7): 788-99, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21291393

RESUMO

As the molecular etiology of cancer unravels, revealing the heterogeneous nature of the malignancy, multi-target drug treatments are more frequently advocated. Such therapeutic avenues often target kinases, the basic signal transducers in the cell. Because kinases share common evolutionary backgrounds, they also share many structural attributes, making it difficult for molecular targeted therapy to distinguish between paralogs. Thus, kinase inhibitors (KIs) tend to have undesired cross-reactivities, resulting in potentially lethal side effects. The health risks are obviously higher in these multi-pronged treatments when contrasted with the effects of more selective therapeutic agents. Using a nonconserved physicochemical biomarker, we present a rationally designed molecular filter that enables the control of specificity and the development of adjuvant drugs to edit out the side effects of the primary therapeutic agent. These editors work by overlapping therapeutically with the primary drug in cancer cells, while interfering with toxicity-related signaling pathways recruited by the primary drug in off-target cells. We then examine the possible application of these filtering methods to specifically target kinases when they present idiosyncratic cancer-related mutations. Such applications open the door to engineer personalized drugs tailored to the genetic makeup of the patient. These various methods of enhancing efficacy and safety show some degree of modularity, allowing drug designers to utilize multiple techniques and various drug combinations to create the safest and most powerful treatment for any given therapeutic scenario.


Assuntos
Biomarcadores/química , Conformação Molecular/efeitos dos fármacos , Terapia de Alvo Molecular/métodos , Fosfotransferases/antagonistas & inibidores , Inibidores de Proteínas Quinases/síntese química , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Desenho de Fármacos , Sinergismo Farmacológico , Humanos , Ligação de Hidrogênio/efeitos dos fármacos , Camundongos , Mutação , Neoplasias/tratamento farmacológico , Fosfotransferases/genética , Fosfotransferases/metabolismo , Medicina de Precisão/métodos , Ligação Proteica/efeitos dos fármacos , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Fatores de Risco , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
10.
Arch Biochem Biophys ; 507(2): 212-8, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21185805

RESUMO

Harmine is a ß-carboline alkaloid. The compound is a potent inhibitor of dual-specificity tyrosine phosphorylation-regulated kinase 1A (Dyrk1A), a kinase implicated in Down syndrome. In this study, we show that harmine functions as an ATP-competitive inhibitor against Dyrk1A. Our conclusion is supported by kinetic analysis of harmine inhibition as well as by the characterization of a Dyrk1A mutation conferring significant resistance to harmine. The mutation, V306A, is located next to the highly conserved D307 residue in kinases known to coordinate the phosphate groups of ATP through a Mg²+ ion. The V306A mutation offers harmine resistance by differentially altering Dyrk1A affinity for harmine and ATP. The V306A mutation causes no apparent alteration to Dyrk1A activity except for the reduction in ATP affinity. This deficiency could be fully compensated by supplying ATP with a concentration in the physiological range. Our results reveal that harmine inhibits Dyrk1A activity by interacting with residues in the ATP-binding pocket and displacing ATP. Our results also suggest that harmine will be a good lead compound for further designing of selective ATP-competitive Dyrk1A inhibitors through exploration of the ATP-binding pocket of Dyrk1A.


Assuntos
Trifosfato de Adenosina/metabolismo , Ligação Competitiva , Harmina/metabolismo , Harmina/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Animais , Ligação Competitiva/efeitos dos fármacos , Harmina/análogos & derivados , Cinética , Camundongos , Mutagênese Sítio-Dirigida , Mutação , Células NIH 3T3 , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Especificidade por Substrato , Quinases Dyrk
11.
Ann Nucl Med ; 25(2): 117-24, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21058048

RESUMO

OBJECTIVE: A radioiodinated analog of PD153035 (m-IPQ) was evaluated as a potential epidermal growth factor receptor tyrosine kinase (EGFR-TK) activity imaging ligand for SPECT. METHODS: The 50% inhibition concentration (IC50) value of m-IPQ for EGFR-TK phosphorylation inhibition was evaluated and compared to various EGFR-TK inhibitors. [(125)I]m-IPQ was synthesized by iododestannylation reaction. Biodistribution study of [(125)I]m-IPQ was conducted in normal mice and tumor-bearing mice. The selectivity and binding characteristics (B(max) and K(d)) were analyzed. RESULTS: The quinazoline derivative m-IPQ was found to have high inhibitory potency (IC50: 50.5 ± 3.5 nM) and selectivity toward EGFR-TK. In vivo biodistribution studies of [(125)I]m-IPQ demonstrated its rapid clearance and low retention in normal tissue. On the other hand, high tumor uptake was observed. However, the increase in [(125)I]m-IPQ uptake in the stomach as a deiodination parameter was found. Thus, [(125)I]m-IPQ showed low in vivo stability. The selectivity toward EGFR-TK of m-IPQ was confirmed by the pretreatment experiment with EGFR-TK specific inhibitors, PD153035, Genistein. [(125)I]m-IPQ bound to single population of binding sites with high affinity and kinetic parameter. In addition, [(125)I]m-IPQ was bound to EGFR-TK according to the amount of EGFR-TK expression in the tumor. CONCLUSIONS: [(125)I]m-IPQ showed a relatively high tumor accumulation with selective EGFR-TK binding. Moreover, the tumor uptake of [(125)I]m-IPQ might be reflected in the amount of EGFR-TK expression in the tumor. These good characteristics of [(125)I]m-IPQ suggested that a ¹²³I-labeled counterpart, [¹²³I]m-IPQ, would have great potential for EGFR-TK imaging with SPECT. However, the in vivo stability of this compound needs to improve.


Assuntos
Receptores ErbB/metabolismo , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/metabolismo , Quinazolinas/química , Quinazolinas/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único , Animais , Linhagem Celular Tumoral , Receptores ErbB/antagonistas & inibidores , Humanos , Radioisótopos do Iodo/química , Ligantes , Masculino , Camundongos , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacocinética , Quinazolinas/farmacologia , Especificidade por Substrato
12.
Curr Med Chem ; 17(25): 2804-21, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20586715

RESUMO

Protein kinases represent an attractive target in oncology drug discovery. Most of kinase inhibitors are ATP-competitive and are called type I inhibitors. The ATP-binding pocket is highly conserved among members of the kinase family and it is difficult to find selective agents. Moreover, the ATP-competitive inhibitors must compete with high intracellular ATP levels leading to a discrepancy between IC50s measured by biochemical versus cellular assays. The non-ATP competitive inhibitors, called type II and type III inhibitors, offer the possibility to overcome these problems. These inhibitors act by inducing a conformational shift in the target enzyme such that the kinase is no longer able to function. In the DFG-out form, the phenylalanine side chain moves to a new position. This movement creates a hydrophobic pocket available for occupation by the inhibitor. Some common features are present in these inhibitors. They contain a heterocyclic system that forms one or two hydrogen bonds with the kinase hinge residue. They also contain a hydrophobic moiety that occupies the pocket formed by the shift of phenylalanine from the DFG motif. Moreover, all the inhibitors bear a hydrogen bond donor-acceptor pair, usually urea or amide, that links the hinge-binding portion to the hydrophobic moiety and interacts with the allosteric site. Examples of non ATP-competitive inhibitors are available for various kinases. In this review small molecules capable of inducing the DFG-out conformation are reported, especially focusing on structural feature, SAR and biological properties.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Desenho de Fármacos , Ligação de Hidrogênio , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Oligopeptídeos/metabolismo , Fosfotransferases/química , Inibidores de Proteínas Quinases/análogos & derivados , Proteínas Quinases , Estrutura Terciária de Proteína
13.
Org Biomol Chem ; 8(7): 1629-39, 2010 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-20237675

RESUMO

Efficient strategies for the introduction of arginine residues featuring acetylene or azide moieties in their side chains are described. The substituents are introduced in a way that maintains the basicity of the guanidine moiety. The methodology can be used e.g. for non-invasive labeling of arginine-containing peptides. Its applicability is demonstrated by the introduction of 'click' handles into a Protein Kinase C (PKC) pseudosubstrate peptide, and the subsequent preparation and evaluation of a novel bisubstrate-based inhibitor based on such a peptide.


Assuntos
Arginina/análogos & derivados , Peptídeos/química , Inibidores de Proteínas Quinases/análogos & derivados , Sequência de Aminoácidos , Arginina/síntese química , Ciclização , Dados de Sequência Molecular , Peptídeos/síntese química , Inibidores de Proteínas Quinases/síntese química
14.
Ann Hematol ; 89(7): 653-62, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20119833

RESUMO

Treatment of acute myeloid leukemia (AML) remains challenging with many patients harboring unfavorable prognostic parameters such as FLT3 internal tandem duplication (FLT3-ITD) mutations leading to a constitutively activated FLT3-receptor tyrosine kinase (RTK). Activation of proteins by phosphorylation of tyrosine residues is a common mechanism in leukemia development. Therefore, specific tyrosine kinase inhibitors (TKI) have been developed for AML therapy and are currently under investigation. The staurosporine derivate PKC412 (Midostaurin) was found to be an effective inhibitor of the FLT3-RTK and is currently undergoing clinical trials for FLT3-mutated AML patients. Since resistance towards TKIs has been observed in vitro and in clinical trials, we have generated a PKC412-resistant clone (MV4-11r) of the human myelomonoblastic cell line MV4-11, which carries a homozygous FLT3-ITD mutation. MV4-11r displayed higher vitality after addition of PKC412 compared with MV4-11 with a pronounced reduction of apoptotic cells. Cytogenetic characterization revealed the acquisition of additional aberrations in the resistant cell line such as clonal alterations at chromosome 13q with additional FLT3 signals. Microarray analysis revealed significant expression changes in several genes prior to and after incubation with PKC412. The expression status of candidate genes being regulated by FLT-ITD like JAG1, p53, MCL-1, C-KIT, and FLT3/-L was confirmed by real-time PCR. In summary, resistance against PKC412 appears to be mediated by up-regulation of anti-apoptotic genes and down-regulation of proapoptotic signals as well as genes that are involved in normal and malignant hematopoiesis.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Leucemia Mieloide Aguda/enzimologia , Mutação , Inibidores de Proteínas Quinases/farmacologia , Estaurosporina/análogos & derivados , Tirosina Quinase 3 Semelhante a fms/metabolismo , Linhagem Celular Tumoral , Cromossomos Humanos Par 13/genética , Cromossomos Humanos Par 13/metabolismo , Ensaios Clínicos como Assunto , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica/genética , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Inibidores de Proteínas Quinases/análogos & derivados , Estaurosporina/farmacologia , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Tirosina Quinase 3 Semelhante a fms/genética
15.
J Chem Inf Model ; 49(12): 2650-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19995092

RESUMO

f152A1 was isolated from a fermentation broth of Curvularia verruculosa and characterized as a potent inhibitor of TNFalpha transcription, with anti-inflammatory activity. f152A1 and several analogues displayed inhibitory activity against the MAP kinases ERK2 and MEK1 in in vitro kinase assays. Through SAR studies on f152A1 and analogues prepared via total synthesis, we have identified structural features that contribute to inhibitory activity. To rationalize these results and to aid in the discovery process, a combination of high temperature molecular dynamics and MOPAC AM1 semiempirical molecular orbital method studies was used in studies that yielded a postulated active conformation, M1(8). This active conformation M1(8) reflects a high degree of conformational similarity among f152A1 and its more potent analogues. In view of the highly reactive cis-enone moiety in the flexible 14-membered resorcylic acid lactone ring of f152A1, the chemical reactivities of the enone moieties in various analogues were assessed by molecular orbital calculations. The enone reactivity analyses suggested that these inhibitors were prone to Michael addition at the alpha,beta-unsaturated ketone moiety and might chemically react with cysteine residues in the ATP-binding site of MAP kinases. Reactivity of the cis-enone moiety and the M1(8) conformation make important contributions to the inhibitory activity of MAP kinases.


Assuntos
Lactonas/química , Lactonas/farmacologia , MAP Quinase Quinase 1/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Modelos Moleculares , Inibidores de Proteínas Quinases/farmacologia , Sequência de Aminoácidos , Descoberta de Drogas , Humanos , Hidróxidos/química , MAP Quinase Quinase 1/química , Proteína Quinase 1 Ativada por Mitógeno/química , Conformação Molecular , Dados de Sequência Molecular , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/química , Estereoisomerismo , Temperatura
16.
J Med Chem ; 52(14): 4380-90, 2009 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-19555113

RESUMO

Cdc7 kinase has recently emerged as an attractive target for cancer therapy and low-molecular-weight inhibitors of Cdc7 kinase have been found to be effective in the inhibition of tumor growth in animal models. In this paper, we describe synthesis and structure-activity relationships of new 1H-pyrrolo[2,3-b]pyridine derivatives identified as inhibitors of Cdc7 kinase. Progress from (Z)-2-phenyl-5-(1H-pyrrolo[2,3-b]pyridin-3-ylmethylene)-3,5-dihydro-4H-imidazol-4-one (1) to [(Z)-2-(benzylamino)-5-(1H-pyrrolo[2,3-b]pyridin-3-ylmethylene)-1,3-thiazol-4(5H)-one] (42), a potent ATP mimetic inhibitor of Cdc7 kinase with IC(50) value of 7 nM, is also reported.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Piridinas/síntese química , Piridinas/farmacologia , Proteínas de Ciclo Celular/química , Linhagem Celular , Humanos , Modelos Moleculares , Conformação Molecular , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/química , Piridinas/química , Relação Estrutura-Atividade
17.
J Med Chem ; 52(14): 4466-80, 2009 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-19522465

RESUMO

Inhibition of tumor angiogenesis through blockade of the vascular endothelial growth factor (VEGF) signaling pathway is a new treatment modality in oncology. Preclinical findings suggest that blockade of additional pro-angiogenic kinases, such as fibroblast and platelet-derived growth factor receptors (FGFR and PDGFR), may improve the efficacy of pharmacological cancer treatment. Indolinones substituted in position 6 were identified as selective inhibitors of VEGF-, PDGF-, and FGF-receptor kinases. In particular, 6-methoxycarbonyl-substituted indolinones showed a highly favorable selectivity profile. Optimization identified potent inhibitors of VEGF-related endothelial cell proliferation with additional efficacy on pericyctes and smooth muscle cells. In contrast, no direct inhibition of tumor cell proliferation was observed. Compounds 2 (BIBF 1000) and 3 (BIBF 1120) are orally available and display encouraging efficacy in in vivo tumor models while being well tolerated. The triple angiokinase inhibitor 3 is currently in phase III clinical trials for the treatment of nonsmall cell lung cancer.


Assuntos
Indóis/síntese química , Indóis/farmacologia , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Administração Oral , Animais , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto , Descoberta de Drogas , Feminino , Humanos , Indóis/química , Indóis/uso terapêutico , Concentração Inibidora 50 , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/uso terapêutico , Especificidade por Substrato
18.
Bioorg Med Chem Lett ; 19(8): 2333-7, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19286381

RESUMO

2,7-Diamino-thiazolo[4,5-d]pyrimidine analogues were synthesized as novel epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors. Representative compounds showed potent and selective EGFR inhibitory activities and inhibited in vitro cellular proliferation in EGFR-overexpressing human tumor cells. The synthesis and preliminary biological, physical, and pharmacokinetic evaluation of these thiazolopyrimidine compounds are reported.


Assuntos
Antineoplásicos/síntese química , Receptores ErbB/antagonistas & inibidores , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/síntese química , Pirimidinas/síntese química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia
19.
J Med Chem ; 51(17): 5229-42, 2008 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-18698753

RESUMO

Cyclin-dependent kinases (CDKs) and casein kinases 1 (CK1) are involved in the two key molecular features of Alzheimer's disease, production of amyloid-beta peptides (extracellular plaques) and hyper-phosphorylation of Tau (intracellular neurofibrillary tangles). A series of 2,6,9-trisubstituted purines, structurally related to the CDK inhibitor roscovitine, have been synthesized. They mainly differ by the substituent on the C-6 position. These compounds were screened for kinase inhibitory activities and antiproliferative effects. Several biaryl derivatives displayed potent inhibition of both CDKs and CK1. In particular, derivative 13a was a potent inhibitor of CDK1/cyclin B (IC 50: 220 nM), CDK5/p25 (IC 50: 80 nM), and CK1 (IC 50: 14 nM). Modeling of these molecules into the ATP-binding pocket of CK1delta provided a rationale for the increased selectivity toward this kinase. 13a was able to prevent the CK1-dependent production of amyloid-beta in a cell model. CDK/CK1 dual-specificity inhibitors may have important applications in Alzheimer's disease and cancers.


Assuntos
Caseína Quinase I/antagonistas & inibidores , Quinases Ciclina-Dependentes/antagonistas & inibidores , Purinas/química , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/biossíntese , Animais , Sítios de Ligação , Linhagem Celular , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Roscovitina , Relação Estrutura-Atividade
20.
J Mol Graph Model ; 27(3): 244-54, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18585943

RESUMO

One of the most intensely studied tyrosine kinases is the epidermal growth factor receptor (EGFR). The tyrosine kinase receptors are known to be over expressed in some solid tumors and non-small cell lung cancers, causing differential susceptibility to the quinazoline inhibitors. In this study we have taken SYK tyrosine kinase coordinates from PDB database to model two new EGFR receptors with these mutations G695S and L834R and conducted all the docking studies of the inhibitors, also evaluated these two models for quality of structure using PROCHECK. Seven quinazoline analogues (gefitinib, erlotinib, CI-1033, and EKB-569 and other analogues) were selected for comparisons among the two new models. This study determined the receptor/inhibitor interactions, at that active domain binding sites consisting of 15 amino acids. We were able to calculate the energy data for each of the seven inhibitors. This data has been important in interpreting the affinity between the inhibitors evaluated against the three models of EGFR (wild-type and two mutated types). "Affinity"-based studies have indicated the order of response based on docking energy levels (Van der Waals and electrostatic interactions). The active ATP binding sites consisting of 15 amino acid residues were identified and the total energy (E(total)) which showed the affinity between the inhibitor molecules and the receptor (Van der Waals and electrostatic interactions). The selection of the quinazoline analogues was purely on their emergence as possible candidates in the drug discovery areas. This study describes the successful application of these models that we constructed for molecular docking studies to rationally design compounds predicted to bind favorably to the modeled EGFR catalytic sites.


Assuntos
Receptores ErbB/antagonistas & inibidores , Receptores ErbB/química , Modelos Moleculares , Proteínas Mutantes/química , Inibidores de Proteínas Quinases/análogos & derivados , Quinazolinas/química , Quinazolinas/farmacologia , Homologia Estrutural de Proteína , Sequência de Aminoácidos , Aminoácidos , Sítios de Ligação , Domínio Catalítico , Ligantes , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Estrutura Secundária de Proteína , Alinhamento de Sequência , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA