Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Molecules ; 29(8)2024 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-38675594

RESUMO

Cancer is a serious threat to human life and social development and the use of scientific methods for cancer prevention and control is necessary. In this study, HQSAR, CoMFA, CoMSIA and TopomerCoMFA methods are used to establish models of 65 imidazo[4,5-b]pyridine derivatives to explore the quantitative structure-activity relationship between their anticancer activities and molecular conformations. The results show that the cross-validation coefficients q2 of HQSAR, CoMFA, CoMSIA and TopomerCoMFA are 0.892, 0.866, 0.877 and 0.905, respectively. The non-cross-validation coefficients r2 are 0.948, 0.983, 0.995 and 0.971, respectively. The externally validated complex correlation coefficients r2pred of external validation are 0.814, 0.829, 0.758 and 0.855, respectively. The PLS analysis verifies that the QSAR models have the highest prediction ability and stability. Based on these statistics, virtual screening based on R group is performed using the ZINC database by the Topomer search technology. Finally, 10 new compounds with higher activity are designed with the screened new fragments. In order to explore the binding modes and targets between ligands and protein receptors, these newly designed compounds are conjugated with macromolecular protein (PDB ID: 1MQ4) by molecular docking technology. Furthermore, to study the nature of the newly designed compound in dynamic states and the stability of the protein-ligand complex, molecular dynamics simulation is carried out for N3, N4, N5 and N7 docked with 1MQ4 protease structure for 50 ns. A free energy landscape is computed to search for the most stable conformation. These results prove the efficient and stability of the newly designed compounds. Finally, ADMET is used to predict the pharmacology and toxicity of the 10 designed drug molecules.


Assuntos
Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Inibidores de Proteínas Quinases , Piridinas , Relação Quantitativa Estrutura-Atividade , Piridinas/química , Piridinas/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Humanos , Aurora Quinases/antagonistas & inibidores , Aurora Quinases/química , Aurora Quinases/metabolismo , Imidazóis/química , Imidazóis/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia
2.
Commun Biol ; 3(1): 701, 2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33219324

RESUMO

Mitosis has been validated by numerous anti-cancer drugs as being a druggable process, and selective inhibition of parasite proliferation provides an obvious opportunity for therapeutic intervention against malaria. Mitosis is controlled through the interplay between several protein kinases and phosphatases. We show here that inhibitors of human mitotic kinases belonging to the Aurora family inhibit P. falciparum proliferation in vitro with various potencies, and that a genetic selection for mutant parasites resistant to one of the drugs, Hesperadin, identifies a resistance mechanism mediated by a member of a different kinase family, PfNek1 (PF3D7_1228300). Intriguingly, loss of PfNek1 catalytic activity provides protection against drug action. This points to an undescribed functional interaction between Ark and Nek kinases and shows that existing inhibitors can be used to validate additional essential and druggable kinase functions in the parasite.


Assuntos
Aurora Quinases , Epistasia Genética , Indóis/farmacologia , Quinase 1 Relacionada a NIMA , Plasmodium falciparum , Sulfonamidas/farmacologia , Aurora Quinases/antagonistas & inibidores , Aurora Quinases/química , Aurora Quinases/metabolismo , Epistasia Genética/efeitos dos fármacos , Epistasia Genética/genética , Humanos , Quinase 1 Relacionada a NIMA/química , Quinase 1 Relacionada a NIMA/genética , Quinase 1 Relacionada a NIMA/metabolismo , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo
3.
Commun Biol ; 2: 477, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31886415

RESUMO

The mitochondrion is an organelle that was derived from an endosymbiosis. Although regulation of mitochondrial growth by the host cell is necessary for the maintenance of mitochondria, it is unclear how this regulatory mechanism was acquired. To address this, we studied the primitive unicellular red alga Cyanidioschyzon merolae, which has the simplest eukaryotic genome and a single mitochondrion. Here we show that the C. merolae Aurora kinase ortholog CmAUR regulates mitochondrial division through phosphorylation of mitochondrial division ring components. One of the components, the Drp1 ortholog CmDnm1, has at least four sites phosphorylated by CmAUR. Depletion of the phosphorylation site conserved among eukaryotes induced defects such as mitochondrial distribution on one side of the cell. Taken together with the observation that human Aurora kinase phosphorylates Drp1 in vitro, we suggest that the phosphoregulation is conserved from the simplest eukaryotes to mammals, and was acquired at the primitive stage of endosymbiosis.


Assuntos
Aurora Quinases/genética , Aurora Quinases/metabolismo , Evolução Biológica , Mitocôndrias/genética , Mitocôndrias/metabolismo , Rodófitas/genética , Rodófitas/metabolismo , Aurora Quinases/química , Mitose , Fosforilação , Rodófitas/enzimologia , Especificidade por Substrato
4.
OMICS ; 23(10): 486-495, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31549911

RESUMO

Thyroid cancer (TC) is a very common malignancy worldwide. Chief among the innovative molecular drug targets for TC are epigenetic modifications. Increased telomerase activity in cancer cells makes telomerase a novel target for epigenetic anticancer drug innovation. Recently, telomerase reverse transcriptase (TERT) gene promoter (TERTp) mutations (C228T and C250T) were reported at high frequency in TC cell lines and tumor biopsies. In this study, three representative TC cell lines, mutant TERTp (TPC1), mutant BRAF/TERTp (KTC2), and wild-type TERTp (WRO), were screened with a drug library composed of 51 epigenetic drugs: 14 Aurora kinase inhibitors; 23 histone deacetylase inhibitors; 5 sirtuin modifiers; 3 hypoxia-inducible factor inhibitors; 2 DNA methyltransferase inhibitors; 2 histone methyltransferase inhibitors, a histone demethylase inhibitor, and a bromodomain inhibitor. Effects of the drugs on cell growth at 48 and 72 h were compared. PF-03814735, a small-molecule inhibitor of Aurora kinase A (IC50 = 0.8 nM) and B (IC50 = 5 nM), was the most potent on KTC2 cells, whereas CUDC-101, a multitarget inhibitor, was effective on both WRO and KTC2 cells. Notably, PF-03814735 was found to be the most effective epigenetic drug on cell lines harboring the C228T mutation. In conclusion, these new findings offer specific guidance on dose and time course selection to design novel therapeutic interventions against TC using PF-03814735, and specifically target cells carrying the TERTpC228T mutation. In a larger context of drug discovery science, these findings inform new strategies to forecast optimal treatment regimens for TC, particularly with Aurora kinase inhibitors and in ways guided by epigenetic drug design.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Aurora Quinases/antagonistas & inibidores , Aurora Quinases/química , Desenho de Fármacos , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Biomarcadores Tumorais , Linhagem Celular Tumoral , Epigênese Genética/efeitos dos fármacos , Humanos , Mutação , Telomerase/genética , Neoplasias da Glândula Tireoide
5.
PLoS Negl Trop Dis ; 13(5): e0007425, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31095613

RESUMO

BACKGROUND: The tumor-like growth of the metacestode larvae of the tapeworm E. multilocularis causes human alveolar echinococcosis, a severe disease mainly affecting the liver. The germinative cells, a population of adult stem cells, are crucial for the larval growth and development of the parasite within the hosts. Maintenance of the germinative cell pools relies on their abilities of extensive proliferation and self-renewal, which requires accurate control of the cell division cycle. Targeting regulators of the cell division progression may impair germinative cell populations, leading to impeded parasite growth. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we describe the characterization of EmAURKA and EmAURKB, which display significant similarity to the members of Aurora kinases that are essential mitotic kinases and play key roles in cell division. Our data suggest that EmAURKA and EmAURKB are actively expressed in the germinative cells of E. multilocularis. Treatment with low concentrations of MLN8237, a dual inhibitor of Aurora A and B, resulted in chromosomal defects in the germinative cells during mitosis, while higher concentrations of MLN8237 caused a failure in cytokinesis of the germinative cells, leading to multinucleated cells. Inhibition of the activities of Aurora kinases eventually resulted in depletion of the germinative cell populations in E. multilocularis, which in turn caused larval growth inhibition of the parasite. CONCLUSIONS/SIGNIFICANCE: Our data demonstrate the vital roles of Aurora kinases in the regulation of mitotic progression and maintenance of the germinative cells in E. multilocularis, and suggest Aurora kinases as promising druggable targets for the development of novel chemotherapeutics against human alveolar echinococcosis.


Assuntos
Aurora Quinases/metabolismo , Equinococose/parasitologia , Echinococcus multilocularis/enzimologia , Echinococcus multilocularis/crescimento & desenvolvimento , Proteínas de Helminto/metabolismo , Sequência de Aminoácidos , Animais , Aurora Quinases/antagonistas & inibidores , Aurora Quinases/química , Aurora Quinases/genética , Azepinas/farmacologia , Clonagem Molecular , Echinococcus multilocularis/efeitos dos fármacos , Echinococcus multilocularis/genética , Proteínas de Helminto/antagonistas & inibidores , Proteínas de Helminto/química , Proteínas de Helminto/genética , Humanos , Larva/enzimologia , Larva/genética , Larva/crescimento & desenvolvimento , Mitose , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Alinhamento de Sequência
6.
Int J Mol Sci ; 19(12)2018 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-30562979

RESUMO

Members of the aurora kinase family are Ser/Thr kinases involved in regulating mitosis. Multiple promising clinical trials to target aurora kinases are in development. To discover flavones showing growth inhibitory effects on cancer cells, 36 flavone derivatives were prepared, and their cytotoxicity was measured using a long-term clonogenic survival assay. Their half-maximal growth inhibitory effects against HCT116 human colon cancer cells were observed at the sub-micromolar level. Pharmacophores were derived based on three-dimensional quantitative structure⁻activity calculations. Because plant-derived flavones inhibit aurora kinase B, we selected 5-methoxy-2-(2-methoxynaphthalen-1-yl)-4H-chromen-4-one (derivative 31), which showed the best half-maximal cell growth inhibitory effect, and tested whether it can inhibit aurora kinases in HCT116 colon cancer cells. We found that derivative 31 inhibited the phosphorylation of aurora kinases A, aurora kinases B and aurora kinases C, suggesting that derivative 31 is a potential pan-aurora kinase inhibitor. The results of our analysis of the binding modes between derivative 31 and aurora A and aurora B kinases using in-silico docking were consistent with the pharmacophores proposed in this study.


Assuntos
Apoptose/efeitos dos fármacos , Aurora Quinases/antagonistas & inibidores , Neoplasias do Colo/enzimologia , Flavonas/farmacologia , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Proteínas de Neoplasias/antagonistas & inibidores , Aurora Quinases/química , Aurora Quinases/genética , Aurora Quinases/metabolismo , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Flavonas/síntese química , Flavonas/química , Células HCT116 , Humanos , Simulação de Acoplamento Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
7.
Chromosoma ; 126(1): 93-103, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27106516

RESUMO

The coordination between late mitotic events such as poleward chromosome motion, spindle elongation, DNA decondensation, and nuclear envelope reformation (NER) is crucial for the completion of chromosome segregation at the anaphase-telophase transition. Mitotic exit is driven by a decrease of Cdk1 kinase activity and an increase of PP1/PP2A phosphatase activities. More recently, Aurora kinases have also emerged as master regulators of late mitotic events and cytokinesis. Aurora A is mainly associated with spindle poles throughout mitosis and midbody during telophase, whereas Aurora B re-localizes from centromeres in early mitosis to the spindle midzone and midbody as cells progress from anaphase to the completion of cytokinesis. Functional studies, together with the identification of a phosphorylation gradient during anaphase, established Aurora B as a major player in the organization of the spindle midzone and in the spatiotemporal coordination between chromosome segregation and NER. Aurora A has been less explored, but a cooperative role in spindle midzone stability has also been proposed, implying that both Aurora A and B contribute to accurate chromosome segregation during mitotic exit. Here, we review the roles of the Aurora kinases in the regulation of late mitotic events and discuss how they work together with other mitotic players to ensure an error-free mitosis.


Assuntos
Aurora Quinases/metabolismo , Mitose , Animais , Aurora Quinases/química , Aurora Quinases/genética , Cromossomos/genética , Cromossomos/metabolismo , Humanos , Fosforilação , Ligação Proteica , Proteólise , Transdução de Sinais , Fuso Acromático/metabolismo , Especificidade por Substrato
8.
Cell Chem Biol ; 23(10): 1193-1205, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27693059

RESUMO

Allostery is a phenomenon observed in many proteins where binding of a macromolecular partner or a small-molecule ligand at one location leads to specific perturbations at a site not in direct contact with the region where the binding occurs. The list of proteins under allosteric regulation includes AGC protein kinases. AGC kinases have a conserved allosteric site, the phosphoinositide-dependent protein kinase 1 (PDK1)-interacting fragment (PIF) pocket, which regulates protein ATP-binding, activity, and interaction with substrates. In this study, we identify small molecules that bind to the ATP-binding site and affect the PIF pocket of AGC kinase family members, PDK1 and Aurora kinase. We describe the mechanistic details and show that although PDK1 and Aurora kinase inhibitors bind to the conserved ATP-binding site, they differentially modulate physiological interactions at the PIF-pocket site. Our work outlines a strategy for developing bidirectional small-molecule allosteric modulators of protein kinases and other signaling proteins.


Assuntos
Trifosfato de Adenosina/metabolismo , Regulação Alostérica/efeitos dos fármacos , Indazóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirimidinas/farmacologia , Sítio Alostérico/efeitos dos fármacos , Aurora Quinases/antagonistas & inibidores , Aurora Quinases/química , Aurora Quinases/metabolismo , Sítios de Ligação/efeitos dos fármacos , Células HEK293 , Humanos , Indazóis/química , Simulação de Acoplamento Molecular , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Pirimidinas/química , Piruvato Desidrogenase Quinase de Transferência de Acetil
9.
Mol Cancer Ther ; 15(10): 2388-2398, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27496137

RESUMO

Although the MAPK pathway is frequently deregulated in cancer, inhibitors targeting RAF or MEK have so far shown clinical activity only in BRAF- and NRAS-mutant melanoma. Improvements in efficacy may be possible by combining inhibition of mitogenic signal transduction with inhibition of cell-cycle progression. We have studied the preclinical pharmacology of BI 847325, an ATP-competitive dual inhibitor of MEK and Aurora kinases. Potent inhibition of MEK1/2 and Aurora A/B kinases by BI 847325 was demonstrated in enzymatic and cellular assays. Equipotent effects were observed in BRAF-mutant cells, whereas in KRAS-mutant cells, MEK inhibition required higher concentrations than Aurora kinase inhibition. Daily oral administration of BI 847325 at 10 mg/kg showed efficacy in both BRAF- and KRAS-mutant xenograft models. Biomarker analysis suggested that this effect was primarily due to inhibition of MEK in BRAF-mutant models but of Aurora kinase in KRAS-mutant models. Inhibition of both MEK and Aurora kinase in KRAS-mutant tumors was observed when BI 847325 was administered once weekly at 70 mg/kg. Our studies indicate that BI 847325 is effective in in vitro and in vivo models of cancers with BRAF and KRAS mutation. These preclinical data are discussed in the light of the results of a recently completed clinical phase I trial assessing safety, tolerability, pharmacokinetics, and efficacy of BI 847325 in patients with cancer. Mol Cancer Ther; 15(10); 2388-98. ©2016 AACR.


Assuntos
Antineoplásicos/farmacologia , Aurora Quinases/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Aurora Quinases/química , Aurora Quinases/metabolismo , Ligação Competitiva , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/química , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Modelos Moleculares , Conformação Molecular , Ligação Proteica , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/química , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Cancer Res Clin Oncol ; 142(9): 1995-2012, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26932147

RESUMO

Compounds that affect enzymatic function of kinases are valuable for the understanding of the complex biochemical processes in cells. Aurora kinases (AURKs) play a key role in the control of the mitosis. These kinases are frequently deregulated in different human cancers: overexpression, amplifications, translocations and deletions were reported in many cancer cell lines as well as patient tissues. These findings steered a rigorous hunt for small-molecule AURK inhibitors not only for research purposes as well as for therapeutic uses. In this review, we describe a number of AURK inhibitors and their use in cancer research and/or therapy. We hope to assist researchers and clinicians in deciding which inhibitor is most appropriate for their specific purpose. The review will also provide a broad overview of the clinical studies performed with some of these inhibitors (if such studies have been performed).


Assuntos
Aurora Quinases/antagonistas & inibidores , Pesquisa Biomédica/métodos , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Animais , Aurora Quinases/química , Humanos , Modelos Moleculares , Inibidores de Proteínas Quinases/química
11.
Arch Pharm (Weinheim) ; 349(3): 193-201, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26773437

RESUMO

A series of benzimidazole-based quinazoline derivatives with different substitutions of primary and secondary amines at the C2 position (1-12) were evaluated for their Aurora kinase inhibitory activities. All compounds except for 3 and 6 showed good activity against Aurora kinase inhibitors, with IC50 values in the range of 0.035-0.532 µM. The ligand efficiency (LE) of the compounds with Aurora A kinase was also determined. The structure-activity relationship and the quantitative structure-activity relationship revealed that the Aurora inhibitory activities of these derivatives primarily depend on the different substitutions of the amine present at the C2 position of the quinazoline core. Molecular docking studies in the active binding site also provided theoretical support for the experimental biological data acquired. The current study identifies a novel class of Aurora kinase inhibitors, which can further be used for the treatment of cancer.


Assuntos
Aurora Quinases/antagonistas & inibidores , Benzimidazóis/química , Quinazolinas/química , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/química , Aurora Quinases/química , Benzimidazóis/síntese química , Simulação de Acoplamento Molecular , Relação Quantitativa Estrutura-Atividade , Quinazolinas/síntese química
12.
Bioorg Med Chem Lett ; 25(19): 4203-9, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26296477

RESUMO

Introduction of a 1-benzyl-1H-pyrazol-4-yl moiety at C7 of the imidazo[4,5-b]pyridine scaffold provided 7a which inhibited a range of kinases including Aurora-A. Modification of the benzyl group in 7a, and subsequent co-crystallisation of the resulting analogues with Aurora-A indicated distinct differences in binding mode dependent upon the pyrazole N-substituent. Compounds 7a and 14d interact with the P-loop whereas 14a and 14b engage with Thr217 in the post-hinge region. These crystallographic insights provide options for the design of compounds interacting with the DFG motif or with Thr217.


Assuntos
Aurora Quinases/antagonistas & inibidores , Aurora Quinases/química , Imidazóis/síntese química , Imidazóis/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/síntese química , Pirazóis/farmacologia , Piridinas/síntese química , Piridinas/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cristalização , Relação Dose-Resposta a Droga , Humanos , Imidazóis/química , Camundongos , Microssomos Hepáticos/química , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Estrutura Molecular , Pirazóis/química , Piridinas/química , Relação Estrutura-Atividade
13.
Curr Cancer Drug Targets ; 15(5): 375-93, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25895501

RESUMO

Aurora kinase A, B and C, are key regulators of mitosis and are over expressed in many of the human cancers, making them an ideal drug target for cancer chemotherapy. Currently, over a dozen of Aurora kinase inhibitors are in various phases of clinical development. The majority of the inhibitors (VX-680/MK-0457, PHA-739358, CYC116, SNS-314, AMG 900, AT-9283, SCH- 1473759, ABT-348, PF-03814735, R-763/AS-703569, KW-2449 and TAK-901) are pan-selective (isoform non-selective) and few are Aurora A (MLN8054, MLN8237, VX-689/MK5108 and ENMD 2076) and Aurora B (AZD1152 and GSK1070916) sub-type selective. Despite the intensive research efforts in the past decade, no Aurora kinase inhibitor has reached the market. Recent evidence suggests that the sub-type selective Aurora kinase A inhibitor could possess advantages over pan-selective Aurora inhibitors, by avoiding Aurora B mediated neutropenia. However, sub-type selective Aurora kinase A inhibitor design is very challenging due to the similarity in the active site among the isoforms. Structural biology and computational aspects pertaining to the design of Aurora kinase inhibitors were analyzed and found that a possible means to develop sub-type selective inhibitor is by targeting Aurora A specific residues (Leu215, Thr217 and Arg220) or Aurora B specific residues (Arg159, Glu161 and Lys164), near the solvent exposed region of the protein. Particularly, a useful strategy for the design of sub-type selective Aurora A inhibitor could be by targeting Thr217 residue as in the case of MLN8054. Further preclinical and clinical studies with the sub-type selective Aurora inhibitors could help bring them to the market for the treatment of cancer.


Assuntos
Aurora Quinases/antagonistas & inibidores , Aurora Quinases/química , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Animais , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/química , Aurora Quinase A/metabolismo , Aurora Quinase B/antagonistas & inibidores , Aurora Quinase B/química , Aurora Quinase B/metabolismo , Aurora Quinases/metabolismo , Benzamidas/química , Benzamidas/metabolismo , Benzamidas/farmacologia , Sítios de Ligação/fisiologia , Humanos , Indazóis/química , Indazóis/metabolismo , Indazóis/farmacologia , Piperazinas/química , Piperazinas/metabolismo , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/metabolismo , Estrutura Secundária de Proteína , Pirazóis/química , Pirazóis/metabolismo , Pirazóis/farmacologia
14.
J Med Chem ; 58(1): 362-75, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25369539

RESUMO

The Aurora family of serine/threonine kinases is essential for mitosis. Their crucial role in cell cycle regulation and aberrant expression in a broad range of malignancies have been demonstrated and have prompted intensive search for small molecule Aurora inhibitors. Indeed, over 10 of them have reached the clinic as potential anticancer therapies. We report herein the discovery and optimization of a novel series of tricyclic molecules that has led to SAR156497, an exquisitely selective Aurora A, B, and C inhibitor with in vitro and in vivo efficacy. We also provide insights into its mode of binding to its target proteins, which could explain its selectivity.


Assuntos
Antineoplásicos/farmacologia , Aurora Quinases/antagonistas & inibidores , Benzimidazóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quinolonas/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/química , Aurora Quinase A/metabolismo , Aurora Quinase B/antagonistas & inibidores , Aurora Quinase B/química , Aurora Quinase B/metabolismo , Aurora Quinase C/antagonistas & inibidores , Aurora Quinase C/química , Aurora Quinase C/metabolismo , Aurora Quinases/química , Aurora Quinases/metabolismo , Benzimidazóis/química , Benzimidazóis/metabolismo , Feminino , Células HCT116 , Humanos , Camundongos SCID , Modelos Químicos , Modelos Moleculares , Estrutura Molecular , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Ligação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína , Quinolonas/química , Quinolonas/metabolismo , Células Sf9 , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
PLoS One ; 9(12): e113773, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25485544

RESUMO

The present study examines the conformational transitions occurring among the major structural motifs of Aurora kinase (AK) concomitant with the DFG-flip and deciphers the role of non-covalent interactions in rendering specificity. Multiple sequence alignment, docking and structural analysis of a repertoire of 56 crystal structures of AK from Protein Data Bank (PDB) has been carried out. The crystal structures were systematically categorized based on the conformational disposition of the DFG-loop [in (DI) 42, out (DO) 5 and out-up (DOU) 9], G-loop [extended (GE) 53 and folded (GF) 3] and αC-helix [in (CI) 42 and out (CO) 14]. The overlapping subsets on categorization show the inter-dependency among structural motifs. Therefore, the four distinct possibilities a) 2W1C (DI, CI, GE) b) 3E5A (DI, CI, GF) c) 3DJ6 (DI, CO, GF) d) 3UNZ (DOU, CO, GF) along with their co-crystals and apo-forms were subjected to molecular dynamics simulations of 40 ns each to evaluate the variations of individual residues and their impact on forming interactions. The non-covalent interactions formed by the 157 AK co-crystals with different regions of the binding site were initially studied with the docked complexes and structure interaction fingerprints. The frequency of the most prominent interactions was gauged in the AK inhibitors from PDB and the four representative conformations during 40 ns. Based on this study, seven major non-covalent interactions and their complementary sites in AK capable of rendering specificity have been prioritized for the design of different classes of inhibitors.


Assuntos
Aurora Quinases/química , Desenho de Fármacos , Inibidores de Proteínas Quinases/química , Sequência de Aminoácidos , Aurora Quinases/antagonistas & inibidores , Sítios de Ligação , Modelos Moleculares , Conformação Molecular , Dados de Sequência Molecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Inibidores de Proteínas Quinases/farmacologia , Alinhamento de Sequência , Relação Estrutura-Atividade
16.
Bioorg Med Chem ; 21(14): 4250-8, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23719279

RESUMO

Due to toxicity problems, various plant-derived compounds have been screened to find the chemotherapeutic agents. As anticancer therapeutic agents, chalcones have advantages such as poor interaction with DNA and low risk of mutagenesity. Chromenones show anticancer activities too. Therefore, hybrids of chalcone and chromenone may be potent chemotherapeutic agents. We prepared 16 synthetic chromenylchalcones and applied a clonogenic long-term survival assay method for them on HCT116 human colorectal cancer cell lines. One of chromenylchalcones tested here, chromenylchalcone 11, showed IC50 of 93.1nM which can be competed with the IC50 values of well-known flavonoids such as catechin gallate and epicatechin gallate. Further biological experiments including cell cycle analysis, apoptosis assay, Western blot analysis, and immunofluorescent microscopy were carried out for this compound. In addition, in vitro kinases binding assay performed to explain its molecular mechanism demonstrated the compound inhibited aurora kinases. The binding modes between chromenylchalcone 11 and aurora kinases were elucidated using in silico docking experiments. These findings could be used for designing cancer therapeutic or preventive plant-derived chromenylchalcone agents.


Assuntos
Antineoplásicos/síntese química , Aurora Quinases/química , Benzopiranos/síntese química , Chalconas/síntese química , Neoplasias do Colo/tratamento farmacológico , Antineoplásicos/química , Antineoplásicos/farmacologia , Aurora Quinases/metabolismo , Benzopiranos/metabolismo , Benzopiranos/farmacologia , Sítios de Ligação , Western Blotting , Catequina/química , Catequina/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Chalconas/química , Chalconas/metabolismo , Chalconas/farmacologia , Humanos , Concentração Inibidora 50 , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA