Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Curr Med Chem ; 27(40): 6787-6814, 2020.
Article in English | MEDLINE | ID: mdl-31580244

ABSTRACT

Due to the three domains of the colchicine-site which is conducive to the combination with small molecule compounds, colchicine-site on the tubulin has become a common target for antitumor drug development, and accordingly, a large number of tubulin inhibitors binding to the colchicine-site have been reported and evaluated over the past years. In this study, tubulin inhibitors targeting the colchicine-site and their application as antitumor agents were reviewed based on the literature from 2015 to 2019. Tubulin inhibitors were classified into ten categories according to the structural features, including colchicine derivatives, CA-4 analogs, chalcone analogs, coumarin analogs, indole hybrids, quinoline and quinazoline analogs, lignan and podophyllotoxin derivatives, phenothiazine analogs, N-heterocycle hybrids and others. Most of them displayed potent antitumor activity, including antiproliferative effects against Multi-Drug-Resistant (MDR) cell lines and antivascular properties, both in vitro and in vivo. In this review, the design, synthesis and the analysis of the structure-activity relationship of tubulin inhibitors targeting the colchicine-site were described in detail. In addition, multi-target inhibitors, anti-MDR compounds, and inhibitors bearing antitumor activity in vivo are further listed in tables to present a clear picture of potent tubulin inhibitors, which could be beneficial for medicinal chemistry researchers.


Subject(s)
Tubulin Modulators/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Tumor , Colchicine , Drug Screening Assays, Antitumor , Humans , Structure-Activity Relationship , Tubulin/metabolism , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use
2.
Bioorg Chem ; 92: 103219, 2019 11.
Article in English | MEDLINE | ID: mdl-31476616

ABSTRACT

Twenty-four 1,2-diarylbenzimidazole derivatives were designed, synthesized and biologically evaluated. It turned out that most of them were potential anticancer drugs. Among them, compound c24 showed the highest anti-tumor activity (GI50 = 0.71-2.41 µM against HeLa, HepG2, A549 and MCF-7 cells), and low toxicity to normal cells (CC50 > 100 µM against L02 cells). In the microtubule binding assay, c24 showed the most potent inhibition of microtubule polymerization (IC50 = 8.47 µM). The binding ability of compound c24 to tubulin crystal was verified by molecular docking simulation experiment. Further studies on HepG2 and HeLa cells showed that compound c24 could cause mitotic arrest of tumor cells to G2/M phase then inducing apoptosis. To sum up, compound c24 is a promising microtubule assembly inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Molecular Docking Simulation , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Polymerization/drug effects , Quantitative Structure-Activity Relationship , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
3.
Expert Opin Ther Pat ; 29(9): 675-688, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31370713

ABSTRACT

Introduction: RAF kinase inhibitors block and regulate RAS/RAF/MEK/ERK signaling, which is a key to tumor treatment. At present, although RAF kinase inhibitors have good efficacy, there are few such drugs with low toxicity, and thus, it is urgent to find novel RAF kinase inhibitors associated with higher activity and fewer adverse reactions. This review highlights the anti-tumor effects of several published RAF kinase inhibitors and might be helpful in providing new ideas for the development of novel drug candidates in the future. Areas covered: This article covers the pertinent literature published on RAF kinase inhibitors from 2010 to 2018, as well as the potential use of these compounds as therapeutics for cancer. Expert opinion: To date, many RAF kinase inhibitors with different structures have been studied, many of which have prominent inhibitory activities toward RAF kinase. Further, the specificity of these drugs offers hope for the targeted therapy of tumors. Although RAF kinase inhibition has achieved promising results for the treatment of many cancers, overcoming limitations associated with drug resistance and safety comprises a new direction for the optimization and improvement of RAF kinase inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , raf Kinases/antagonists & inhibitors , Animals , Drug Development/methods , Drug Resistance, Neoplasm , Humans , Molecular Targeted Therapy , Neoplasms/enzymology , Patents as Topic , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , raf Kinases/metabolism
4.
Chem Biol Drug Des ; 94(5): 1894-1904, 2019 09.
Article in English | MEDLINE | ID: mdl-31106514

ABSTRACT

Several novel cycloalkyl-fused 2,3-diaryl pyrazole derivatives were designed, synthesized, and evaluated as potential anti-tubulin agents. Compound A10 exhibited the most potent antiproliferative activity against a panel of cancer lines (IC50  = 0.78-2.42 µM) and low cytotoxicity against 293T & L02 (CC50 values of 131.74 and 174.89 µM, respectively). Moreover, A10 displayed inhibition of tubulin polymerization in vitro, arrested the G2/M phase of the cell cycle, changed morphology of tubulin, increased intracellular reactive oxygen species, and induced apoptosis of HeLa cells. Docking simulation and 3D-QSAR models were performed to elaborate on the anti-tubulin mechanism of the derivatives. The inhibition of monoclonal colony formation provided more intuitional data to verify the possibility of A10 as a novel tubulin assembling inhibitor.


Subject(s)
Antineoplastic Agents/chemical synthesis , Hydrocarbons, Cyclic/chemical synthesis , Hydrocarbons, Cyclic/metabolism , Pyrazoles/chemical synthesis , Tubulin Modulators/chemical synthesis , Tubulin/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Hydrocarbons, Cyclic/chemistry , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protein Conformation , Pyrazoles/pharmacology , Quantitative Structure-Activity Relationship , Reactive Oxygen Species/metabolism , Tubulin Modulators/pharmacology
5.
Eur J Med Chem ; 163: 896-910, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30580241

ABSTRACT

In current work, a class of novel 4,5-dihydro-1H-pyrazole-1-carboxylate derivatives (E01-E28) were designed, synthesized and evaluated. Among them, the most potent compound E24 exhibited comparable activity against a panel of cancer cells (GI50 ranging 0.05-0.98 µM) and tubulin polymerization inhibition (IC50 = 1.49 µM) with reference drug CA-4(P) (GI50 ranging 0.019-0.32 µM, IC50 = 2.18 µM). The following assays indicated that compound E24 disturbed the dynamics of tubulin catastrophe and rescue, which triggered G2/M arrest, leading to ROS accumulation, cleavage of PARP and apoptosis. Molecular dynamics simulation validated that compound E24 could tightly bind into tubulin heterodimers with ß Lys 254 and ß Cys 241 of tubulin in the docking pose. Metabolic stability and pharmacokinetics parameters were also determined. The half time (t1/2) displayed species differences in three microsomes. The plasma elimination half-life (t1/2), peak plasma concentration (Cmax), mean retention time (MRT), the area under the curve (AUC0-∞) and distribution volume (Vz) of E24 after intravenous administration were 0.90 ± 0.22 h, 594.50 ± 97.23 ng/mL, 1.09 ± 0.22 h, 413.67 ± 105.64 ng/mL*h and 5.03 ± 1.82 L/kg, respectively. In HeLa-xenografts, compound E24 exhibited obvious antitumor efficacy via the suppression of tumor growth without weight loss of body or organ. In brief, compound E24 might be a hopeful candidate with excellent properties for oncotherapy as tubulin polymerization inhibitor.


Subject(s)
Antineoplastic Agents/chemical synthesis , Polymerization/drug effects , Tubulin Modulators/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Drug Screening Assays, Antitumor , HeLa Cells , Heterografts , Humans , Mice , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Tubulin/metabolism , Tubulin Modulators/chemical synthesis
6.
Eur J Med Chem ; 156: 722-737, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-30041136

ABSTRACT

In the present work, twenty-five nicotinoyl pyrazoline derivates bearing N-methyl indole moiety have been designed and synthesized. The biological evaluation of these compounds as tubulin assembly inhibitors revealed that most of them were potential antitumor agents. Among them, compound 28 exhibited most potency against cancer cell line panels (GI50 = 29-90 nM for HeLa, HepG2 and MCF-7 cells) without toxicity to non-tumor cells (CC50 > 300 µM for 293 T cell), bound to the colchicine site of tubulin and displayed excellent inhibitory activity in tubulin assembly assay (IC50 = 1.6 µM, better than CA-4). Molecular dynamics simulation was carried out to validate the docking pose of compound 28 with tubulin crystalline. Further investigation on HepG2 and HeLa cells demonstrated that compound 28 could cause mitosis arrest to G2/M phase, and subsequently induced cell apoptosis. The efficiency in vivo of compound 28 was also evaluated on HeLa-Xenograft nude mice, and the relative tumor inhibition ration was up to 61.52% without noticeable weight loss and tissue damage (examined by H&E staining), which was comparable to CA-4 (inhibited 59.92%). In brief, compound 28 is a promising candidate for tumor therapy as tubulin assembly inhibitor.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Pyrazoles/chemistry , Pyrazoles/therapeutic use , Tubulin Modulators/chemistry , Tubulin Modulators/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Drug Design , Female , HEK293 Cells , HeLa Cells , Humans , Indoles/chemistry , Indoles/pharmacology , Indoles/therapeutic use , Mice, Nude , Mitosis/drug effects , Models, Molecular , Neoplasms/metabolism , Neoplasms/pathology , Pyrazoles/pharmacology , Tubulin Modulators/pharmacology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
7.
Chem Biol Drug Des ; 90(1): 112-118, 2017 07.
Article in English | MEDLINE | ID: mdl-28032450

ABSTRACT

A series of new 1-phenylsulphonyl-2-(1-methylindol-3-yl)-benzimidazole derivatives were designed, synthesized and evaluated as potential inhibitors of tubulin polymerization and anthropic cancer cell lines. Among them, compound 33 displayed the most potent tubulin polymerization inhibitory activity in vitro (IC50  = 1.41 µM) and strong antiproliferative activities against A549, Hela, HepG2 and MCF-7 cell lines in vitro with GI50 value of 1.6, 2.7, 2.9 and 4.3 µM, respectively, comparable with the positive control colchicine (GI50 value of 4.1, 7.2, 9.5 and 14.5 µM, respectively) and CA-4 (GI50 value of 2.2, 4.3, 6.4 and 11.4 µM, respectively). Simultaneously, we evaluated that compound 33 could effectively induce apoptosis of A549 associated with G2/M phase cell cycle arrest. Immunofluorescence microscopy also clearly indicated compound 33 a potent antimicrotubule agent. Docking simulation showed that compound 33 could bind tightly with the colchicine-binding site and act as a tubulin inhibitor. Three-dimensional-QSAR model was also built to provide more pharmacophore understanding that could be used to design new agents with more potent tubulin assembling inhibitory activity in the future.


Subject(s)
Benzimidazoles/chemistry , Tubulin Modulators/chemical synthesis , Tubulin/metabolism , A549 Cells , Animals , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Binding Sites , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Cells, Cultured , Colchicine/chemistry , Colchicine/metabolism , Colchicine/pharmacology , Crystallography, X-Ray , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Mice , Molecular Docking Simulation , Protein Structure, Tertiary , Quantitative Structure-Activity Relationship , Tubulin/chemistry , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
8.
ChemMedChem ; 11(13): 1446-58, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27159418

ABSTRACT

A series of 1-methyl-1H-indole-pyrazoline hybrids were designed, synthesized, and biologically evaluated as potential tubulin polymerization inhibitors. Among them, compound e19 [5-(5-bromo-1-methyl-1H-indol-3-yl)-3-(3,4,5-trimethoxyphenyl)-4,5-dihydro-1H-pyrazole-1-carboxamide] showed the most potent inhibitory effect on tubulin assembly (IC50 =2.12 µm) and in vitro growth inhibitory activity against a panel of four human cancer cell lines (IC50 values of 0.21-0.31 µm). Further studies confirmed that compound e19 can induce HeLa cell apoptosis, cause cell-cycle arrest in G2 /M phase, and disrupt the cellular microtubule network. These studies, along with molecular docking and 3D-QSAR modeling, provide an important basis for further optimization of compound e19 as a potential anticancer agent.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Pyrazoles/pharmacology , Tubulin Modulators/pharmacology , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints , Humans , Indoles/chemical synthesis , Molecular Docking Simulation , Pyrazoles/chemical synthesis , Quantitative Structure-Activity Relationship , Tubulin/chemistry , Tubulin/metabolism , Tubulin Modulators/chemical synthesis
9.
Eur J Med Chem ; 99: 125-37, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26070164

ABSTRACT

A series of 1-benzene acyl-2-(1-methylindol-3-yl)-benzimidazole derivatives were designed, synthesized and evaluated as potential tubulin polymerization inhibitors and for the cytotoxicity against anthropic cancer cell lines. Among the novel compounds, compound 11f was demonstrated the most potent tubulin polymerization inhibitory activity (IC50 = 1.5 µM) and antiproliferative activity against A549, HepG2 and MCF-7 (GI50 = 2.4, 3.8 and 5.1 µM, respectively), which was compared with the positive control colchicine and CA-4. We also evaluated that compound 11f could effectively induce apoptosis of A549 associated with G2/M phase cell cycle arrest. Docking simulation and 3D-QSAR model in these studies provided more information that could be applied to design new molecules with more potent tubulin inhibitory activity.


Subject(s)
Benzene/chemical synthesis , Benzene/pharmacology , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Drug Design , Indoles/chemical synthesis , Indoles/pharmacology , Molecular Docking Simulation , Protein Multimerization/drug effects , Tubulin/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Benzene/chemistry , Benzene/metabolism , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Cell Division/drug effects , Cell Line, Tumor , Chemistry Techniques, Synthetic , Drug Screening Assays, Antitumor , Humans , Indoles/chemistry , Indoles/metabolism , Protein Structure, Quaternary , Quantitative Structure-Activity Relationship , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacology
10.
Eur J Med Chem ; 93: 291-9, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25703297

ABSTRACT

A series of novel 5-phenyl-1H-pyrazol derivatives (5a-5x) containing cinnamamide moiety were synthesized and their biological activities as potential tubulin polymerization inhibitors were evaluated. Among them, compound 5j exhibited the most potent inhibitory activity with an IC50 value of 1.02 µM for tubulin, which was superior to that of Colchicine (IC50 = 1.34 µM). Docking simulation was performed to insert compound 5j into the crystal structure of tubulin at colchicine binding site to determine the probable binding model. 3D-QSAR model was also built to provide more pharmacophore understanding that could be used to design new agents with more potent tubulin inhibitory activity.


Subject(s)
Cinnamates/chemistry , Cinnamates/pharmacology , Pyrazoles/chemistry , Quantitative Structure-Activity Relationship , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Catalytic Domain , Cell Line, Tumor , Chemistry Techniques, Synthetic , Cinnamates/chemical synthesis , Cinnamates/metabolism , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Tubulin/chemistry , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/metabolism
11.
Bioorg Med Chem ; 22(21): 6201-8, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25267006

ABSTRACT

A series of novel 5-phenyl-1H-pyrazole derivatives (5 a-5 u) containing niacinamide moiety were synthesized and evaluated for biological activity as potential BRAF(V600E) inhibitors. Among them, compound 5h exhibited the most potent inhibitory activity with an IC50 value of 0.33 µM for BRAF(V600E). Antiproliferative assay results indicated that compound 5h has better antiproliferative activity against WM266.4 and A375 in vitro with IC50 value of 2.63 and 3.16 µM, respectively, being comparable with the positive control vemurafenib. Molecular docking of 5h into the BRAF(V600E) active site was performed to determine the probable binding mode. Furthermore, molecular docking and 3D QSAR study by means of DS 3.5 (Discovery Studio 3.5, Accelrys, Co. Ltd) explored the binding modes and the structure and activity relationship (SAR) of these derivatives.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyrazoles/chemistry , Pyrazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins B-raf/metabolism , Pyrazoles/chemical synthesis , Quantitative Structure-Activity Relationship , Structure-Activity Relationship
12.
Bioorg Med Chem ; 22(15): 4312-22, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24909678

ABSTRACT

A series of 1,3,4-thiadiazol-2-amide derivatives (6a-w) were designed and synthesized as potential inhibitors of tubulin polymerization and as anticancer agents. The in vitro anticancer activities of these compounds were evaluated against three cancer cell lines by the MTT method. Among all the designed compounds, compound 6f exhibited the most potent anticancer activity against A549, MCF-7 and HepG2 cancer cell lines with IC50 values of 0.03 µM, 0.06 µM and 0.05 µM, respectively. Compound 6f also exhibited significant tubulin polymerization inhibitory activity (IC50=1.73 µM), which was superior to the positive control. The obtained results, along with a 3D-QSAR study and molecular docking that were used for investigating the probable binding mode, could provide an important basis for further optimization of compound 6f as a novel anticancer agent.


Subject(s)
Amides/chemistry , Amides/pharmacology , Antineoplastic Agents/chemical synthesis , Tubulin Modulators/chemical synthesis , Tubulin Modulators/pharmacology , Amides/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , G2 Phase Cell Cycle Checkpoints/drug effects , Hep G2 Cells , Humans , M Phase Cell Cycle Checkpoints/drug effects , MCF-7 Cells , Molecular Conformation , Molecular Docking Simulation , Protein Structure, Tertiary , Quantitative Structure-Activity Relationship , Tubulin Modulators/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...