Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Expert Opin Ther Targets ; 28(3): 193-220, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38618889

ABSTRACT

INTRODUCTION: Microtubules play a vital role in cancer therapeutics. They are implicated in tumorigenesis, thus inhibiting tubulin polymerization in cancer cells, and have now become a significant target for anticancer drug development. A plethora of drug molecules has been crafted to influence microtubule dynamics and presently, numerous tubulin inhibitors are being investigated. This review discusses the recently developed inhibitors including natural products, and also examines the preclinical and clinical data of some potential molecules. AREA COVERED: The current review article summarizes the development of tubulin inhibitors while detailing their specific binding sites. It also discusses the newly designed inhibitors that may be useful in the treatment of solid tumors. EXPERT OPINION: Microtubules play a crucial role in cellular processes, especially in cancer therapy where inhibiting tubulin polymerization holds promise. Ongoing trials signify a commitment to revolutionizing cancer treatment and exploring targeted therapies. Challenges in microtubule modulation, like resistance and off-target effects, demand focused efforts, emphasizing combination therapies and personalized treatments. Beyond microtubules, promising avenues in cancer research include immunotherapy, genomic medicine, CRISPR gene editing, liquid biopsies, AI diagnostics, and stem cell therapy, showcasing a holistic approach for future advancements.


Subject(s)
Antineoplastic Agents , Drug Development , Microtubules , Molecular Targeted Therapy , Neoplasms , Tubulin Modulators , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Tubulin Modulators/pharmacology , Antineoplastic Agents/pharmacology , Animals , Microtubules/drug effects , Microtubules/metabolism , Drug Design , Biological Products/pharmacology , Tubulin/metabolism
2.
Fitoterapia ; 173: 105781, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38128619

ABSTRACT

Six anthraquinones were isolated from Morinda scabrida Craib, an unexplored species of Morinda found in the tropical forest of Thailand. All six anthraquinones showed cytotoxicity against A549 lung cancer cells, with the most active compound, nordamnacanthal (MS01), exhibiting the IC50 value of 16.3 ± 2.5 µM. The cytotoxic effect was dose-dependent and led to cell morphological changes characteristic of apoptosis. In addition, flow cytometric analysis showed dose-dependent apoptosis induction and the G2/M phase cell cycle arrest, which was in agreement with the tubulin polymerization inhibitory activity of MS01. Molecular docking analysis illustrated the binding between MS01 and the α/ß-tubulin heterodimer at the colchicine binding site, and UV-visible absorption spectroscopy revealed the DNA binding capacity of MS01.


Subject(s)
Lung Neoplasms , Morinda , Humans , Molecular Structure , Morinda/chemistry , Cell Proliferation , Cell Line, Tumor , Polymerization , Lung Neoplasms/drug therapy , Molecular Docking Simulation , Tubulin/chemistry , Tubulin/metabolism , Anthraquinones/pharmacology , Tubulin Modulators/pharmacology , Tubulin Modulators/chemistry , Tubulin Modulators/metabolism
3.
Comput Math Methods Med ; 2022: 4004068, 2022.
Article in English | MEDLINE | ID: mdl-35075369

ABSTRACT

Microtubules play a critical role in mitosis and cell division and are regarded as an excellent target for anticancer therapy. Although microtubule-targeting agents have been widely used in the clinical treatment of different human cancers, their clinical application in cancer therapy is limited by both intrinsic and acquired drug resistance and adverse toxicities. In a previous work, we synthesized compound 9IV-c, ((E)-2-(3,4-dimethoxystyryl)-6,7,8-trimethoxy-N-(3,4,5-trimethoxyphenyl)quinoline-4-amine) that showed potent activity against multiple human tumor cell lines, by targeting spindle formation and/or the microtubule network. Accordingly, in this study, to identify potent tubulin inhibitors, at first, molecular docking and molecular dynamics studies of compound 9IV-c were performed into the colchicine binding site of tubulin; then, a pharmacophore model of the 9IV-c-tubulin complex was generated. The pharmacophore model was then validated by Güner-Henry (GH) scoring methods and receiver operating characteristic (ROC) analysis. The IBScreen database was searched by using this pharmacophore model as a screening query. Finally, five retrieved compounds were selected for molecular docking studies. These efforts identified two compounds (b and c) as potent tubulin inhibitors. Investigation of pharmacokinetic properties of these compounds (b and c) and compound 9IV-c displayed that ligand b has better drug characteristics compared to the other two ligands.


Subject(s)
Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Tumor , Colchicine/chemistry , Colchicine/pharmacology , Computational Biology , Computer Simulation , Databases, Pharmaceutical , Drug Design , Drug Evaluation, Preclinical , Humans , Ligands , Microtubules/chemistry , Microtubules/drug effects , Molecular Docking Simulation , Molecular Dynamics Simulation , Tubulin/chemistry , Tubulin Modulators/chemical synthesis , User-Computer Interface
4.
Cancer Lett ; 526: 168-179, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34838691

ABSTRACT

Bladder cancers, and specifically urothelial carcinoma, have few effective treatment options, and tumors typically develop resistance against standard of care chemotherapies leading to significant mortality. The development of alternative therapies with increased selectivity and improved tolerability would significantly impact this patient population. Here, we investigate a novel colchicine derivative, CR42-24, with increased selectivity for the ßIII tubulin subtype as a treatment for urothelial carcinoma. ßIII tubulin is a promising target due to its low expression in healthy tissues and its clinical association with poor prognosis. This study demonstrated that CR42-24 is selectively cytotoxic to several cancer cell lines at low nanomolar IC50, with high activity in bladder cancer cell lines both in vitro and in vivo. CR42-24 monotherapy in an aggressive urothelial carcinoma xenograft model results in effective control when treated early. We observed significant ablation of large tumors and patient-derived xenografts at low doses with excellent tolerability. CR42-24 was highly synergistic in combination with the standard of care chemotherapies gemcitabine and cisplatin, further increasing its therapeutic potential as a novel treatment for urothelial carcinoma.


Subject(s)
Carcinoma, Transitional Cell/drug therapy , Colchicine/pharmacology , Urinary Bladder Neoplasms/drug therapy , Animals , Cell Line, Tumor , Disease Models, Animal , HeLa Cells , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Tubulin Modulators/pharmacology , Xenograft Model Antitumor Assays
5.
Eur J Pharmacol ; 914: 174687, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34883072

ABSTRACT

BACKGROUND: Trans-cinnamaldehyde (TCA) is a main compound of Cinnamomum cassia, used in traditional Chinese medicine to treat many ailments. Increasing evidence has demonstrated the therapeutic effects of TCA in cardiovascular diseases. PURPOSE: The present study aimed to determine whether TCA exerts antihypertrophic effects in vitro and in vivo and to elucidate the underlying mechanisms of these effects. METHODS: Neonatal rat cardiac myocytes (NRCMs) and adult mouse cardiac myocytes (AMCMs) were treated with 50 µΜ phenylephrine (PE) for 48 h. Tubulin detyrosination, store-operated Ca2+ entry (SOCE), stromal interaction molecule-1 (STIM1)/Orai1 translocation, and calcineurin/nuclear factor of activated T-cells (NFAT) signaling pathways were analyzed in NRCMs. Meanwhile, tubulin detyrosination, junctophilin-2, T-tubule distribution pattern, Ca2+ handling, and sarcomere shortening were observed in AMCMs. Male C57BL/6 mice were stimulated with PE (70 mg/kg per day) with or without TCA treatment for 2 weeks. Cardiac hypertrophy and tubulin detyrosination were also assessed. RESULTS: TCA was confirmed to alleviate cardiac hypertrophy induced by PE stimulation in vitro and in vivo. PE-induced cardiac hypertrophy was associated with excessive tubulin detyrosination and overexpression of vasohibin 1 (VASH1) and small vasohibin binding protein (SVBP), two key proteins responsible for tubulin detyrosination. These effects were largely blocked by TCA administration. PE treatment also enhanced SOCE with massive translocation of STIM1 and Orai1, Ca2+ mishandling, reduced sarcomere shortening, junctophilin-2, and T-tubule redistribution, all of which were significantly ameliorated by TCA administration. CONCLUSION: Our study indicated that the therapeutic effects of TCA against cardiac hypertrophy may be associated with its ability to reduce tubulin detyrosination.


Subject(s)
Acrolein/analogs & derivatives , Cardiomegaly , Microtubules , Myocytes, Cardiac , Tubulin/metabolism , Acrolein/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Membrane Proteins/metabolism , Mice , Microtubules/drug effects , Microtubules/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Rats , Signal Transduction/drug effects , Treatment Outcome , Tubulin Modulators/pharmacology
6.
Bioorg Chem ; 118: 105486, 2022 01.
Article in English | MEDLINE | ID: mdl-34801948

ABSTRACT

The colchicine binding site of tubulin is a promising target for discovering novel antitumor agents which exert the antiangiogenic effect and are not susceptible to multidrug resistance. For identifying novel tubulin inhibitors, structure-based virtual screening was applied to identify hit 9 which displayed moderate tubulin polymerization inhibition and broad-spectrum in vitro antitumor activity. Structural optimization was performed, and biological assay revealed analog E27 displayed the best antitumor activity with IC50 values ranging from 7.81 µM to 10.36 µM, and improved tubulin polymerization inhibitory activity (IC50 = 16.1 µM). It significantly inhibited cancer cell migration and invasion, induced cell apoptosis and arrested the cell cycle at G2/M phase. Moreover, the apoptotic effect of E27 is related to the increased ROS level, the decrease of MMP, and the abnormal expression of apoptosis-related proteins. Taken together, these results suggested E27 was a promising lead compound for discovering novel tubulin-targeted antitumor agents.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Structure , Polymerization/drug effects , Structure-Activity Relationship , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
7.
Drug Des Devel Ther ; 15: 4195-4211, 2021.
Article in English | MEDLINE | ID: mdl-34675483

ABSTRACT

BACKGROUND: Fenugreek, also known as Trigonella foenum-graecum L, is a natural plant that belongs to the Fabaceae family and has been known as a promising source of bioactive compounds. It has been widely used as traditional medicine since it has shown to lower blood glucose, manage cholesterol levels and further aid in the prevention and treatment of cancer. Herein, we aim to evaluate the anticancer activity of methanolic fenugreek seed extract against several cancer cell lines. METHODS: We sought to investigate the phytochemical classes present in multiple fenugreek seeds extracts using HPLC-DAD followed by LC/MS, predict and investigate anticancer activity using PASS online webserver, the CellTiter-Glo assay, evaluate ADME properties, and perform molecular docking for all bioactive compounds via Maestro software. RESULTS: Multiple extracts exhibited distinct phytochemical classes that demonstrated different biological activities. Fenugreek methanolic extract contains flavonoid chemical class, which showed the highest anticancer activity against the HCT8 cell line of colorectal cancer (IC50 of 8.83 µg/mL), followed by KAIMRC1 breast cancer cell line (IC50 of 35.06 µg/mL), HL60 leukemia cell line (37.80 µg/mL), MDA-MB-231 breast cancer cell line (38.51 µg/mL), and lastly, HCT116 colorectal cancer cell line with IC50 of 56.03 µg/mL. In contrast, the chloroform extract was inactive. The molecular docking study for all the bioactive compounds suggested that flavonoids F6 (-9.713 and -12.132), F7 (-10.166 and -12.411), and F11 (-10.084 and -13.516) possess the highest docking scores through SP and XP scores, respectively. CONCLUSION: The obtained results confirm that the bioactive compounds present in fenugreek seeds exhibit anticancer activity against several cancer cells that can mediate via tubulin polymerization inhibition. Although our study has evaluated the anticancer potential of Trigonella foenum-graecum as a promising natural source for new anticancer agents, fenugreek biological activity needs further research and investigations on their mechanism of action and toxicity profile.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Neoplasms/drug therapy , Plant Extracts/pharmacology , Tubulin Modulators/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Chromatography, High Pressure Liquid , Chromatography, Liquid , Humans , Inhibitory Concentration 50 , Mass Spectrometry , Molecular Docking Simulation , Neoplasms/pathology , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Trigonella/chemistry , Tubulin/drug effects , Tubulin/metabolism , Tubulin Modulators/administration & dosage , Tubulin Modulators/chemistry
8.
Biomed Res Int ; 2021: 6480804, 2021.
Article in English | MEDLINE | ID: mdl-34485522

ABSTRACT

In this study, we aimed to develop a pharmacophore-based three-dimensional quantitative structure activity relationship (3D-QSAR) for a set including sixty-two cytotoxic quinolines (1-62) as anticancer agents with tubulin inhibitory activity. A total of 279 pharmacophore hypotheses were generated based on the survival score to build QSAR models. A six-point pharmacophore model (AAARRR.1061) was identified as the best model which consisted of three hydrogen bond acceptors (A) and three aromatic ring (R) features. The model showed a high correlation coefficient (R 2 = 0.865), cross-validation coefficient (Q 2 = 0.718), and F value (72.3). The best pharmacophore model was then validated by the Y-Randomization test and ROC-AUC analysis. The generated 3D contour maps were used to reveal the structure activity relationship of the compounds. The IBScreen database was screened against AAARRR.1061, and after calculating ADMET properties, 10 compounds were selected for further docking study. Molecular docking analysis showed that compound STOCK2S-23597 with the highest docking score (-10.948 kcal/mol) had hydrophobic interactions and can form four hydrogen bonds with active site residues.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Tubulin/chemistry , Antineoplastic Agents/chemistry , Catalytic Domain , Computational Chemistry/methods , Computer Simulation , Drug Evaluation, Preclinical/methods , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Molecular Conformation , Molecular Docking Simulation/methods , Neoplasms/metabolism , Neoplasms/pathology , Quantitative Structure-Activity Relationship , Tubulin/metabolism
9.
Bioorg Chem ; 110: 104795, 2021 05.
Article in English | MEDLINE | ID: mdl-33730670

ABSTRACT

Virtual screening of commercially available molecular entities by using CDRUG, structure-based virtual screening, and similarity identified eight new derivatives of 3-phenyl-1H-indole-2-carbohydrazide with anti-proliferative activities. The molecules were tested experimentally for inhibition of tubulin polymerisation, which revealed furan-3-ylmethylene-3-phenyl-1H-indole-2-carbohydrazide (27a) as the most potent candidate. Molecule 27a was able to induce G2/M phase arrest in A549 cell line, similar to other tubulin inhibitors. Synthetic modifications of 27a were focussed on small substitutions on the furan ring, halogenation at R1 position and alteration of furyl connectivity. Derivatives 27b, 27d and 27i exhibited the strongest tubulin inhibition activities and were comparable to 27a. Bromine substitution at R1 position showed most prominent anticancer activities; derivatives 27b-27d displayed the strongest activities against HuCCA-1 cell line and were more potent than doxorubicin and the parent molecule 27a with IC50 values <0.5 µM. Notably, 27b with a 5-methoxy substitution on furan displayed the strongest activity against HepG2 cell line (IC50 = 0.34 µM), while 27d displayed stronger activity against A549 cell line (IC50 = 0.43 µM) compared to doxorubicin and 27a. Fluorine substitutions at the R1 position tended to show more modest anti-tubulin and anticancer activities, and change of 2-furyl to 3-furyl was tolerable. The new derivatives, thiophenyl 26, displayed the strongest activity against A549 cell line (IC50 = 0.19 µM), while 1-phenylethylidene 21b and 21c exhibited more modest anticancer activities with unclear mechanisms of action; 26 and 21c demonstrated G2/M phase arrest, but showed weak tubulin inhibitory properties. Molecular docking suggests the series inhibit tubulin at the colchicine site, in agreement with the experimental findings. The calculated molecular descriptors indicated that the molecules obey Lipinski's rule which suggests the molecules are drug-like structures.


Subject(s)
Antineoplastic Agents/pharmacology , Hydrazines/pharmacology , Indoles/pharmacology , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Humans , Hydrazines/chemical synthesis , Hydrazines/chemistry , Indoles/chemical synthesis , Indoles/chemistry , Molecular Structure , Structure-Activity Relationship , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
10.
BMC Cancer ; 21(1): 91, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33482772

ABSTRACT

BACKGROUND: Recent years, survival rates of human with high-risk acute myeloid leukaemia (AML) have not raised substantially. This research aimed to investigate the role of 4'-O-Methylbroussochalcone B, for the treatment of human AML. METHODS: Firstly, we evaluated the effects of six chalcones on AML cells activity by MTT assay. Immunofluorescence staining, tubulin polymerization assay and N,N'-ethylenebis (iodoacetamide) (EBI) competition assay were performed on ML-2 cells. Transwell and apoptosis assay were also utilized in ML-2 cells and OCI-AML5 cells. The expressions of migration-related proteins, apoptosis-related proteins and Wnt/ß-catenin pathway were detected by Western Blot. RESULTS: The results found six chalcones exhibited the anti-proliferative activity against different AML cell lines. Based on the results of immunofluorescence staining, tubulin polymerization assay and EBI competition assay, 4'-O-Methylbroussochalcone B was discovered to be a novel colchicine site tubulin polymerization inhibitor. 4'-O-Methylbroussochalcone B could induce apoptosis, inhibit proliferation and migration of ML-2 cells and OCI-AML5 cells. The cells were arrested in the G2-M phase by the treatment of 4'-O-Methylbroussochalcone B. In addition, 4'-O-Methylbroussochalcone B regulated MAPK and Wnt/ß-catenin pathways in AML cells. CONCLUSION: 4'-O-Methylbroussochalcone B might inhibit proliferation and migration of the AML cells by MAPK and Wnt/ß-catenin pathways as a tubulin polymerization inhibitor. It is promising for 4'-O-Methylbroussochalcone B to become a new drug to treat AML.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Movement , Cell Proliferation , Chalcone/chemistry , Chalcones/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Tubulin Modulators/pharmacology , Tubulin/chemistry , Apoptosis , Fabaceae/chemistry , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Plant Extracts/pharmacology , Polymerization , Seeds/chemistry , Tumor Cells, Cultured , Wnt Signaling Pathway
11.
Invest New Drugs ; 39(2): 386-393, 2021 04.
Article in English | MEDLINE | ID: mdl-32997210

ABSTRACT

Coptis chinensis Franch. has been extensively used in traditional Chinese medicine. The chemical structure of oxyepiberberine, as an alkaloid isolated from Coptis chinensis Franch., has been previously studied. However, anti-cancer effects and underlying mechanisms of oxyepiberberine need to be explored. This study aimed to investigate the anti-cancer effects and underlying mechanisms of oxyepiberberine on LS-1034 human colon cancer cells. The anti-proliferative effects of six derivatives of oxyepiberberine on colon cancer cells were assessed. Among six derivatives, oxyepiberberine showed the greatest anti-proliferative effect on LS-1034 cells with an IC50 value of 1.36 µM. Oxyepiberberine also induced apoptosis and inhibited migration of LS-1034 cells in a concentration-dependent manner. Importantly, oxyepiberberine was identified as a potent tubulin polymerization inhibitor. The tubulin polymerization inhibitory effects of oxyepiberberine in a concentration-dependent manner with an IC50 value of 1.26 µM were observed. A xenograft mouse model of colon cancer showed that oxyepiberberine could suppress tumor growth without an obvious toxicity. Conclusion Oxyepiberberine was found as a novel tubulin polymerization inhibitor, and it could be a promising agent to treat colon cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Berberine Alkaloids/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Tubulin Modulators/pharmacology , Animals , Cell Line, Tumor , Coptis chinensis , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Xenograft Model Antitumor Assays
12.
Biochem Biophys Res Commun ; 534: 330-336, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33272565

ABSTRACT

Microtubule is a key component of cytoskeleton and has been considered as an important target for the treatment of cancer. In particular, the tubulin taxane-site inhibitors such as taxol analogs and epothilones have achieved great success in clinical trials. However, the structural basis of many taxane-site inhibitors is still lacking in exploring their mechanism of action. We here reported crystal complex structures for three taxane-site inhibitors, Ixabepilone, Epothilone B, and Epothilone D, which were determined to 2.4 Å, 2.4 Å, and 2.85 Å, respectively. The crystal structures revealed that these taxane-site inhibitors possess similar binding modes to that of Epothilone A at the taxane site, e.g. making critical hydrogen-bonding interactions with multiple residues on the M-loop, which facilitating the tubulin polymerization. Furthermore, we summarized the binding modes of almost all taxane-site inhibitors and identified novel taxane-site ligands with simpler chemical structures through virtual screening. On this basis, new derivatives with higher binding affinity to tubulin were designed and developed, which can form additional hydrogen bond interactions with tubulin. Overall, this work determined the mechanism of action of epothilones and provided a structural basis to design reasonably novel taxane-site inhibitors with simpler structure and improved pharmacokinetic properties.


Subject(s)
Epothilones/chemistry , Epothilones/pharmacology , Microtubules/drug effects , Microtubules/metabolism , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Tubulin/chemistry , Tubulin/metabolism , Binding Sites , Crystallography, X-Ray , Drug Design , Drug Evaluation, Preclinical , Epothilones/pharmacokinetics , Humans , Models, Molecular , Molecular Docking Simulation , Tubulin Modulators/pharmacokinetics , User-Computer Interface
13.
Med Chem ; 17(6): 611-622, 2021.
Article in English | MEDLINE | ID: mdl-31951171

ABSTRACT

BACKGROUND: Owing to its potential to interfere in microtubule dynamics in the mitotic phase of cell cycle and selectively induce apoptosis in cancer cells without affecting normal cells, noscapine and its synthetic analogues have been investigated by other research groups in different cell lines for their capability to be used as anti-cancer agents. OBJECTIVE: The present study is focused on the investigation of the mode of binding of noscapinoids with tubulin, prediction of target binding affinities and mapping of their spatial fingerprints (shape and electrostatic). METHODS: Molecular docking assisted alignment based 3D-QSAR was used on a dataset (43 molecules) having an inhibitory activity (IC50 = 1.2-250 µM) against human lymphoblast (CEM) cell line. RESULTS AND CONCLUSION: Key amino acid residues of target tubulin were mapped for the binding of most potent noscapine analogue (Compound 11) and were compared with noscapine. Spatial fingerprints of noscapinoids for favorable tubulin inhibitory activity were generated and are proposed herewith for further pharmacophoric amendments of noscapine analogues to design and develop novel potent noscapine based anti-cancer agents that may enter into drug development pipeline.


Subject(s)
Noscapine/chemistry , Noscapine/pharmacology , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Tubulin/metabolism , Cell Line , Drug Evaluation, Preclinical , Humans , Molecular Docking Simulation , Noscapine/metabolism , Protein Conformation , Quantitative Structure-Activity Relationship , Tubulin/chemistry , Tubulin Modulators/metabolism
14.
Theranostics ; 10(10): 4308-4322, 2020.
Article in English | MEDLINE | ID: mdl-32292496

ABSTRACT

Rationale: The dual-targeted drug delivery system was designed for enhancing permeation of the blood-brain barrier (BBB) and providing an anti-glioma effect. As transferrin receptor (TfR) is over-expressed by the brain capillary endothelial (hCMEC/D3) and glioma cells, a mouse monoclonal antibody, RI7217, with high affinity and selectivity for TfR, was used to study the brain targeted drug delivery system. Muscone, an ingredient of traditional Chinese medicine (TCM) musk, was used as the "guide" drug to probe the permeability of the BBB for drug delivery into the cerebrospinal fluid. This study investigated the combined effects of TCM aromatic resuscitation and modern receptor-targeted technology by the use of muscone/RI7217 co-modified docetaxel (DTX) liposomes for enhanced drug delivery to the brain for anti-glioma effect. Methods: Cellular drug uptake from the formulations was determined using fluorescence microscopy and flow cytometry. The drug penetrating ability into tumor spheroids were visualized using confocal laser scanning microscopy (CLSM). In vivo glioma-targeting ability of formulations was evaluated using whole-body fluorescent imaging system. The survival curve study was performed to evaluate the anti-glioma effect of the formulations. Results: The results showed that muscone and RI7217 co-modified DTX liposomes enhanced uptake into both hCMEC/D3 and U87-MG cells, increased penetration to the deep region of U87-MG tumor spheroids, improved brain targeting in vivo and prolonged survival time of nude mice bearing tumor. Conclusion: Muscone and RI7217 co-modified DTX liposomes were found to show improved brain targeting and enhanced the efficacy of anti-glioma drug treatment in vivo.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Neoplasms/pathology , Cycloparaffins/pharmacology , Glioma/drug therapy , Liposomes/pharmacokinetics , Animals , Antigens, CD/chemistry , Antigens, CD/pharmacology , Blood-Brain Barrier/metabolism , Case-Control Studies , Cell Line, Tumor , Cell Survival/drug effects , Cycloparaffins/administration & dosage , Cycloparaffins/cerebrospinal fluid , Docetaxel/pharmacology , Drug Delivery Systems , Drug Therapy, Combination/methods , Glioma/metabolism , Liposomes/chemistry , Medicine, Chinese Traditional/adverse effects , Medicine, Chinese Traditional/methods , Mice , Mice, Nude , Permeability/drug effects , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism , Tubulin Modulators/pharmacology
15.
Bioorg Chem ; 95: 103565, 2020 01.
Article in English | MEDLINE | ID: mdl-31927336

ABSTRACT

A novel series of chalcone derivatives containing diaryl ether moiety (5a-5p) were designed, synthesized and evaluated their anti-tubulin polymerization activities and anticancer activities. Among them, compound 5b with 4-methoxy substitution on right aromatic ring was found to be most active on MCF-7, HepG2 and HCT116 cancer cell lines, with IC50 values of 3.44 ± 0.19, 4.64 ± 0.23, and 6.31 ± 0.27 µM, respectively. In vitro tubulin polymerization assay showed that 5b could effectively inhibit tubulin polymerization. Further mechanism studies revealed that 5b could induce G2/M phase arrest and cell apoptosis. Molecular docking studies revealed that 5b interact and bind at the colchicine binding site of the tubulin.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Chalcones/pharmacology , Drug Design , Polymerization/drug effects , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Tubulin/chemistry , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcones/chemistry , Drug Evaluation, Preclinical , Humans , Molecular Docking Simulation , Structure-Activity Relationship , Tubulin Modulators/chemical synthesis
16.
Phytomedicine ; 67: 153152, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31887479

ABSTRACT

BACKGROUND: Lung cancer is the leading cause of cancer-related deaths worldwide. Several targets have been identified for lung cancer therapy, amongst which 'Microtubule' and its dynamics are the most widely studied and used in therapy. Tubulin-microtubule polymer dynamics are highly sought after targets in the field of anti-cancer drug designing. Natural compounds are important sources for developing anticancer therapeutics owing to their efficacy and lower cytotoxicity. Evidence suggested that therapeutic targeting of microtubule by natural compounds is amongst the most widely used interventions in numerous cancer therapies including lung cancer. PURPOSE: To determine the efficacy of apocynin (a natural compound) in suppressing the progression of lung carcinoma both in vitro and in vivo, along with the identification of targets and the underlying mechanism for developing a novel therapeutic approach. METHODS: We have demonstrated themicrotubule depolymerizing role of apocynin by established protocols in cellular and cell-free system. The efficacy of apocynin to inhibit lung carcinoma progression was studied on A549 cells.The tumoricidal ability of apocynin was studied in BALB/c mice model as well.Mice were classified into 4 groups namely-group II mice as tumor control; group III-IV mice asalso tumor-induced but treated with differential apocynin doses whereas group I mice were kept as normal. RESULTS: Apocynin, showed selective cytotoxicity towards lung cancer cells rather than normal lung fibroblast cells. Apocynin inhibited oncogenic properties including growth, proliferation (p < 0.05), colony formation (p < 0.05), invasion (p < 0.05) and spheroid formation (p < 0.05) in lung cancer cells. Apart from other established properties, apocynin was found to be a novel and potent component to bind with tubulin and depolymerize cellular microtubule network. Apocynin mediated cellular microtubule depolymerization was the driving mechanism to trigger autophagy-mediated apoptotic cell death (p < 0.05) which in turn retarded lung cancer progression. Furthermore, apocynin showed tumoricidal characteristics to inhibit lung tumorigenesis in mice as well. CONCLUSION: Targeting tubulin-microtubule equilibrium with apocynin could be the key regulator to catastrophe cellular catabolic processes to mitigate lung carcinoma. Thus, apocynin could be a potential therapeutic agent for lung cancer treatment.


Subject(s)
Acetophenones/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Lung Neoplasms/drug therapy , Tubulin Modulators/pharmacology , A549 Cells , Acetophenones/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Male , Mice, Inbred BALB C , Microtubules/metabolism , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Tubulin/chemistry , Tubulin/metabolism , Tubulin Modulators/chemistry
17.
Molecules ; 24(17)2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31480625

ABSTRACT

Tubulin inhibitors have been considered as potential drugs for cancer therapy. However, their drug resistance and serious side-effects are the main reasons for clinical treatment failure. Therefore, there is still an urgent need to develop effective therapeutic drugs. Herein, a structure-based pharmacophore model was developed based on the co-crystallized structures of the tubulin with a high resolution. The model including one hydrogen-bond acceptor feature, two aromatic features, and one hydrophobic feature was further validated using the Gunner-Henry score method. Virtual screening was performed by an integrated protocol that combines drug-likeness analysis, pharmacophore mapping, and molecular docking approaches. Finally, five hits were selected for biological evaluation. The results indicated that all these hits at the concentration of 40 µM showed an inhibition of more than 50% against five human tumor cells (MCF-7, U87MG, HCT-116, MDA-MB-231, and HepG2). Particularly, hit 1 effectively inhibited the proliferation of these tumor cells, with inhibition rates of more than 80%. The results of tubulin polymerization and colchicine-site competition assays suggested that hit 1 significantly inhibited tubulin polymerization by binding to the colchicine site. Thus, hit 1 could be used as a potential chemotherapeutic agent for cancer treatment. This work also demonstrated the potential of our screening protocol to identify biologically active compounds.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Drug Evaluation, Preclinical , Quantitative Structure-Activity Relationship , Tubulin Modulators/pharmacology , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/metabolism , Humans , Ligands , Models, Molecular , Polymerization , Tubulin/metabolism
18.
Molecules ; 24(15)2019 Jul 30.
Article in English | MEDLINE | ID: mdl-31366123

ABSTRACT

Colchicine was extracted from Gloriosa superba seeds using the Super Critical Fluid (CO2) Extraction (SCFE) technology. The seeds were purified upto 99.82% using column chromatography. Colchicine affinity was further investigated for anticancer activity in six human cancer cell lines, i.e., A549, MCF-7, MDA-MB231, PANC-1, HCT116, and SiHa. Purified colchicine showed the least cell cytotoxicity and antiproliferation and caused no G2/M arrest at clinically acceptable concentrations. Mitotic arrest was observed in only A549 and MDA-MB231 cell lines at 60nM concentration. Our finding indicated the possible use of colchicine at a clinically acceptable dose and provided insight into the science behind microtubule destabilization. However, more studies need to be conducted beforethese findings could be established.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Chromatography, Supercritical Fluid/methods , Colchicaceae/chemistry , Colchicine/pharmacology , Seeds/chemistry , Tubulin Modulators/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Carbon Dioxide/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromatography, Liquid , Colchicine/isolation & purification , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Plant Extracts/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tubulin Modulators/isolation & purification
19.
Int J Mol Sci ; 20(11)2019 May 28.
Article in English | MEDLINE | ID: mdl-31141929

ABSTRACT

Deoxypodophyllotoxin (DPT) is a cyclolignan compound that exerts anti-cancer effects against various types of cancers. DPT induces apoptosis and inhibits the growth of breast, brain, prostate, gastric, lung, and cervical tumors. In this study, we sought to determine the effect of DPT on cell proliferation, apoptosis, motility, and tumorigenesis of three colorectal cancer (CRC) cell lines: HT29, DLD1, and Caco2. DPT inhibited the proliferation of these cells. Specifically, the compound-induced mitotic arrest in CRC cells by destabilizing microtubules and activating the mitochondrial apoptotic pathway via regulation of B-cell lymphoma 2 (Bcl-2) family proteins (increasing Bcl-2 associated X (BAX) and decreasing B-cell lymphoma-extra-large (Bcl-xL)) ultimately led to caspase-mediated apoptosis. In addition, DPT inhibited tumorigenesis in vitro, and in vivo skin xenograft experiments revealed that DPT significantly decreased tumor size and tumor weight. Taken together, our results suggest DPT to be a potent compound that is suitable for further exploration as a novel chemotherapeutic for human CRC.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinogenesis/drug effects , Colorectal Neoplasms/drug therapy , Podophyllotoxin/analogs & derivatives , Tubulin Modulators/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Caco-2 Cells , Colorectal Neoplasms/metabolism , Drugs, Chinese Herbal , HT29 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Microtubules/drug effects , Microtubules/metabolism , Podophyllotoxin/pharmacology , Podophyllotoxin/therapeutic use , Tubulin Modulators/therapeutic use
20.
Curr Top Med Chem ; 19(13): 1092-1120, 2019.
Article in English | MEDLINE | ID: mdl-31109275

ABSTRACT

BACKGROUND: Tubulin polymerization inhibitors interfere with microtubule assembly and their functions lead to mitotic arrest, therefore they are attractive target for design and development of novel anticancer compounds. OBJECTIVE: The proposed novel and effective structures following the use of three-dimensionalquantitative structure activity relationship (3D-QSAR) pharmacophore based virtual screening clearly demonstrate the high efficiency of this method in modern drug discovery. METHODS: Combined computational approach was applied to extract the essential 2D and 3D features requirements for higher activity as well as identify new anti-tubulin agents. RESULTS: The best quantitative pharmacophore model, Hypo1, exhibited good correlation of 0.943 (RMSD=1.019) and excellent predictive power in the training set compounds. Generated model AHHHR, was well mapped to colchicine site and three-dimensional spatial arrangement of their features were in good agreement with the vital interactions in the active site. Total prediction accuracy (0.92 for training set and 0.86 for test set), enrichment factor (4.2 for training set and 4.5 for test set) and the area under the ROC curve (0.86 for training set and 0.94 for the test set), the developed model using Extended Class FingerPrints of maximum diameter 4 (ECFP_4) was chosen as the best model. CONCLUSION: Developed computational platform provided a better understanding of requirement features for colchicine site inhibitors and we believe the results of this study might be useful for the rational design and optimization of new inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Colchicine/pharmacology , Drug Discovery , Quantitative Structure-Activity Relationship , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/chemical synthesis , Colchicine/chemistry , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Polymerization/drug effects , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL