Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Biol Rep ; 48(11): 7443-7456, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34716862

ABSTRACT

BACKGROUND: Survival rate of patients affected with anaplastic thyroid carcinoma (ATC) is less than 5% with current treatment. In ATC, BRAFV600E mutation is the major mutation that results in the transformation of normal cells in to an undifferentiated cancer cells via aberrant molecular signaling mechanisms. Although vemurufenib is a selective oral drug for the BRAFV600E mutant kinase with a response rate of nearly 50% in metastatic melanoma, our study has showed resistance to this drug in ATC. Hence the rationale of the study is to explore combinational therapeutic effect to improve the efficacy of vemurafenib along with metformin. Metformin, a diabetic drug is an AMPK activator and has recently proved to be involved in preventing or treating several types of cancer. METHODS AND RESULTS: Using iGEMDock software, a protein-ligand interaction was successful between Metformin and TSHR (receptor present in the thyroid follicular cells). Our study demonstrates that combination of vemurufenib with metformin has synergistic anti-cancer effects which was evaluated through MTT assay (cytotoxicity), colony formation assay (antiproliferation evaluation) and suppressed the progression of ATC cells growth by inducing significant apoptosis, proven by Annexin V-FITC assay (Early Apoptosis Detection). Downregulation of ERK signaling, upregulation of AMPK pathway and precision in epithelial-mesenchymal transition (EMT) pathway which were assessed by RT-PCR and Western blot provide the evidence that the combination of drugs involved in the precision of altered molecular signaling Further our results suggest that Metformin act as a demethylating agent in anaplastic thyroid cancer cells by inducing the expression of NIS and TSHR. Our study for the first time explored cAMP signaling in ATC wherein cAMP signaling is downregulated due to decrease in intracellular cAMP level upon metformin treatment. CONCLUSION: To conclude, our findings demonstrate novel therapeutic targets and treatment strategies for undifferentiated ATC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Neoplasm Proteins , Receptors, Thyrotropin , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Drug Synergism , Humans , Metformin/chemistry , Metformin/pharmacology , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/metabolism , Thyroid Carcinoma, Anaplastic/chemistry , Thyroid Carcinoma, Anaplastic/drug therapy , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Neoplasms/chemistry , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Vemurafenib/chemistry , Vemurafenib/pharmacology
2.
Bioengineered ; 12(1): 2970-2983, 2021 12.
Article in English | MEDLINE | ID: mdl-34252351

ABSTRACT

The aim of this research was to screen the ZINC15 database to select lead compounds and drug candidates which can inhibit B-RAF (V600E). In order to identify drugs potentially inhibited B-RAF (V600E), numerous modules of Discovery Studio 4.5 were employed. Structure-based screening using LibDock was carried out followed by ADME (absorption, distribution, metabolism, excretion) and toxicity prediction. CDOCKER was performed to demonstrate the binding affinity and mechanism between ligands and B-RAF(V600E). To evaluate whether ligand-receptor complexes were stable, molecular dynamics were employed. Two novel natural compounds (ZINC000100168592 and ZINC000049784088) from ZINC15 database were found binding to B-RAF(V600E) with more favorable interaction energy in comparison with the reference drug Vemurafenib. Also, they were predicted with less ames mutagenicity, rodent carcinogenicity, non-developmental toxic potential and tolerance to cytochrome P450 2D6 (CYP2D6). The molecular dynamics simulation analysis indicated that the compound-B-RAF(V600E) complexes had more favorable potential energy compared with Vemurafenib and they can exist in natural environments stably. The result of this study shows that ZINC000100168592 and ZINC000049784088 are ideal leading potential compounds to inhibit B-RAF(V600E). The findings of this study and these selected drug candidates greatly contributed to the medication design and improvement of B-RAF(V600E) and other proteins.


Subject(s)
Drug Discovery , Molecular Dynamics Simulation , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Animals , Databases, Chemical , Humans , Mice , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Rats , Vemurafenib/chemistry , Vemurafenib/metabolism , Vemurafenib/toxicity
3.
Molecules ; 26(7)2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33806033

ABSTRACT

The photochemical behavior of the photosensitive first-line anticancer drug vemurafenib (VFB) is of great interest due to the impact of such behavior on its pharmacological activity. In this work, we computationally elucidated the mechanism of the photoinduced release of VFB from the 4,5-dimethoxy-2-nitrobenzene (DMNB) photoprotecting group by employing various density functional theory (DFT)/time-dependent DFT (TD-DFT) approaches. The computational investigations included a comparative assessment of the influence of the position of the photoprotecting group as a substituent on the thermodynamics and kinetics of the photouncaging reactions of two VFB-DMNB prodrugs, namely pyrrole (NP) and sulfonamide (NS). With the aid of the DFT calculations concerning the activation energy barrier (∆G‡), the obtained results suggest that the step of the photoinduced intramolecular proton transfer of the DMNB moiety is not detrimental concerning the overall reaction profile of the photouncaging reaction of both prodrugs. However, the obtained results suggested that the position of the substitution position of the DMNB photoprotecting group within the prodrug structure has a substantial impact on the photouncaging reaction. In particular, the DMNB-Ns-VFB prodrug exhibited a notable increase in ∆G‡ for the key step of ring opining within the DMNB moiety indicative of potentially hindered kinetics of the photouncaging process compared with DMNB-Np-VFB. Such an increase in ∆G‡ may be attributed to the electronic influence of the NP fragment of the prodrug. The results reported herein elaborate on the mechanism of the photoinduced release of an important anticancer drug from photoprotecting groups with the aim of enhancing our understanding of the photochemical behavior of such photosensitive pharmaceutical materials at the molecular level.


Subject(s)
Computer Simulation , Models, Chemical , Vemurafenib/chemistry , Kinetics , Thermodynamics
4.
Proc Natl Acad Sci U S A ; 117(49): 31105-31113, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229534

ABSTRACT

Kinase-targeted therapies have the potential to improve the survival of patients with cancer. However, the cancer-specific spectrum of kinase alterations exhibits distinct functional properties and requires mutation-oriented drug treatments. Besides post-translational modifications and diverse intermolecular interactions of kinases, it is the distinct disease mutation which reshapes full-length kinase conformations, affecting their activity. Oncokinase mutation profiles differ between cancer types, as it was shown for BRAF in melanoma and non-small-cell lung cancers. Here, we present the target-oriented application of a kinase conformation (KinCon) reporter platform for live-cell measurements of autoinhibitory kinase activity states. The bioluminescence-based KinCon biosensor allows the tracking of conformation dynamics of full-length kinases in intact cells and real time. We show that the most frequent BRAF cancer mutations affect kinase conformations and thus the engagement and efficacy of V600E-specific BRAF inhibitors (BRAFi). We illustrate that the patient mutation harboring KinCon reporters display differences in the effectiveness of the three clinically approved BRAFi vemurafenib, encorafenib, and dabrafenib and the preclinical paradox breaker PLX8394. We confirmed KinCon-based drug efficacy predictions for BRAF mutations other than V600E in proliferation assays using patient-derived lung cancer cell lines and by analyzing downstream kinase signaling. The systematic implementation of such conformation reporters will allow to accelerate the decision process for the mutation-oriented RAF-kinase cancer therapy. Moreover, we illustrate that the presented kinase reporter concept can be extended to other kinases which harbor patient mutations. Overall, KinCon profiling provides additional mechanistic insights into full-length kinase functions by reporting protein-protein interaction (PPI)-dependent, mutation-specific, and drug-driven changes of kinase activity conformations.


Subject(s)
Lung Neoplasms/drug therapy , Protein Conformation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , A549 Cells , Carbamates/chemistry , Carbamates/pharmacology , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation/drug effects , Oximes/chemistry , Oximes/pharmacology , Phosphotransferases/antagonists & inhibitors , Phosphotransferases/ultrastructure , Protein Kinase Inhibitors/chemistry , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/ultrastructure , Sulfonamides/chemistry , Sulfonamides/pharmacology , Vemurafenib/chemistry , Vemurafenib/pharmacology
5.
J Enzyme Inhib Med Chem ; 35(1): 1712-1726, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32962435

ABSTRACT

A series of imidazothiazole derivatives possessing potential activity against melanoma cells were investigated for molecular mechanism of action. The target compounds were tested against V600E-B-RAF and RAF1 kinases. Compound 1zb is the most potent against both kinases with IC50 values 0.978 and 8.2 nM, respectively. It showed relative selectivity against V600E mutant B-RAF kinase. Compound 1zb was also tested against four melanoma cell lines and exerted superior potency (IC50 0.18-0.59 µM) compared to the reference standard drug, sorafenib (IC50 1.95-5.45 µM). Compound 1zb demonstrated also prominent selectivity towards melanoma cells than normal skin cells. It was further tested in whole-cell kinase assay and showed in-cell V600E-B-RAF kinase inhibition with IC50 of 0.19 µM. Compound 1zb induces apoptosis not necrosis in the most sensitive melanoma cell line, UACC-62. Furthermore, molecular dynamic and 3D-QSAR studies were done to investigate the binding mode and understand the pharmacophoric features of this series of compounds.


Subject(s)
Antineoplastic Agents/chemistry , Melanoma/diet therapy , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Thiazoles/chemistry , Antineoplastic Agents/pharmacology , Carbamates/chemistry , Carbamates/pharmacology , Cell Line, Tumor , Cell Proliferation , Drug Screening Assays, Antitumor , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Molecular Dynamics Simulation , Oximes/chemistry , Oximes/pharmacology , Protein Kinase Inhibitors/pharmacology , Quantitative Structure-Activity Relationship , Sorafenib/chemistry , Sorafenib/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Thiazoles/pharmacology , Vemurafenib/chemistry , Vemurafenib/pharmacology
6.
J Med Chem ; 63(8): 4069-4080, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32223235

ABSTRACT

BRAF is among the most frequently mutated oncogenes in human cancers. Multiple small molecule BRAF kinase inhibitors have been approved for treating melanoma carrying BRAF-V600 mutations. However, the benefits of BRAF kinase inhibitors are generally short-lived. Small molecule-mediated targeted protein degradation has recently emerged as a novel pharmaceutical strategy to remove disease proteins through hijacking the cellular ubiquitin proteasome system (UPS). In this study, we developed thalidomide-based heterobifunctional compounds that induced selective degradation of BRAF-V600E, but not the wild-type BRAF. Downregulation of BRAF-V600E suppressed the MEK/ERK kinase cascade in melanoma cells and impaired cell growth in culture. Abolishing the interaction between degraders and cereblon or blocking the UPS significantly impaired the activities of these degraders, validating a mechanistic role of UPS in mediating targeted degradation of BRAF-V600E. These findings highlight a new approach to modulate the functions of oncogenic BRAF mutants and provide a framework to treat BRAF-dependent human cancers.


Subject(s)
Drug Discovery/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins B-raf/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Humans , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Vemurafenib/chemistry , Vemurafenib/metabolism , Vemurafenib/pharmacology
7.
Biochem Biophys Res Commun ; 524(1): 28-35, 2020 03 26.
Article in English | MEDLINE | ID: mdl-31980175

ABSTRACT

BRAFV600E mutation is frequently observed in melanoma, and contributes to tumor malignancy. Despite inhibition of BRAF causes a profound cell growth inhibition and a strong clinical benefit in BRAFV600E melanoma, acquired drug resistance is still the major hurdle. In this study, we demonstrate that BRAFV600E drives cell growth and glycolysis in melanoma cells but does so by a previously unappreciated mechanism that involves direct induction of Skp2. Skp2 is highly expressed in melanoma tissues and particularly in tissues with BRAFV600E mutation. The inhibition of BRAFV600E by either siRNA or inhibitor vemurafenib suppressed Skp2 expression and cell growth. Mechanistic study shows that BRAFV600E suppression of Skp2 is dependent on c-Myc transcription factor via specifically bounding to the E-box region on SKP2 promoter. Further, the overexpression of Skp2 resulted in a markedly increase in cell growth, cell cycle progression and glycolysis which were repressed by BRAFV600E inhibition. Supporting the biological significance, Skp2 is specifically correlated with poor patient outcome in BRAFV600E but did not in BRAFWT melanomas. Thus, as a downstream target of BRAFV600E, Skp2 is critical for responses to BRAF inhibition, indicating targeting Skp2 might be a promising strategy for the treatment of BRAFi resistant melanomas.


Subject(s)
Melanoma/metabolism , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Vemurafenib/chemistry , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Humans , Mutation , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins B-raf/genetics , RNA, Small Interfering/metabolism , Vemurafenib/metabolism
8.
Mol Cancer Ther ; 18(5): 937-946, 2019 05.
Article in English | MEDLINE | ID: mdl-30926642

ABSTRACT

Protein tyrosine kinase 6 (PTK6, also called BRK) is overexpressed and activated in human prostate cancer. Loss of the tumor suppressor PTEN, a frequent event in prostate cancer, leads to PTK6 activation at the plasma membrane and its oncogenic signaling. The small molecule inhibitor vemurafenib, also known as PLX4032, and its tool analog PLX4720 were designed to inhibit constitutively active BRAF V600E, yet they also have potent effects against PTK6. Vemurafenib is used in the treatment of metastatic melanoma, but its efficacy in prostate cancer has not been assessed. When activated at the plasma membrane, PTK6 promotes signaling through FAK, EGFR, and ERK1/2, and we show this can be blocked by vemurafenib. In addition, PTK6-mediated cell growth, migration, and invasion are inhibited upon vemurafenib administration. Using a flank xenograft model, vemurafenib treatment reduced tumor burden. Using saturation transfer difference NMR and molecular docking, we demonstrate that vemurafenib binds in the active site of PTK6, inhibiting its activation. These structural studies provide insight into the PTK6-vemurafenib complex, which can be utilized for further refinement chemistry, whereas functional studies demonstrate that active PTK6 is a viable drug target in prostate cancer.


Subject(s)
Neoplasm Proteins/chemistry , Prostatic Neoplasms/drug therapy , Protein-Tyrosine Kinases/chemistry , Vemurafenib/pharmacology , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , ErbB Receptors/genetics , Focal Adhesion Kinase 1/genetics , Heterografts , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Molecular Docking Simulation , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction/drug effects , Vemurafenib/chemistry
9.
Chem Biol Drug Des ; 93(2): 177-187, 2019 02.
Article in English | MEDLINE | ID: mdl-30225883

ABSTRACT

BRAF kinase is an essential target for anti-cancer drug development. Emergence of the ß3-αC loop deletion mutation (ΔNVTAP) in BRAF kinase frequently occurred in human cancers seriously compromises the therapeutic efficacy of some BRAF kinase inhibitors, such as dabrafenib and vemurafenib. However, the mechanism of this resistance is still not well understood. In this study, the influence of the ß3-αC deletion mutation on the binding profiles of three BRAF kinase inhibitors (AZ628, dabrafenib, and vemurafenib) with BRAFV600E or BRAFΔNVTAP was explored by conventional molecular dynamics (MD) simulations and binding free energy calculations. The simulation results indicated that the ß3-αC deletion mutation enhances the flexibility of the αC helix and alters their conformations, which amplify the conformational entropy change (-TΔS) and weaken the interactions between the inhibitors and BRAF. The further per-residue binding free energy decomposition analysis revealed that the ΔNVTAP mutation changed the contributions of a few key residues to the bindings of dabrafenib or vemurafenib, such as L57, L66, W83, C84, F135, G145, and F147, but did not have obvious impact on the contributions of these residues to AZ628. Our results provide valuable clues to understand the mechanisms of drug resistance conferred by the ß3-αC deletion mutation.


Subject(s)
Imidazoles/chemistry , Oximes/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Vemurafenib/chemistry , Binding Sites , Drug Resistance, Neoplasm , Humans , Imidazoles/metabolism , Molecular Dynamics Simulation , Mutation , Oximes/metabolism , Principal Component Analysis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Thermodynamics , Vemurafenib/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...