Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Med Chem ; 65(2): 1418-1444, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34672584

ABSTRACT

Phosphoinositide-3-kinase γ (PI3Kγ) is highly expressed in immune cells and promotes the production and migration of inflammatory mediators. The inhibition of PI3Kγ has been shown to repolarize the tumor immune microenvironment to a more inflammatory phenotype, thereby controlling immune suppression in cancer. Herein, we report the structure-based optimization of an early lead series of pyrazolopyrimidine isoindolinones, which culminated in the discovery of highly potent and isoform-selective PI3Kγ inhibitors with favorable drug-like properties. X-ray cocrystal structure analysis, molecular docking studies, and detailed structure-activity relationship investigations resulted in the identification of the optimal amide and isoindolinone substituents to achieve a desirable combination of potency, selectivity, and metabolic stability. Preliminary in vitro studies indicate that inhibition of PI3Kγ with compound 56 results in a significant immune response by increasing pro-inflammatory cytokine gene expression in M1 macrophages.


Subject(s)
Amides/chemistry , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Drug Design , Drug Discovery , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Animals , Humans , Male , Molecular Docking Simulation , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
2.
Mol Divers ; 25(3): 1271-1282, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34160714

ABSTRACT

Nowadays, more and more attention has been attracted to develop selective PI3Kγ inhibitors, but the unique structural features of PI3Kγ protein make it a very big challenge. In the present study, a virtual screening strategy based on machine learning with multiple PI3Kγ protein structures was developed to screen novel PI3Kγ inhibitors. First, six mainstream docking programs were chosen to evaluate their scoring power and screening power; CDOCKER and Glide show satisfactory reliability and accuracy against the PI3Kγ system. Next, virtual screening integrating multiple PI3Kγ protein structures was demonstrated to significantly improve the screening enrichment rate comparing to that with an individual protein structure. Last, a multi-conformational Naïve Bayesian Classification model with the optimal docking programs was constructed, and it performed a true capability in the screening of PI3Kγ inhibitors. Taken together, the current study could provide some guidance for the docking-based virtual screening to discover novel PI3Kγ inhibitors.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/chemistry , Machine Learning , Models, Molecular , Molecular Conformation , Phosphoinositide-3 Kinase Inhibitors/chemistry , Binding Sites , Databases, Pharmaceutical , Drug Discovery , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Protein Binding , ROC Curve , Structure-Activity Relationship
3.
Elife ; 102021 03 04.
Article in English | MEDLINE | ID: mdl-33661099

ABSTRACT

Class I Phosphoinositide 3-kinases (PI3Ks) are master regulators of cellular functions, with the class IB PI3K catalytic subunit (p110γ) playing key roles in immune signalling. p110γ is a key factor in inflammatory diseases and has been identified as a therapeutic target for cancers due to its immunomodulatory role. Using a combined biochemical/biophysical approach, we have revealed insight into regulation of kinase activity, specifically defining how immunodeficiency and oncogenic mutations of R1021 in the C-terminus can inactivate or activate enzyme activity. Screening of inhibitors using HDX-MS revealed that activation loop-binding inhibitors induce allosteric conformational changes that mimic those in the R1021C mutant. Structural analysis of advanced PI3K inhibitors in clinical development revealed novel binding pockets that can be exploited for further therapeutic development. Overall, this work provides unique insights into regulatory mechanisms that control PI3Kγ kinase activity and shows a framework for the design of PI3K isoform and mutant selective inhibitors.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/genetics , Immunologic Deficiency Syndromes/genetics , Mutation , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Humans
4.
J Mol Biol ; 433(8): 166838, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33539876

ABSTRACT

Phosphatidylinositol-3-kinases (PI3Ks) are lipid kinases that phosphorylate phosphatidylinositol 4,5-bisphosphate to generate a key lipid second messenger, phosphatidylinositol 3,4,5-bisphosphate. PI3Kα and PI3Kγ require activation by RAS proteins to stimulate signaling pathways that control cellular growth, differentiation, motility and survival. Intriguingly, RAS binding to PI3K isoforms likely differ, as RAS mutations have been identified that discriminate between PI3Kα and PI3Kγ, consistent with low sequence homology (23%) between their RAS binding domains (RBDs). As disruption of the RAS/PI3Kα interaction reduces tumor growth in mice with RAS- and epidermal growth factor receptor driven skin and lung cancers, compounds that interfere with this key interaction may prove useful as anti-cancer agents. However, a structure of PI3Kα bound to RAS is lacking, limiting drug discovery efforts. Expression of full-length PI3K isoforms in insect cells has resulted in low yield and variable activity, limiting biophysical and structural studies of RAS/PI3K interactions. This led us to generate the first RBDs from PI3Kα and PI3Kγ that can be expressed at high yield in bacteria and bind to RAS with similar affinity to full-length PI3K. We also solved a 2.31 Å X-ray crystal structure of the PI3Kα-RBD, which aligns well to full-length PI3Kα. Structural differences between the PI3Kα and PI3Kγ RBDs are consistent with differences in thermal stability and may underly differential RAS recognition and RAS-mediated PI3K activation. These high expression, functional PI3K RBDs will aid in interrogating RAS interactions and could aid in identifying inhibitors of this key interaction.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Protein Interaction Domains and Motifs , Animals , Antineoplastic Agents/pharmacology , Class I Phosphatidylinositol 3-Kinases , Class Ib Phosphatidylinositol 3-Kinase/drug effects , Class Ib Phosphatidylinositol 3-Kinase/genetics , Drug Discovery , Humans , Mice , Mutation , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Protein Binding , Protein Conformation , Protein Isoforms , Sequence Alignment , Signal Transduction , ras Proteins/chemistry , ras Proteins/genetics , ras Proteins/metabolism
5.
J Mol Biol ; 432(22): 5849-5859, 2020 11 06.
Article in English | MEDLINE | ID: mdl-32918948

ABSTRACT

PI3K lipid kinases signal through the PI3K/Akt pathway, regulating cell growth and proliferation. While the structural features that distinguish between the active and inactive states of protein kinases are well established, that has not been the case for lipid kinases, and neither was the structural mechanism controlling the switch between the two states. Class I PI3Ks are obligate heterodimers with catalytic and regulatory subunits. Here, we analyze PI3K crystal structures. Structures with the nSH2 (inactive state) are featured by collapsed activation loop (a-loop) and an IN kinase domain helix 11 (kα11). In the active state, the a-loop is extended and kα11 in the OUT conformation. Our analysis suggests that the nSH2 domain in the regulatory subunit regulates activation, catalysis and autoinhibition through the a-loop. Inhibition, activation and catalytic scenarios are shared by class IA PI3Ks; the activation is mimicked by oncogenic mutations and the inhibition offers an allosteric inhibitor strategy.


Subject(s)
Lipids/chemistry , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Animals , Class I Phosphatidylinositol 3-Kinases/chemistry , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Enzyme Activation , Humans , Mice , Models, Molecular , Phosphatidylinositol 3-Kinase/chemistry , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Protein Binding , Protein Conformation , Protein Domains , Protein Kinases/metabolism
6.
Anticancer Res ; 40(8): 4547-4556, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32727785

ABSTRACT

BACKGROUND/AIM: Phosphatidyl-inositol-3-kinase (PI3K), a cancer therapeutic target, has been exploited for cancer therapy. The natural compounds flavonoids have increasingly been shown to possess anticancer activity. The current study aimed to explore all known flavonoids for their ability to inhibit PI3Kγ. MATERIALS AND METHODS: Virtual screening of flavonoids using molecular docking to the ATP binding site of PI3Kγ was performed. The top 10 scoring flavonoids were selected for pose analysis and binding strength scores. RESULTS: Molecular docking revealed that the 10 selected flavonoids might inhibit PI3Kγ kinase activity. Literature search did not identify studies reporting a bioassay activity for any of these compounds. CONCLUSION: All 10 selected flavonoids are potential PI3Kγ kinase inhibitors and anticancer agents. Interestingly, one of the 10 least scoring flavonoids has been reported to be inactive, as expected, and thus validating the accuracy of the results.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Flavonoids/pharmacology , Neoplasms/enzymology , Binding Sites , Computer Simulation , Down-Regulation , Drug Screening Assays, Antitumor , Flavonoids/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Neoplasms/drug therapy
7.
Commun Biol ; 3(1): 267, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32461675

ABSTRACT

Development of novel PI3K inhibitors is an important strategy to overcome their resistance and poor tolerability in clinical trials. The quassinoid family member Brusatol shows specific inhibitory activity against hematologic malignancies. However, the mechanism of its anti-cancer activity is unknown. We investigated the anti-cancer activity of Brusatol on multiple hematologic malignancies derived cell lines. The results demonstrated that the PI3Kγ isoform was identified as a direct target of Brusatol, and inhibition was dramatically reduced on cells with lower PI3Kγ levels. Novel synthetic analogs were also developed and tested in vitro and in vivo. They shared comparable or superior potency in their ability to inhibit malignant hematologic cell lines, and in a xenograft transplant mouse model. One unique analog had minimal toxicity to normal human cells and in a mouse model. These new analogs have enhanced potential for development as a new class of PI3K inhibitors for treatment of hematologic malignancies.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/genetics , Hematologic Neoplasms/drug therapy , Quassins/pharmacology , Animals , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Hematologic Neoplasms/genetics , Heterografts , Isoenzymes , Male , Mice , Mice, Inbred NOD , Transplantation, Heterologous
8.
Mol Cancer Ther ; 19(3): 755-764, 2020 03.
Article in English | MEDLINE | ID: mdl-31974273

ABSTRACT

Macrophages (MΦ) play a critical role in tumor growth, immunosuppression, and inhibition of adaptive immune responses in cancer. Hence, targeting signaling pathways in MΦs that promote tumor immunosuppression will provide therapeutic benefit. PI3Kγ has been recently established by our group and others as a novel immuno-oncology target. Herein, we report that an MΦ Syk-PI3K axis drives polarization of immunosuppressive MΦs that establish an immunosuppressive tumor microenvironment in in vivo syngeneic tumor models. Genetic or pharmacologic inhibition of Syk and/or PI3Kγ in MΦs promotes a proinflammatory MΦ phenotype, restores CD8+ T-cell activity, destabilizes HIF under hypoxia, and stimulates an antitumor immune response. Assay for transposase-accessible Chromatin using Sequencing (ATAC-seq) analyses on the bone marrow-derived macrophages (BMDM) show that inhibition of Syk kinase promotes activation and binding of NF-κB motif in SykMC-KO BMDMs, thus stimulating immunostimulatory transcriptional programming in MΦs to suppress tumor growth. Finally, we have developed in silico the "first-in-class" dual Syk/PI3K inhibitor, SRX3207, for the combinatorial inhibition of Syk and PI3K in one small molecule. This chemotype demonstrates efficacy in multiple tumor models and represents a novel combinatorial approach to activate antitumor immunity.


Subject(s)
Carcinoma, Lewis Lung/immunology , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Colonic Neoplasms/immunology , Macrophages/immunology , Melanoma, Experimental/immunology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Syk Kinase/antagonists & inhibitors , Animals , Apoptosis , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/enzymology , Carcinoma, Lewis Lung/pathology , Cell Proliferation , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Cytokines/metabolism , Humans , Immune Tolerance , Immunosuppression Therapy , Macrophages/drug effects , Macrophages/metabolism , Melanoma, Experimental/drug therapy , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Future Med Chem ; 11(16): 2151-2169, 2019 08.
Article in English | MEDLINE | ID: mdl-31538525

ABSTRACT

Recently, PI3Kγ, a vital kinase, which involved in numerous intracellular signaling pathways, has been considered as a promising drug target for the treatment of immune diseases and certain cancers. Before the 21st century, few selective PI3Kγ inhibitors were discovered because no non-conserved structure in the ATP binding sites of PI3Kγ had been found. Since the discovery of the non-ATP binding pocket, the reported structures of potent and selective PI3Kγ inhibitors have become more diverse, and one compound (IPI549) has entered Phase I clinical trial. This review centers on a general overview of PI3Kγ inhibitors in clinical and preclinical as well as further therapeutic applications in human diseases.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/immunology , Inflammation/drug therapy , Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors/chemistry , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Animals , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Clinical Trials as Topic , Drug Development , Drug Discovery , Humans , Inflammation/immunology , Models, Molecular , Neoplasms/immunology , Phosphoinositide-3 Kinase Inhibitors/pharmacology
10.
PLoS One ; 14(6): e0219180, 2019.
Article in English | MEDLINE | ID: mdl-31247018

ABSTRACT

The PI3K-AKT-mTOR pathway is often a commonly disrupted pathway in human cancer and, therefore, it is widely exploited for cancer therapy. The inhibitors for the important proteins of the pathway including PI3K and mTOR have been increasingly designed. The dual inhibitors targeting PI3K and mTOR both have proven to be more effective than those targeting single protein only. An orally-active compound XL765 is well established as PI3K/mTOR dual inhibitor and have shown in vitro and in vivo anticancer activity against a variety of cancer types and is undergoing clinical trials. The present study explored the exact binding pose and the the interactive forces holding XL765 within the active sites of PI3Kγ and mTOR using molecular docking analyses. The XL765 interacting residues of both the proteins were delineated and the degree of participation in binding was estimated by various methods. In the process, among the interacting residues of PI3Kγ, the Lys-890 and the Met-953 were recognized as the key residues involved in XL765 binding. While, in mTOR case, the Trp-2239 was recognized as the key residue playing role in the XL765 binding. In order to explore the better inhibitors, the study also generated combinatorial chemical library by modifying the scaffold considered from XL765. The virtual screening of the generated compound library led to identification of six novel promising compounds proposed as PI3K/mTOR dual inhibitors. Thus, the present work will through light on the drug inhibitory mechanism of XL765 for PI3K and mTOR, and will also assist in designing novel efficacious drug candidates.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Quinoxalines/pharmacology , Sulfonamides/pharmacology , TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/metabolism , Binding Sites , Catalytic Domain , Computer Simulation , Drug Screening Assays, Antitumor , Humans , Lysine/metabolism , Methionine/metabolism , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Quinoxalines/chemistry , Sulfonamides/chemistry
11.
Cancer Lett ; 442: 91-103, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30391357

ABSTRACT

Phosphoinositide 3-kinase gamma isoform (PI3Kγ) plays a critical role in myeloid-derived cells of the immunosuppressive tumor microenvironment. IPI-549, a recently discovered small molecule selective PI3Kγ inhibitor, is currently under immuno-oncology clinical trials in combination with nivolumab, an anti-PD-1 monoclonal antibody immune checkpoint blocker. The purpose of this study is to investigate whether IPI-549 could reverse P-glycoprotein (P-gp)-mediated MDR when combined with chemotherapeutic substrates of P-gp. Cytotoxicity assays showed that IPI-549 reverses P-gp-mediated MDR in SW620/Ad300 and LLC-PK-MDR1 cells. IPI-549 increases the amount of intracellular paclitaxel and inhibits the efflux of paclitaxel out of SW620/Ad300 cells. ABCB1-ATPase assay showed that IPI-549 stimulates the activity of ABCB1-ATPase. IPI-549 does not alter the expression and does not affect the subcellular localization of P-gp in SW620/Ad300 cells. The combination of IPI-549 with paclitaxel showed that IPI-549 potentiates the anti-tumor effects of paclitaxel in P-gp-overexpressing MDR SW620/Ad300 xenograft tumors. With clinical trials beginning to add newly approved immune checkpoint-based immunotherapy into standard-of-care immunogenic chemotherapy to improve patient outcomes, our findings support the rationale of adding IPI-549 to both the chemotherapeutic and immunotherapeutic aspects of cancer combination treatment strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Paclitaxel/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , ATP Binding Cassette Transporter, Subfamily B/agonists , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Colonic Neoplasms/enzymology , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Humans , LLC-PK1 Cells , Male , Mice, Nude , Molecular Docking Simulation , Neoplasm Proteins/metabolism , Paclitaxel/metabolism , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/chemistry , Signal Transduction/drug effects , Swine , Xenograft Model Antitumor Assays
12.
Chem Biol Drug Des ; 93(5): 818-831, 2019 05.
Article in English | MEDLINE | ID: mdl-30582283

ABSTRACT

The phosphoinositide 3-kinase γ (PI3Kγ) has been verified to be a potential drug target for the treatments of various human physical disorders. Although received lots of attention, the development of PI3Kγ-selective inhibitors is still a challenging subject because of its unique protein structural features. Aiming to uncover the interaction mechanism between the selective inhibitors and PI3Kγ, a series of benzothiazole and thiazolopiperidine PI3Kγ isoform-selective inhibitors were studied with an integrated in silico strategy by combining molecular docking, molecular dynamic simulations, binding free energy calculations, and decomposition analysis. Firstly, three molecular docking models, including rigid receptor docking, induced fit docking (IFD), and quantum mechanical-polarized ligand docking, were respectively, built, and the IFD preliminarily predicted the docking poses of all studied inhibitors and roughly analyzed the binding mechanism. Secondly, four binding complexes with representative inhibitors were selected to perform molecular dynamic simulations and free energy calculations. The predicted binding energies were consistent with the experimental bioactivities and different binding patterns between potent and weak inhibitors were uncovered. Finally, through the Molecular Mechanics/Generalized Born Surface Area binding free energy decomposition, residue-inhibitor interactions spectra were obtained and several key residues contributing to favorable binding were highlighted, which provides valuable information for rational PI3Kγ inhibitor design and modification.


Subject(s)
Benzothiazoles/chemistry , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Enzyme Inhibitors/chemistry , Piperidines/chemistry , Benzothiazoles/metabolism , Binding Sites , Catalytic Domain , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Enzyme Inhibitors/metabolism , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Piperidines/metabolism , Thermodynamics
13.
ChemMedChem ; 11(14): 1517-30, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27310202

ABSTRACT

The phosphoinositide 3-kinase (PI3K) pathway is aberrantly activated in many disease states, including tumor cells, either by growth factor receptor tyrosine kinases or by the genetic mutation and amplification of key pathway components. A variety of PI3K isoforms play differential roles in cancers. As such, the development of PI3K inhibitors from novel compound classes should lead to differential pharmacological and pharmacokinetic profiles and allow exploration in various indications, combinations, and dosing regimens. A screening effort aimed at the identification of PI3Kγ inhibitors for the treatment of inflammatory diseases led to the discovery of the novel 2,3-dihydroimidazo[1,2-c]quinazoline class of PI3K inhibitors. A subsequent lead optimization program targeting cancer therapy focused on inhibition of PI3Kα and PI3Kß. Herein, initial structure-activity relationship findings for this class and the optimization that led to the identification of copanlisib (BAY 80-6946) as a clinical candidate for the treatment of solid and hematological tumors are described.


Subject(s)
Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Quinazolines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Drug Discovery , Humans , Hydrogen Bonding , Imidazoles/chemical synthesis , Imidazoles/chemistry , Molecular Docking Simulation , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Quinazolines/chemical synthesis , Quinazolines/chemistry , Structure-Activity Relationship
14.
Curr Comput Aided Drug Des ; 11(4): 304-20, 2015.
Article in English | MEDLINE | ID: mdl-26603929

ABSTRACT

The PI3K pathway is a signal transduction process including oncogenes and receptor tyrosine kinase regulating cellular functions i.e., survival, protein synthesis, and metabolism. In the present work, we have investigated the role of water molecules on inhibitor's binding orientation in crystal structures of PI3K pathway targets using molecular docking approach. AutoDock v4.2 docking software was employed to dock PI3Kγ and its known inhibitors viz., wortmannin, quercetin, myricetin and pyridyl-triazine. Besides, serpentine was also docked on the same binding pocket, subsequently its anticancer activity was evaluated through in vitro experiment. Docking studies have been performed in the presence as well as in absence of water molecules at the binding pocket, and results were compared with crystallographic structural data. The comparison was done on the basis of binding energy, RMSD, inhibition constant (Ki), conserved and bridging water molecules, and found that, while considering water molecules during docking experiments, it increases the binding affinity of PI3K inhibitors.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Water/metabolism , Androstadienes/chemistry , Androstadienes/pharmacology , Binding Sites , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Flavonoids/chemistry , Flavonoids/pharmacology , Humans , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Binding , Quercetin/chemistry , Quercetin/pharmacology , Software , Triazines/chemistry , Triazines/pharmacology , Water/chemistry , Wortmannin
15.
Biochem J ; 469(1): 59-69, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26173259

ABSTRACT

Class IB phosphoinositide 3-kinases γ (PI3Kγ) are second-messenger-generating enzymes downstream of signalling cascades triggered by G-protein-coupled receptors (GPCRs). PI3Kγ variants have one catalytic p110γ subunit that can form two different heterodimers by binding to one of a pair of non-catalytic subunits, p87 or p101. Growing experimental data argue for a different regulation of p87-p110γ and p101-p110γ allowing integration into distinct signalling pathways. Pharmacological tools enabling distinct modulation of the two variants are missing. The ability of an anti-p110γ monoclonal antibody [mAb(A)p110γ] to block PI3Kγ enzymatic activity attracted us to characterize this tool in detail using purified proteins. In order to get insight into the antibody-p110γ interface, hydrogen-deuterium exchange coupled to MS (HDX-MS) measurements were performed demonstrating binding of the monoclonal antibody to the C2 domain in p110γ, which was accompanied by conformational changes in the helical domain harbouring the Gßγ-binding site. We then studied the modulation of phospholipid vesicles association of PI3Kγ by the antibody. p87-p110γ showed a significantly reduced Gßγ-mediated phospholipid recruitment as compared with p101-p110γ. Concomitantly, in the presence of mAb(A)p110γ, Gßγ did not bind to p87-p110γ. These data correlated with the ability of the antibody to block Gßγ-stimulated lipid kinase activity of p87-p110γ 30-fold more potently than p101-p110γ. Our data argue for differential regulatory functions of the non-catalytic subunits and a specific Gßγ-dependent regulation of p101 in PI3Kγ activation. In this scenario, we consider the antibody as a valuable tool to dissect the distinct roles of the two PI3Kγ variants downstream of GPCRs.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Class Ib Phosphatidylinositol 3-Kinase , GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , Animals , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Deuterium Exchange Measurement , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/chemistry , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/metabolism , HEK293 Cells , Humans , Sf9 Cells , Spodoptera
16.
Immunol Cell Biol ; 93(8): 735-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25753393

ABSTRACT

Phosphoinositide 3-kinase γ (PI3Kγ) consists of the catalytic subunit p110γ that forms a mutually exclusive heterodimer with one of the two adaptor subunits, p101 or p84. Although activation of PI3Kγ is necessary for cell migration downstream of G-protein-coupled receptor engagement, particularly within the immune system, aberrant PI3Kγ signalling has been associated with transformation, increased migration and the progression of multiple cancer types. Regulation of PI3Kγ signal activation and duration is critical to controlling and maintaining coordinated cellular migration; however, the mechanistic basis for this is not well understood. We have recently demonstrated that, in contrast to the tumour-promoting potential of p110γ and p101, p84 possesses tumour-suppressor activity, suggesting a negative regulatory role within PI3Kγ signalling. The present study investigated the role of p84 phosphorylation in the context of PI3Kγ signalling, cell migration and p84-mediated tumour suppression. Two putative phosphorylation sites were characterised within p84, Ser358 and Thr607. Expression of wild-type p84 reduced the oncogenic potential of MDA.MB.231 cells and inhibited metastatic lung colonisation in vivo, effects that were dependent on Thr607. Furthermore, loss of Thr607 enhanced migration of MDA.MB.231 cells in vitro and prevented the induction of p84/p110γ dimers. The dimerisation of wild-type p84 with p110γ was not detected at the plasma membrane, indicating an inhibitory interaction preventing PI3Kγ lipid-kinase activity. In contrast, Ser358 phosphorylation was not determined to be critical for p84 activity in the context of migration. Our findings suggest that p84 binding to p110γ may represent a novel negative feedback signal that terminates PI3Kγ activity.


Subject(s)
Cell Movement , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Multiprotein Complexes/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Cell Line , Chemokine CXCL12/pharmacology , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/genetics , Female , Gene Expression , Humans , Immunoprecipitation , Mice , Molecular Sequence Data , Mutation , Phosphorylation , Protein Binding , Protein Multimerization , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled , Recombinant Fusion Proteins , Signal Transduction/drug effects
17.
J Med Chem ; 58(1): 517-21, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-24754609

ABSTRACT

Phosphoinositide 3-kinase γ (PI3Kγ) is an attractive target to potentially treat a range of disease states. Herein, we describe the evolution of a reported phenylthiazole pan-PI3K inhibitor into a family of potent and selective benzothiazole inhibitors. Using X-ray crystallography, we discovered that compound 22 occupies a previously unreported hydrophobic binding cleft adjacent to the ATP binding site of PI3Kγ, and achieves its selectivity by exploiting natural sequence differences among PI3K isoforms in this region.


Subject(s)
Benzothiazoles/pharmacology , Enzyme Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Benzothiazoles/chemistry , Benzothiazoles/metabolism , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Molecular Structure , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
18.
PLoS One ; 9(2): e87309, 2014.
Article in English | MEDLINE | ID: mdl-24586269

ABSTRACT

Plumbagin (5-hydroxy-2-methyl-1,4-naphthoquinone) is a naphthoquinone derivative from the roots of plant Plumbago zeylanica and belongs to one of the largest and diverse groups of plant metabolites. The anticancer and antiproliferative activities of plumbagin have been observed in animal models as well as in cell cultures. Plumbagin exerts inhibitory effects on multiple cancer-signaling proteins, however, the binding mode and the molecular interactions have not yet been elucidated for most of these protein targets. The present study is the first attempt to provide structural insights into the binding mode of plumbagin to five cancer signaling proteins viz. PI3Kγ, AKT1/PKBα, Bcl-2, NF-κB, and Stat3 using molecular docking and (un)binding simulation analysis. We validated plumbagin docking to these targets with previously known important residues. The study also identified and characterized various novel interacting residues of these targets which mediate the binding of plumbagin. Moreover, the exact modes of inhibition when multiple mode of inhibition existed was also shown. Results indicated that the engaging of these important interacting residues in plumbagin binding leads to inhibition of these cancer-signaling proteins which are key players in the pathogenesis of cancer and thereby ceases the progression of the disease.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Naphthoquinones/chemistry , Animals , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Databases, Pharmaceutical , Humans , Ligands , Mice , Models, Molecular , Molecular Conformation , NF-kappa B/chemistry , NF-kappa B/metabolism , Naphthoquinones/metabolism , Naphthoquinones/pharmacology , Protein Binding , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/metabolism
19.
Proc Natl Acad Sci U S A ; 110(47): 18862-7, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24190998

ABSTRACT

Phosphoinositide 3-kinase gamma (PI3Kγ) has profound roles downstream of G-protein-coupled receptors in inflammation, cardiac function, and tumor progression. To gain insight into how the enzyme's activity is shaped by association with its p101 adaptor subunit, lipid membranes, and Gßγ heterodimers, we mapped these regulatory interactions using hydrogen-deuterium exchange mass spectrometry. We identify residues in both the p110γ and p101 subunits that contribute critical interactions with Gßγ heterodimers, leading to PI3Kγ activation. Mutating Gßγ-interaction sites of either p110γ or p101 ablates G-protein-coupled receptor-mediated signaling to p110γ/p101 in cells and severely affects chemotaxis and cell transformation induced by PI3Kγ overexpression. Hydrogen-deuterium exchange mass spectrometry shows that association with the p101 regulatory subunit causes substantial protection of the RBD-C2 linker as well as the helical domain of p110γ. Lipid interaction massively exposes that same helical site, which is then stabilized by Gßγ. Membrane-elicited conformational change of the helical domain could help prepare the enzyme for Gßγ binding. Our studies and others identify the helical domain of the class I PI3Ks as a hub for diverse regulatory interactions that include the p101, p87 (also known as p84), and p85 adaptor subunits; Rab5 and Gßγ heterodimers; and the ß-adrenergic receptor kinase.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Models, Molecular , Phosphatidylinositol 3-Kinases/metabolism , Protein Conformation , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Chemotaxis , Class Ib Phosphatidylinositol 3-Kinase/genetics , Deuterium Exchange Measurement , Enzyme Activation , HEK293 Cells , Humans , Mass Spectrometry , Mice , Microscopy, Confocal , NIH 3T3 Cells , Receptors, G-Protein-Coupled/agonists , Signal Transduction/genetics , ras Proteins/metabolism
20.
J Chem Inf Model ; 53(10): 2587-612, 2013 Oct 28.
Article in English | MEDLINE | ID: mdl-24050502

ABSTRACT

The mammalian target of rapamycin (mTOR) has an important role in cell growth, proliferation, and survival. mTOR is frequently hyperactivated in cancer, and therefore, it is a clinically validated target for cancer therapy. In this study, we combined exhaustive pharmacophore modeling and quantitative structure-activity relationship (QSAR) analysis to explore the structural requirements for potent mTOR inhibitors employing 210 known mTOR ligands. Genetic function algorithm (GFA) coupled with k nearest neighbor (kNN) and multiple linear regression (MLR) analyses were employed to build self-consistent and predictive QSAR models based on optimal combinations of pharmacophores and physicochemical descriptors. Successful pharmacophores were complemented with exclusion spheres to optimize their receiver operating characteristic curve (ROC) profiles. Optimal QSAR models and their associated pharmacophore hypotheses were validated by identification and experimental evaluation of several new promising mTOR inhibitory leads retrieved from the National Cancer Institute (NCI) structural database. The most potent hit illustrated an IC50 value of 48 nM.


Subject(s)
Algorithms , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Models, Chemical , Protein Kinase Inhibitors/chemistry , TOR Serine-Threonine Kinases/chemistry , Bayes Theorem , Computer Simulation , Crystallography, X-Ray , Databases, Chemical , Drug Discovery , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Inhibitory Concentration 50 , Ligands , Linear Models , Phosphoinositide-3 Kinase Inhibitors , Quantitative Structure-Activity Relationship , ROC Curve , TOR Serine-Threonine Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...