Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Xenobiotica ; 54(2): 64-74, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38197324

ABSTRACT

Glioblastoma multiforme (GBM) is the most common primary brain tumour in adults. Available treatments have not markedly improved patient survival in the last twenty years. However, genomic investigations have showed that the PI3K pathway is frequently altered in this glioma, making it a potential therapeutic target.Paxalisib is a brain penetrant PI3K/mTOR inhibitor (mouse Kp,uu 0.31) specifically developed for the treatment of GBM. We characterised the preclinical pharmacokinetics and efficacy of paxalisib and predicted its pharmacokinetics and efficacious dose in humans.Plasma protein binding of paxalisib was low, with the fraction unbound ranging from 0.25 to 0.43 across species. The hepatic clearance of paxalisib was predicted to be low in mice, rats, dogs and humans, and high in monkeys, from hepatocytes incubations. The plasma clearance was low in mice, moderate in rats and high in dogs and monkeys. Oral bioavailability ranged from 6% in monkeys to 76% in rats.The parameters estimated from the pharmacokinetic/pharmacodynamic modelling of the efficacy in the subcutaneous U87 xenograft model combined with the human pharmacokinetics profile predicted by PBPK modelling suggested that a dose of 56 mg may be efficacious in humans. Paxalisib is currently tested in Phase III clinical trials.


Subject(s)
Phosphatidylinositol 3-Kinases , Protein Kinase Inhibitors , Humans , Rats , Mice , Animals , Dogs , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Phosphoinositide-3 Kinase Inhibitors/metabolism , Brain/metabolism , TOR Serine-Threonine Kinases/metabolism
2.
Cell Mol Neurobiol ; 43(7): 3623-3637, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37314618

ABSTRACT

The disruption of the blood-brain barrier (BBB) is hypothesized to be involved in the progression of anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis, but its mechanism is still unclear. Recently, the phosphatidylinositol 3-kinase (PI3K)/threonine kinase (Akt) pathway is involved in the regulation of the BBB in various diseases. This study is aimed to investigate the mechanism of BBB damage and neurobehavior changes in anti-NMDAR encephalitis mice. Female C57BL/6J mice were actively immunized to establish an anti-NMDAR encephalitis mouse model and evaluate the neurobehavior changes of mice. To study its potential mechanism, LY294002 (PI3K inhibitor, 8 mg/kg) and Recilisib (PI3K agonist, 10 mg/kg) were treated by intraperitoneal injection, respectively. Anti-NMDAR encephalitis mice showed neurological deficits, increased BBB permeability, open endothelial tight junctions (TJs), and decreased expression of TJ-related proteins zonula occludens (ZO)-1 and Claudin-5. However, administration of PI3K inhibitor significantly reduced the expression of p-PI3K and p-Akt, improved neurobehavior function, decreased BBB permeability, and upregulated the expressions of ZO-1 and Claudin-5. Furthermore, PI3K inhibition reversed the decline of NMDAR NR1 in the membranes of hippocampal neurons, which reduced the loss of neuron-specific nucleoprotein (NeuN) and microtubule-associated protein 2 (MAP2). In contrast, administration of the PI3K agonist Recilisib showed a tendency to exacerbate BBB breakdown and neurological deficits. Our results showed that the activation of PI3K/Akt, along with the changes in TJ-related proteins ZO-1 and Claudin-5, may be closely related to BBB damage and neurobehavior changes in anti-NMDAR encephalitis mice. PI3K inhibition attenuates BBB disruption and neuronal damage in mice, thereby improving neurobehavior.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis , Blood-Brain Barrier , Mice , Female , Animals , Blood-Brain Barrier/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/metabolism , Claudin-5/metabolism , Mice, Inbred C57BL , Signal Transduction , Tight Junctions/metabolism , Phosphoinositide-3 Kinase Inhibitors/metabolism
3.
Nat Commun ; 13(1): 245, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017531

ABSTRACT

About 15-20% of breast cancer (BCa) is triple-negative BCa (TNBC), a devastating disease with limited therapeutic options. Aberrations in the PI3K/PTEN signaling pathway are common in TNBC. However, the therapeutic impact of PI3K inhibitors in TNBC has been limited and the mechanism(s) underlying this lack of efficacy remain elusive. Here, we demonstrate that a large subset of TNBC expresses significant levels of MAPK4, and this expression is critical for driving AKT activation independent of PI3K and promoting TNBC cell and xenograft growth. The ability of MAPK4 to bypass PI3K for AKT activation potentially provides a direct mechanism regulating tumor sensitivity to PI3K inhibition. Accordingly, repressing MAPK4 greatly sensitizes TNBC cells and xenografts to PI3K blockade. Altogether, we conclude that high MAPK4 expression defines a large subset or subtype of TNBC responsive to MAPK4 blockage. Targeting MAPK4 in this subset/subtype of TNBC both represses growth and sensitizes tumors to PI3K blockade.


Subject(s)
Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , Triple Negative Breast Neoplasms/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, SCID , Mitogen-Activated Protein Kinases/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphoinositide-3 Kinase Inhibitors/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
4.
Eur J Med Chem ; 229: 113996, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34802837

ABSTRACT

Established roles for PI3K and MAPK signaling pathways in tumorigenesis has prompted extensive research towards the discovery of small-molecule inhibitors as cancer therapeutics. However, significant compensatory regulation exists between these two signaling cascades, leading to redundancy among survival pathways. Consequently, initial clinical trials aimed at either PI3K or MEK inhibition alone have proven ineffective and highlight the need for development of targeted and innovative therapeutic combination strategies. We designed a series of PI3K inhibitor derivatives wherein a single morpholine group of the PI3K inhibitor ZSTK474 was substituted with a variety of 2-aminoethyl functional groups. Analogs with pendant hydroxyl or methoxy groups maintained low nanomolar inhibition towards PI3Kα, PI3Kγ, and PI3Kδ isoforms in contrast to those with pendant amino groups which were significantly less inhibitory. Synthesis of prototype PI3K/MEK bifunctional inhibitors (6r, 6s) was guided by the structure-activity data, where a MEK-targeting inhibitor was tethered directly via a short PEG linker to the triazine core of the PI3K inhibitor analogs. These compounds (6r, 6s) displayed nanomolar inhibition towards PI3Kα, δ, and MEK (IC50 ∼105-350 nM), and low micromolar inhibition for PI3Kß and PI3Kγ (IC50 ∼1.5-3.9 µM) in enzymatic inhibition assays. Cell viability assays demonstrated superior anti-proliferative activity for 6s over 6r in three tumor-derived cell lines (A375, D54, SET-2), which correlated with inhibition of downstream AKT and ERK1/2 phosphorylation. Compounds 6r and 6s also demonstrated in vivo tolerability with therapeutic efficacy through reduction of kinase activation and amelioration of disease phenotypes in the JAK2V617F mutant myelofibrosis mouse cancer model. Taken together, these results support further structure optimization of 6r and 6s as promising leads for combination therapy in human cancer as a new class of PI3K/MEK bifunctional inhibitors.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Morpholines/chemistry , Phosphatidylinositol 3-Kinases/chemistry , Phosphoinositide-3 Kinase Inhibitors/chemistry , Triazines/chemistry , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Docking Simulation , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/metabolism , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Primary Myelofibrosis/drug therapy , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Structure-Activity Relationship , Triazines/metabolism , Triazines/therapeutic use
5.
Nat Commun ; 12(1): 7190, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34907165

ABSTRACT

Interrogation of cellular metabolism with high-throughput screening approaches can unravel contextual biology and identify cancer-specific metabolic vulnerabilities. To systematically study the consequences of distinct metabolic perturbations, we assemble a comprehensive metabolic drug library (CeMM Library of Metabolic Drugs; CLIMET) covering 243 compounds. We, next, characterize it phenotypically in a diverse panel of myeloid leukemia cell lines and primary patient cells. Analysis of the drug response profiles reveals that 77 drugs affect cell viability, with the top effective compounds targeting nucleic acid synthesis, oxidative stress, and the PI3K/mTOR pathway. Clustering of individual drug response profiles stratifies the cell lines into five functional groups, which link to specific molecular and metabolic features. Mechanistic characterization of selective responses to the PI3K inhibitor pictilisib, the fatty acid synthase inhibitor GSK2194069, and the SLC16A1 inhibitor AZD3965, bring forth biomarkers of drug response. Phenotypic screening using CLIMET represents a valuable tool to probe cellular metabolism and identify metabolic dependencies at large.


Subject(s)
Leukemia, Myeloid/metabolism , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Survival/drug effects , Cluster Analysis , Fatty Acids/biosynthesis , Genotype , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/pathology , Monocarboxylic Acid Transporters/genetics , Phenotype , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Pyrimidinones/metabolism , Pyrimidinones/pharmacology , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Signal Transduction , Small Molecule Libraries/classification , Symporters/genetics , Systems Analysis , Thiophenes/metabolism , Thiophenes/pharmacology , Triazoles/metabolism , Triazoles/pharmacology , Tumor Cells, Cultured
6.
J Med Chem ; 64(18): 13780-13792, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34510892

ABSTRACT

Optimization of a previously reported lead series of PI3Kδ inhibitors with a novel binding mode led to the identification of a clinical candidate compound 31 (GSK251). Removal of an embedded Ames-positive heteroaromatic amine by reversing a sulfonamide followed by locating an interaction with Trp760 led to a highly selective compound 9. Further optimization to avoid glutathione trapping, to enhance potency and selectivity, and to optimize an oral pharmacokinetic profile led to the discovery of compound 31 (GSK215) that had a low predicted daily dose (45 mg, b.i.d) and a rat toxicity profile suitable for further development.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology , Animals , Crystallography, X-Ray , Female , Male , Mice, Inbred BALB C , Molecular Structure , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors/metabolism , Protein Binding , Rats, Wistar , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/metabolism
8.
Respir Res ; 22(1): 238, 2021 Aug 26.
Article in English | MEDLINE | ID: mdl-34446024

ABSTRACT

BACKGROUND: Childhood asthma is a common respiratory disease characterized by airway inflammation. Tumor necrosis factor-α-induced protein 8-like 2 (TIPE2) has been found to be involved in the progression of asthma. This study aimed to explore the role of TIPE2 in the regulation of airway smooth muscle cells (ASMCs), which are one of the main effector cells in the development of asthma. MATERIALS AND METHODS: ASMCs were transfected with pcDNA3.0-TIPE2 or si-TIPE2 for 48 h and then treated with platelet-derived growth factor (PDGF)-BB. Cell proliferation of ASMCs was measured using the MTT assay. Cell migration of ASMCs was determined by a transwell assay. The mRNA expression levels of calponin and smooth muscle protein 22α (SM22α) were measured using qRT-PCR. The levels of TIPE2, calponin, SM22α, PI3K, p-PI3K, Akt, and p-Akt were detected by Western blotting. RESULTS: Our results showed that PDGF-BB treatment significantly reduced TIPE2 expression at both the mRNA and protein levels in ASMCs. Overexpression of TIPE2 inhibited PDGF-BB-induced ASMC proliferation and migration. In addition, overexpression of TIPE2 increased the expression of calponin and SM22α in PDGF-BB-stimulated ASMCs. However, an opposite effect was observed with TIPE2 knockdown. Furthermore, TIPE2 overexpression blocked PDGF-BB-induced phosphorylation of PI3K and Akt, whereas the expression of p-PI3K and p-Akt were aggravated by TIPE2 knockdown. Additionally, the effects of TIPE2 overexpression and TIPE2 knockdown were altered by IGF-1 and LY294002 treatments, respectively. CONCLUSIONS: Our findings demonstrate that TIPE2 inhibits PDGF-BB-induced ASMC proliferation, migration, and phenotype switching via the PI3K/Akt signaling pathway. Thus, TIPE2 may be a potential therapeutic target for the treatment of asthma.


Subject(s)
Becaplermin/toxicity , Intracellular Signaling Peptides and Proteins/biosynthesis , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Airway Remodeling/drug effects , Airway Remodeling/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Mice, Inbred C57BL , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Phenotype , Phosphoinositide-3 Kinase Inhibitors/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Trachea/cytology , Trachea/drug effects , Trachea/metabolism
9.
ACS Chem Biol ; 16(9): 1644-1653, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34397208

ABSTRACT

Covalent inhibition is a powerful strategy to develop potent and selective small molecule kinase inhibitors. Targeting the conserved catalytic lysine is an attractive method for selective kinase inactivation. We have developed novel, selective inhibitors of phosphoinositide 3-kinase δ (PI3Kδ) which acylate the catalytic lysine, Lys779, using activated esters as the reactive electrophiles. The acylating agents were prepared by adding the activated ester motif to a known selective dihydroisobenzofuran PI3Kδ inhibitor. Three esters were designed, including an acetate ester which was the smallest lysine modification evaluated in this work. Covalent binding to the enzyme was characterized by intact protein mass spectrometry of the PI3Kδ-ester adducts. An enzymatic digest coupled with tandem mass spectrometry identified Lys779 as the covalent binding site, and a biochemical activity assay confirmed that PI3Kδ inhibition was a direct result of covalent lysine acylation. These results indicate that a simple chemical modification such as lysine acetylation is sufficient to inhibit kinase activity. The selectivity of the compounds was evaluated against lipid kinases in cell lysates using a chemoproteomic binding assay. Due to the conserved nature of the catalytic lysine across the kinome, we believe the covalent inhibition strategy presented here could be applicable to a broad range of clinically relevant targets.


Subject(s)
Acrylamides/chemistry , Adenine/analogs & derivatives , Afatinib/chemistry , Aniline Compounds/chemistry , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Lysine/chemistry , Phosphoinositide-3 Kinase Inhibitors/chemistry , Piperidines/chemistry , Acetylation , Acrylamides/metabolism , Adenine/chemistry , Adenine/metabolism , Afatinib/metabolism , Amino Acid Sequence , Aniline Compounds/metabolism , Catalysis , Catalytic Domain , Class I Phosphatidylinositol 3-Kinases/metabolism , Humans , Mass Spectrometry , Molecular Docking Simulation , Phosphoinositide-3 Kinase Inhibitors/metabolism , Piperidines/metabolism , Protein Binding , Protein Conformation , Substrate Specificity
10.
Eur J Med Chem ; 223: 113661, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34237636

ABSTRACT

Based on indole scaffold, a potent and selective phosphoinositide 3-kinase delta (PI3Kδ) inhibitor, namely FD223, was developed by the bioisosteric replacement drug discovery approach and studied for the treatment of acute myeloid leukemia (AML). In vitro studies revealed that FD223 displays high potency (IC50 = 1 nM) and selectivity (29-51 fold over other PI3K isoforms) against PI3Kδ, and exhibits efficient inhibition of the proliferation of AML cell lines (MOLM-16, HL-60, EOL-1 and KG-1) by suppressing p-AKT Ser473 thus causing G1 phase arrest during the cell cycle. Further given the favorable pharmacokinetic (PK) profiles of FD223, in vivo studies were evaluated using xenograft model in nude mice, confirming its significant antitumor efficacy meanwhile with no observable toxicity. All these results are comparable to the positive group of Idelalisib (CAL-101), indicating that FD223 has potential for further development as a promising PI3Kδ inhibitor for the treatment of leukemia such as AML.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Drug Design , Indoles/chemistry , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Half-Life , Humans , Indoles/metabolism , Indoles/pharmacology , Indoles/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Nude , Molecular Docking Simulation , Phosphoinositide-3 Kinase Inhibitors/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Transplantation, Heterologous
11.
J Med Chem ; 64(11): 7331-7340, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33876637

ABSTRACT

Aberrant activation of the PI3K pathway has been intensively targeted for cancer therapeutics for decades, leading to more than 40 PI3K inhibitors advanced into clinical trials. However, it is increasingly noticed that PI3K inhibitors often showed limited efficacy as well as a number of serious on-target adverse effects during the clinical development. In this work, we designed and synthesized a novel photocaged PI3K inhibitor 1, which could be readily activated by UV irradiation to release a highly potent PI3K inhibitor 2. Upon UV irradiation, the photocaged inhibitor 1 demonstrated remarkably enhanced antiproliferative activity against multiple cancer cell lines and significant efficacy in the patient-derived tumor organoid model. Furthermore, 1 also showed favorable anticancer activity in an in vivo zebrafish xenograft model. Taken together, the photocaged PI3K inhibitor 1 represents a promising avenue for novel therapeutics toward precise cancer treatment.


Subject(s)
Drug Design , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Animals , Binding Sites , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Dogs , Drug Stability , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Molecular Dynamics Simulation , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/chemistry , Phosphoinositide-3 Kinase Inhibitors/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rats , Signal Transduction/drug effects , Ultraviolet Rays , Xenograft Model Antitumor Assays , Zebrafish/metabolism
12.
BMC Complement Med Ther ; 21(1): 83, 2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33663486

ABSTRACT

BACKGROUND: Vasculogenic mimicry (VM) is characterized by the formation of tubular structure inside the tumor stroma. It has been shown that a small fraction of cancer cells, namely cancer stem cells (CSCs), could stimulate the development of vascular units in the tumor niche, leading to enhanced metastasis to the remote sites. This study aimed to study the inhibitory effect of phytocompound, Thymoquinone (TQ), on human breast MDA-MB-231 cell line via monitoring Wnt/PI3K signaling pathway. METHODS: MDA-MB-231 CSCs were incubated with different concentrations of TQ for 48 h. The viability of CSCs was determined using the MTT assay. The combination of TQ and PI3K and Wnt3a inhibitors was examined in CSCs. By using the Matrigel assay, we measured the tubulogenesis capacity. The percent of CD24- CSCs and Rhodamine 123 efflux capacity was studied using flow cytometry analysis. Protein levels of Akt, p-Akt, Wnt3a, vascular endothelial-cadherin (VE-cadherin), and matrix metalloproteinases-2 and -9 (MMP-2 and -9) were detected by western blotting. RESULTS: TQ decreased the viability of CSCs in a dose-dependent manner. The combination of TQ with PI3K and Wnt3a inhibitors reduced significantly the survival rate compared to the control group (p < 0.05). TQ could blunt the stimulatory effect of vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), fibroblast growth factor (FGF) on CSCs (p < 0.05). The vasculogenic capacity of CSCs was reduced after being-exposed to TQ (p < 0.05). Western blotting revealed the decrease of CSCs metastasis by suppressing MMP-2 and -9. The protein level of VE-cadherin was also diminished in TQ-treated CSCs as compared to the control cell (p < 0.05), indicating inhibition of mesenchymal-endothelial transition (MendT). TQ could suppress Wnt3a and PI3K, which coincided with the reduction of the p-Akt/Akt ratio. TQ had the potential to decrease the number of CD24- CSCs and Rhodamine 123 efflux capacity after 48 h. CONCLUSION: TQ could alter the vasculogenic capacity and mesenchymal-epithelial transition of human breast CSCs in vitro. Thus TQ together with anti-angiogenic therapies may be a novel therapeutic agent in the suppression of VM in breast cancer.


Subject(s)
Benzoquinones/pharmacology , Breast Neoplasms/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Phosphoinositide-3 Kinase Inhibitors/metabolism , Wnt3A Protein/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Neoplastic Stem Cells
13.
Chem Biol Drug Des ; 97(6): 1158-1169, 2021 06.
Article in English | MEDLINE | ID: mdl-33657663

ABSTRACT

Phosphatidylinositol-3-kinase (PI3K) is important for cell proliferation, differentiation, and apoptosis, and the diverse physiological roles of different PI3K isoforms have highlighted the significance of the development of PI3Kδ inhibitors. A large number of PI3Kδ inhibitors have been reported after the FDA approval of Idelalisib, but the clinical use of Idelalisib was limited because of its serious side effects. Therefore, great efforts have been made on the development of PI3Kδ inhibitors with higher selectivity and lower toxicity, but there is no new PI3Kδ inhibitor coming into the market so far. Even so, as the first listed PI3K inhibitor, Idelalisib could be used as an effective tool to investigate the selective inhibition mechanism of PI3Kδ. Thus, in this study, a modeling strategy integrated 3D-QSAR, pharmacophore model, and molecular dynamics simulation was employed to reveal the key chemical characteristics of Idelalisib analogs and the binding pattern between the inhibitors and PI3Kδ. First, the CoMFA model with high statistical significance was built to reveal the general structure-activity relationships. And then, a reliable pharmacophore model with a robust discrimination capability was constructed to expound the main chemical characteristics of the PI3Kδ inhibitors. Finally, molecular dynamics simulation was conducted to explore the binding modes and some key residues refer to δ-selective binding were highlighted with binding-free energy calculation. In summary, these models and results would provide some effective help for the discovery or the rational design of novel PI3Kδ inhibitors.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/chemistry , Molecular Dynamics Simulation , Phosphoinositide-3 Kinase Inhibitors/chemistry , Purines/chemistry , Quinazolinones/chemistry , Area Under Curve , Binding Sites , Class I Phosphatidylinositol 3-Kinases/metabolism , Humans , Phosphoinositide-3 Kinase Inhibitors/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Purines/metabolism , Quantitative Structure-Activity Relationship , Quinazolinones/metabolism , ROC Curve , Static Electricity , Thermodynamics
14.
J Med Chem ; 63(23): 14700-14723, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33297683

ABSTRACT

PI3Kδ inhibitors have been approved for B-cell malignancies like CLL, small lymphocytic lymphoma, and so forth. However, currently available PI3Kδ inhibitors are nonoptimal, showing weakness against at least one of the several important properties: potency, isoform selectivity, and/or pharmacokinetic profile. To come up with a PI3Kδ inhibitor that overcomes all these deficiencies, a pharmacophoric expansion strategy was employed. Herein, we describe a systematic transformation of a "three-blade propeller" shaped lead, 2,3-disubstituted quinolizinone 11, through a 1,2-disubstituted quinolizinone 20 to a novel "four-blade propeller" shaped 1,2,3-trisubstituted quinolizinone 34. Compound 34 has excellent potency, isoform selectivity, metabolic stability across species, and exhibited a favorable pharmacokinetic profile. Compound 34 also demonstrated a differentiated efficacy profile in human germinal center B and activated B cell-DLBCL cell lines and xenograft models. Compound 34 qualifies for further evaluation as a candidate for monotherapy or in combination with other targeted agents in DLBCLs and other forms of iNHL.


Subject(s)
Antineoplastic Agents/therapeutic use , Class I Phosphatidylinositol 3-Kinases/therapeutic use , Hematologic Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Quinolizines/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/chemical synthesis , Class I Phosphatidylinositol 3-Kinases/metabolism , Class I Phosphatidylinositol 3-Kinases/pharmacokinetics , Dogs , Drug Discovery , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Molecular Docking Simulation , Molecular Structure , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacokinetics , Quinolizines/chemical synthesis , Quinolizines/metabolism , Quinolizines/pharmacokinetics , RAW 264.7 Cells , Rats, Sprague-Dawley , Structure-Activity Relationship , Xenograft Model Antitumor Assays
15.
Sci Rep ; 10(1): 20548, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33239624

ABSTRACT

Acidosis is associated with E. coli induced pyelonephritis but whether bacterial cell wall constituents inhibit HCO3 transport in the outer medullary collecting duct from the inner stripe (OMCDi) is not known. We examined the effect of lipopolysaccharide (LPS), on HCO3 absorption in isolated perfused rabbit OMCDi. LPS caused a ~ 40% decrease in HCO3 absorption, providing a mechanism for E. coli pyelonephritis-induced acidosis. Monophosphoryl lipid A (MPLA), a detoxified TLR4 agonist, and Wortmannin, a phosphoinositide 3-kinase inhibitor, prevented the LPS-mediated decrease, demonstrating the role of TLR4-PI3-kinase signaling and providing proof-of-concept for therapeutic interventions aimed at ameliorating OMCDi dysfunction and pyelonephritis-induced acidosis.


Subject(s)
Bicarbonates/metabolism , Lipopolysaccharides/metabolism , Renal Reabsorption/drug effects , Acidosis, Renal Tubular/metabolism , Acidosis, Renal Tubular/physiopathology , Animals , Bicarbonates/chemistry , Escherichia coli/metabolism , Female , Kidney/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Collecting/metabolism , Lipid A/analogs & derivatives , Lipid A/metabolism , Lipopolysaccharides/pharmacology , Loop of Henle/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors/metabolism , Pyelonephritis/metabolism , Rabbits , Signal Transduction/drug effects
16.
Drugs ; 80(16): 1685-1697, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32894420

ABSTRACT

Approximately 70% of invasive breast cancers have some degree of dependence on the estrogen hormone for cell proliferation and growth. These tumors have estrogen and/or progesterone receptors (ER/PR+), generally referred to as hormone receptor positive (HR+) tumors, as indicated by the presence of positive staining and varying intensity levels of estrogen and/or progesterone receptors on immunohistochemistry. Therapies that inhibit ER signaling pathways, such as aromatase inhibitors (letrozole, anastrozole, exemestane), selective ER modulators (tamoxifen), and ER down-regulators (fulvestrant), are the mainstays of treatment for hormone-receptor-positive breast cancers. However, de novo or acquired resistance to ER targeted therapies is present in many tumors, leading to disease progression. The PI3K/AKT/mTOR pathway is implicated in sustaining endocrine resistance and has become the target of many new drugs for ER+ breast cancer. This article reviews the function of the phosphoinositide 3-kinase (PI3K)/AKT/mTOR pathway and the various classes of PI3K pathway inhibitors that have been developed to disrupt this pathway signaling for the treatment of hormone-receptor-positive breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Neoplasm Recurrence, Local/drug therapy , Signal Transduction/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , DNA Mutational Analysis , Drug Resistance, Neoplasm/genetics , Female , Humans , Mutation , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Phosphoinositide-3 Kinase Inhibitors/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome
17.
Eur J Med Chem ; 208: 112865, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32987316

ABSTRACT

Inhibition of PI3Kδ has been proved to be an efficacious strategy for the treatment of hematological malignancies where the PI3K/Akt signaling pathway is hyperactive. Herein, a series of quinazoline derivatives bearing acrylamide fragment were prepared using skeleton-deconstruction strategy. The preliminary bioactivity evaluation resulted in the discovery of lead compound 15c. Compound 15c exhibited excellent enzyme activity against PI3Kδ (IC50 = 27.5 nM) compared with BEZ235 as well as the significant anti-proliferation activities. With the high selectivity over other PI3K isoforms and potent effects on PI3K/Akt pathway, 15c can be identified as a promising PI3Kδ inhibitor worthy of further profiling.


Subject(s)
Antineoplastic Agents/pharmacology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases/metabolism , Drug Screening Assays, Antitumor , Humans , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Structure , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors/metabolism , Protein Binding , Quinazolines/chemical synthesis , Quinazolines/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
18.
Int J Mol Sci ; 21(12)2020 Jun 25.
Article in English | MEDLINE | ID: mdl-32630372

ABSTRACT

Oncogenic activation of the phosphatidylinositol-3-kinase (PI3K), protein kinase B (PKB/AKT), and mammalian target of rapamycin (mTOR) pathway is a frequent event in prostate cancer that facilitates tumor formation, disease progression and therapeutic resistance. Recent discoveries indicate that the complex crosstalk between the PI3K-AKT-mTOR pathway and multiple interacting cell signaling cascades can further promote prostate cancer progression and influence the sensitivity of prostate cancer cells to PI3K-AKT-mTOR-targeted therapies being explored in the clinic, as well as standard treatment approaches such as androgen-deprivation therapy (ADT). However, the full extent of the PI3K-AKT-mTOR signaling network during prostate tumorigenesis, invasive progression and disease recurrence remains to be determined. In this review, we outline the emerging diversity of the genetic alterations that lead to activated PI3K-AKT-mTOR signaling in prostate cancer, and discuss new mechanistic insights into the interplay between the PI3K-AKT-mTOR pathway and several key interacting oncogenic signaling cascades that can cooperate to facilitate prostate cancer growth and drug-resistance, specifically the androgen receptor (AR), mitogen-activated protein kinase (MAPK), and WNT signaling cascades. Ultimately, deepening our understanding of the broader PI3K-AKT-mTOR signaling network is crucial to aid patient stratification for PI3K-AKT-mTOR pathway-directed therapies, and to discover new therapeutic approaches for prostate cancer that improve patient outcome.


Subject(s)
Phosphatidylinositol 3-Kinase/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Androgen Antagonists/therapeutic use , Cell Line, Tumor , Cell Proliferation , Humans , Male , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Recurrence, Local/genetics , Oncogenes , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/metabolism , Prostatic Neoplasms/genetics , Receptors, Androgen/metabolism , Wnt Signaling Pathway/physiology
19.
Pharmacol Res ; 158: 104868, 2020 08.
Article in English | MEDLINE | ID: mdl-32407961

ABSTRACT

Hepatocellular carcinoma (HCC) is a lethal malignancy with limited treatment options. The tyrosine kinase receptor EphB4 promotes oncogenesis and tumor development and progression. Its inhibition is regarded as an effective strategy for the treatment of solid tumors. In the present study, we identified cantharidin as a novel EphB4 inhibitor for HCC treatment and evaluated the underlying molecular pharmacological mechanisms of action. We observed increased expression levels of EphB4 in HCC patients and a positive correlation between EphB4 and p-JAK2 levels in HCC patient samples. Knockdown of EphB4 using small interfering RNA decreased the expression levels of p-JAK2 and p-STAT3 in HCC cells, suggesting JAK2/STAT3 being a novel downstream signaling target of EphB4. Cell viability experiments revealed that the anti-cancer effect of cantharidin was positively correlated with EphB4 expression levels in HCC cell lines. We confirmed the potent antiproliferative activity of cantharidin on HepG2 cells with high expression of EphB4 and tumor xenograft. Molecular docking assay, immunoblotting assay and quantitative reverse transcription PCR assay indicated that cantharidin bound to EphB4, and thereby resulted in EphB4 suppression at mRNA and protein levels. Hep3B and SMMC-7721 cells were with low expression of EphB4. In EphB4-/HepG2, EphB4+/HepG2, and EphB4+/Hep3B cells, EphB4 knockdown alleviated the cantharidin-induced decrease in cell viability and colony formation ability and increase in apoptosis in HepG2 cells, while its overexpression exacerbated these effects in Hep3B cells and increased the apoptosis of HepG2 cells. In nude mouse models, cantharidin suppressed tumor growth more effectively in EphB4+/SMMC-7721 xenografts than in wild-type SMMC-7721 xenografts. Underlying mechanistic study showed that by targeting EphB4, cantharidin blocked a novel target, the downstream JAK2/STAT3 pathway, and the previously known target, the PI3K/Akt signaling, resulting in intrinsic apoptosis. These results indicated that cantharidin may be a potential candidate for HCC treatment by regulating the EphB4 signaling pathway.


Subject(s)
Cantharidin/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Phosphoinositide-3 Kinase Inhibitors/metabolism , Receptor, EphB4/antagonists & inhibitors , Receptor, EphB4/metabolism , Animals , Cantharidin/pharmacology , Cantharidin/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Hep G2 Cells , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Liver Neoplasms/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Protein Structure, Secondary , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptor, EphB4/chemistry , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
20.
Eur J Med Chem ; 197: 112309, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32375077

ABSTRACT

Using a rational design strategy for isoform-selective inhibition of PI3Kα, two series of novel 2,3,4,5-tetra-substituted thiophene derivatives containing either diaryl urea or N-Acylarylhydrazone scaffold were designed and synthesized. The most promising compound 12k was demonstrated to bear nanomolar PI3Kα inhibitory potency with 12, 28, 30, 196-fold selectivity against isoforms ß, γ, δ and mTOR. Besides, it also showed good developability profiles in cell-based proliferation against a panel of human tumor cells as well as ADME assays. We herein report on their design, synthesis, SAR and potential developability properties.


Subject(s)
Antineoplastic Agents/pharmacology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Thiophenes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacokinetics , Protein Binding , Structure-Activity Relationship , TOR Serine-Threonine Kinases/antagonists & inhibitors , Thiophenes/chemical synthesis , Thiophenes/metabolism , Thiophenes/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...