Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 729
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 181(7): 1518-1532.e14, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32497502

ABSTRACT

The rise of antibiotic resistance and declining discovery of new antibiotics has created a global health crisis. Of particular concern, no new antibiotic classes have been approved for treating Gram-negative pathogens in decades. Here, we characterize a compound, SCH-79797, that kills both Gram-negative and Gram-positive bacteria through a unique dual-targeting mechanism of action (MoA) with undetectably low resistance frequencies. To characterize its MoA, we combined quantitative imaging, proteomic, genetic, metabolomic, and cell-based assays. This pipeline demonstrates that SCH-79797 has two independent cellular targets, folate metabolism and bacterial membrane integrity, and outperforms combination treatments in killing methicillin-resistant Staphylococcus aureus (MRSA) persisters. Building on the molecular core of SCH-79797, we developed a derivative, Irresistin-16, with increased potency and showed its efficacy against Neisseria gonorrhoeae in a mouse vaginal infection model. This promising antibiotic lead suggests that combining multiple MoAs onto a single chemical scaffold may be an underappreciated approach to targeting challenging bacterial pathogens.


Subject(s)
Gram-Negative Bacteria/drug effects , Pyrroles/metabolism , Pyrroles/pharmacology , Quinazolines/metabolism , Quinazolines/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Female , Folic Acid/metabolism , Gram-Positive Bacteria/drug effects , HEK293 Cells , Humans , Male , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Ovariectomy , Proteomics , Pseudomonas aeruginosa/drug effects
2.
Bioorg Med Chem Lett ; 96: 129518, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37838344

ABSTRACT

The NLRP3 inflammasome is a multiprotein complex that plays a critical role in activating the immune system in response to danger signals. Small molecule agonists of NLRP3 may offer clinical benefits in cancer immunology either as a monotherapy or in combination with checkpoint blockade, where it is hypothesised that their application can help to initiate an antitumor immune response. In this study, we report the discovery of quinazolines and 8-azaquinazolines as NLRP3 agonists and their chemical optimization to afford compounds with oral bioavailability in mice. We confirm that these compounds engage the NLRP3 inflammasome by verifying their dependence upon lipopolysaccharide (LPS) priming for cytokine release and the activation of Caspase-1. We further demonstrate pathway engagement through loss of activity in an NLRP3-knockout THP1 cell line. Based on their pharmacokinetic profile and biological activity, these compounds represent valuable tools to evaluate the therapeutic potential of NLRP3 activation in a pre-clinical setting.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Mice , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Biological Availability , Quinazolines/pharmacology , Quinazolines/metabolism , Macrophages/metabolism , Caspase 1/metabolism , Lipopolysaccharides/pharmacology , Interleukin-1beta/metabolism
3.
Int J Mol Sci ; 24(21)2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37958505

ABSTRACT

Arsenic is a carcinogenic metalloid toxicant widely found in the natural environment. Acute or prolonged exposure to arsenic causes a series of damages to the organs, mainly the liver, such as hepatomegaly, liver fibrosis, cirrhosis, and even hepatocellular carcinoma. Therefore, it is imperative to seek drugs to prevent arsenic-induced liver injury. Quinazolines are a class of nitrogen heterocyclic compounds with biological and pharmacological effects in vivo and in vitro. This study was designed to investigate the ameliorating effects of quinazoline derivatives on arsenic-induced liver injury and its molecular mechanism. We investigated the mechanism of the quinazoline derivative KZL-047 in preventing and ameliorating arsenic-induced liver injury in vitro by cell cycle and apoptosis. We performed real-time fluorescence quantitative polymerase chain reaction (qPCR) and Western blotting combined with molecular docking. In vivo, the experiments were performed to investigate the mechanism of KZL-047 in preventing and ameliorating arsenic-induced liver injury using arsenic-infected mice. Physiological and biochemical indices of liver function in mouse serum were measured, histopathological changes in liver tissue were observed, and immunohistochemical staining was used to detect changes in the expression of RecQ-family helicases in mouse liver tissue. The results of in vitro experiments showed that sodium arsenite (SA) inhibited the proliferation of L-02 cells, induced apoptosis, blocked the cell cycle at the G1 phase, and decreased the expression of RecQ family helicase; after KZL-047 treatment in arsenic-induced L-02 cells, the expression of RecQ family helicase was upregulated, and the apoptosis rate was slowed, leading to the restoration of the cell viability level. KZL-047 inhibited arsenic-induced oxidative stress, alleviated oxidative damage and lipid peroxidation in vivo, and ameliorated arsenic toxicity-induced liver injury. KZL-047 restored the expression of RecQ family helicase proteins, which is consistent with the results of in vitro studies. In summary, KZL-047 can be considered a potential candidate for the treatment of arsenic-induced liver injury.


Subject(s)
Arsenic , Arsenites , Chemical and Drug Induced Liver Injury, Chronic , Mice , Animals , Arsenic/toxicity , Arsenic/metabolism , RecQ Helicases/metabolism , Quinazolines/pharmacology , Quinazolines/metabolism , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Molecular Docking Simulation , Liver/metabolism , Oxidative Stress , Liver Cirrhosis/metabolism , Arsenites/toxicity
4.
Molecules ; 28(5)2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36903565

ABSTRACT

Sapitinib (AZD8931, SPT) is a tyrosine kinase inhibitor of the epidermal growth factor receptor (EGFR) family (pan-erbB). In multiple tumor cell lines, STP has been shown to be a much more potent inhibitor of EGF-driven cellular proliferation than gefitinib. In the current study, a highly sensitive, rapid, and specific LC-MS/MS analytical method for the estimation of SPT in human liver microsomes (HLMs) was established with application to metabolic stability assessment. The LC-MS/MS analytical method was validated in terms of linearity, selectivity, precision, accuracy, matrix effect, extraction recovery, carryover, and stability following the FDA guidelines for bioanalytical method validation. SPT was detected using electrospray ionization (ESI) as an ionization source under multiple reaction monitoring (MRM) in the positive ion mode. The IS-normalized matrix factor and extraction recovery were acceptable for the bioanalysis of SPT. The SPT calibration curve was linear, from 1 ng/mL to 3000 ng/mL HLM matrix samples, with a linear regression equation of y = 1.7298x + 3.62941 (r2 = 0.9949). The intraday and interday accuracy and precision values of the LC-MS/MS method were -1.45-7.25% and 0.29-6.31%, respectively. SPT and filgotinib (FGT) (internal standard; IS) were separated through the use of an isocratic mobile phase system with a Luna 3 µm PFP(2) column (150 × 4.6 mm) stationary phase column. The limit of quantification (LOQ) was 0.88 ng/mL, confirming the LC-MS/MS method sensitivity. The intrinsic clearance and in vitro half-life of STP were 38.48 mL/min/kg and 21.07 min, respectively. STP exhibited a moderate extraction ratio that revealed good bioavailability. The literature review demonstrated that the current analytical method is the first developed LC-MS/MS method for the quantification of SPT in an HLM matrix with application to SPT metabolic stability evaluation.


Subject(s)
Microsomes, Liver , Tandem Mass Spectrometry , Humans , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Microsomes, Liver/metabolism , Quinazolines/metabolism , Reproducibility of Results
5.
Cell Mol Biol (Noisy-le-grand) ; 68(4): 108-112, 2022 Apr 30.
Article in English | MEDLINE | ID: mdl-35988274

ABSTRACT

In this study, the antitumor effects of tubulin-binding agent MPC-6827 on HeLa, MCF-7 and A549 cell lines originated from cervix carcinoma, metastatic breast adenocarcinoma and adenocarcinomic human alveolar basal epithelial cells respectively were determined. Cell index, BrdU labelling index, mitotic index and apoptotic index were evaluated in experiments. In cell index experiment 2 nM, 4 nM, 6 nM, 8 nM, 10 nM MPC-6827 applied to three cell lines. These parameters showed that 4 nM was the optimum concentration for HeLa and A549 cells, while 2 nM was the optimum concentration for MCF-7 cells. The use of optimum concentrations for each cell line has shown that while there was a significant decrease in mitotic index, BrdU labelling index, there was a significant increase in apoptotic index.


Subject(s)
Antineoplastic Agents , Neoplasms , Quinazolines , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Bromodeoxyuridine/pharmacology , Cell Line, Tumor/drug effects , Cell Proliferation , Female , HeLa Cells , Humans , Neoplasms/drug therapy , Quinazolines/metabolism , Quinazolines/pharmacology , Tubulin/metabolism , Tubulin/pharmacology
6.
Addict Biol ; 27(1): e13060, 2022 01.
Article in English | MEDLINE | ID: mdl-34013595

ABSTRACT

The epigenetic enzyme G9a is a histone methyltransferase that dimethylates lysine 9 on histone H3 (H3K9me2), and in the adult nucleus accumbens (NAc), G9a regulates multiple behaviors associated with substance use disorder. We show here that chronic intermittent ethanol (CIE) exposure in male mice reduced both G9a and H3K9me2 levels in the adult NAc, but not dorsal striatum. Viral-mediated reduction of G9a in the NAc had no effects on baseline volitional ethanol drinking or escalated alcohol drinking produced by CIE exposure; however, NAc G9a was required for stress-regulated changes in ethanol drinking, including potentiated alcohol drinking produced by activation of the kappa-opioid receptor. In addition, we observed that chronic systemic administration of a G9a inhibitor, UNC0642, also blocked stress-potentiated alcohol drinking. Together, our findings suggest that chronic alcohol use, similar to other abused substances, produces a NAc-selective reduction in G9a levels that serves to limit stress-regulated alcohol drinking. Moreover, our findings suggest that pharmacological inhibition of G9a might provide a novel therapeutic approach to treat stress-induced alcohol drinking, which is a major trigger of relapse in individuals suffering from AUD.


Subject(s)
Alcohol Drinking/metabolism , Histone Methyltransferases/metabolism , Quinazolines/metabolism , Stress, Psychological/metabolism , Animals , Epigenesis, Genetic , Ethanol , Histones/metabolism , Male , Mice , Nucleus Accumbens/metabolism
7.
Int J Mol Sci ; 23(24)2022 Dec 11.
Article in English | MEDLINE | ID: mdl-36555369

ABSTRACT

Quinazoline derivatives have various pharmacological activities and are widely used in clinical practice. Here, we reviewed the proposed mechanisms of the physiological activity of the quinazoline derivative EVP4593 and perspectives for its clinical implication. We summarized the accumulated data about EVP4593 and focused on its activities in different models of Huntington's disease (HD), including patient-specific iPSCs-based neurons. To make a deeper insight into its neuroprotective role in HD treatment, we discussed the ability of EVP4593 to modulate calcium signaling and reduce the level of the huntingtin protein. Moreover, we described possible protective effects of EVP4593 in other pathologies, such as oncology, cardiovascular diseases and parasite invasion. We hope that comprehensive analyses of the molecular mechanisms of EVP4593 activity will allow for the expansion of the scope of the EVP4593 application.


Subject(s)
Huntington Disease , Humans , Huntington Disease/metabolism , Neurons/metabolism , Quinazolines/pharmacology , Quinazolines/therapeutic use , Quinazolines/metabolism , Phenyl Ethers/pharmacology , Huntingtin Protein/metabolism
8.
Int J Mol Sci ; 23(14)2022 Jul 11.
Article in English | MEDLINE | ID: mdl-35886992

ABSTRACT

Chikungunya virus (CHIKV) has repeatedly spread via the bite of an infected mosquito and affected more than 100 countries. The disease poses threats to public health and the economy in the infected locations. Many efforts have been devoted to identifying compounds that could inhibit CHIKV. Unfortunately, successful clinical candidates have not been found yet. Computations through the simulating recognition process were performed on complexation of the nsP3 protein of CHIKV with the structures of triply conjugated drug lead candidates. The outcomes provided the aid on rational design of functionalized quinazoline-(α-substituted coumarin)-arylsulfonate compounds to inhibit CHIKV in Vero cells. The molecular docking studies showed a void space around the ß carbon atom of coumarin when a substituent was attached at the α position. The formed vacancy offered a good chance for a Michael addition to take place owing to steric and electronic effects. The best conjugate containing a quinazolinone moiety exhibited potency with EC50 = 6.46 µM, low toxicity with CC50 = 59.7 µM, and the selective index (SI) = 9.24. Furthermore, the corresponding 4-anilinoquinazoline derivative improved the anti-CHIKV potency to EC50 = 3.84 µM, CC50 = 72.3 µM, and SI = 18.8. The conjugate with 4-anilinoquinazoline exhibited stronger binding affinity towards the macro domain than that with quinazolinone via hydrophobic and hydrogen bond interactions.


Subject(s)
Chikungunya virus , Animals , Antiviral Agents/chemistry , Arylsulfonates/metabolism , Arylsulfonates/pharmacology , Chlorocebus aethiops , Computer-Aided Design , Coumarins/pharmacology , Molecular Docking Simulation , Quinazolines/metabolism , Quinazolines/pharmacology , Quinazolinones/pharmacology , Vero Cells , Virus Replication
9.
Int J Mol Sci ; 23(5)2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35269887

ABSTRACT

Mesenchymal stem cells (MSCs) are considered to be a powerful tool in the treatment of various diseases. Scientists are particularly interested in the possibility of using MSCs in cancer therapy. The research carried out so far has shown that MSCs possess both potential pro-oncogenic and anti-oncogenic properties. It has been confirmed that MSCs can regulate tumor cell growth through a paracrine mechanism, and molecules secreted by MSCs can promote or block a variety of signaling pathways. These findings may be crucial in the development of new MSC-based cell therapeutic strategies. The abilities of MSCs such as tumor tropism, deep migration and immune evasion have evoked considerable interest in their use as tumor-specific vectors for small-molecule anticancer agents. Studies have shown that MSCs can be successfully loaded with chemotherapeutic drugs such as gemcitabine and paclitaxel, and can release them at the site of primary and metastatic neoplasms. The inhibitory effect of MSCs loaded with anti-cancer agents on the proliferation of cancer cells has also been observed. However, not all known chemotherapeutic agents can be used in this approach, mainly due to their cytotoxicity towards MSCs and insufficient loading and release capacity. Quinazoline derivatives appear to be an attractive choice for this therapeutic solution due to their biological and pharmacological properties. There are several quinazolines that have been approved for clinical use as anticancer drugs by the US Food and Drug Administration (FDA). It gives hope that the synthesis of new quinazoline derivatives and the development of methods of their application may contribute to the establishment of highly effective therapies for oncological patients. However, a deeper understanding of interactions between MSCs and tumor cells, and the exploration of the possibilities of using quinazoline derivatives in MSC-based therapy is necessary to achieve this goal. The aim of this review is to discuss the prospects for using MSC-based cell therapy in cancer treatment and the potential use of quinazolines in this procedure.


Subject(s)
Antineoplastic Agents , Mesenchymal Stem Cells , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Humans , Mesenchymal Stem Cells/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Quinazolines/metabolism , Quinazolines/pharmacology , Quinazolines/therapeutic use
10.
Bioorg Med Chem Lett ; 49: 128212, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34153471

ABSTRACT

In this study, a series of multifunctional hybrids (6a-6l) against Alzheimer's disease were designed and obtained by conjugating the pharmacophores of deoxyvasicinone and indole. These analogs of deoxyvasicinone-indole were evaluated as inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), and as inhibitors of amyloid aggregation (Aß1-42) for treatment of Alzheimer's disease (AD). Subsequently, AChE induced Aß aggregation inhibition test was also performed for selected compounds. Biological activity results demonstrated that compound 6b was the most potent and balanced dual ChEs inhibitor with IC50 values 0.12 µM and 0.15 µM for eeAChE and eqBuChE, respectively. Kinetic analysis and docking study indicated that compound 6b was a mixed-type inhibitor for both AChE and BuChE. Compound 6b also found to be the best inhibitors of self-induced Aß1-42 aggregation with IC50 values of 1.21 µM. Compound 6b also afforded excellent inhibition of AChE-induced Aß1-42 aggregation by 81.1%. Overall, these results indicate that 6b may be considered as lead compound for the development of highly effective anti-AD drugs.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/pharmacology , Indoles/pharmacology , Neuroprotective Agents/pharmacology , Quinazolines/pharmacology , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Amyloid beta-Peptides/metabolism , Animals , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/metabolism , Catalytic Domain , Cell Line, Tumor , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/toxicity , Drug Design , Electrophorus , Horses , Indoles/chemical synthesis , Indoles/metabolism , Indoles/toxicity , Kinetics , Mice , Molecular Docking Simulation , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Neuroprotective Agents/toxicity , Peptide Fragments/metabolism , Protein Binding , Protein Multimerization/drug effects , Quinazolines/chemical synthesis , Quinazolines/metabolism , Quinazolines/toxicity , Rats
11.
Int J Mol Sci ; 22(10)2021 May 19.
Article in English | MEDLINE | ID: mdl-34069531

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the most common neurodegenerative disease, and it manifests as progressive memory loss and cognitive decline. However, there are no effective therapies for AD, which is an urgent problem to solve. Evodiamine, one of the main bioactive ingredients of Evodia rutaecarpa, has been reported to ameliorate blood-brain barrier (BBB) permeability and improve cognitive impairment in ischemia and AD mouse models. However, whether evodiamine alleviates tauopathy remains unclear. This study aimed to examine whether evodiamine ameliorates tau phosphorylation and cognitive deficits in AD models. METHODS: A protein phosphatase 2A inhibitor, okadaic acid (OA), was used to induce tau phosphorylation to mimic AD-like models in neuronal cells. Protein expression and cell apoptosis were detected using Western blotting and flow cytometry, respectively. Spatial memory/cognition was assessed using water maze, passive avoidance tests, and magnetic resonance imaging assay in OA-induced mice models, and brain slices were evaluated further by immunohistochemistry. RESULTS: The results showed that evodiamine significantly reduced the expression of phosphor-tau, and further decreased tau aggregation and neuronal cell death in response to OA treatment. This inhibition was found to be via the inhibition of glycogen synthase kinase 3ß, cyclin-dependent kinase 5, and mitogen-activated protein kinase pathways. In vivo results indicated that evodiamine treatment ameliorated learning and memory impairments in mice, whereas Western blotting and immunohistochemical analysis of the mouse brain also confirmed the neuroprotective effects of evodiamine. CONCLUSIONS: Evodiamine can decrease the neurotoxicity of tau aggregation and exhibit a neuroprotective effect. Our results demonstrate that evodiamine has a therapeutic potential for AD treatment.


Subject(s)
Quinazolines/pharmacology , Tauopathies/drug therapy , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Animals , Apoptosis/drug effects , Brain/metabolism , Cell Line , Cognition/drug effects , Cognition/physiology , Cognition Disorders/metabolism , Disease Models, Animal , Humans , Male , Maze Learning/drug effects , Mice , Mice, Inbred ICR , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Okadaic Acid/adverse effects , Okadaic Acid/pharmacology , Phosphorylation , Quinazolines/metabolism , Spatial Memory/drug effects , tau Proteins/drug effects , tau Proteins/metabolism
12.
Molecules ; 26(16)2021 Aug 21.
Article in English | MEDLINE | ID: mdl-34443658

ABSTRACT

In recent decades, fungi-derived naturally occurring quinazolines have emerged as potential drug candidates. Nevertheless, most studies are conducted for bioactivity assays, and little is known about their absorption, distribution, metabolism, and elimination (ADME) properties. To perform metabolic studies, the synthesis of the naturally occurring quinazolinone, fiscalin B (1), and its chloro derivative, 4-((1H-indol-3-yl)methyl)-8,10-dichloro-1-isobutyl-1,2-dihydro-6H-pyrazino[2,1-b]quinazoline-3,6(4H)-dione (2), disclosed as an antibacterial agent, was performed in a gram scale using a microwave-assisted polycondensation reaction with 22% and 17% yields, respectively. The structure of the non-natural (+)-fiscalin B was established, for the first time, by X-ray crystallography as (1R,4S)-1, and the absolute configuration of the naturally occurring fiscalin B (-)-1 was confirmed by comparison of its calculated and experimental electronic circular dichroism (ECD) spectra as (1S,4R)-1. in vitro metabolic studies were monitored for this class of natural products for the first time by ultra-high-performance liquid chromatography (UHPLC) coupled with high-resolution mass spectrometry (HRMS). The metabolic characteristics of 1 and 2 in human liver microsomes indicated hydration and hydroxylation mass changes introduced to the parent drugs.


Subject(s)
Anti-Bacterial Agents/metabolism , Biological Products/metabolism , Metabolome/genetics , Pyrazines/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Chromatography, High Pressure Liquid , Circular Dichroism , Crystallography, X-Ray , Fungi/drug effects , Humans , Indoles/chemical synthesis , Indoles/chemistry , Indoles/metabolism , Mass Spectrometry , Molecular Structure , Pyrazines/chemical synthesis , Pyrazines/chemistry , Quinazolines/chemical synthesis , Quinazolines/chemistry , Quinazolines/metabolism , Stereoisomerism
13.
J Biol Chem ; 294(27): 10428-10437, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31118272

ABSTRACT

RET is a transmembrane growth factor receptor. Aberrantly activated RET is found in several types of human cancer and is a target for treating RET aberration-associated cancer. Multiple clinically relevant RET protein-tyrosine kinase inhibitors (TKIs) have been identified, but how TKIs bind to RET is unknown except for vandetanib. Nintedanib is a RET TKI that inhibits the vandetanib-resistant RET(G810A) mutant. Here, we determined the X-ray co-crystal structure of RET kinase domain-nintedanib complex to 1.87 Å resolution and a RET(G810A) kinase domain crystal structure to 1.99 Å resolution. We also identified a vandetanib-resistant RET(L881V) mutation previously found in familial medullary thyroid carcinoma. Drug-sensitivity profiling of RET(L881V) revealed that it remains sensitive to nintedanib. The RET-nintedanib co-crystal structure disclosed that Leu-730 in RET engages in hydrophobic interactions with the piperazine, anilino, and phenyl groups of nintedanib, providing a structural basis for explaining that the p.L730V mutation identified in nine independently isolated cell lines resistant to nintedanib. Comparisons of RET-nintedanib, RET(G810A), and RET-vandetanib crystal structures suggested that the solvent-front Ala-810 makes hydrophobic contacts with a methyl group and aniline in nintedanib and blocks water access to two oxygen atoms of vandetanib, resulting in an energetic penalty for burying polar groups. Of note, even though the p.L881V mutation did not affect sensitivity to nintedanib, RET(L881V) was resistant to nintedanib analogs lacking a phenyl group. These results provide structural insights into resistance of RET mutants against the TKIs nintedanib and vandetanib.


Subject(s)
Indoles/chemistry , Piperidines/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-ret/chemistry , Quinazolines/chemistry , Animals , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Drug Resistance, Neoplasm , Humans , Hydrophobic and Hydrophilic Interactions , Indoles/metabolism , Mice , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Piperidines/metabolism , Protein Kinase Inhibitors/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Quinazolines/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
14.
Cell Biol Int ; 44(9): 1890-1899, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32437063

ABSTRACT

Adjuvant chemotherapy in combination with surgery is expected to be a curative strategy for gastric cancer. However, drug resistance remains an obstacle in effective chemotherapy. Therefore, understanding the potential mechanisms of chemotherapy induced gastric cancer cell death is of great importance. We demonstrated that BIX-01294 (BIX) at low concentration could induce autophagic flux by converting LC3B-I to LC3B-II and directly activate autophagy associated cell death in gastric cancer cell lines at high concentration. BIX at low concentration could help obtain sensitivity of gastric cancer cells to chemotherapy with significantly reduced cell viability. Interestingly, BIX combined Cis (BIX + Cis) treated SGC-7901 cells display pyroptosis related cell death with large bubbles blown around the membrane, significantly decreased cell viability, elevated lactate dehydrogenase release and increased percentage of propidium iodide and Annexin-V double positive cells. Furthermore, the cleavage of gasdermin E (GSDME) and caspase-3 but not GSDMD was detected by immunoblotting and the knockout of GSDME switched pyroptosis into apoptosis in the BIX + Cis combined treated group. Furthermore, the deficiency of Beclin-1 to inhibit BIX induced autophagic flux completely blocked BIX + Cis combined treated induced cell pyroptosis related cell death. Additionally, BIX + Cis in vivo treatment could inhibit tumor growth, which could be reversed by the deficiency of Beclin-1 and be delayed by the deficiency of GSDME. In conclusion, our data was the first to reveal that BIX enhanced the anticancer chemotherapy effect by induced GSDME-mediated pyroptosis through the activation of autophagic flux in gastric cancer cells.


Subject(s)
Azepines/pharmacology , Pyroptosis/physiology , Quinazolines/pharmacology , Stomach Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Autophagy , Azepines/metabolism , Cell Death , Cell Line, Tumor , Cell Survival , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/metabolism , Pyroptosis/drug effects , Quinazolines/metabolism , Stomach Neoplasms/metabolism
15.
Bioorg Med Chem Lett ; 30(2): 126719, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31784319

ABSTRACT

Cytochrome P450 isozyme 1A2 (CYP1A2) is one main xenobiotic metabolizing enzyme in humans. It has been associated with the bioactivation of procarcinogens, including 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a tobacco specific and potent pulmonary carcinogen. This work describes the computational design and in-silico screening of potential CYP1A2 inhibitors, their chemical synthesis, and enzymatic characterization with the ultimate aim of assessing their potential as cancer chemopreventive agents. To achieve this, a combined classifiers model was used to screen a library of quinazoline-based molecules against known CYP1A2 inhibitors, non-inhibitors, and substrates to predict which quinazoline candidates had a better probability as an inhibitor. Compounds with high probability of CYP1A2 inhibition were further computationally evaluated via Glide docking. Candidates predicted to have selectivity and high binding affinity for CYP1A2 were synthesized and assayed for their enzymatic inhibition of CYP1A2, leading to the discovery of novel and potent quinazoline-based CYP1A2 inhibitors.


Subject(s)
Cytochrome P-450 CYP1A2/chemistry , Drug Design , Quinazolines/chemistry , Binding Sites , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1A2 Inhibitors/chemical synthesis , Cytochrome P-450 CYP1A2 Inhibitors/metabolism , Humans , Molecular Docking Simulation , Protein Structure, Tertiary , Quinazolines/metabolism , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 30(11): 127159, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32247733

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO) are promising drug development targets due to their implications in pathologies such as cancer and neurodegenerative diseases. The search for IDO1 inhibitor has been intensely pursued but there is a paucity of potent TDO and IDO1/TDO dual inhibitors. Natural product tryptanthrin has been confirmed to bear IDO1 and/or TDO inhibitory activities. Herein, twelve novel tryptanthrin derivatives were synthesized and evaluated for the IDO1 and TDO inhibitory potency. All of the compounds were found to be IDO1/TDO dual inhibitors, in particular, compound 9a and 9b bore IDO1 inhibitory activity similar to that of INCB024360, and compound 5a and 9b had remarkable TDO inhibitory activity superior to that of the well-known TDO inhibitor LM10. This work enriches the collection of IDO1/TDO dual inhibitors and provides chemical molecules for potential development into drugs.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Quinazolines/chemistry , Tryptophan Oxygenase/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inhibitory Concentration 50 , Quinazolines/metabolism , Solubility , Structure-Activity Relationship , Tryptophan Oxygenase/metabolism
17.
Bioorg Med Chem Lett ; 30(23): 127602, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33038544

ABSTRACT

G-protein coupled receptor kinase 2 (GRK2), which is upregulated in the failing heart, appears to play a critical role in heart failure (HF) progression in part because enhanced GRK2 activity promotes dysfunction of ß-adrenergic signaling and myocyte death. An orally bioavailable GRK2 inhibitor could offer unique therapeutic outcomes that cannot be attained by current heart failure treatments that directly target GPCRs or angiotensin-converting enzyme. Herein, we describe the discovery of a potent, selective, and orally bioavailable GRK2 inhibitor, 8h, through high-throughput screening, hit-to-lead optimization, structure-based design, molecular modelling, synthesis, and biological evaluation. In the cellular target engagement assays, 8h enhances isoproterenol-mediated cyclic adenosine 3',5'-monophosphate (cAMP) production in HEK293 cells overexpressing GRK2. Compound 8h was further evaluated in a human stem cell-derived cardiomyocyte (HSC-CM) contractility assay and potentiated isoproterenol-induced beating rate in HSC-CMs.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Animals , Enzyme Assays , G-Protein-Coupled Receptor Kinase 2/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Molecular Docking Simulation , Molecular Structure , Myocytes, Cardiac/drug effects , Phthalazines/chemical synthesis , Phthalazines/pharmacokinetics , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/chemical synthesis , Quinazolines/metabolism , Quinazolines/pharmacokinetics , Structure-Activity Relationship
18.
Bioorg Med Chem Lett ; 30(4): 126811, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31787491

ABSTRACT

This Letter details our efforts to develop new M4 PAM scaffolds with improved pharmacological properties. This endeavor involved replacing the 3,4-dimethylpyridazine core with two novel cores: a 2,3-dimethyl-2H-indazole-5-carboxamide core or a 1-methyl-1H-benzo[d][1,2,3]triazole-6-carboxamide core. Due to shallow SAR, these cores were further evolved into two unique tricyclic cores: an 8,9-dimethyl-8H-pyrazolo[3,4-h]quinazoline core and an 1-methyl-1H-[1,2,3]triazolo[4,5-h]quinazoline core. Both tricyclic cores displayed low nanomolar potency against both human and rat M4.


Subject(s)
Pyridazines/chemistry , Quinazolines/chemistry , Receptor, Muscarinic M4/chemistry , Triazoles/chemistry , Allosteric Regulation , Animals , Drug Design , Half-Life , Humans , Inhibitory Concentration 50 , Pyridazines/metabolism , Pyridazines/pharmacokinetics , Quinazolines/metabolism , Quinazolines/pharmacokinetics , Rats , Receptor, Muscarinic M4/metabolism , Structure-Activity Relationship , Triazoles/metabolism , Triazoles/pharmacokinetics
19.
Inorg Chem ; 59(20): 14879-14890, 2020 Oct 19.
Article in English | MEDLINE | ID: mdl-33003697

ABSTRACT

Ispinesib is a potent inhibitor of kinesin spindle protein (KSP), which has been identified as a promising target for antimitotic anticancer drugs. Herein, we report the synthesis of half-sandwich complexes of Ru, Os, Rh, and Ir bearing the ispinesib-derived N,N-bidentate ligands (R)- and (S)-2-(1-amino-2-methylpropyl)-3-benzyl-7-chloroquinazolin-4(3H)-one and studies on their chemical and biological properties. Using the enantiomerically pure (R)- and (S)-forms of the ligand, depending on the organometallic moiety, either the SM,R or RM,S diastereomers, respectively, were observed in the molecular structures of the Ru- and Os(cym) (cym = η6-p-cymene) compounds, whereas the RM,R or SM,S diastereomers were found for the Rh- and Ir(Cp*) (Cp* = η5-pentamethylcyclopentadienyl) derivatives. However, density functional theory (DFT) calculations suggest that the energy difference between the diastereomers is very small, and therefore a mixture of both will be present in solution. The organometallics exhibited varying antiproliferative activity in a series of human cancer cell lines, with the complexes featuring the (R)-enantiomer of the ligand being more potent than the (S)-configured counterparts. Notably, the Rh and Ir complexes demonstrated high KSP inhibitory activity, even at 1 nM concentration, which was independent of the chirality of the ligand, whereas the Ru and especially the Os derivatives were much less active.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Coordination Complexes/pharmacology , Kinesins/antagonists & inhibitors , Quinazolines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antioxidants/chemical synthesis , Antioxidants/metabolism , Antioxidants/pharmacology , Benzamides/chemical synthesis , Benzamides/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/metabolism , Drug Screening Assays, Antitumor , Humans , Kinesins/metabolism , Ligands , Metals, Heavy/chemistry , Molecular Docking Simulation , Protein Binding , Quinazolines/chemical synthesis , Quinazolines/metabolism , Stereoisomerism
20.
Bioorg Med Chem ; 28(21): 115720, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33065445

ABSTRACT

An organic small-molecular drug, 4-(1H-indol-3-yl)-2-(p-tolyl)quinazoline-3-oxide 1a was synthesized. It was employed to investigate the binding interaction and mechanism with human serum albumin (HSA). The experimental results indicated that the fluorescence quenching of HSA by 1a is a static quenching process and formation 1a-HSA complex. The site competition experiments revealed that the combination of 1a on HSA are hydrophobic interactions in the IIA domain and hydrogen bonds in IIIA domain of HSA, and the hydrophobic interactions of 1a on HSA are stronger than that of hydrogen bonds. These results were also confirmed by molecular docking theoretic analysis and ANS-hydrophobic fluorescent probe experiment. Synchronous fluorescence experiments showed that the polarity of HSA microenvironment was increase in the interaction process of 1a with HSA. The results of binding distance explored indicated that the combination distance between 1a and HSA is 3.63 nm, which is between 0.5R0 and 1.5R0, revealing the energy transfer between HSA and 1a is non-radiative. These results are very helpful for people to screen out high efficient indoloquinazoline drugs.


Subject(s)
Quinazolines/chemistry , Serum Albumin, Human/metabolism , Binding Sites , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation , Protein Binding , Quinazolines/metabolism , Serum Albumin, Human/chemistry , Temperature , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL