Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
1.
J Med Chem ; 66(14): 9561-9576, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37199108

ABSTRACT

Tau accumulation is one of the predominant neuropathological biomarkers for in vivo diagnosis of Alzheimer's disease due to its high correlation with disease progression. In this study, we focused on the structure-activity relationship study of the substituent effect on the aza-fused tricyclic core imidazo[1,2-h][1,7]naphthyridine to screen 18F-labeled Tau tracers. Through a series of autoradiographic studies and biological evaluations, 4-[18F]fluorophenyl-substituted tracer [18F]13 ([18F]FPND-4) was identified as a promising candidate with high affinity to native Tau tangles (IC50 = 2.80 nM), few appreciable binding to Aß plaques and MAO-A/B. Validated by dynamic positron emission tomography (PET) imaging in rodents and rhesus monkey, [18F]13 displayed desirable brain uptake (SUV = 1.75 at 2 min), fast clearance (brain2min/60min = 5.9), minimal defluorination, and few off-target binding, which met the requirements of a Tau-specific PET radiotracer.


Subject(s)
Alzheimer Disease , Neurofibrillary Tangles , Humans , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Radiopharmaceuticals , Positron-Emission Tomography/methods , Alzheimer Disease/metabolism , Brain/metabolism , Monoamine Oxidase/metabolism , Naphthyridines/metabolism , tau Proteins/metabolism
2.
Chem Biodivers ; 20(6): e202300410, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37088929

ABSTRACT

Two new naphthyridine compounds, 4-methoxycarbonyl-5-oxo-1,6-naphthyridine (1) and 5-methoxycarbonyl-4-oxo-1,6-naphthyridine (2) were obtained from the MeOH extracts of sponge Aaptos suberitoides. Their structures were determined by spectroscopic methods, including HR-ESI-MS, 1D-NMR (1 H-NMR, 13 C-NMR), 2D-NMR (COSY, HSQC, HMBC). The structure of compound 1 was further confirmed via single crystal X-ray diffraction analysis. Compound 1 was found to reduce NO production in LPS-induced RAW 264.7 macrophages with IC50 value of 0.15 mM. In addition, it decreased the mRNA expression levels of pro-inflammatory mediators, such as the tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1ß (IL-1ß), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX2) in LPS-induced macrophages. It also decreased the protein expression of iNOS and COX-2 in LPS-induced macrophages. Mechanistic studies further revealed that compound 1 inhibited the mitogen-activated protein kinase (MAPK), and activated the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) signaling pathways in LPS-induced RAW 264.7 macrophages.


Subject(s)
Lipopolysaccharides , Mitogen-Activated Protein Kinases , Animals , Mice , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/pharmacology , Lipopolysaccharides/pharmacology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , RAW 264.7 Cells , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , Signal Transduction , Macrophages , Naphthyridines/pharmacology , Naphthyridines/metabolism , Nitric Oxide Synthase Type II/metabolism , Heme Oxygenase-1/metabolism , Nitric Oxide/metabolism
3.
Toxicol In Vitro ; 82: 105374, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35537566

ABSTRACT

Drug-induced liver injury (DILI) is an adverse hepatic reaction and a serious concern for public healthcare systems and pharmaceutical companies. DILI is frequently caused by a combination of direct toxic stresses and subsequent immune damage to hepatocytes. However, little is known about the mechanism by which drugs facilitate the activation of the innate immune system. Here, we aimed to decipher the inflammatory events in trovafloxacin (TVX)-induced reactions using liver macrophages. We showed that proinflammatory M1-like macrophages mainly contributed to hepatotoxicity mediated by TVX, a DILI drug. Additionally, transcriptome results showed that the interferon type I pathway, cytokines, and apoptosis pathway were involved in the initiation of synergistic effects resulting in TVX-induced liver injury. We hypothesized that DILI drugs could drive liver injury by altering the activation and phenotype of hepatic macrophages. Furthermore, drug treatment-induced transcriptional changes such as Traf1 and 2, Socs3, and Hbegf in macrophage polarization could be used to assess drug-specific immune-mediated reactions. Therefore, we proposed that transcriptional change in the genes related to macrophage polarization index could be an indicator to reflect the severity of DILI in a preclinical setting during drug development.


Subject(s)
Chemical and Drug Induced Liver Injury , Drug-Related Side Effects and Adverse Reactions , Chemical and Drug Induced Liver Injury/metabolism , Fluoroquinolones , Humans , Inflammation/chemically induced , Inflammation/metabolism , Liver/metabolism , Macrophages , Naphthyridines/metabolism , Naphthyridines/toxicity
4.
Nat Commun ; 12(1): 7085, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34873166

ABSTRACT

Antibiotic resistance is becoming one of the major crises, among which hydrolysis reaction is widely employed by bacteria to destroy the reactive pharmacophore. Correspondingly, antibiotic producer has canonically co-evolved this approach with the biosynthetic capability for self-resistance. Here we discover a self-defense strategy featuring with reductive inactivation of hemiaminal pharmacophore by short-chain dehydrogenases/reductases (SDRs) NapW and homW, which are integrated with the naphthyridinomycin biosynthetic pathway. We determine the crystal structure of NapW·NADPH complex and propose a catalytic mechanism by molecular dynamics simulation analysis. Additionally, a similar detoxification strategy is identified in the biosynthesis of saframycin A, another member of tetrahydroisoquinoline (THIQ) antibiotics. Remarkably, similar SDRs are widely spread in bacteria and able to inactive other THIQ members including the clinical anticancer drug, ET-743. These findings not only fill in the missing intracellular events of temporal-spatial shielding mode for cryptic self-resistance during THIQs biosynthesis, but also exhibit a sophisticated damage-control in secondary metabolism and general immunity toward this family of antibiotics.


Subject(s)
Bacteria/metabolism , Bacterial Proteins/metabolism , Biosynthetic Pathways , Molecular Dynamics Simulation , Tetrahydroisoquinolines/metabolism , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Bacteria/genetics , Bacterial Proteins/genetics , Biocatalysis , Chromatography, High Pressure Liquid , Drug Resistance, Microbial/genetics , Humans , Isoquinolines/chemistry , Isoquinolines/metabolism , Mass Spectrometry/methods , Molecular Structure , NADP/chemistry , NADP/metabolism , Naphthyridines/chemistry , Naphthyridines/metabolism , Oxidation-Reduction , Oxidoreductases/genetics , Oxidoreductases/metabolism , Tetrahydroisoquinolines/chemistry
5.
Bioorg Med Chem Lett ; 49: 128314, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34391891

ABSTRACT

A series of IDO1 inhibitors containing a decahydroquinoline, decahydro-1,6-naphthyridine, or octahydro-1H-pyrrolo[3,2-c]pyridine scaffold were identified with good cellular and human whole blood activity against IDO1. These inhibitors contain multiple chiral centers and all diastereomers were separated. The absolute stereochemistry of each isomers were not determined. Compounds 15 and 27 stood out as leads due to their good cellular as well as human whole blood IDO1 inhibition activity, low unbound clearance, and reasonable mean residence time in rat cassette PK studies.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Naphthyridines/pharmacology , Pyrroles/pharmacology , Quinolines/pharmacology , Animals , Catalytic Domain , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , HeLa Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Molecular Docking Simulation , Naphthyridines/chemical synthesis , Naphthyridines/metabolism , Naphthyridines/pharmacokinetics , Pyrroles/chemical synthesis , Pyrroles/metabolism , Pyrroles/pharmacokinetics , Quinolines/chemical synthesis , Quinolines/metabolism , Quinolines/pharmacokinetics , Rats , Stereoisomerism , Structure-Activity Relationship
6.
J Med Chem ; 64(8): 5082-5098, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33834781

ABSTRACT

Multifunctional entities have recently been attractive for the development of anticancer chemotherapeutic drugs. However, such entities with concurrent CK2 along with cancer stem cell (CSC) inhibitory activities are rare in a single small molecule. Herein, a series of 5-(3-chlorophenylamino)benzo[c][2,6]naphthyridine derivatives were synthesized using a known CK2 inhibitor, silmitasertib (CX-4945), as the lead compound. Among the resulting compounds, 1c exhibited stronger CK2 inhibitory activity with higher Clk2/CK2 selectivity than CX-4945. Significantly, 1c could modulate the Akt1(ser129)-GSK-3ß(ser9)-Wnt/ß-catenin signaling pathway and inhibit the expression of the stemness marker ALDH1A1, CSC surface antigens, and stem genes, showing potent CSC inhibitory activity. Moreover, 1c also displayed superior pharmacokinetics and antitumor activity compared with CX-4945 sodium salt, without obvious toxicity. The favorable antiproliferative and antitumor activity of 1c, its high inhibitory selectivity for CK2, and its potent inhibition of cancer cell stemness make this molecule a candidate for the treatment of cancer.


Subject(s)
Antineoplastic Agents/chemistry , Casein Kinase II/antagonists & inhibitors , Naphthyridines/chemistry , Protein Kinase Inhibitors/chemistry , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Binding Sites , Casein Kinase II/metabolism , Cell Cycle Checkpoints/drug effects , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Nude , Molecular Docking Simulation , Naphthyridines/metabolism , Naphthyridines/pharmacology , Naphthyridines/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/chemistry , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Wnt Signaling Pathway/drug effects
7.
Eur J Med Chem ; 216: 113270, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33765486

ABSTRACT

ß-Site amyloid precursor protein cleaving enzyme 1 (BACE1) has been pursued as a prime target for the treatment of Alzheimer's disease (AD). In this report, we describe the discovery of BACE1 inhibitors with a 1-amino-3,4-dihydro-2,6-naphthyridine scaffold. Leveraging known inhibitors 2a and 2b, we designed the naphthyridine-based compounds by removing a structurally labile moiety and incorporating pyridine rings, which showed increased biochemical and cellular potency, along with reduced basicity on the amidine moiety. Introduction of a fluorine atom on the pyridine culminated in compound 11 which had improved cellular activity as well as further reduced basicity and demonstrated a robust and sustained cerebrospinal fluid (CSF) Aß reduction in dog. The crystal structure of compound 11 bound to BACE1 confirmed van der Waals interactions between the fluorine on the pyridine and Tyr71 in the flap.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Naphthyridines/chemistry , Protease Inhibitors/chemistry , Pyridines/chemistry , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Aspartic Acid Endopeptidases/metabolism , Binding Sites , Crystallography, X-Ray , Dogs , Drug Evaluation, Preclinical , Half-Life , Humans , Microsomes/metabolism , Molecular Dynamics Simulation , Naphthyridines/metabolism , Naphthyridines/pharmacology , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Rats , Static Electricity , Structure-Activity Relationship
8.
Eur J Med Chem ; 215: 113273, 2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33601310

ABSTRACT

In this study, a series of pyrrolo [2,3-d]pyrimidine derivatives containing 1,8-naphthyridine-4-one fragment were synthesized and their biological activity were tested. Most of the target compounds displayed moderate to excellent activity against one or more cancer cell lines and low activity against human normal cell LO2 in vitro. The most promising compound 51, of which the IC50 values were 0.66 µM, 0.38 µM and 0.44 µM against cell lines A549, Hela and MCF-7, shown more remarkable activity and better apoptosis effect than the positive control Cabozantinib. The structure-activity relationships (SARs) indicated that double-EWGs (such as R3 = 2-Cl-4-CF3) on the terminal phenyl rings was a key factor in improving the biological activity. In addition, the further research on compound 51 mainly included c-Met kinase activity and selectivity, concentration dependence, and molecular docking.


Subject(s)
Antineoplastic Agents/pharmacology , Naphthyridines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Anilides/metabolism , Anilides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Naphthyridines/chemical synthesis , Naphthyridines/metabolism , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Pyridines/metabolism , Pyridines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Pyrroles/chemical synthesis , Pyrroles/metabolism , Structure-Activity Relationship
9.
Int J Biol Macromol ; 174: 1-10, 2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33476619

ABSTRACT

A series of 11-substituted sampangine derivatives have been designed, synthesized, and tested for their ability to inhibit cholinesterase. Their chelating ability and selectivity for Cu2+ over other biologically relevant metal ions were demonstrated by isothermal titration calorimetry. Their blood-brain barrier permeability was also tested by parallel artificial membrane permeation assay. Among the synthesized derivatives, compound 11 with the strong anti-acetylcholinesterase activity, high blood-brain barrier penetration ability and high binding affinity to Cu2+ was selected for further research. Western blotting analysis, transmission electron microscopy, DCFH-DA assay and paralysis experiment indicated that compound 11 suppressed the formation of Cu2+-Aß complexes, alleviated the Cu2+ induced neurotoxicity and inhibited the production of ROS catalyzed by Cu2+ in Aß42 transgenic C. elegans. Moreover, compound 11 also inhibited the expressions of proinflammatory cytokines, such as NO, TNF-α, IL-6 and IL-1ß, induced by Cu2+ + Aß1-42 in BV2 microglial cells. In general, this work provided new insights into the design and development of potent metal-chelating agents for AD treatment.


Subject(s)
Alkaloids/chemistry , Alkaloids/metabolism , Cholinesterase Inhibitors/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/metabolism , Naphthyridines/chemistry , Naphthyridines/metabolism , Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Animals , Animals, Genetically Modified , Blood-Brain Barrier/metabolism , Caenorhabditis elegans/metabolism , Calorimetry/methods , Cell Line , Chelating Agents/chemistry , Cholinesterase Inhibitors/chemical synthesis , Cholinesterases/metabolism , Copper/chemistry , Cytokines , Inflammation , Microglia , Oxidative Stress/drug effects , Peptide Fragments/metabolism , Protein Aggregates , Reactive Oxygen Species/metabolism
10.
Med Chem ; 17(4): 299-309, 2021.
Article in English | MEDLINE | ID: mdl-31660840

ABSTRACT

New chemical agents that could combat increasing antibiotic resistance are urgently needed. In this mini-review, an old but highly relevant RNA sequence which is crucial for the continuation of bacterial life-cycle is covered. Some of the most significant advances of the last decade in sensing and targeting the bacterial rRNA A-site: a well-validated binding site of proverbially known aminoglycoside antibiotics are described. Some of the major advances in direct sensing of the bacterial decoding side (A-site) are described and also new fluorescent molecules that are capable of detecting lead compounds through high-throughput assays by displacement of fluorescent probe molecules are highlighted. Lastly, some of the recently discovered non-aminoglycoside small molecule binders of bacterial rRNA A-site as a new class of molecules that could provide future scaffolds and molecules for developing new antibacterial agents have been discussed.


Subject(s)
Anti-Bacterial Agents/metabolism , RNA, Bacterial/metabolism , RNA, Ribosomal/metabolism , Aminoglycosides/chemical synthesis , Aminoglycosides/metabolism , Anti-Bacterial Agents/chemical synthesis , Bacteria/chemistry , Bacteria/drug effects , Binding Sites , Fluorescent Dyes/chemistry , Naphthyridines/metabolism , Peptide Nucleic Acids/metabolism , Spiro Compounds/metabolism
11.
J Med Chem ; 63(21): 12786-12798, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33119282

ABSTRACT

CK2α is a ubiquitous, well-studied kinase that is a target for small-molecule inhibition, for treatment of cancers. While many different classes of adenosine 5'-triphosphate (ATP)-competitive inhibitors have been described for CK2α, they tend to suffer from significant off-target activity and new approaches are needed. A series of inhibitors of CK2α has recently been described as allosteric, acting at a previously unidentified binding site. Given the similarity of these inhibitors to known ATP-competitive inhibitors, we have investigated them further. In our thorough structural and biophysical analyses, we have found no evidence that these inhibitors bind to the proposed allosteric site. Rather, we report crystal structures, competitive isothermal titration calorimetry (ITC) and NMR, hydrogen-deuterium exchange (HDX) mass spectrometry, and chemoinformatic analyses that all point to these compounds binding in the ATP pocket. Comparisons of our results and experimental approach with the data presented in the original report suggest that the primary reason for the disparity is nonspecific inhibition by aggregation.


Subject(s)
Protein Kinase Inhibitors/chemistry , Adenosine Triphosphate/metabolism , Allosteric Regulation , Allosteric Site , Binding, Competitive , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Casein Kinase II/metabolism , Crystallography, X-Ray , Deuterium Exchange Measurement , Humans , Ligands , Molecular Dynamics Simulation , Naphthyridines/chemistry , Naphthyridines/metabolism , Nuclear Magnetic Resonance, Biomolecular , Phenazines , Protein Binding , Protein Kinase Inhibitors/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
12.
Bioorg Med Chem ; 28(24): 115819, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33120078

ABSTRACT

The exploitation of GLU988 and LYS903 residues in PARP1 as targets to design isoquinolinone (I & II) and naphthyridinone (III) analogues is described. Compounds of structure I have good biochemical and cellular potency but suffered from inferior PK. Constraining the linear propylene linker of structure I into a cyclopentene ring (II) offered improved PK parameters, while maintaining potency for PARP1. Finally, to avoid potential issues that may arise from the presence of an anilinic moiety, the nitrogen substituent on the isoquinolinone ring was incorporated as part of the bicyclic ring. This afforded a naphthyridinone scaffold, as shown in structure III. Further optimization of naphthyridinone series led to identification of a novel and highly potent PARP1 inhibitor 34, which was further characterized as preclinical candidate molecule. Compound 34 is orally bioavailable and displayed favorable pharmacokinetic (PK) properties. Compound 34 demonstrated remarkable antitumor efficacy both as a single-agent as well as in combination with chemotherapeutic agents in the BRCA1 mutant MDA-MB-436 breast cancer xenograft model. Additionally, compound 34 also potentiated the effect of agents such as temozolomide in breast cancer, pancreatic cancer and Ewing's sarcoma models.


Subject(s)
Antineoplastic Agents/chemistry , Naphthyridines/chemistry , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Quinolones/chemistry , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Half-Life , Humans , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Naphthyridines/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Quinolones/metabolism , Structure-Activity Relationship , Transplantation, Heterologous
13.
Nat Commun ; 11(1): 4659, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32938936

ABSTRACT

The αvß6 integrin plays a key role in the activation of transforming growth factor-ß (TGFß), a pro-fibrotic mediator that is pivotal to the development of idiopathic pulmonary fibrosis (IPF). We identified a selective small molecule αvß6 RGD-mimetic, GSK3008348, and profiled it in a range of disease relevant pre-clinical systems. To understand the relationship between target engagement and inhibition of fibrosis, we measured pharmacodynamic and disease-related end points. Here, we report, GSK3008348 binds to αvß6 with high affinity in human IPF lung and reduces downstream pro-fibrotic TGFß signaling to normal levels. In human lung epithelial cells, GSK3008348 induces rapid internalization and lysosomal degradation of the αvß6 integrin. In the murine bleomycin-induced lung fibrosis model, GSK3008348 engages αvß6, induces prolonged inhibition of TGFß signaling and reduces lung collagen deposition and serum C3M, a marker of IPF disease progression. These studies highlight the potential of inhaled GSK3008348 as an anti-fibrotic therapy.


Subject(s)
Butyrates/pharmacology , Idiopathic Pulmonary Fibrosis/drug therapy , Integrins/antagonists & inhibitors , Naphthyridines/pharmacology , Pyrazoles/pharmacology , Pyrrolidines/pharmacology , Administration, Inhalation , Animals , Antigens, Neoplasm/metabolism , Bleomycin/toxicity , Butyrates/administration & dosage , Butyrates/metabolism , Butyrates/pharmacokinetics , Collagen/metabolism , Disease Models, Animal , Epithelial Cells/drug effects , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/pathology , Integrins/metabolism , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Naphthyridines/administration & dosage , Naphthyridines/metabolism , Naphthyridines/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Pyrrolidines/administration & dosage , Pyrrolidines/metabolism , Pyrrolidines/pharmacokinetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Tomography, Emission-Computed, Single-Photon , Transforming Growth Factor beta/metabolism , Translational Research, Biomedical
14.
Eur J Med Chem ; 208: 112719, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32865176

ABSTRACT

Integrins αvß5 and αvß3 are closely related, proangiogenic members of the wider RGD-binding integrin family. Due to their high sequence homology, the development of αvß5-selective compounds has remained elusive to synthetic and medicinal chemists. Herein, we describe a survey of SAR around a series of amide-containing 3-aryl-succinamic acid-based RGD mimetics. This resulted in the discovery of α,α,α-trifluorotolyl 12 which exhibits 800 × selectivity for αvß5versus αvß3 with a pyrrolidine amide linker that confers selectivity for αvß5 by positioning a key aryl ring in the SDL of αvß5 with good complementarity; binding in this mode is disfavoured in αvß3 due to clashes with key residues in the ß3-subunit. Compound 12 exhibits selective inhibition by a cell adhesion assay, high passive permeability and solubility which enables potential use of this inhibitor as an αvß5-selective in vitro tool compound.


Subject(s)
Amides/pharmacology , Pyrrolidines/pharmacology , Receptors, Vitronectin/antagonists & inhibitors , Amides/chemical synthesis , Amides/metabolism , Cell Adhesion/drug effects , Humans , K562 Cells , Molecular Docking Simulation , Molecular Structure , Naphthyridines/chemical synthesis , Naphthyridines/metabolism , Naphthyridines/pharmacology , Protein Binding , Pyrrolidines/chemical synthesis , Pyrrolidines/metabolism , Receptors, Vitronectin/metabolism , Stereoisomerism , Structure-Activity Relationship
15.
Eur J Med Chem ; 208: 112785, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32898795

ABSTRACT

As a privileged scaffold, the quinazoline ring is widely used in the development of EGFR inhibitors, while few quinazoline-based MET inhibitors are reported. In our ongoing efforts to develop new MET-targeted anticancer drug candidates, a series of quinazoline-based 1,6-naphthyridinone derivatives were designed, synthesized, and evaluated for their biological activities. The preliminary SARs studies indicate that the quinazoline scaffold was also acceptable for the block A of class II MET inhibitors. The further pharmacokinetic studies led to the identification of the most promising compound 22a with favorable in vitro potency (MET, IC50 = 9.0 nM), human microsomal metabolic stability (t1/2 = 621.2 min) and oral bioavailability (F = 42%). Moreover, 22a displayed good in vivo antitumor efficacy (IR of 81% in 75 mg/kg) in MET-positive human glioblastoma U-87 MG xenograft model. These positive results indicated that 22a is a potential new MET-targeted antitumor drug lead, which is worthy of further development.


Subject(s)
Antineoplastic Agents/therapeutic use , Glioblastoma/drug therapy , Naphthyridines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinazolines/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Female , Humans , Mice, Nude , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Structure , Naphthyridines/chemical synthesis , Naphthyridines/metabolism , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins c-met/metabolism , Quinazolines/chemical synthesis , Quinazolines/metabolism , Rats , Structure-Activity Relationship , Thermodynamics , Xenograft Model Antitumor Assays
16.
Bioorg Med Chem Lett ; 30(23): 127517, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32911078

ABSTRACT

A series of Torin2, a second-generation ATP-competitive inhibitor, analogues were biologically characterized to identify their potential for ATR and mTOR kinase inhibition. Compound SPK 98 was observed to inhibit ATR/mTOR kinase selectively over ATM kinase in HCT 116 cell line. In addition to that, SPK 98 on 30 min incubation with human, mice and rat liver microsomes showed improved properties with an increased half-life (a maximum T ½ of 157 min) and internal clearance in mouse as compared to Torin2. Further, SPK 98 was also noticed to indulge in inducing premature chromatin condensation as a result of ATR/mTOR kinase inhibition at 50 nM. In a nutshell, our work presents the identification and characterization of SPK 98, a small molecule inhibitor, which exhibits improved specific inhibition for ATR at a lower concentration than Torin2.


Subject(s)
Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Naphthyridines/pharmacology , Photosensitizing Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/metabolism , Chromatin/metabolism , DNA/radiation effects , DNA Damage/radiation effects , HCT116 Cells , Humans , Mice , Microsomes, Liver/metabolism , Naphthyridines/metabolism , Photosensitizing Agents/metabolism , Protein Kinase Inhibitors/metabolism , Protein Stability , Rats , Signal Transduction/drug effects , Ultraviolet Rays
17.
J Med Chem ; 63(17): 9045-9069, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32691589

ABSTRACT

Pan-bromodomain and extra terminal (BET) inhibitors interact equipotently with all eight bromodomains of the BET family of proteins. They have shown profound efficacy in vitro and in vivo in oncology and immunomodulatory models, and a number of them are currently in clinical trials where significant safety signals have been reported. It is therefore important to understand the functional contribution of each bromodomain to assess the opportunity to tease apart efficacy and toxicity. This article discloses the in vitro and cellular activity profiles of GSK789, a potent, cell-permeable, and highly selective inhibitor of the first bromodomains of the BET family.


Subject(s)
Naphthyridines/chemistry , Transcription Factors/antagonists & inhibitors , ATPases Associated with Diverse Cellular Activities/antagonists & inhibitors , ATPases Associated with Diverse Cellular Activities/metabolism , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Binding Sites , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Half-Life , Humans , Molecular Dynamics Simulation , Naphthyridines/metabolism , Naphthyridines/pharmacology , Protein Domains , Quinolones/chemistry , Quinolones/metabolism , Quinolones/pharmacology , Transcription Factors/metabolism
18.
Bioorg Med Chem ; 28(12): 115555, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32503697

ABSTRACT

New N-substituted-3-phenyl-1,6-naphthyridinone derivatives are designed and synthesized, based on structural modification of our previously reported compound 3. Extensive enzyme-based SAR studies and PK evaluation led to the discovery of compound 4r, with comparable c-Met potency to that of Cabozantinib and high VEGFR-2 selectivity, while Cabozantinib displayed no VEGFR-2 selectivity. More importantly, at oral doses of 45 mg/kg (Q.D.), compound 4r exhibits significant tumor growth inhibition (93%) in a U-87MG human gliobastoma xenograft model. The promising selectivity against VEGFR-2 and excellent tumor growth inhibition of compound 4r suggest that it could be used as a new lead molecule for further discovery of selective type II c-Met inhibitors.


Subject(s)
Drug Design , Naphthyridines/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinolines/chemistry , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Molecular Docking Simulation , Naphthyridines/metabolism , Naphthyridines/pharmacology , Naphthyridines/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/metabolism , Structure-Activity Relationship , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Alcohol Clin Exp Res ; 44(2): 354-367, 2020 02.
Article in English | MEDLINE | ID: mdl-31840823

ABSTRACT

Alcohol use disorder (AUD) is a chronic, relapsing disorder that is characterized by the compulsive use of alcohol despite numerous health, social, and economic consequences. Initially, the use of alcohol is driven by positive reinforcement. Over time, however, alcohol use can take on a compulsive quality that is driven by the desire to avoid the negative consequences of abstinence, including negative affect and heightened stress/anxiety. This transition from positive reinforcement- to negative reinforcement-driven consumption involves the corticotropin-releasing factor (CRF) system, although mounting evidence now suggests that the CRF system interacts with other neural systems to ultimately produce behaviors that are symptomatic of compulsive alcohol use, such as the hypocretin (Hcrt) system. Hypocretins are produced exclusively in the hypothalamus, but Hcrt neurons project widely throughout the brain and reach regions that perform regulatory functions for numerous behavioral and physiological responses-including the infralimbic cortex (IL) of the medial prefrontal cortex (mPFC). Although the entire mPFC undergoes neuroadaptive changes following prolonged alcohol exposure, the IL appears to undergo more robust changes compared with other mPFC substructures. Evidence to date suggests that the IL is likely involved in EtOH-seeking behavior, but ambiguities with respect to the specific role of the IL in this regard make it difficult to draw definitive conclusions. Furthermore, the manner in which CRF interacts with Hcrt in this region as it pertains to alcohol-seeking behavior is largely unknown, although immunohistochemical and electrophysiological experiments have shown that CRF and Hcrt directly interact in the mPFC, suggesting that the interaction between CRF and Hcrt in the IL may be critically important for the development and subsequent maintenance of compulsive alcohol seeking. This review aims to consolidate recent literature regarding the role of the IL in alcohol-seeking behavior and to discuss evidence that supports a functional interaction between Hcrt and CRF in the IL.


Subject(s)
Alcoholism/metabolism , Compulsive Behavior/metabolism , Corticotropin-Releasing Hormone/metabolism , Drug-Seeking Behavior/physiology , Orexins/metabolism , Prefrontal Cortex/metabolism , Alcoholism/drug therapy , Animals , Benzoxazoles/metabolism , Benzoxazoles/pharmacology , Benzoxazoles/therapeutic use , Compulsive Behavior/drug therapy , Drug-Seeking Behavior/drug effects , Humans , Naphthyridines/metabolism , Naphthyridines/pharmacology , Naphthyridines/therapeutic use , Prefrontal Cortex/drug effects , Protein Binding/physiology , Urea/analogs & derivatives , Urea/metabolism , Urea/pharmacology , Urea/therapeutic use
20.
J Biol Chem ; 294(37): 13545-13559, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31341017

ABSTRACT

The homeodomain-interacting protein kinase (HIPK) family is comprised of four nuclear protein kinases, HIPK1-4. HIPK proteins phosphorylate a diverse range of transcription factors involved in cell proliferation, differentiation, and apoptosis. HIPK2, thus far the best-characterized member of this largely understudied family of protein kinases, plays a role in the activation of p53 in response to DNA damage. Despite this tumor-suppressor function, HIPK2 is also found overexpressed in several cancers, and its hyperactivation causes chronic fibrosis. There are currently no structures of HIPK2 or of any other HIPK kinase. Here, we report the crystal structure of HIPK2's kinase domain bound to CX-4945, a casein kinase 2α (CK2α) inhibitor currently in clinical trials against several cancers. The structure, determined at 2.2 Å resolution, revealed that CX-4945 engages the HIPK2 active site in a hybrid binding mode between that seen in structures of CK2α and Pim1 kinases. The HIPK2 kinase domain crystallized in the active conformation, which was stabilized by phosphorylation of the activation loop. We noted that the overall kinase domain fold of HIPK2 closely resembles that of evolutionarily related dual-specificity tyrosine-regulated kinases (DYRKs). Most significant structural differences between HIPK2 and DYRKs included an absence of the regulatory N-terminal domain and a unique conformation of the CMGC-insert region and of a newly defined insert segment in the αC-ß4 loop. This first crystal structure of HIPK2 paves the way for characterizing the understudied members of the HIPK family and for developing HIPK2-directed therapies for managing cancer and fibrosis.


Subject(s)
Carrier Proteins/chemistry , Protein Serine-Threonine Kinases/chemistry , Amino Acid Sequence , Binding Sites , Carrier Proteins/classification , Carrier Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Evolution, Molecular , Humans , Molecular Dynamics Simulation , Naphthyridines/chemistry , Naphthyridines/metabolism , Phenazines , Phylogeny , Protein Binding , Protein Serine-Threonine Kinases/classification , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL