Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(11): 9431-9446, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38818879

ABSTRACT

Synthetic lethality has recently emerged as a new approach for the treatment of mutated genes that were previously considered undruggable. Targeting methionine adenosyltransferase 2A (MAT2A) in cancers with deletion of the methylthioadenosine phosphorylase (MTAP) gene leads to synthetic lethality and thus has attracted significant interest in the field of precise anticancer drug development. Herein, we report the discovery of a series of novel MAT2A inhibitors featuring a pyrazolo[3,4-c]quinolin-4-one skeleton based on structure-based drug design. Further optimization led to compound 39, which has a high potency for inhibiting MAT2A and a remarkable selectivity for MTAP-deleted cancer cell lines. Compound 39 has a favorable pharmacokinetic profile with high plasma exposure and oral bioavailability, and it exhibits significant efficacy in xenograft MTAP-depleted models. Moreover, 39 demonstrates excellent brain exposure with a Kpuu of 0.64 in rats.


Subject(s)
Brain , Drug Design , Enzyme Inhibitors , Methionine Adenosyltransferase , Methionine Adenosyltransferase/antagonists & inhibitors , Methionine Adenosyltransferase/metabolism , Humans , Animals , Structure-Activity Relationship , Rats , Brain/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/chemical synthesis , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Mice , Male , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
2.
Eur J Med Chem ; 205: 112537, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32768738

ABSTRACT

The chemokine receptor CXCR4 has been proposed as a drug target based on its important functions in HIV infection, inflammation/autoimmune diseases and cancer metastasis. Herein we report the design, synthesis and evaluation of novel CXCR4 antagonists based on a pyrrolidine scaffold. The structural exploration/optimization identified numerous potent CXCR4 antagonists, represented by compound 46, which displayed potent binding affinity to CXCR4 receptor (IC50 = 79 nM competitively displacing fluorescent 12G5 antibody) and inhibited CXCL12 induced cytosolic calcium flux (IC50 = 0.25 nM). Moreover, in a transwell invasion assay, compound 46 significantly mitigated CXCL12/CXCR4 mediated cell migration. Compound 46 exhibited good physicochemical properties (MW 367, logD7.4 1.12, pKa 8.2) and excellent in vitro safety profiles (e.g., hERG patch clamp IC50 > 30 µM and minimal CYP isozyme inhibition). Importantly, 46 displayed much improved metabolic stability in human and rat liver microsomes. Lastly, 46 demonstrated marked efficacy in a cancer metastasis model in mice. These results strongly support 46 as a prototypical lead for the development of promising CXCR4 antagonists as clinical candidates.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Drug Design , Pyrrolidines/chemical synthesis , Pyrrolidines/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Calcium/metabolism , Cell Line, Tumor , Chemistry Techniques, Synthetic , Cytosol/drug effects , Cytosol/metabolism , Humans , Mice , Neoplasm Metastasis , Pyrrolidines/chemistry , Rats
3.
ChemMedChem ; 15(13): 1150-1162, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32391652

ABSTRACT

Chemokine receptor CXCR4 and its natural ligand CXCL12 (also known as stromal cell-derived factor-1, or SDF-1) regulate a broad range of physiological functions. Dysregulation of the CXCL12/CXCR4 axis is involved in numerous pathological conditions such as HIV infection, inflammation and cancer. Herein, we report the design, synthesis, and characterization of novel CXCR4 antagonists based on cyclic amine scaffolds. Compound 24 was identified as a potent CXCR4 receptor antagonist (competitive inhibition of 12G5 binding, IC50 =24 nM; functional inhibition of CXCL12-induced cytosolic calcium increase, IC50 =0.1 nM). In addition, compound 24 potently inhibited cell migration in CXCR4/CXCL12-mediated chemotaxis in a matrigel invasion assay. The absolute configuration of compound 24 was elucidated by X-ray crystallography.


Subject(s)
Amines/pharmacology , Drug Design , Receptors, CXCR4/antagonists & inhibitors , Amines/chemical synthesis , Amines/chemistry , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Mice , Molecular Structure , Receptors, CXCR4/metabolism , Structure-Activity Relationship
4.
Bioorg Chem ; 99: 103824, 2020 06.
Article in English | MEDLINE | ID: mdl-32334192

ABSTRACT

The chemokine receptor CXCR4 has been explored as a drug target due to its involvement in pathological conditions such as HIV infection and cancer metastasis. Here we report the structure-activity relationship study of novel CXCR4 antagonists based on an aminoquinoline template. This template is devoid of the chiral center in the classical tetrahydroquinoline (THQ) ring moiety and therefore can be easily synthesized. A number of potent CXCR4 antagonists were identified, exemplified by compound 3, which demonstrated excellent binding affinity with CXCR4 receptor (IC50 = 57 nM) and inhibited CXCL12 induced cytosolic calcium increase (IC50 = 0.24 nM). Furthermore, compound 3 potently inhibited CXLC12/CXCR4 mediated cell migration in a transwell invasion assay. The simplified synthetic approach combined with good physicochemical properties (e.g. MW 362, clogP 2.1, PSA 48, pKa 7.0 for compound 3) demonstrate the potential of this aminoquinoline template as a novel scaffold to develop CXCR4 antagonists.


Subject(s)
Aminoquinolines/pharmacology , Drug Design , Receptors, CXCR4/antagonists & inhibitors , Aminoquinolines/chemical synthesis , Aminoquinolines/chemistry , Cell Line , Dose-Response Relationship, Drug , Humans , Molecular Structure , Receptors, CXCR4/metabolism , Structure-Activity Relationship
5.
Eur J Med Chem ; 187: 111914, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31806538

ABSTRACT

Structural optimization of aminopyrimidine-based CXCR4 antagonists is reported. The optimization is guided by molecular docking studies based on available CXCR4-small molecule crystal complex. The optimization identifies a number of compounds with improved receptor binding affinity and functional activity exemplified by compound 23 (inhibition of APC-conjugate clone 12G5 for CXCR4 binding in a cell based assay: IC50 = 8.8 nM; inhibition of CXCL12 induced cytosolic calcium increase: IC50 = 0.02 nM). In addition, compound 23 potently inhibits CXCR4/CXLC12 mediated chemotaxis in a matrigel invasion assay. Furthermore, compound 23 exhibits good physicochemical properties (MW 367, clogP 2.1, PSA 48, pKa 7.2) and in vitro safety profiles (marginal/moderate inhibition of CYP isozymes and hERG). These results represent significant improvement over the initial hit from scaffold hybridization and suggest that compound 23 can be used as a starting point to support lead optimization.


Subject(s)
Pyrimidines/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Receptors, CXCR4/metabolism , Structure-Activity Relationship
6.
Cell Death Dis ; 10(7): 493, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31235688

ABSTRACT

Necroptosis is a form of regulated necrosis controlled by receptor-interacting kinase 1 (RIPK1 or RIP1), RIPK3 (RIP3), and pseudokinase mixed lineage kinase domain-like protein (MLKL). Increasing evidence suggests that necroptosis is closely associated with pathologies including inflammatory diseases, neurodegenerative diseases, and cancer metastasis. Herein, we discovered the small-molecule PK6 and its derivatives as a novel class of necroptosis inhibitors that directly block the kinase activity of RIPK1. Optimization of PK6 led to PK68, which has improved efficacy for the inhibition of RIPK1-dependent necroptosis, with an EC50 of around 14-22 nM in human and mouse cells. PK68 efficiently blocks cellular activation of RIPK1, RIPK3, and MLKL upon necroptosis stimuli. PK68 displays reasonable selectivity for inhibition of RIPK1 kinase activity and favorable pharmacokinetic properties. Importantly, PK68 provides strong protection against TNF-α-induced systemic inflammatory response syndrome in vivo. Moreover, pre-treatment of PK68 significantly represses metastasis of both melanoma cells and lung carcinoma cells in mice. Together, our study demonstrates that PK68 is a potent and selective inhibitor of RIPK1 and also highlights its great potential for use in the treatment of inflammatory disorders and cancer metastasis.


Subject(s)
Enzyme Inhibitors/therapeutic use , Necroptosis/drug effects , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Systemic Inflammatory Response Syndrome/drug therapy , Systemic Inflammatory Response Syndrome/metabolism , Tumor Necrosis Factor-alpha/toxicity , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , HT29 Cells , Humans , Male , Melanoma, Experimental , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Necrosis/metabolism , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Systemic Inflammatory Response Syndrome/chemically induced , U937 Cells
7.
Eur J Med Chem ; 172: 1-15, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-30939349

ABSTRACT

The Hedgehog (Hh) pathway plays a critical role during embryonic development by controlling cell patterning, growth and migration. In adults, the function of Hh pathway is curtailed to tissue repair and maintenance. Aberrant reactivation of Hh signaling has been linked to tumorigenesis in various cancers, such as basal cell carcinoma (BCC) and medulloblastoma. The Smoothened (Smo) receptor, a key component of the Hh pathway which is central to the signaling transduction, has emerged as an attractive therapeutic target for the treatment of human cancers. Taking advantage of the availability of several crystal structures of Smo in complex with different antagonists, we have previously conducted a molecular docking-based virtual screening to identify several compounds which exhibited significant inhibitory activity against the Hh pathway activation (IC50 < 10 µM) in a Gli-responsive element (GRE) reporter gene assay. The most potent compound (ChemDiv ID C794-1677: 47 nM) showed comparable Hh signaling inhibition to the marketed drug vismodegib (46 nM). Herein, we report our structural optimization based on the virtual screening hit C794-1677. Our efforts are aimed to improve potency, decrease cLogP, and remove potentially metabolic labile/toxic pyrrole and aniline functionalities presented in C794-1677. The optimization led to the identification of numerous potent compounds exemplified by 25 (7.1 nM), which was 7 folds more potent compared with vismodegib. In addition, 25 was much less lipophilic compared with C794-1677 and devoid of the potentially metabolic labile/toxic pyrrole and aniline functional groups. Furthermore, 25 exhibited promising efficacy in inhibiting Gli1 mRNA expression in NIH3T3 cells with either wildtype Smo or D473H Smo mutant. These results represented significant improvement over the virtual screening hit C794-1677 and suggested that compound 25 can be used as a good starting point to support lead optimization.


Subject(s)
Anilides/pharmacology , Computer Simulation , Drug Evaluation, Preclinical , Pyridines/pharmacology , Smoothened Receptor/antagonists & inhibitors , Anilides/chemistry , Animals , Dose-Response Relationship, Drug , Mice , Molecular Docking Simulation , Molecular Structure , NIH 3T3 Cells , Pyridines/chemistry , Structure-Activity Relationship
8.
Acta Pharmacol Sin ; 40(10): 1292-1302, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31015738

ABSTRACT

Microglia, the brain-resident macrophage, is known as the innate immune cell type in the central nervous system. Microglia is also the major cellular component of tumor mass of gliomas that plays a key role in glioma development. Mutations of isocitrate dehydrogenases 1 and 2 (IDH1/2) frequently occur in gliomas, which leads to accumulation of oncometabolic product 2-hydroxyglutarate (2HG). Moreover, IDH1/2 mutations were found to correlate with better prognosis in glioma patients. In the present study, we investigated the effects of the 2HG on microglial inflammatory activation. We showed that the conditioned media (CM) from GL261 glioma cells stimulated the activation of BV-2 microglia cells, evidenced by markedly increased expression of interleukin-6 (IL-6), IL-1ß, tumor necrosis factor-α (TNF-α), CCL2 (C-C motif chemokine ligand 2) and CXCL10 (C-X-C motif chemokine 10). CM-induced expression of proinflammatory genes was significantly suppressed by pretreatment with a synthetic cell-permeable 2HG (1 mM) or a nuclear factor-κB (NF-κB) inhibitor BAY11-7082 (10 µM). In lipopolysaccharide (LPS)- or TNF-α-stimulated BV-2 microglia cells and primary microglia, pretreatment with 2HG (0.25-1 mM) dose-dependently suppressed the expression of proinflammatory genes. We further demonstrated that 2HG significantly suppressed LPS-induced phosphorylation of IκB kinase α/ß (IKKα/ß), IκBα and p65, IκB degradation, and nuclear translocation of p65 subunit of NF-κB, as well as NF-κB transcriptional activity. Similarly, ectopic expression of mutant isocitrate dehydrogenase 1 (IDH1) (R132H) significantly decreased TNF-α-induced activation of NF-κB signaling pathway. Finally, we revealed that activation of adenosine 5'-monophosphate-activated protein kinase (AMPK) and subsequent inhibition of mammalian target of rapamycin (mTOR) signaling contributed to the inhibitory effect of 2HG on NF-κB signaling pathway in BV-2 cells. Taken together, these results, for the first time, show that oncometabolite 2HG inhibits microglial activation through affecting AMPK/mTOR/NF-κB signaling pathway and provide evidence that oncometabolite 2HG may regulate glioma development via modulating microglial activation in tumor microenvironment.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Glutarates/pharmacology , Microglia/drug effects , NF-kappa B/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Microglia/metabolism , NF-kappa B/metabolism , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
9.
ACS Chem Neurosci ; 10(2): 783-791, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30199223

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease with significant unmet medical needs. The current dopamine-centered treatments aim to restore motor functions of patients without slowing the disease progression. Long-term usage of these drugs is associated with diminished efficacy, motor fluctuation, and dyskinesia. Furthermore, the nonmotor features associated with PD such as sleep disorder, pain, and psychiatric symptoms are poorly addressed by the dopaminergic treatments. Adenosine receptor A2A antagonists have emerged as potential treatment for PD in the past decade. Here we summarize the recent work (2015-2018) on adenosine receptor A2A antagonists and discuss the challenge and opportunity for the treatment of PD.


Subject(s)
Adenosine A2 Receptor Antagonists/therapeutic use , Drug Development/methods , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists/chemistry , Animals , Drug Development/trends , Humans , Parkinson Disease/pathology , Treatment Outcome
10.
Anticancer Drugs ; 29(10): 995-1003, 2018 11.
Article in English | MEDLINE | ID: mdl-30106753

ABSTRACT

The hedgehog-smoothened (HH/SMO) pathway has been proposed as a potential therapeutic target for hematological malignancies. Our previous studies designed a series of HH inhibitors with novel scaffolds distinctive from vismodegib, the first Food and Drug Administration-approved HH inhibitor for the treatment of basal-cell carcinoma and medulloblastoma. In the present study, we evaluated these HH inhibitors against blood cancers and found that HH78 displayed potent activity in suppressing the HH signaling pathway. HH78 competitively bound to SMO and suppressed the transcriptional activity of GLI by the luciferase reporter gene assay and the measurement of HH/SMO-downregulated genes, including cyclin D2, cyclin E, PTCH1, PTCH2, and GLI. HH78 at low micromolar concentrations induced significant cancer cell apoptosis showed by increased caspase-3 activation, annexin V-staining and downregulated prosurvival proteins, including c-Myc, Bcl-2, Mcl-1, and Bcl-xL. In contrast, vismodegib did not show any effects on these apoptotic events. HH78 also suppressed the activation of the AKT/mTOR pathway, which cross-talks with the HH/SMO pathway. Finally, HH78 inhibited the growth of human leukemia K562 in nude mice xenografts with no overt toxicity. Collectively, the present study identified a novel HH inhibitor with great potential for the treatment of hematological malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Hematologic Neoplasms/drug therapy , Smoothened Receptor/antagonists & inhibitors , Anilides/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Dose-Response Relationship, Drug , Down-Regulation/genetics , Hematologic Neoplasms/pathology , Humans , K562 Cells , Mice , Mice, Nude , Pyridines/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
11.
Eur J Med Chem ; 155: 34-48, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29857275

ABSTRACT

The Hedgehog (Hh) signaling pathway plays a critical role in controlling patterning, growth and cell migration during embryonic development. Aberrant activation of Hh signaling has been linked to tumorigenesis in various cancers, such as basal cell carcinoma (BCC) and medulloblastoma. As a key member of the Hh pathway, the Smoothened (Smo) receptor, a member of the G protein-coupled receptor (GPCR) family, has emerged as an attractive therapeutic target for the treatment and prevention of human cancers. The recent determination of several crystal structures of Smo in complex with different antagonists offers the possibility to perform structure-based virtual screening for discovering potent Smo antagonists with distinct chemical scaffolds. In this study, based on the two Smo crystal complexes with the best capacity to distinguish the known Smo antagonists from decoys, the molecular docking-based virtual screening was conducted to identify promising Smo antagonists from ChemDiv library. A total of 21 structurally novel and diverse compounds were selected for experimental testing, and six of them exhibited significant inhibitory activity against the Hh pathway activation (IC50 < 10 µM) in a GRE (Gli-responsive element) reporter gene assay. Specifically, the most potent compound (compound 20: 47 nM) showed comparable Hh signaling inhibition to vismodegib (46 nM). Compound 20 was further confirmed to be a potent Smo antagonist in a fluorescence based competitive binding assay. Optimization using substructure searching method led to the discovery of 12 analogues of compound 20 with decent Hh pathway inhibition activity, including four compounds with IC50 lower than 1 µM. The important residues uncovered by binding free energy calculation (MM/GBSA) and binding free energy decomposition were highlighted and discussed. These findings suggest that the novel scaffold afforded by compound 20 can be used as a good starting point for further modification/optimization and the clarified interaction patterns may also guide us to find more potent Smo antagonists.


Subject(s)
Boron Compounds/pharmacology , Drug Discovery , Fluorescent Dyes/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Veratrum Alkaloids/pharmacology , Animals , Boron Compounds/chemical synthesis , Boron Compounds/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Mice , Molecular Docking Simulation , Molecular Structure , NIH 3T3 Cells , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship , Veratrum Alkaloids/chemical synthesis , Veratrum Alkaloids/chemistry
12.
Eur J Med Chem ; 149: 30-44, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29494843

ABSTRACT

The important roles of the CXCL12/CXCR4 axis in numerous pathogenic pathways involving HIV infection and cancer metastasis make the CXCR4 receptor an attractive target for the development of therapeutic agents. Through scaffold hybridization of a few known CXCR4 antagonists, a series of novel aminopyrimidine derivatives was developed. Compound 3 from this new scaffold demonstrates excellent binding affinity with CXCR4 receptor (IC50 = 54 nM) and inhibits CXCL12 induced cytosolic calcium increase (IC50 = 2.3 nM). Furthermore, compound 3 possesses good physicochemical properties (MW 353, clogP 2.0, PSA 48, pKa 6.7) and exhibits minimal hERG and CYP isozyme (e.g. 3A4, 2D6) inhibition. Collectively, these results strongly support further optimization of this novel scaffold to develop better CXCR4 antagonists.


Subject(s)
Drug Design , Pyrimidines/chemistry , Receptors, CXCR4/antagonists & inhibitors , Calcium/metabolism , Chemokine CXCL12/physiology , Humans , Protein Binding , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Structure-Activity Relationship
13.
Eur J Med Chem ; 149: 110-121, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29499483

ABSTRACT

Embryonic stem cell pathways such as hedgehog and Wnt pathways are central to the tumorigenic properties of cancer stem cells (CSC). Since CSCs are characterized by their ability to self-renew, form differentiated progeny, and develop resistance to anticancer therapies, targeting the Wnt and hedgehog signaling pathways has been an important strategy for cancer treatment. Although molecules targeting either Wnt or hedgehog are common, to the best of our knowledge, those targeting both pathways have not been documented. Here we report a small molecule (compound 1) that inhibits both Wnt (IC50 = 0.5 nM) and hedgehog (IC50 = 71 nM) pathways based on reporter gene assays. We further identified that the molecular target of 1 for Wnt pathway inhibition was porcupine (a member of the membrane-bound O-acyltransferase family of proteins), a post-translational modification node in Wnt signaling; while the target of 1 mitigating hedgehog pathway was Smoothened, a key G protein coupled receptor (GPCR) mediating hedgehog signal transduction. Preliminary analysis of structure-activity-relationship identified key functional elements for hedgehog/Wnt inhibition. In in vivo studies, compound 1 demonstrated good oral exposure and bioavailability while eliciting no overt toxicity in mice. An important consideration in cancer treatment is the potential therapeutic escape through compensatory activation of an interconnected pathway when only one signaling pathway is inhibited. Toward this end, compound 1 may not only lead to the development of new therapeutics for Wnt and hedgehog related cancers, but may also help to develop potential cancer treatment which needs to target Wnt and hedgehog signaling simultaneously.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Hedgehog Proteins/drug effects , Signal Transduction/drug effects , Wnt Signaling Pathway/drug effects , Acyltransferases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Hedgehog Proteins/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Mice , Smoothened Receptor/antagonists & inhibitors , Structure-Activity Relationship
14.
Front Pharmacol ; 8: 741, 2017.
Article in English | MEDLINE | ID: mdl-29163149

ABSTRACT

The neuromodulator hydrogen sulfide (H2S) was shown to exert neuroprotection in different models of Parkinson's disease (PD) via its anti-inflammatory and anti-apoptotic properties. In this study, we evaluated the effect of an H2S slow-releasing compound GYY4137 (GYY) on a mouse PD model induced by acute injection with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). GYY was intraperitoneally (i.p.) injected once daily into male C57BL/6J mice 3 days before and 2 weeks after MPTP (14 mg/kg, four times at 2-h intervals, i.p.) administration. Saline was given as a control. Behavioral tests (rotarod, balance beam, and grid walking) showed that 50 mg/kg GYY significantly ameliorated MPTP-caused motor impairments. At lower doses (12.5 and 25 mg/kg) GYY exhibited a less obvious effect. Consistent with this, immunohistochemistry and western blot analysis demonstrated that 50 mg/kg GYY attenuated the loss of tyrosine hydroxylase (TH) positive neurons in the substantia nigra and the decrease of TH expression in the striatum of MPTP-treated mice. Moreover, at this regimen GYY relieved the nitrative stress, as indicated by the decreases in nitric oxide (NO) generation and neuronal NO synthase (nNOS) upregulation elicited by MPTP in the striatum. The suppression of GYY on nNOS expression was verified in vitro, and the results further revealed that Akt activation may participate in the inhibition by GYY on nNOS upregulation. More important, GYY reduced the nitrated modification of α-synuclein, a PD-related protein, in MPTP-induced mice. Overall, our findings suggest that GYY attenuated dopaminergic neuron degeneration and reduced α-synuclein nitration in the midbrain, thus exerting neuroprotection in MPTP-induced mouse model of PD.

15.
ACS Chem Neurosci ; 8(9): 1980-1994, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28618224

ABSTRACT

Medulloblastoma is one of the most prevalent brain tumors in children. Aberrant hedgehog (Hh) pathway signaling is thought to be involved in the initiation and development of medulloblastoma. Vismodegib, the first FDA-approved cancer therapy based on inhibition of aberrant hedgehog signaling, targets smoothened (Smo), a G-protein coupled receptor (GPCR) central to the Hh pathway. Although vismodegib exhibits promising therapeutic efficacy in tumor treatment, concerns have been raised from its nonlinear pharmacokinetic (PK) profiles at high doses partly due to low aqueous solubility. Many patients experience adverse events such as muscle spasms and weight loss. In addition, drug resistance often arises among tumor cells during treatment with vismodegib. There is clearly an urgent need to explore novel Smo antagonists with improved potency and efficacy. Through a scaffold hopping strategy, we have identified a series of novel tetrahydropyrido[4,3-d]pyrimidine derivatives, which exhibited effective inhibition of Hh signaling. Among them, compound 24 is three times more potent than vismodegib in the NIH3T3-GRE-Luc reporter gene assay. Compound 24 has a lower melting point and much greater solubility compared with vismodegib, resulting in linear PK profiles when dosed orally at 10, 30, and 100 mg/kg in rats. Furthermore, compound 24 showed excellent PK profiles with a 72% oral bioavailability in beagle dogs. Compound 24 demonstrated overall favorable in vitro safety profiles with respect to CYP isoform and hERG inhibition. Finally, compound 24 led to significant regression of subcutaneous tumor generated by primary Ptch1-deficient medulloblastoma cells in SCID mouse. In conclusion, tetrahydropyrido[4,3-d]pyrimidine derivatives represent a novel set of Smo inhibitors that could potentially be utilized to treat medulloblastoma and other Hh pathway related malignancies.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Anilides/chemistry , Anilides/pharmacokinetics , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , CHO Cells , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/metabolism , Cricetulus , Dogs , Drug Design , Female , Humans , Male , Medulloblastoma/drug therapy , Medulloblastoma/metabolism , Mice , Mice, Inbred ICR , Mice, SCID , Mice, Transgenic , NIH 3T3 Cells , Neoplasm Transplantation , Patched-1 Receptor/deficiency , Patched-1 Receptor/genetics , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Random Allocation , Rats, Sprague-Dawley , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism , Structure-Activity Relationship , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism
16.
Oncotarget ; 8(14): 23277-23288, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28177895

ABSTRACT

Hepatocellular carcinoma (HCC) is a common malignancy found worldwide and is associated with a high incidence of metastasis and vascular invasion. Elucidating the molecular mechanisms that underlie HCC tumorigenesis and progression is necessary for the development of novel therapeutics. By analyzing the Cancer Genome Atlas Network (TCGA) dataset, we identified Thrombospondin 4 (THBS4) is significantly overexpressed in HCC samples and is correlated with prognosis. Overexpression of THBS4 was also highly correlated with vascular invasion of advanced HCC. While THBS4 is often overexpressed in HCC it has also been shown to inhibit tumor growth by mediating cell-to-cell and cell-to-matrix interactions. Here, we identified that knockdown of THBS4 inhibits migration and invasion of HCC cells and inhibits HCC induced angiogenesis. MiRNAs are crucial regulators of multiple cellular processes, and aberrant expression of miRNAs has been observed to effect cancer development and progression. We further found that miR-142 is an upstream regulator of THBS4 in HCC cells. Moreover, miR-142 was significantly down-regulated in HCC tissue samples and correlated with overexpression of THBS4. Overexpression of miR-142 inhibited invasion and angiogenesis of HCC cells and re-expression of THBS4 overcame these effects of miR-142 expression. Stable over-expression of miR-142 significantly inhibited tumour growth in a xenograft tumour model through inhibiting THBS4 expression and tumor angiogenesis. In conclusion, our findings indicate that loss of miR-142 results in the over-expression of THBS4, which enhances HCC migration and vascular invasion. Thus, targeting THBS4 or miR-142 may provide a promising therapeutic strategy for treatment of advanced HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Thrombospondins/biosynthesis , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Heterografts , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , MicroRNAs/genetics , Neoplasm Invasiveness , Survival Rate , Thrombospondins/genetics
17.
Bioorg Med Chem ; 24(22): 5861-5872, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27692509

ABSTRACT

The Wnt signaling pathway is an essential signal transduction pathway which leads to the regulation of cellular processes such as proliferation, differentiation and migration. Aberrant Wnt signaling is known to have an association with multiple cancers. Porcupine is an enzyme that catalyses the addition of palmitoleate to a serine residue in Wnt proteins, a process which is required for the secretion of Wnt proteins. Here we report the synthesis and structure-activity-relationship of the novel porcupine inhibitors based on a 'reversed' amide scaffold. The leading compound 53 was as potent as the clinical compound LGK974 in a cell based STF reporter gene assay. Compound 53 potently inhibited the secretion of Wnt3A, therefore was confirmed to be a porcupine inhibitor. Furthermore, compound 53 showed excellent chemical and plasma stabilities. However, the clearance of compound 53 in liver microsomal tests was moderate to high, and the solubility of compound 53 was suboptimal. Collective efforts toward further optimization of this novel tricyclic template to develop better porcupine inhibitors will be subsequently undertaken and reported in due course.


Subject(s)
Amides/chemistry , Amides/pharmacology , Drug Design , Membrane Proteins/antagonists & inhibitors , Acyltransferases , Amides/chemical synthesis , Dose-Response Relationship, Drug , Humans , Membrane Proteins/metabolism , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Solubility , Structure-Activity Relationship , Wnt Signaling Pathway/drug effects , Wnt3A Protein/metabolism
18.
ACS Chem Neurosci ; 7(11): 1575-1584, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27569066

ABSTRACT

Adenosine receptor A2A antagonists have emerged as potential treatment for Parkinson's disease in the past decade. We have recently reported a series of adenosine receptor antagonists using heterocycles as bioisosteres for a potentially unstable acetamide. These compounds, while showing excellent potency and ligand efficiency, suffered from moderate cytochrome P450 inhibition and high clearance. Here we report a new series of adenosine receptor A2A antagonists based on a 4-amino-5-carbonitrile pyrimidine template. Compounds from this new template exhibit excellent potency and ligand efficiency with low cytochrome P450 inhibition. Although the clearance remains moderate to high, the leading compound, when dosed orally as low as 3 mg/kg, demonstrated excellent efficacy in the haloperidol induced catalepsy rat model for Parkinson's disease.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Antiparkinson Agents/pharmacology , Pyrimidines/pharmacology , Adenosine A2 Receptor Antagonists/chemical synthesis , Adenosine A2 Receptor Antagonists/pharmacokinetics , Animals , Antiparkinson Agents/chemical synthesis , Antiparkinson Agents/pharmacokinetics , Drug Design , Drug Evaluation, Preclinical , Haloperidol , Humans , Mice , Microsomes, Liver/drug effects , Molecular Docking Simulation , Molecular Structure , Parkinsonian Disorders/drug therapy , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Rats , Structure-Activity Relationship
19.
Oncol Rep ; 36(3): 1325-32, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27430152

ABSTRACT

Nutlin-3, a small molecule regulator of the tumor suppressor p53, targets the interaction between p53 and murine double minute 2 (MDM2) thereby promoting stabilization of p53 and subsequent p53­dependent induction of apoptosis and cell cycle arrest. Recent studies have demonstrated that Nutlin­3 plays a critical role in regulating tumor cell migration, invasion, metastasis, and drug resistance. Although these studies identified various biological functions of Nutlin­3, our understanding of the exact molecular mechanisms of Nutlin­3­mediated antitumor activity remains incomplete. In this study, we elucidated a role of Nutlin­3 in reversing the epithelial­mesenchymal transition (EMT) in gemcitabine-resistant (GR) hepatocellular carcinoma (HCC) cells. We assessed the effect of Nutlin­3 treatment on cell growth, migration, and invasion in both parental HCC cells and GR HCC cells. Moreover, we detected the expression of EMT markers in GR HCC cells treated with Nutlin­3 by real­time RT­PCR and western blot analysis, respectively. We found that Nutlin-3 inhibited cell migration and invasion in the GR HCC cells. Additionally, Nutlin­3 treatment increased E-cadherin protein levels, but decreased the protein levels of vimentin, Snail and Slug in the GR HCC cells. Furthermore, we found that Smad2 was highly expressed in the GR HCC cells compared with their parental HCC cells, and Nutlin-3 treatment downregulated Smad2 expression in the GR HCC cells. Depletion of Smad2 retarded cell migration and regulated the expression of EMT markers in GR HCC cells similarly to Nutlin­3 treatment. Our findings highlight an important role of Nutlin­3 in reversing EMT in GR cells through regulation of Smad2 expression, suggesting that Nutlin-3 could be a potential agent for the treatment of HCC patients with gemcitabine resistance.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Imidazoles/pharmacology , Liver Neoplasms/drug therapy , Piperazines/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cadherins/genetics , Carcinoma, Hepatocellular/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Deoxycytidine/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Neoplasm Invasiveness/genetics , Smad2 Protein/genetics , Snail Family Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Vimentin/genetics , Gemcitabine
20.
J Agric Food Chem ; 64(28): 5742-50, 2016 Jul 20.
Article in English | MEDLINE | ID: mdl-27362917

ABSTRACT

Rhein is a major component of the many medicinal herbs such as rhubarb. Despite wide use, intoxication cases associated with rhein-containing herbs are often reported. The present work aimed to investigate if rhein was subject to metabolic activation leading to toxicity. Upon incubations with different species of liver microsomes, three monoglucuronides were identified, corresponding to two hydroxyl glucuronides and one acyl glucuronide via the carboxyl group, respectively. Further study revealed that rhein acyl glucuronide was chemically reactive, and showed cytotoxicity toward hepatocarcinoma cells. In addition, significant species differences in glucuronidation of rhein were observed between laboratory animals and humans. Reaction phenotyping experiments demonstrated that rhein acyl glucuronide was catalyzed predominantly by uridine 5'-diphospho-glucuronosyltransferase 1A1, 1A9, and 2B7. Taken together, the present study confirmed that rhein could be metabolically activated via the formation of acyl glucuronide, especially in human.


Subject(s)
Anthraquinones/metabolism , Drugs, Chinese Herbal/metabolism , Plant Extracts/metabolism , Rheum/chemistry , Activation, Metabolic , Animals , Anthraquinones/chemistry , Anthraquinones/toxicity , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/toxicity , Glucuronosyltransferase/metabolism , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Structure , Plant Extracts/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL