Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Med Chem ; 66(16): 11282-11293, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37535016

ABSTRACT

2'-Deoxy-2'-ß-fluoroadenosines bearing 4'-azido or 4'-ethynyl groups designed for the treatment of HIV-1 infection have been synthesized. All these compounds possess nanomolar anti-HIV-1 activity, with the 4'-ethynyl-2-fluoroadenosine analog 1c (CL-197) being the most potent compound with low cytotoxicity (EC50 = 0.9 nM, CC50 > 100 µM). It also shows potent inhibitory activities on drug resistant and clinical HIV-1 strains. Oral administration of 1c to Beagle dogs resulted in high levels of its bioactive form 1c-TP in peripheral blood mononuclear cells, the HIV-1 target cells, where the resulting triphosphate exhibited a long-term intracellular retention and could prevent HIV-1 infection for an extended time. 1c displayed low in vivo toxicity and favorable pharmacokinetics profiles in Sprague-Dawley rats. The preclinical data support further development of 1c as a highly potent and orally bioavailable clinical candidate to treat HIV-1 infection. Currently, CL-197 is in clinical trials in China (registration number: CXHL2200529).


Subject(s)
Anti-HIV Agents , HIV Infections , HIV-1 , Rats , Animals , Dogs , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Leukocytes, Mononuclear , Rats, Sprague-Dawley , HIV Infections/drug therapy
2.
Phytomedicine ; 116: 154848, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37163901

ABSTRACT

BACKGROUND: Hepatitis B virus (HBV) infection remains a major global health burden, due to the increasing risk of complications, such as cirrhosis and hepatocellular carcinoma. Novel anti-HBV agents are critical required. Our previous study suggested that Artemisia argyi essential oil (AAEO) significantly inhibited the replication of HBV DNA and especially the secretion of hepatitis B antigen in vitro. PURPOSE: The aim of this study was to prepare AAEO loaded nanostructured lipid carriers (AAEO-NLCs) for the delivery of AAEO to the liver, investigated the therapeutic benefits of AAEO-NLCs against HBV in a duck HBV (DHBV) model and explored its potential mechanism. STUDY DESIGN AND METHODS: AAEO-NLCs were prepared by hot homogenization and ultrasonication method. The DHBV-infected ducks were treated with AAEO (4 mg/kg), AAEO-NLCs (0.8, 4, and 20 mg/kg of AAEO), and lamivudine (20 mg/kg) for 15 days. The DHBV DNA levels in the serum and liver were measured by quantitative Real-Time PCR. Pharmacokinetics and liver distribution were performed in rats after oral administration of AAEO-NLCs and AAEO suspension. The potential antiviral mechanism and active compounds of AAEO were investigated by network pharmacology and molecular docking. RESULTS: AAEO-NLCs markedly inhibited the replication of DHBV DNA in a dose-dependent manner and displayed a low virologic rebound following withdrawal the treatment in DHBV-infected ducks. Moreover, AAEO-NLCs led to a more pronounced reduction in viral DNA levels than AAEO suspension. Further investigations of pharmacokinetics and liver distribution in rats confirmed that NLCs improved the oral bioavailability and increased the liver exposure of AAEO. The potential mechanisms of AAEO against HBV explored by network pharmacology were associated with signaling pathways related to immune response, such as tumor necrosis factor, nuclear factor kappa B, and sphingolipid signaling pathways. Furthermore, a total of 16 potential targets were obtained, including prostaglandin-endoperoxide synthase-2 (PTGS2), caspase-3, progesterone receptor, etc. Compound-target docking results confirmed that four active compounds of AAEO had strong binding interactions with the active sites of PTGS2. CONCLUSIONS: AAEO-NLCs displayed potent anti-HBV activity with improved oral bioavailability and liver exposure of AAEO. Thus, it may be a potential therapeutic strategy for the treatment of HBV infection.


Subject(s)
Artemisia , Hepatitis B Virus, Duck , Liver Neoplasms , Oils, Volatile , Rats , Animals , Molecular Docking Simulation , Oils, Volatile/pharmacology , Network Pharmacology , Cyclooxygenase 2 , Antiviral Agents/pharmacology , Hepatitis B virus/genetics , Hepatitis B Virus, Duck/genetics
3.
J Med Chem ; 65(13): 9493-9505, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35776695

ABSTRACT

A series of new O-(substituted benzyl) phosphoramidate prodrugs of tenofovir for the treatment of hepatitis B virus (HBV) infections have been designed and synthesized. An investigation of structure-activity relationships revealed that the compound bearing an o-methylbenzyl group (1a) has the most potent in vitro anti-HBV activity. This prodrug (1a) was well-tolerated in KM mice via intragastric administration at a dosage of up to 1.5 g/kg. In DHBV-infected ducks, prodrug 1a displayed a good inhibitory effect on the viral DNA replication in both the serum and the liver in a time- and dose-dependent manner and did not cause any necrosis, hemorrhage, or inflammatory response in the animal livers. Further investigation demonstrated that prodrug 1a achieved a higher exposure of the bioactive metabolite (tenofovir diphosphate, TFV-DP) in the liver, the target organ for the treatment of HBV infection, than tenofovir alafenamide fumarate (TAF) did at an equimolar dose.


Subject(s)
Hepatitis B , Prodrugs , Alanine/pharmacology , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , DNA Replication , DNA, Viral , Hepatitis B/drug therapy , Hepatitis B virus/metabolism , Mice , Prodrugs/pharmacology , Prodrugs/therapeutic use , Tenofovir/metabolism , Tenofovir/pharmacology , Tenofovir/therapeutic use , Virus Replication
5.
Genes Genomics ; 44(1): 123-131, 2022 01.
Article in English | MEDLINE | ID: mdl-34697761

ABSTRACT

BACKGROUND: Previously, we published that 4'-azid-2'-deoxy-2'-fluorarabinoside (FNC), a novel cytosine nucleoside analog, has good anti-viral and anti-tumor activity. OBJECTIVE: This study aimed to further explore the role and molecular mechanism of FNC in non-small cell lung cancer (NSCLC). METHODS: FNC was tested in the NSCLC H460 cell line, the Lewis mouse model, and the H460 cell xenograft model. The effects of FNC were assessed by cell viability, transwell migration, and wound scratch analyses of cell migration and invasion. Apoptosis was assessed by flow cytometry. Proteins expression was assessed by western blot and immunohistochemistry staining (IHC). RESULTS: FNC inhibits the proliferation and metastasis of H460 cells in a time- and dose-dependent manner. FNC treatment showed efficacy and low toxicity in the Lewis mouse lung cancer model as well as in the H460 cell xenograft model. Further, FNC induced H460 cell apoptosis through the activation of the mitochondrial pathway. Notably, FNC inhibited invasion by increasing E-cadherin protein and reducing the protein expression of VEGF, MMP-2, MMP-9, and CD31. CONCLUSION: FNC inhibits NSCLC by activating the mitochondrial apoptosis pathway and regulating the expressions of multiple proteins related to cell adhesion and invasion, highlighting its potential as an NSCLC therapeutic.


Subject(s)
Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cytosine Nucleotides/pharmacology , Lung Neoplasms/drug therapy , Neoplasms, Experimental/drug therapy , Xenograft Model Antitumor Assays/methods , Animals , Autophagy-Related Proteins/metabolism , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytosine Nucleotides/chemistry , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Signal Transduction/drug effects
6.
Front Cell Dev Biol ; 9: 664791, 2021.
Article in English | MEDLINE | ID: mdl-34268303

ABSTRACT

The tumor metastasis is the major hurdle for the treatment of advanced hepatocellular carcinoma (HCC), due in part to the lack of effective systemic treatments. DEPDC1, a novel oncoantigen upregulated in HCC, is thought to be a molecular-target for novel therapeutic drugs. Artemisia argyi is a traditional Chinese medicine with anti-inflammatory and anti-tumor activities. This study investigated the potential therapeutic benefits of Artemisia argyi essential oil (AAEO) in suppressing metastasis of HCC by targeting DEPDC1. Assessment of AAEO cytotoxicity was performed by MTT assay. Anti-metastatic effects of AAEO were investigated in vitro using wound healing and transwell assays. The HepG2 cells were transduced with lentiviral vector containing luciferase (Luc). A metastasis model of nude mice was established by tail vein injection of HepG2-Luc cells. The nude mice were treated with AAEO (57.5, 115, and 230 mg/kg) or sorafenib (40 mg/kg). Metastasis of HCC cells was monitored via in vivo bioluminescence imaging. After treatment for 21 days, tissues were collected for histological examination and immunohistochemistry analysis. Gene and protein levels were determined by real-time quantitative PCR and western blotting. The results revealed that AAEO significantly inhibits the migration and invasion in vitro in a concentration-dependent manner. In vivo assays further confirmed that AAEO markedly inhibits HCC metastasis into lung, brain, and femur tissues and exhibits low toxicity. Our results suggested that AAEO significantly downregulates the mRNA and protein expression of DEPDC1. Also, AAEO attenuated Wnt/ß-catenin signaling through reduction of Wnt1 and ß-catenin production. Moreover, AAEO prevented epithelial-mesenchymal transition (EMT) by downregulation of vimentin and upregulation of E-cadherin. Furthermore, we found that DEPDC1 promoted HCC migration and invasion via Wnt/ß-catenin signaling pathway and EMT. These results demonstrate that AAEO effectively inhibits HCC metastasis via attenuating Wnt/ß-catenin signaling and inhibiting EMT by suppressing DEPDC1 expression. Thus, AAEO likely acts as a novel inhibitor of the DEPDC1 dependent Wnt/ß-catenin signaling pathway.

7.
Int Immunopharmacol ; 97: 107690, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33940323

ABSTRACT

It has recently been shown that expression levels of tissue factor (TF) are high in the serum and peripheral blood mononuclear cells of patients with asthma. However, whether TF impacts airway inflammation and remodelling in asthma remains unknown. The aim of this study was to investigate the effect of TF in asthma airway inflammation and remodelling using a house dust mite (HDM)-induced chronic asthma model and human bronchial epithelial (16HBE) cells. A chronic asthma model was constructed in BALB/c mice by the intranasal instillation of HDM. Mice were treated with short hairpin TF (shTF), and airway inflammation and remodelling features of asthma and epithelial-mesenchymal transition (EMT) were assessed. 16HBE cells were induced by transforming growth factor-ß1 (TGF-ß1) and HDM in the presence or absence of shTF; then, EMT markers and invasion and migration ability were determined. TF expression increased in the lung tissue and 16HBE cells when exposed to HDM. TF downregulation in the lung significantly reduced airway hyperresponsiveness, eosinophil inflammation, the EMT process, and levels of interleukin (IL)-4, IL-6, IL-13, and TGF-ß1 in bronchoalveolar lavage fluid of asthmatic mice. Moreover, TF downregulation inhibited migration and incursion and decreased the expression levels of fibronectin 1 and TGF-ß1, but increased the expression of E-cadherin in HDM- and TGF-ß1-stimulated 16HBE cells. These results demonstrated that TF promoted airway pathological features by enhancing the EMT of bronchial epithelial cells both in vitro and in mice with house dust mite-induced asthma.


Subject(s)
Allergens/immunology , Asthma/immunology , Dermatophagoides pteronyssinus/immunology , Thromboplastin/metabolism , Airway Remodeling/immunology , Animals , Asthma/pathology , Bronchi/cytology , Bronchi/immunology , Bronchi/pathology , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/immunology , HEK293 Cells , Humans , Mice , Specific Pathogen-Free Organisms , Thromboplastin/genetics , Up-Regulation/immunology
8.
Acta Virol ; 65(1): 58-67, 2021.
Article in English | MEDLINE | ID: mdl-33827222

ABSTRACT

Coxsackie virus B3 (CVB3) is believed to be a major cause of viral myocarditis, with virus-induced apoptosis playing an important role in pathogenesis. The purpose of this study was to characterize the antiviral activity of a novel fluoronucleoside analogue, N-cyclopropyl-4'-azido-2'-deoxy-2'-fluoro-ß-D-cytidine (NCC), against CVB3 in vitro and in vivo, and to establish whether NCC inhibits apoptosis in infected cells. In this study, HeLa cells infected with CVB3 were treated with NCC. Cell viability and apoptosis were examined. Caspase-3 and Bcl-2 levels were monitored by real-time RT PCR and Western blot analysis. For in vivo studies, BALB/c mice infected with CVB3 were treated with NCC daily. Serum markers of myocardial injury and histological studies were measured to examine myocardial injury on day 8 post-infection. To measure apoptosis, levels of Bcl-2 and caspase-3 were examined by immunohistochemistry and real-time RT-PCR. We found that NCC inhibited virus-mediated cytopathic effects in HeLa cells with an EC50 of 116.60 ± 0.32 µM. In infected mice, administration of NCC (2 mg/kg) decreased the activities of serum creatine kinase and lactic dehydrogenase, inhibited the replication of CVB3 and alleviated damage to the heart. Importantly, NCC suppressed CVB3-induced apoptosis in HeLa cells and affected the expression of apoptosis-related factors in infected mice. Together, our results demonstrate that NCC exerts significant antiviral activities against CVB3. We conclude that NCC is a potential therapeutic agent for the treatment of viral myocarditis. Keywords: coxsackie virus B3; viral myocarditis; N-cyclopropyl-4'-azido-2'-deoxy-2'-fluoro-ß-D-cytidine; apoptosis.


Subject(s)
Coxsackievirus Infections , Myocarditis , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Coxsackievirus Infections/drug therapy , Enterovirus B, Human , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Myocarditis/drug therapy , Virus Replication
9.
J Med Chem ; 64(2): 925-937, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33459024

ABSTRACT

Osimertinib is a highly potent and selective third-generation epidermal growth factor receptor (EGFR) inhibitor, which provides excellent clinical benefits and is now a standard-of-care therapy for advanced EGFR mutation-positive non-small-cell lung cancer (NSCLC). However, AZ5104, a primary toxic metabolite of osimertinib, has caused unwanted toxicities. To address this unmet medical need, we initiated an iterative program focusing on structural optimizations of osimertinib and preclinical characterization, leading to the discovery of a highly potent, selective, and orally efficacious deuterated EGFR-targeting clinical candidate, dosimertinib. Preclinical studies revealed that dosimertinib demonstrated robust in vivo antitumor efficacy and favorable PK profiles, but with lower toxicity than osimertinib. These preclinical data support further clinical development of dosimertinib for the treatment of NSCLC. Dosimertinib has received official approval in China to initiate the phase I clinical trial (registration numbers: CXHL2000060 and CXHL2000061).


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Acrylamides/metabolism , Aniline Compounds/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation , Dogs , Drug Discovery , ErbB Receptors/drug effects , Humans , Indoles , Lung Neoplasms/genetics , Mice , Microsomes, Liver/metabolism , Mutation/genetics , Pyrimidines , Rats , Signal Transduction/drug effects , Structure-Activity Relationship , Xenograft Model Antitumor Assays
10.
Article in English | MEDLINE | ID: mdl-33160078

ABSTRACT

BACKGROUND: Lipids participate in many important biological functions through energy storage, material transport, signal transduction, and molecular recognition processes. Studies have reported that asthmatic patients have abnormal lipid metabolism. However, there are limited studies on the characterization of lipid metabolism in asthmatic patients by lipidomics. METHODS: We characterized the plasma lipid profile of 28 healthy controls and 33 outpatients with asthma (18 mild, 15 moderate) by liquid chromatography mass spectrometry/mass spectrometry-based lipidomics. RESULTS: We determined 1338 individual lipid species in the plasma. Significant changes were identified in ten lipid species in asthmatic patients than in healthy controls (all P < 0.05). Phosphatidylethanolamine (PE) (18:1p/22:6), PE (20:0/18:1), PE (38:1), sphingomyelin (SM) (d18:1/18:1), and triglyceride (TG) (16:0/16:0/18:1) positively correlated with the severity of asthma (all P < 0.05). Phosphatidylinositol (PI) (16:0/20:4), TG (17:0/18:1/18:1), phosphatidylglycerol (PG) (44:0), ceramide (Cer) (d16:0/27:2), and lysophosphatidylcholine (LPC) (22:4) negatively correlated with the severity of asthma (all P < 0.05). Correlation analysis showed a significant correlation between all ten lipid species (all P < 0.05). From the area under the curve of the receiver operating characteristic curve analysis, PE (38:1) was the major lipid metabolite that distinguished asthmatic patients from healthy controls, and may be considered a potential lipid biomarker. PE (20:0/18:1) and TG (16:0/16:0/18:1) might be related to IgE levels in asthmatic patients. CONCLUSIONS: Our results indicated the presence of abnormal lipid metabolism, which correlated with the severity and IgE levels in asthmatic patients.


Subject(s)
Asthma/diagnosis , Lipid Metabolism , Lipids/blood , Adolescent , Adult , Aged , Asthma/blood , Asthma/immunology , Asthma/metabolism , Biomarkers/blood , Biomarkers/metabolism , Case-Control Studies , Chromatography, High Pressure Liquid/methods , Female , Healthy Volunteers , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Lipidomics/methods , Male , Middle Aged , Severity of Illness Index , Tandem Mass Spectrometry/methods , Young Adult
11.
J Med Chem ; 63(15): 8554-8566, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32678592

ABSTRACT

In preclinical and phase I and II clinical studies, 2'-deoxy-2'-ß-fluoro-4'-azidocytidine (FNC) displays a potent and long-lasting inhibition of HIV-1 infection. To investigate its mechanism of action, we compared it with the well-documented lamivudine (3TC). Pharmacokinetic studies revealed that the intracellular retention of FNC triphosphate in peripheral blood mononuclear cells was markedly longer than that of the 3TC triphosphate. FNC selectively enters and is retained in HIV target cells, where it exerts long-lasting prevention of HIV-1 infection. In addition to inhibition of HIV-1 reverse transcription, FNC also restores A3G expression in CD4+ T cells in FNC-treated HIV-1 patients. FNC binds to the Vif-E3 ubiquitin ligase complex, enabling A3G to avoid Vif-induced ubiquitination and degradation. These data reveal the mechanisms underlying the superior anti-HIV potency and long-lasting action of FNC. Our results also suggest a potential clinical application of FNC as a long-lasting pre-exposure prophylactic agent capable of preventing HIV infection.


Subject(s)
Anti-HIV Agents/therapeutic use , Azides/therapeutic use , Deoxycytidine/analogs & derivatives , HIV Infections/drug therapy , HIV-1/drug effects , Lamivudine/therapeutic use , Animals , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/pharmacology , Azides/pharmacokinetics , Azides/pharmacology , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , HIV Infections/metabolism , HIV-1/physiology , Humans , Lamivudine/pharmacokinetics , Lamivudine/pharmacology , Macaca mulatta , Models, Molecular , Reverse Transcriptase Inhibitors/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcriptase Inhibitors/therapeutic use , Ubiquitination/drug effects
12.
Oral Dis ; 26(1): 182-192, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31696592

ABSTRACT

This study aims to investigate the levels of SLIT3 in gingival crevicular fluid (GCF) of healthy and periodontal disease subjects, and their correlations to periodontal disease. A total of 45 periodontal patients and 45 periodontally healthy volunteers were enrolled. The clinical parameters, radiographic bone loss and the levels of SLIT3, receptor activator of NF-κB ligand (RANKL) and osteoprotegerin (OPG) in GCF were measured. The prevalences of Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia in subgingival plaque were also analyzed. The expression of SLIT3 and RANKL was detected in the periodontium of experimental periodontitis in rats and lipopolysaccharide (LPS)-induced mouse macrophage. The total amounts and concentrations of SLIT3 and RANKL were significantly higher in periodontitis than those in healthy, while the level of OPG was significantly lower (p < .05). Significant positive correlations were observed between the level of GCF SLIT3 and clinical attachment level and radiographic bone loss (p < .05). There existed a significant positive correlation between SLIT3 and RANKL (p < .05). Increased expression of SLIT3 and RANKL was observed in the periodontium of periodontal rats. SLIT3 expression was induced by LPS stimulation in macrophages. These results suggest that SLIT3 may act as a diagnostic indicator of periodontal disease and should be further investigated.


Subject(s)
Gingival Crevicular Fluid/chemistry , Membrane Proteins/metabolism , Periodontitis/metabolism , Adult , Animals , Dental Plaque/microbiology , Female , Humans , Male , Mice , Osteoprotegerin/metabolism , Periodontium/metabolism , Porphyromonas gingivalis/isolation & purification , RANK Ligand/metabolism , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Tannerella forsythia/isolation & purification , Treponema denticola/isolation & purification
13.
Bioorg Med Chem Lett ; 29(11): 1291-1297, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30962085

ABSTRACT

Hepatitis B virus (HBV) is a global health problem requiring more efficient and better tolerated anti-HBV agent. In this paper, a series of novel 2'-deoxy-2'-fluoro-2'-C-methyl-ß-d-arabinofuranosyl 8-azanebularine analogues (1 and 2a) and N4-substituted 8-azaadenosine derivatives (2b-g) were designed, synthesized and screened for in vitro anti-HBV activity. Two concise and practical synthetic routes were developed toward the structural motif construction of 2'-deoxy-2'-fluoro-2'-C-methyl-ß-d-arabinofuranosyl 8-azainosine from the ribonolactone 3 under mild conditions. The in vitro anti-HBV screening results showed that these 8-azanebularine analogues had a significant inhibitory effect on the expression of HBV antigens and HBV DNA at a concentration of 20 µM. Among them, halogen-substituted 8-azaadenosine derivative 2g displayed activities comparable to that of 3TC. In particular, 2g retained excellent activity against lamivudine-resistant HBV mutants.


Subject(s)
Antiviral Agents/pharmacology , Drug Design , Hepatitis B virus/drug effects , Purine Nucleosides/pharmacology , Ribonucleosides/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , DNA, Viral/drug effects , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Microbial Sensitivity Tests , Molecular Structure , Purine Nucleosides/chemical synthesis , Purine Nucleosides/chemistry , Ribonucleosides/chemical synthesis , Ribonucleosides/chemistry , Structure-Activity Relationship
14.
Eur J Pharm Sci ; 128: 290-298, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30557605

ABSTRACT

We recently reported that the absolute oral bioavailability of flavonoid Morin was extremely low at 0.45%, resulting in unsatisfied therapeutic efficacy in vivo. The present study was aimed to systemically assess the pre-absorption risks of Morin for rationale formulation design. Physicochemical properties of Morin were evaluated using in vitro assays including water solubility and stability in simulated gastric, intestinal fluids, followed by permeability tests in Caco-2 cells. The results suggested that both poor solubility and low membrane permeability were rate-limiting steps for oral absorption of Morin. Pharmacokinetic studies were performed via a series of administration routes in a dual-vein cannulated rat model. In contrast to high bioavailability (92.92%) and negligible metabolites of Morin in intraportal administered rats, Morin exhibited low bioavailability (5.28%) and considerable amount of metabolites in rats following intraduodenal administration, suggested that intestinal first-pass metabolism made a major contribution to its poor oral absorption. Morin-phospholipid complex loaded self-emulsifying drug delivery system (MPC-SENDDS) exhibited comparable plasma concentration of metabolites to parent drug after oral administration, indicated that MPC-SNEDDS failed to bypass first-pass metabolism and therefore showed compromised oral absorption enhancement. The present study could promote the development of more efficient oral formulations of Morin with optimized absorption enhancement.


Subject(s)
Antioxidants/pharmacokinetics , Flavonoids/pharmacokinetics , Administration, Oral , Animals , Antioxidants/administration & dosage , Area Under Curve , Biological Availability , Biological Products , Caco-2 Cells , Flavonoids/administration & dosage , Half-Life , Humans , Male , Permeability , Rats , Rats, Wistar
15.
Braz J Infect Dis ; 22(6): 477-486, 2018.
Article in English | MEDLINE | ID: mdl-30586543

ABSTRACT

Antiviral drug resistance is the most important factor contributing to treatment failure using nucleos(t)ide analogs such as lamivudine for chronic infection with hepatitis B virus (HBV). Development of a system supporting efficient replication of clinically resistant HBV strains is imperative, and new antiviral drugs are needed urgently to prevent selection of drug-resistant HBV mutants. A novel fluorinated cytidine analog, NCC (N-cyclopropyl-4'-azido-2'-deoxy-2'-fluoro-ß-d-cytidine), was recently shown to strongly inhibit human HBV in vitro and in vivo. This study was designed to evaluate the antiviral activity of NCC against lamivudine-resistant HBV. We generated a stable cell line encoding the major pattern of lamivudine-resistant mutations rtL180M/M204V and designated it "HepG2.RL1". Immuno-transmission electron microscopic examination and enzyme-linked immunosorbent assay were used to detect secretion of HBV-specific particles and antigens. Quantification of extracellular DNA and intracellular DNA of HepG2.RL1 cells by quantitative real-time polymerase chain reaction revealed >625-fold and >5556-fold increases in the 50% inhibitory concentration of lamivudine, respectively, compared with that for the wild-type virus. The results showed that NCC inhibited DNA replication and HBeAg production in wild-type or lamivudine-resistant HBV in a dose-dependent manner. In conclusion, screening for antiviral compounds active against lamivudine-resistant HBV can be carried out with relative ease using hepG2.RL1 cells. NCC is a potential antiviral agent against wild-type HBV and clinical lamivudine-resistant HBV and deserves evaluation for the treatment of HBV infection.


Subject(s)
Antiviral Agents/pharmacology , Cytidine/analogs & derivatives , Drug Resistance, Viral/drug effects , Hepatitis B virus/drug effects , Lamivudine/pharmacology , Virus Replication/drug effects , Cell Line , DNA, Viral/chemistry , Female , Hepatitis B virus/isolation & purification , Hepatitis B virus/physiology , Hepatocytes/virology , Humans , Microbial Sensitivity Tests , Middle Aged , Mutation
16.
Braz. j. infect. dis ; 22(6): 477-486, Nov.-Dec. 2018. tab, graf
Article in English | LILACS | ID: biblio-984016

ABSTRACT

ABSTRACT Antiviral drug resistance is the most important factor contributing to treatment failure using nucleos(t)ide analogs such as lamivudine for chronic infection with hepatitis B virus (HBV). Development of a system supporting efficient replication of clinically resistant HBV strains is imperative, and new antiviral drugs are needed urgently to prevent selection of drug-resistant HBV mutants. A novel fluorinated cytidine analog, NCC (N-cyclopropyl-4′-azido-2′-deoxy-2′-fluoro-β-d-cytidine), was recently shown to strongly inhibit human HBV in vitro and in vivo. This study was designed to evaluate the antiviral activity of NCC against lamivudine-resistant HBV. We generated a stable cell line encoding the major pattern of lamivudine-resistant mutations rtL180M/M204V and designated it "HepG2.RL1". Immuno-transmission electron microscopic examination and enzyme-linked immunosorbent assay were used to detect secretion of HBV-specific particles and antigens. Quantification of extracellular DNA and intracellular DNA of HepG2.RL1 cells by quantitative real-time polymerase chain reaction revealed >625-fold and >5556-fold increases in the 50% inhibitory concentration of lamivudine, respectively, compared with that for the wild-type virus. The results showed that NCC inhibited DNA replication and HBeAg production in wild-type or lamivudine-resistant HBV in a dose-dependent manner. In conclusion, screening for antiviral compounds active against lamivudine-resistant HBV can be carried out with relative ease using hepG2.RL1 cells. NCC is a potential antiviral agent against wild-type HBV and clinical lamivudine-resistant HBV and deserves evaluation for the treatment of HBV infection.


Subject(s)
Humans , Female , Middle Aged , Antiviral Agents/pharmacology , Virus Replication/drug effects , Hepatitis B virus/drug effects , Lamivudine/pharmacology , Cytidine/analogs & derivatives , DNA, Viral/chemistry , Microbial Sensitivity Tests , Cell Line , Hepatitis B virus/isolation & purification , Hepatitis B virus/physiology , Hepatocytes/virology , Drug Resistance, Viral/drug effects , Mutation
17.
Pharmazie ; 73(9): 503-507, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30223932

ABSTRACT

The purpose of the present study was to dig into recent studies designed to characterize the impacts of 2'-deoxy-2'-ß-fluoro-4'-azidocytidine (FNC) on P-glycoprotein (P-gp), multidrug resistance-associated protein 2 (MRP2) and breast cancer resistance protein (BCRP). Specifically, the modulation effects of FNC on P-gp, MRP2 and BCRP protein expressions were assessed by western blot methods. 5 (and 6)-Carboxy-2',7'-dichloroflourescein (CDF) and BODIPY-prazosin were used to provide indications of alterations of MRP2 and BCRP activities. The effects of P-gp, MRP2 and BCRP on FNC were evaluated in the in situ single-pass intestinal perfusion model. The results showed that FNC at higher concentrations and with longer incubation times can upregulate the protein expression of P-gp, MRP2 and BCRP in Caco-2 cells. The upregulated proteins were also functionally active, as revealed by a lower degree of CDF and BODIPY-prazosin uptake by the cell monolayers. The intestinal absorptive coefficient (Peff) was observed to significantly increase with the inhibitors of P-gp, MRP2 and BCRP. These results suggested that FNC could modulate the expressions and functions of P-gp, MRP2 and BCRP, while P-gp, MRP2 and BCRP were involved in the efflux transport of FNC. The inductive effects of FNC on P-gp, MRP2 and BCRP suggested the possibility of FNC to contribute to the inter- and intra-individual variability of itself, as well as to alter the absorption of other drugs that may be administered concomitantly.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Azides/pharmacology , Deoxycytidine/analogs & derivatives , Multidrug Resistance-Associated Proteins/genetics , Neoplasm Proteins/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Anti-HIV Agents/pharmacology , Biological Transport , Caco-2 Cells , Deoxycytidine/pharmacology , Drug Interactions , Humans , Intestinal Absorption , Male , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
18.
Drug Deliv ; 25(1): 1117-1126, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29781332

ABSTRACT

Asthma is one of the most prevalent chronic inflammatory diseases of lung. Current asthma therapy using inhaled corticosteroid often results in undesired treatment outcome due to poor compliance and drugs' lack of tissue specificity. N,N,N'-trimethyl-N'-(2-hydroxyl-3-methyl-5-123Iiodobenzyl)-1,3-propanediamine (HIPD), a phenolic propanediamine derivative, has been used as an imaging agent for localized pulmonary diseases. Inspired by this, N,N,N'-trimethyl-N'-(4-hydroxyl-benzyl)-1,3-propanediamine (TPD), a new HIPD analog, was proposed as a lung-targeting ligand and covalently conjugated to an anti-inflammatory compound Rhein for asthma therapy. Cellular uptake efficiency of TPD-Rhein by A549 cells was significantly enhanced compared with Rhein. The enhanced cellular uptake was mainly mediated by organic cation transporters (OCTs) in an active manner, showing concentration- and energy-dependent. After systemic administration in rats, TPD-Rhein specifically distributed to lungs, displaying the highest Cmax and AUC0-t values of all tested tissues and resulting in a 13-fold increase in Cmax and a 103-fold increase in AUC0-t for lung compared with Rhein. Also, TPD-Rhein remarkably decreased serum histamine levels, serum IL-5 levels as well as bronchoalveolar lavage fluid IL-5 levels in lungs of asthmatic rats challenged by ovalbumin (OVA). Accordingly, histological examinations demonstrated that TPD-Rhein attenuated lung inflammation in rats, with no apparent toxicity against major organs. Together, phenolic propanediamine-based lung-targeting approach represents an efficient and safe strategy for asthma therapy.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Asthma/drug therapy , Sulfhydryl Compounds/pharmacology , A549 Cells , Animals , Anthraquinones/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Cell Line, Tumor , Disease Models, Animal , HeLa Cells , Humans , Lung , Male , Mice , Rats , Rats, Wistar
19.
Eur J Med Chem ; 143: 137-149, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29174810

ABSTRACT

Novel drugs are urgently needed to combat hepatitis B virus (HBV) infection due to drug-resistant virus. In this paper, a series of novel 4-monosubstituted 2'-deoxy-2'-ß-fluoro-4'-azido-ß-d-arabinofuranosyl 1,2,3-triazole nucleoside analogues (1a-g) were designed, synthesized and screened for in vitro anti-HBV activity. At 5.0 µM in the cellular model, all the synthetic compounds display activities comparable to that of the positive control, lamivudine at 20 µM. Of the compounds tested, the amide-substituted analogue (1a) shows the most promising anti-HBV activity and low cytotoxicity in the cell model. In particular, it retains excellent activity against lamivudine-resistant HBV mutants. In duck HBV (DHBV)-infected duck models, both the serum and liver DHBV DNA levels (67.4% and 53.3%, respectively) were reduced markedly by the treatment with 1a. Analysis of the structure of HBV polymer/1a-triphosphate (1a-TP) complex shows that 1a-TP is stabilized by specific van der Waals interactions with the enzyme residues arising from 4-amino-1,2,3-triazole and the 4'-azido group.


Subject(s)
Antiviral Agents/pharmacology , Drug Design , Hepatitis B Virus, Duck/drug effects , Hepatitis B virus/drug effects , Hepatitis B/drug therapy , Monosaccharides/pharmacology , Nucleosides/pharmacology , Triazoles/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Ducks , Hep G2 Cells , Hepatitis B virus/genetics , Humans , Models, Molecular , Molecular Structure , Monosaccharides/chemical synthesis , Monosaccharides/chemistry , Nucleosides/chemical synthesis , Nucleosides/chemistry , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , Virus Replication/drug effects
20.
Eur J Med Chem ; 143: 107-113, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29172078

ABSTRACT

A novel ß-D-2'-deoxy-2'-α-fluoro-2'-ß-C-(fluoromethyl)uridine phosphoramidate prodrug (1) has been synthesized. This compound exhibits submicromolar-level antiviral activity in vitro against HCV genotypes 1b, 1a, 2a, and S282T replicons (EC50 = 0.18-1.13 µM) with low cytotoxicity (CC50 > 1000 µM). Administered orally, prodrug 1 is well tolerated at doses of up to 4 g/kg in mice, and produces a high level of the corresponding triphosphate in rat liver.


Subject(s)
Hepacivirus/drug effects , Hepatitis C/drug therapy , Organophosphorus Compounds/pharmacology , Prodrugs/pharmacology , Uridine/analogs & derivatives , Administration, Oral , Animals , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Hepacivirus/genetics , Humans , Liver/drug effects , Liver/virology , Mice , Mice, Inbred Strains , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Prodrugs/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Uridine/administration & dosage , Uridine/chemistry , Uridine/pharmacology , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...