Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Nucleic Acids Res ; 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38742636

RESUMEN

Oral delivery is the most widely used and convenient route of administration of medicine. However, oral administration of hydrophilic macromolecules is commonly limited by low intestinal permeability and pre-systemic degradation in the gastrointestinal (GI) tract. Overcoming some of these challenges allowed emergence of oral dosage forms of peptide-based drugs in clinical settings. Antisense oligonucleotides (ASOs) have also been investigated for oral administration but despite the recent progress, the bioavailability remains low. Given the advancement with highly potent and durable trivalent N-acetylgalactosamine (GalNAc)-conjugated small interfering RNAs (siRNAs) via subcutaneous (s.c.) injection, we explored their activities after oral administration. We report robust RNA interference (RNAi) activity of orally administrated GalNAc-siRNAs co-formulated with permeation enhancers (PEs) in rodents and non-human primates (NHPs). The relative bioavailability calculated from NHP liver exposure was <2.0% despite minimal enzymatic degradation in the GI. To investigate the impact of oligonucleotide size on oral delivery, highly specific GalNAc-conjugated single-stranded oligonucleotides known as REVERSIRs with different lengths were employed and their activities for reversal of RNAi effect were monitored. Our data suggests that intestinal permeability is highly influenced by the size of oligonucleotides. Further improvements in the potency of siRNA and PE could make oral delivery of GalNAc-siRNAs as a practical solution.

2.
Nat Biotechnol ; 40(10): 1500-1508, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35654979

RESUMEN

Therapeutics based on short interfering RNAs (siRNAs) delivered to hepatocytes have been approved, but new delivery solutions are needed to target additional organs. Here we show that conjugation of 2'-O-hexadecyl (C16) to siRNAs enables safe, potent and durable silencing in the central nervous system (CNS), eye and lung in rodents and non-human primates with broad cell type specificity. We show that intrathecally or intracerebroventricularly delivered C16-siRNAs were active across CNS regions and cell types, with sustained RNA interference (RNAi) activity for at least 3 months. Similarly, intravitreal administration to the eye or intranasal administration to the lung resulted in a potent and durable knockdown. The preclinical efficacy of an siRNA targeting the amyloid precursor protein was evaluated through intracerebroventricular dosing in a mouse model of Alzheimer's disease, resulting in amelioration of physiological and behavioral deficits. Altogether, C16 conjugation of siRNAs has the potential for safe therapeutic silencing of target genes outside the liver with infrequent dosing.


Asunto(s)
Precursor de Proteína beta-Amiloide , Tratamiento con ARN de Interferencia , Animales , Ratones , Primates/genética , Primates/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico
3.
Drug Metab Dispos ; 50(6): 781-797, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34154993

RESUMEN

Conjugation of oligonucleotide therapeutics, including small interfering RNAs (siRNAs) or antisense oligonucleotides, to N-acetylgalactosamine (GalNAc) ligands has become the primary strategy for hepatocyte-targeted delivery, and with the recent approvals of GIVLAARI (givosiran) for the treatment of acute hepatic porphyria, OXLUMO (lumasiran) for the treatment of primary hyperoxaluria, and Leqvio (inclisiran) for the treatment of hypercholesterolemia, the technology has been well validated clinically. Although much knowledge has been gained over decades of development, there is a paucity of published literature on the drug metabolism and pharmacokinetic properties of GalNAc-siRNA. With this in mind, the goals of this minireview are to provide an aggregate analysis of these nonclinical absorption, distribution, metabolism, and excretion (ADME) data to build confidence on the translation of these properties to human. Upon subcutaneous administration, GalNAc-conjugated siRNAs are quickly distributed to the liver, resulting in plasma pharmacokinetic (PK) properties that reflect rapid elimination through asialoglycoprotein receptor-mediated uptake from circulation into hepatocytes. These studies confirm that liver PK, including half-life and, most importantly, siRNA levels in RNA-induced silencing complex in hepatocytes, are better predictors of pharmacodynamics (PD) than plasma PK. Several in vitro and in vivo nonclinical studies were conducted to characterize the ADME properties of GalNAc-conjugated siRNAs. These studies demonstrate that the PK/PD and ADME properties of GalNAc-conjugated siRNAs are highly conserved across species, are largely predictable, and can be accurately scaled to human, allowing us to identify efficacious and safe clinical dosing regimens in the absence of human liver PK profiles. SIGNIFICANCE STATEMENT: Several nonclinical ADME studies have been conducted in order to provide a comprehensive overview of the disposition and elimination of GalNAc-conjugated siRNAs and the pharmacokinetic/pharmacodynamic translation between species. These studies demonstrate that the ADME properties of GalNAc-conjugated siRNAs are well correlated and predictable across species, building confidence in the ability to extrapolate to human.


Asunto(s)
Acetilgalactosamina , Porfirias Hepáticas , Acetilgalactosamina/farmacocinética , Receptor de Asialoglicoproteína/metabolismo , Hepatocitos/metabolismo , Humanos , Porfirias Hepáticas/metabolismo , ARN Interferente Pequeño/genética
4.
Drug Metab Dispos ; 49(7): 572-580, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33941543

RESUMEN

Givosiran is an N-acetylgalactosamine-conjugated RNA interference therapeutic that targets 5'-aminolevulinate synthase 1 mRNA in the liver and is currently marketed for the treatment of acute hepatic porphyria. Herein, nonclinical pharmacokinetics and absorption, distribution, metabolism, and excretion properties of givosiran were characterized. Givosiran was completely absorbed after subcutaneous administration with relatively short plasma elimination half-life (t1/2; less than 4 hours). Plasma exposure increased approximately dose proportionally with no accumulation after repeat doses. Plasma protein binding was concentration dependent across all species tested and was around 90% at clinically relevant concentration in human. Givosiran predominantly distributed to the liver by asialoglycoprotein receptor-mediated uptake, and the t1/2 in the liver was significantly longer (∼1 week). Givosiran was metabolized by nucleases, not cytochrome P450 (P450) isozymes, across species with no human unique metabolites. Givosiran metabolized to form one primary active metabolite with the loss of one nucleotide from the 3' end of antisense strand, AS(N-1)3' givosiran, which was equipotent to givosiran. Renal and fecal excretion were minor routes of elimination of givosiran as approximately 10% and 16% of the dose was recovered intact in excreta of rats and monkeys, respectively. Givosiran is not a substrate, inhibitor, or inducer of P450 isozymes, and it is not a substrate or inhibitor of uptake and most efflux transporters. Thus, givosiran has a low potential of mediating drug-drug interactions involving P450 isozymes and drug transporters. SIGNIFICANCE STATEMENT: Nonclinical pharmacokinetics and absorption, distribution, metabolism, and excretion (ADME) properties of givosiran were characterized. Givosiran shows similar pharmacokinetics and ADME properties across rats and monkeys in vivo and across human and animal matrices in vitro. Subcutaneous administration results in adequate exposure of givosiran to the target organ (liver). These studies support the interpretation of toxicology studies, help characterize the disposition of givosiran in humans, and support the clinical use of givosiran for the treatment of acute hepatic porphyria.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Pirrolidinas/farmacocinética , 5-Aminolevulinato Sintetasa/antagonistas & inhibidores , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/farmacocinética , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Femenino , Semivida , Inyecciones Subcutáneas , Eliminación Intestinal , Macaca fascicularis , Masculino , Modelos Animales , Porfobilinógeno Sintasa/deficiencia , Porfirias Hepáticas/tratamiento farmacológico , Pirrolidinas/administración & dosificación , Ratas , Eliminación Renal , Distribución Tisular
5.
Nucleic Acid Ther ; 31(4): 309-315, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33861634

RESUMEN

Serum protein interactions are evaluated during the drug development process since they determine the free drug concentration in blood and thereby can influence the drug's pharmacokinetic and pharmacodynamic properties. While the impact of serum proteins on the disposition of small molecules is well understood, it is not yet well characterized for a new modality, RNA interference therapeutics. When administered systemically, small interfering RNAs (siRNAs) conjugated to the N-acetylgalactosamine (GalNAc) ligand bind to proteins present in circulation. However, it is not known if these protein interactions may impact the GalNAc-conjugated siRNA uptake into hepatocytes mediated through the asialoglycoprotein receptor (ASGPR) and thereby influence the activity of GalNAc-conjugated siRNAs. In this study, we assess the impact of serum proteins on the uptake and activity of GalNAc-conjugated siRNAs in primary human hepatocytes. We found that a significant portion of the GalNAc-conjugated siRNAs is bound to serum proteins. However, ASGPR-mediated uptake and activity of GalNAc-conjugated siRNAs were minimally impacted by the presence of serum relative to their uptake and activity in the absence of serum. Therefore, in contrast to small molecules, serum proteins are expected to have minimal impact on pharmacokinetic and pharmacodynamic properties of GalNAc-conjugated siRNAs.


Asunto(s)
Acetilgalactosamina , Hepatocitos , Receptor de Asialoglicoproteína/genética , Receptor de Asialoglicoproteína/metabolismo , Proteínas Sanguíneas/genética , Hepatocitos/metabolismo , Humanos , Interferencia de ARN , ARN Interferente Pequeño/genética
6.
Bioanalysis ; 11(21): 1927-1939, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31829053

RESUMEN

Aim: The electrophoretic mobility shift assay (EMSA) was evaluated as an alternative to ultrafiltration (UF) to assess plasma protein binding (PPB) of small interfering RNAs (siRNA) and antisense oligonucleotides (ASO). Results & methodology: EMSA analysis showed that PPB depended on siRNA and plasma concentration. Conversely, when analyzed by ultrafiltration, siRNA bound the filtration device nonspecifically and PPB remained >98% across physiologically relevant siRNA concentrations. Using EMSA, siRNA exhibited charge-based interactions with plasma proteins, while ASO remained highly bound to plasma proteins or albumin in the presence of 500 mM salt. Conclusion: PPB characteristics of siRNA and ASO can be distinguished using EMSA. Characterization of siRNA PPB by EMSA enhances our knowledge of siRNA absorption, distribution, metabolism and excretion and advanced development of RNA interference therapeutics.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Ensayo de Cambio de Movilidad Electroforética/métodos , ARN Interferente Pequeño/metabolismo , Unión Proteica
7.
Bioanalysis ; 11(21): 1967-1980, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31829056

RESUMEN

Aim: Advancements in RNA interference therapeutics have triggered development of improved bioanalytical methods for oligonucleotide metabolite profiling and high-throughput quantification in biological matrices. Results & methodology: HPLC coupled with high-resolution mass spectrometry (LC-HRMS) methods were developed to investigate the metabolism of a REVERSIR™ molecule in vivo. Plasma and tissue samples were extracted using solid-phase extraction followed by LC-HRMS analysis for metabolite profiling and quantification. The method was qualified from 10 to 5000 ng/ml (plasma) and 100 to 50000 ng/g (liver and kidney). In rat liver, intra and interday accuracy ranged from 80.9 to 118.5% and 88.4 to 111.9%, respectively, with acceptable precision (<20% CV). Conclusion: The LC-HRMS method can be applied for metabolite profiling and quantification of oligonucleotides in biological matrices.


Asunto(s)
Espectrometría de Masas/métodos , Metabolómica/métodos , Oligonucleótidos/metabolismo , Animales , Secuencia de Bases , Cromatografía Liquida , Hígado/metabolismo , Macaca fascicularis , Oligonucleótidos/sangre , Oligonucleótidos/genética , Oligonucleótidos/orina
8.
Drug Metab Dispos ; 47(10): 1183-1194, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31270142

RESUMEN

Small interfering RNAs (siRNAs) represent a new class of medicines that are smaller (∼16,000 Da) than biologic therapeutics (>150,000 Da) but much larger than small molecules (<900 Da). Current regulatory guidance on drug-drug interactions (DDIs) from the European Medicines Agency, Food and Drug Administration, and Pharmaceutical and Medical Devices Agency provides no recommendations for oligonucleotide therapeutics including siRNAs; therefore, small molecule guidance documents have historically been applied. Over ∼10 years, in vitro DDI investigations with siRNAs conjugated to a triantennary N-acetylgalactosamine [(GalNAc)-siRNA] ligand have been conducted during nonclinical drug development to elucidate the potential clinical DDI liability. GalNAc siRNAs were evaluated as substrates, inhibitors, or inducers of major cytochrome P450s (P450s) and as substrates and inhibitors of transporters. Aggregate analysis of these data demonstrates a low potential for DDI against P450s. Zero of five, 10, and seven are inducers, time-dependent inhibitors, or substrates, respectively, and nine of 12 do not inhibit any P450 isoform evaluated. Three GalNAc siRNAs inhibited CYP2C8 at supratherapeutic concentrations, and one mildly inhibited CYP2B6. The lowest K i value of 28 µM is >3000-fold above the therapeutic clinical C max at steady state, and importantly no clinical inhibition was projected. Of four GalNAc siRNAs tested none were substrates for transporters and one caused inhibition of P-glycoprotein, calculated not to be clinically relevant. The pharmacological basis for DDIs, including consideration of the target and/or off-target profiles for GalNAc siRNAs, should be made as part of the overall DDI risk assessment. If modulation of the target protein does not interfere with P450s or transporters, then in vitro or clinical investigations into the DDI potential of the GalNAc siRNAs are not warranted. SIGNIFICANCE STATEMENT: Recommendations for evaluating DDI potential of small molecule drugs are well established; however, guidance for novel modalities, particularly oligonucleotide-based therapeutics are lacking. Given the paucity of published data in this field, in vitro DDI investigations are often conducted. The aggregate analysis of GalNAc-siRNA data reviewed herein demonstrates that, like new biological entities, these oligonucleotide-based therapeutic drugs are unlikely to result in DDIs; therefore, it is recommended that the need for in vitro or clinical investigations similarly be determined on a case-by-case basis. Given the mechanism of siRNA action, special consideration should be made in cases where there may be a pharmacological basis for DDIs.


Asunto(s)
Acetilgalactosamina/farmacología , Inductores de las Enzimas del Citocromo P-450/farmacología , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Oligonucleótidos/farmacología , ARN Interferente Pequeño/farmacología , Acetilgalactosamina/análogos & derivados , Células Cultivadas , Simulación por Computador , Inductores de las Enzimas del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Pruebas de Enzimas , Hepatocitos , Humanos , Concentración 50 Inhibidora , Proteínas de Transporte de Membrana/agonistas , Proteínas de Transporte de Membrana/genética , Modelos Biológicos , Oligonucleótidos/química , ARN Interferente Pequeño/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
9.
J Agric Food Chem ; 64(27): 5598-606, 2016 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-27321734

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) refers to hepatic pathologies, including simple fatty liver (SFL), nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis, that may progress to hepatocellular carcinoma. These liver disease states may affect the activity and expression levels of drug-metabolizing enzymes, potentially resulting in an alteration in the pharmacokinetics, therapeutic efficacy, and safety of drugs. This study investigated the hepatic cytochrome P450 (CYP) 2B1-modulating effect of a specific NAFLD state in dietary rat models. Sprague-Dawley rats were given a methionine/choline-deficient (MCD) or high-fat (HF) diet to induce NASH and SFL, respectively. The induction of these disease states was confirmed by plasma chemistry and liver histological analysis. Both the protein and mRNA levels of hepatic CYP2B1 were considerably reduced in MCD diet-fed rats; however, they were similar between the HF diet-fed and control rats. Consistently, the enzyme-kinetic and pharmacokinetic parameters for CYP2B1-mediated bupropion metabolism were considerably reduced in MCD diet-fed rats; however, they were also similar between the HF diet-fed and control rats. These results may promote a better understanding of the influence of NAFLD on CYP2B1-mediated metabolism, which could have important implications for the safety and pharmacokinetics of drug substrates for the CYP2B subfamily in patients with NAFLD.


Asunto(s)
Bupropión/administración & dosificación , Deficiencia de Colina/tratamiento farmacológico , Metionina/deficiencia , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Animales , Colina/metabolismo , Deficiencia de Colina/enzimología , Deficiencia de Colina/genética , Deficiencia de Colina/metabolismo , Citocromo P-450 CYP2B1/genética , Citocromo P-450 CYP2B1/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ratas , Ratas Sprague-Dawley
10.
Cancer Lett ; 377(1): 25-31, 2016 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-27105612

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) is widely used for preventing neutropenia during chemotherapy. Polyethylene glycol-conjugated granulocyte colony-stimulating factor (PEG-G-CSF, pegfilgrastim) serves the same purpose but has a longer half-life and greater stability than G-CSF. In this study, we investigated whether 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol, acetylated diglyceride (PLAG), augments the therapeutic effect of pegfilgrastim on chemotherapy-induced neutropenia. We compared neutrophil counts in four groups of mice: control mice, gemcitabine-treated mice, gemcitabine/pegfilgrastim-treated mice, and gemcitabine/pegfilgrastim/PLAG-treated mice. PLAG (50 mg/kg) was orally administered every day during the treatment course. CBC analysis showed that the group treated with PLAG experienced a dramatically increased neutrophil counts on the third day following pegfilgrastim treatment. PLAG had no effect on blood cell apoptosis and neutrophil release from bone marrow. Additionally, pegfilgrastim-induced CXCR2 expression in neutrophils was markedly decreased in PLAG-treated animals. These results suggest that PLAG plays a role in inhibiting neutrophil extravasation, giving rise to an increased number of circulating neutrophils when used with pegfilgrastim during gemcitabine treatment. These data support the potential for PLAG to be used with pegfilgrastim to treat or prevent chemotherapy-induced neutropenia by modulating neutrophil transmigration.


Asunto(s)
Desoxicitidina/análogos & derivados , Diglicéridos/farmacología , Factor Estimulante de Colonias de Granulocitos/farmacología , Neutropenia/tratamiento farmacológico , Neutrófilos/efectos de los fármacos , Animales , Biomarcadores/sangre , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Quimioterapia Combinada , Filgrastim , Recuento de Leucocitos , Ratones Endogámicos BALB C , Neutropenia/sangre , Neutropenia/inducido químicamente , Neutrófilos/metabolismo , Polietilenglicoles , Receptores de Interleucina-8B/sangre , Proteínas Recombinantes/farmacología , Factores de Tiempo , Gemcitabina
11.
PLoS One ; 11(3): e0151758, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27010397

RESUMEN

Increased number of eosinophils in the circulation and sputum is associated with the severity of asthma. The respiratory epithelium produces chemokine (C-C motif) ligands (CCL) which recruits and activates eosinophils. A chemically synthesized monoacetyl-diglyceride, PLAG (1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol) is a major constituent in the antlers of Sika deer (Cervus nippon Temminck) which has been used in oriental medicine. This study was aimed to investigate the molecular mechanism of PLAG effect on the alleviation of asthma phenotypes. A549, a human alveolar basal epithelial cell, and HaCaT, a human keratinocyte, were activated by the treatment of interleukin-4 (IL-4), and the expression of chemokines, known to be effective on the induction of eosinophil migration was analyzed by RT-PCR. The expression of IL-4 induced genes was modulated by the co-treatment of PLAG. Especially, CCL26 expression from the stimulated epithelial cells was significantly blocked by PLAG, which was confirmed by ELISA. The transcriptional activity of signal transducer and activator of transcription 6 (STAT6), activated by IL-4 mediated phosphorylation and nuclear translocation, was down-regulated by PLAG in a concentration-dependent manner. In ovalbumin-induced mouse model, the infiltration of immune cells into the respiratory tract was decreased by PLAG administration. Cytological analysis of the isolated bronchoalveolar lavage fluid (BALF) cells proved the infiltration of eosinophils was significantly reduced by PLAG. In addition, PLAG inhibited the migration of murine bone marrow-derived eosinophils, and human eosinophil cell line, EoL-1, which was induced by the addition of A549 culture medium.


Asunto(s)
Asma/tratamiento farmacológico , Quimiocinas CC/genética , Quimiotaxis/efectos de los fármacos , Diglicéridos/uso terapéutico , Eosinófilos/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Animales , Asma/genética , Asma/inmunología , Línea Celular , Quimiocina CCL26 , Quimiocinas CC/inmunología , Ciervos , Diglicéridos/farmacología , Eosinófilos/citología , Eosinófilos/inmunología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Humanos , Interleucina-4/inmunología , Ratones Endogámicos BALB C
12.
Immune Netw ; 15(3): 150-60, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26140047

RESUMEN

We previously reported that 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) accelerates hematopoiesis and has an improving effect on animal disease models such as sepsis and asthma. The effects of PLAG supplementation on immune modulation were assessed in healthy men and women. The objective was to evaluate the effects of PLAG supplementation on immune regulatory functions such as activities of immune cells and cytokine production. A randomized double blind placebo-controlled trial was conducted. Seventy-five participants were assigned to one of two groups; all participants had an appropriate number of white blood cells on the testing day. The PLAG group (n=27) received oral PLAG supplements and the control group (n=22) received oral soybean oil supplements. IL-4 and IL-6 production by peripheral blood mononuclear cells (PBMC) were lower (p<0.001 and p<0.001, respectively) with PLAG than with soybean oil. However, the production of IL-2 and IFN-γ by PBMC was unaltered with PLAG supplementation. The B cell proliferation decreased significantly in the PLAG group compared to the soybean oil control (p<0.05). The intake of PLAG in healthy adults for 4 weeks was deemed safe. These data suggest that PLAG has an immunomodulatory function that inhibits the excessive immune activity of immunological disorders such as atopic and autoimmune diseases. PLAG could improve the condition of these diseases safely as a health food supplement.

13.
Immune Netw ; 15(2): 100-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25922599

RESUMEN

Controlling balance between T-helper type 1 (Th1) and T-helper type 2 (Th2) plays a pivotal role in maintaining the biological rhythm of Th1/Th2 and circumventing diseases caused by Th1/Th2 imbalance. Interleukin 4 (IL-4) is a Th2-type cytokine and often associated with hypersensitivity-related diseases such as atopic dermatitis and allergies when overexpressed. In this study, we have tried to elucidate the function of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (EC-18) as an essential modulator of Th1/Th2 balance. EC-18 has showed an inhibitory effect on the production of IL-4 in a dose-dependent manner. RT-PCR analysis has proved EC-18 affect the transcription of IL-4. By analyzing the phosphorylation status of Signal transducer and activator of transcription 6 (STAT6), which is a transcriptional activator of IL-4 expression, we discovered that EC-18 induced the decrease of STAT6 activity in several stimulated cell lines, which was also showed in STAT6 reporter analysis. Co-treatment of EC-18 significantly weakened atopy-like phenotypes in mice treated with an allergen. Collectively, our results suggest that EC-18 is a potent Th2 modulating factor by regulating the transcription of IL-4 via STAT6 modulation, and could be developed for immune-modulatory therapeutics.

14.
Planta Med ; 80(7): 561-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24710899

RESUMEN

Puerarin (8-ß-D-glucopyranosyl-7-hydroxy-3-(4-hydroxyphenyl)-4H-1-benzopyran-4-one) is a major pharmacological component of Puerariae Radix, the root of Pueraria lobata. We investigated the effect of puerarin on hepatic cytochrome P450-mediated drug metabolism in rats and humans. The in vitro cytochrome P450 inhibitory effect of puerarin in human and rat liver microsomes was evaluated using the following model cytochrome P450 substrates: phenacetin for CYP1A, diclofenac for CYP2C, dextromethorphan for CYP2D, and testosterone for CYP3A. The in vivo pharmacokinetics of intravenous and oral buspirone, a probe substrate for CYP3A, was studied with single simultaneous intravenous coadministration of puerarin in rats. In the in vitro cytochrome P450 inhibition study, the rate of disappearance of testosterone was significantly reduced in the presence of 10 µM PU, while that of other cytochrome P450 substrates was not significantly affected in both human and rat liver microsomes, suggesting that puerarin inhibits the in vitro hepatic CYP3A-mediated metabolism in the human and rat systems (IC50 = 15.5 ± 3.9 µM). After intravenous administration of buspirone with single simultaneous coadministration of intravenous puerarin at a dose of 10 mg/kg in rats, the total area under the plasma concentration-time curve from time zero to time infinity was increased while time-averaged total body clearance decreased. When buspirone was orally administered in rats with the 10 mg/kg intravenous puerarin coadministration, both total area under the plasma concentration-time curve from time zero to time infinity and the extent of absolute oral bioavailability were significantly increased. Therefore, results of the in vitro microsomal and in vivo pharmacokinetic studies suggest the possible inhibition of hepatic CYP3A-mediated drug metabolism by puerarin administration, potentially leading to metabolism-mediated herb-drug interactions with clinical significance.


Asunto(s)
Buspirona/farmacocinética , Sistema Enzimático del Citocromo P-450/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Interacciones de Hierba-Droga , Isoflavonas/farmacología , Pueraria/química , Administración Intravenosa , Administración Oral , Animales , Área Bajo la Curva , Disponibilidad Biológica , Citocromo P-450 CYP3A/efectos de los fármacos , Citocromo P-450 CYP3A/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/aislamiento & purificación , Humanos , Concentración 50 Inhibidora , Isoflavonas/química , Isoflavonas/aislamiento & purificación , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Raíces de Plantas/química , Ratas , Ratas Sprague-Dawley
15.
Int J Pharm ; 441(1-2): 373-80, 2013 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23174411

RESUMEN

Chitosan oligosaccharide-arachidic acid (CSOAA) conjugate was successfully synthesized and used for the development of self-assembled nanoparticles for doxorubicin (DOX) delivery. The molar substitution of AA on CSO and critical micelle concentration (CMC) of CSOAA were measured. Physicochemical properties of DOX-loaded CSOAA-based nanoparticles, such as particle size, zeta potential and morphology, were also characterized. The DOX-loaded CSOAA-based nanoparticles showed spherical shape with a mean diameter of 130 nm and positive charge. According to the result of in vitro release test, DOX-loaded CSOAA-based nanoparticles exhibited sustained and pH-dependent drug release profiles. The CSOAA showed negligible cytotoxicity in FaDu, human head and neck cancer, cells. Cellular uptake of DOX in FaDu cells was higher in the nanoparticle-treated group compared to the free DOX group. The anti-tumor efficacy of DOX-loaded nanoparticles was also verified in FaDu tumor xenografted mouse model. These results suggested that synthesized amphiphilic CSOAA might be used for the preparation of self-assembled nanoparticles for anti-cancer drug delivery.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Animales , Antibióticos Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Quitosano/química , Preparaciones de Acción Retardada , Doxorrubicina/administración & dosificación , Portadores de Fármacos/química , Ácidos Eicosanoicos/química , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas , Oligosacáridos/química , Tamaño de la Partícula , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Xenobiotica ; 43(4): 355-67, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23020787

RESUMEN

The oral (po) bioavailability of gemifloxacin mesylate in rats and its possible association with efflux transporters was investigated. The apparent permeabilities (Papp) of gemifloxacin across the Caco-2 cell monolayer were 1.20 ± 0.09 × 10(-5) cm/s for apical to basal (absorptive) transport, and 2.13 ± 0.6 × 10(-5) cm/s for basal to apical (secretory) transport for a 5-500 µM concentration range, suggesting the involvement of a carrier-mediated efflux in the secretory transport. The secretory transport in Caco-2 cells was significantly decreased by MRP2 (MK571) and BCRP (Ko143) inhibitors. The secretory transport was distinct in MDCKII/P-gp, MDCKII/MRP2 and MDCKII/BCRP cells, and the affinity was highest for MRP2, followed by BCRP and P-gp. The efflux was significantly decreased by verapamil and Ko143, but not significantly by MK571. The comparative po bioavailability in rats was increased by the preadministration of Ko143 (four-fold), MK571 (two-fold) and verapamil (two-fold). Efflux transporters appeared to significantly limit the bioavailability of gemifloxacin in rats, suggesting their possible contribution to the low bioavailability of the drug in the human (70%).


Asunto(s)
Antibacterianos/metabolismo , Fluoroquinolonas/metabolismo , Absorción Intestinal , Proteínas de Transporte de Membrana/metabolismo , Naftiridinas/metabolismo , Quinolonas/metabolismo , Administración Oral , Animales , Antibacterianos/sangre , Antibacterianos/química , Antibacterianos/farmacocinética , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Perros , Fluoroquinolonas/sangre , Fluoroquinolonas/química , Fluoroquinolonas/farmacocinética , Gemifloxacina , Humanos , Concentración 50 Inhibidora , Cinética , Células de Riñón Canino Madin Darby , Masculino , Naftiridinas/sangre , Naftiridinas/química , Naftiridinas/farmacocinética , Quinolonas/sangre , Quinolonas/química , Quinolonas/farmacocinética , Ratas , Ratas Sprague-Dawley , Verapamilo/farmacología
17.
Pharm Res ; 29(12): 3443-54, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22886625

RESUMEN

PURPOSE: Hyaluronic acid-ceramide (HACE)-based nanoparticles (NPs) were developed for the targeted delivery of doxorubicin (DOX), and their antitumor efficacy for melanoma was evaluated. METHODS: DOX-loaded HACE-based self-assembled NPs were prepared and their physicochemical properties were characterized. The in vitro cytotoxicity of HACE was measured using an MTS-based assay. The cellular uptake efficiency of DOX into mouse melanoma B16F10 cells was assessed by confocal laser scanning microscopy and flow cytometry. Tumor growth and body weight were monitored after the intratumoral and intravenous injection of DOX-loaded NPs into a B16F10 tumor-bearing mouse model. RESULTS: DOX-loaded NPs, with a mean diameter of ~110 nm, a narrow size distribution, and high drug entrapment efficiency, were prepared. A sustained DOX release pattern was shown, and drug release was enhanced at pH 5.5 compared with pH 7.4. The cytotoxicity of HACE to B16F10 cells was negligible. It was assumed that DOX was taken up into the B16F10 cells through receptor-mediated endocytosis. A significant inhibitory effect was observed on tumor growth, without any serious changes in body weight, after the injection of DOX-loaded NPs into the B16F10 tumor-bearing mouse model. CONCLUSIONS: DOX-loaded HACE-based NPs were successfully developed and their antitumor efficacy against B16F10 tumors was demonstrated.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos , Melanoma/tratamiento farmacológico , Nanopartículas/química , Animales , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Ceramidas/química , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapéutico , Femenino , Ácido Hialurónico/química , Melanoma/patología , Ratones , Ratones Endogámicos C57BL
18.
Xenobiotica ; 42(11): 1110-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22747239

RESUMEN

Identifying kinetic determinants of hepatic elimination of drugs would be crucial for better understanding its pharmacokinetics and predicting drug interactions. Present study investigated the kinetics of sinusoidal uptake of docetaxel and its impact on the overall hepatic elimination of docetaxel in rats. The non-renal clearance (CL(NR); hepatic elimination) of docetaxel were significantly reduced by co-administration of intravenous rifampicin, a potent inhibitor of organic anion transporting peptides (OATPs; Oatps), at a dose of 20 mg/kg. Docetaxel uptake into isolated rat hepatocytes was found to be temperature/concentration/energy-dependent, saturable, and reduced by Oatps inhibitors (rifampicin and bromosulfophthalein). Moreover, docetaxel uptake into perfused rat liver was significantly reduced in the presence of 10-µM rifampicin. However, docetaxel metabolism in rat hepatic microsome was not affected by rifampicin at less than 50 µM. Based on the comparison of intrinsic clearances related to hepatic clearance, it can be suggested that sinusoidal uptake could be the rate-determining process in the overall hepatic elimination of docetaxel in rats.


Asunto(s)
Antineoplásicos/farmacocinética , Hígado/metabolismo , Transportadores de Anión Orgánico/metabolismo , Taxoides/farmacocinética , Animales , Docetaxel , Inhibidores Enzimáticos , Hepatocitos/metabolismo , Técnicas In Vitro , Cinética , Masculino , Tasa de Depuración Metabólica , Microsomas Hepáticos/metabolismo , Ratas , Ratas Sprague-Dawley , Rifampin
19.
Int J Pharm ; 434(1-2): 191-8, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22643227

RESUMEN

Phytosphingosine and methyl derivatives are important mediators on cellular processes, and are associated with cell growth and death. The antitumor activity of N,N,N-trimethylphytosphingosine-iodide (TMP) as a novel potent inhibitor of angiogenesis and metastasis was evaluated in B16F10 murine melanoma cells. The results indicated that TMP itself effectively inhibited in vitro cell migration, tube formation, and the expression of angiogenic factors as well as in vivo lung metastasis. However, TMP slightly suppressed in vivo experimental tumor metastasis in its free form and induced side effects including hemolysis and local side effects. Therefore, in an attempt to reduce the toxicity and the undesirable side effects of TMP, a liposomal formulation was prepared and tested for its effectiveness. TMP liposomes retained the effectiveness of TMP in vitro while side effects were reduced, and both in vivo experimental and spontaneous tumor metastasis were significantly suppressed. These results support the conclusion that TMP effectively inhibits in vitro angiogenesis as well as in vivo metastasis, and a liposomal formulation is more efficient delivery system for TMP treatment than solution.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Melanoma Experimental/tratamiento farmacológico , Compuestos de Amonio Cuaternario/farmacología , Esfingosina/análogos & derivados , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/toxicidad , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/toxicidad , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Hemólisis/efectos de los fármacos , Humanos , Liposomas , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Compuestos de Amonio Cuaternario/administración & dosificación , Compuestos de Amonio Cuaternario/toxicidad , Ratas , Esfingosina/administración & dosificación , Esfingosina/farmacología , Esfingosina/toxicidad
20.
Xenobiotica ; 42(7): 603-13, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22233275

RESUMEN

The phenomenon known as multiple-drug resistance, whereby anti-cancer agents are expelled from cancer cells, makes it necessary to develop methods that will reliably increase the accumulation of anti-cancer agents within cancer cells. To accomplish this goal, a new model compound, Val-SN-38, was synthesized by introducing valine to SN-38, an active ingredient of irinotecan. Val-SN-38 improved intracellular accumulation approximately 5-fold in MCF7 cells, compared with SN-38, and rather than changes in membrane permeability, the amino acid transporter ATB(0,+) played a role, whereas the dipeptide transporter PEPT1 did not. Other sodium-dependent amino acid transporters, namely ATA1, ATA2, and ASCT2, were unexpectedly involved in the uptake of Val-SN-38 as well. The efflux of Val-SN-38 by major efflux transporters was variably changed, but not significantly. In summary, the enhanced accumulation of Val-SN-38 in cancer cells was due to augmented uptake via various amino acid transporters. The results of the present study make a compelling argument in favour of a prodrug concept that can improve intracellular accumulation and take advantage of amino acid transporters without significantly inducing multiple-drug resistance.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Antineoplásicos/metabolismo , Camptotecina/análogos & derivados , Valina/metabolismo , Sistema de Transporte de Aminoácidos A/metabolismo , Sistema de Transporte de Aminoácidos ASC/metabolismo , Antineoplásicos/síntesis química , Transporte Biológico , Camptotecina/química , Camptotecina/metabolismo , Ésteres , Células HEK293 , Humanos , Irinotecán , Antígenos de Histocompatibilidad Menor , Profármacos/síntesis química , Profármacos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...