Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Drug Metab Dispos ; 50(3): 214-223, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34937801

RESUMEN

Idasanutlin is a potent inhibitor of the p53-MDM2 interaction that enables reactivation of the p53 pathway, which induces cell cycle arrest and/or apoptosis in tumor cells expressing functional p53. It was investigated for the treatment of solid tumors and several hematologic indications such as relapsed/refractory acute myeloid leukemia, polycythemia vera, or non-Hodgkin lymphoma. For safety reasons, it cannot be given in healthy volunteers for drug-drug interaction (DDI) explorations. This triggered the need for in silico explorations on top of the one available CYP3A clinical DDI study with posaconazole in solid tumor patients. Idasanutlin's clearance is dependent on CYP3A4/2C8 forming its major circulating metabolite M4, with contributions from UGT1A3 and biliary excretion. Idasanutlin and M4 have low permeability, very low clearance, and extremely low unbound fraction in plasma (<0.001), which makes in vitro data showing inhibition on CYP3A4/2C8 enzymes challenging to translate to clinical relevance. Physiologically-based pharmacokinetic models of idasanutlin and M4 have been established to simulate perpetrator and victim DDI scenarios and to evaluate whether further DDI studies in oncology patients are necessary. Modeling indicated that idasanutlin and M4 would show no or weak clinical inhibition of selective CYP3A4/2C8 substrates. Co-administered strong CYP3A and CYP2C8 inhibitors might lead to weak or moderate idasanutlin exposure increases, and the strong inducer rifampicin might cause moderate exposure reduction. As the simulated idasanutlin systemic exposure changes would be within the range of observed intrinsic variability, the target population can take co-medications that are either CYP2C8/3A4 inhibitors or weak/moderate CYP2C8/3A4 inducers without dose adjustment. SIGNIFICANCE STATEMENT: Clinical trials for idasanutlin are restricted to cancer patients, which imposes practical, scientific, and ethical challenges on drug-drug interaction investigations. Furthermore, idasanutlin and its major circulating metabolite have very challenging profiles of absorption, distribution, metabolism and excretion including high protein binding, low permeability and a combination of different elimination pathways each with extremely low clearance. Nonetheless, physiologically-based pharmacokinetic models could be established and applied for drug-drug interaction risk assessment and were especially useful to provide guidance on concomitant medications in patients.


Asunto(s)
Isoenzimas , Leucemia Mieloide Aguda , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Interacciones Farmacológicas , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Modelos Biológicos , Pirrolidinas , Medición de Riesgo , para-Aminobenzoatos
2.
Drug Metab Dispos ; 50(1): 65-75, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34620695

RESUMEN

Small molecules that present complex absorption, distribution, metabolism, and elimination (ADME) properties can be challenging to investigate as potential therapeutics. Acquiring data through standard methods can yield results that are insufficient to describe the in vivo situation, which can affect downstream development decisions. Implementing in vitro-in vivo-in silico strategies throughout the drug development process is effective in identifying and mitigating risks while speeding up their development. Risdiplam (Evrysdi)-an orally bioavailable, small molecule approved by the US Food and Drug Administration and more recently by the European Medicines Agency for the treatment of patients ≥2 months of age with spinal muscular atrophy-is presented here as a case study. Risdiplam is a low-turnover compound whose metabolism is mediated through a non-cytochrome P450 enzymatic pathway. Four main challenges of risdiplam are discussed: predicting in vivo hepatic clearance, determining in vitro metabolites with regard to metabolites in safety testing guidelines, elucidating enzymes responsible for clearance, and estimating potential drug-drug interactions. A combination of in vitro and in vivo results was successfully extrapolated and used to develop a robust physiologically based pharmacokinetic model of risdiplam. These results were verified through early clinical studies, further strengthening the understanding of the ADME properties of risdiplam in humans. These approaches can be applied to other compounds with similar ADME profiles, which may be difficult to investigate using standard methods. SIGNIFICANCE STATEMENT: Risdiplam is the first approved, small-molecule, survival of motor neuron 2 mRNA splicing modifier for the treatment of spinal muscular atrophy. The approach taken to characterize the absorption, distribution, metabolism, and excretion (ADME) properties of risdiplam during clinical development incorporated in vitro-in vivo-in silico techniques, which may be applicable to other small molecules with challenging ADME. These strategies may be useful in improving the speed at which future drug molecules can be developed.


Asunto(s)
Compuestos Azo/metabolismo , Compuestos Azo/farmacocinética , Preparaciones Farmacéuticas/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacocinética , Empalme del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Distribución Tisular , Animales , Humanos , Técnicas In Vitro , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
3.
Xenobiotica ; 46(8): 667-76, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26586447

RESUMEN

1. Idasanutlin (RG7388) is a potent p53-MDM2 antagonist currently in clinical development for treatment of cancer. The purpose of the present studies was to investigate the cause of marked decrease in plasma exposure after repeated oral administration of RG7388 in monkeys and whether the autoinduction observed in monkeys is relevant to humans. 2. In monkey liver and intestinal microsomes collected after repeated oral administration of RG7388 to monkeys, significantly increased activities of homologue CYP3A8 were observed (ex vivo). Investigation using a physiologically based pharmacokinetic (PBPK) model suggested that the loss of exposure was primarily due to induction of metabolism in the gut of monkeys. 3. Studies in monkey and human primary hepatocytes showed that CYP3A induction by RG7388 only occurred in monkey hepatocytes but not in human hepatocytes, which suggests the observed CYP3A induction is monkey specific. 4. The human PK data obtained from the first cohorts confirmed the lack of relevant induction as predicted by the human hepatocytes and the PBPK modelling based on no induction in humans.


Asunto(s)
Antineoplásicos/farmacología , Macaca fascicularis/fisiología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Pirrolidinas/farmacología , para-Aminobenzoatos/farmacología , Animales , Antineoplásicos/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Pirrolidinas/metabolismo , para-Aminobenzoatos/metabolismo
4.
J Pharmacol Exp Ther ; 352(2): 358-67, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25503386

RESUMEN

RO5263397 [(S)-4-(3-fluoro-2-methyl-phenyl)-4,5-dihydro-oxazol-2-ylamine], a new compound that showed promising results in animal models of schizophrenia, is mainly metabolized in humans by N-glucuronidation. Enzyme studies, using the (then) available commercial uridine 5'-diphosphate-glucuronosyltransferases (UGTs), suggested that UGT1A4 is responsible for its conjugation. In the first clinical trial, in which RO5263397 was administered orally to healthy human volunteers, a 136-fold above-average systemic exposure to the parent compound was found in one of the participants. Further administration in this trial identified two more such poor metabolizers, all three of African origin. Additional in vitro studies with recombinant UGTs showed that the contribution of UGT2B10 to RO5263397 glucuronidation is much higher than UGT1A4 at clinically relevant concentrations. DNA sequencing in all of these poor metabolizers identified a previously uncharacterized splice site mutation that prevents assembly of full-length UGT2B10 mRNA and thus functional UGT2B10 protein expression. Further DNA database analyses revealed the UGT2B10 splice site mutation to be highly frequent in individuals of African origin (45%), moderately frequent in Asians (8%) and almost unrepresented in Caucasians (<1%). A prospective study using hepatocytes from 20 individual African donors demonstrated a >100-fold lower intrinsic clearance of RO5263397 in cells homozygous for the splice site variant allele. Our results highlight the need to include UGT2B10 when screening the human UGTs for the enzymes involved in the glucuronidation of a new compound, particularly when there is a possibility of N-glucuronidation. Moreover, this study demonstrates the importance of considering different ethnicities during drug development.


Asunto(s)
Población Negra/genética , Silenciador del Gen , Glucuronosiltransferasa/genética , Oxazoles/farmacocinética , Polimorfismo de Nucleótido Simple , Células Cultivadas , Cromatografía Líquida de Alta Presión , Bases de Datos de Ácidos Nucleicos , Glucurónidos/metabolismo , Humanos , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Oxazoles/administración & dosificación , Especificidad por Sustrato , Espectrometría de Masas en Tándem
5.
J Biol Chem ; 286(22): 19501-10, 2011 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-21460230

RESUMEN

Insulin-like growth factor I (IGF-I) has important anabolic and homeostatic functions in tissues like skeletal muscle, and a decline in circulating levels is linked with catabolic conditions. Whereas IGF-I therapies for musculoskeletal disorders have been postulated, dosing issues and disruptions of the homeostasis have so far precluded clinical application. We have developed a novel IGF-I variant by site-specific addition of polyethylene glycol (PEG) to lysine 68 (PEG-IGF-I). In vitro, this modification decreased the affinity for the IGF-I and insulin receptors, presumably through decreased association rates, and slowed down the association to IGF-I-binding proteins, selectively limiting fast but maintaining sustained anabolic activity. Desirable in vivo effects of PEG-IGF-I included increased half-life and recruitment of IGF-binding proteins, thereby reducing risk of hypoglycemia. PEG-IGF-I was equipotent to IGF-I in ameliorating contraction-induced muscle injury in vivo without affecting muscle metabolism as IGF-I did. The data provide an important step in understanding the differences of IGF-I and insulin receptor contribution to the in vivo activity of IGF-I. In addition, PEG-IGF-I presents an innovative concept for IGF-I therapy in diseases with indicated muscle dysfunction.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/farmacocinética , Músculo Esquelético/metabolismo , Enfermedades Musculoesqueléticas/tratamiento farmacológico , Polietilenglicoles/farmacocinética , Receptor de Insulina/agonistas , Animales , Línea Celular , Perros , Semivida , Humanos , Hipoglucemia/inducido químicamente , Hipoglucemia/metabolismo , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/química , Factor I del Crecimiento Similar a la Insulina/farmacología , Músculo Esquelético/patología , Enfermedades Musculoesqueléticas/metabolismo , Enfermedades Musculoesqueléticas/patología , Polietilenglicoles/química , Polietilenglicoles/farmacología , Receptor de Insulina/metabolismo
6.
Mol Cancer Ther ; 20(11): 2177-2188, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34433660

RESUMEN

BRAF-targeted therapies including vemurafenib (Zelboraf) induce dramatic cancer remission; however, drug resistance commonly emerges. The purpose was to characterize a naturally occurring canine cancer model harboring complex features of human cancer, to complement experimental models to improve BRAF-targeted therapy. A phase I/II clinical trial of vemurafenib was performed in pet dogs with naturally occurring invasive urothelial carcinoma (InvUC) harboring the canine homologue of human BRAF V600E The safety, MTD, pharmacokinetics, and antitumor activity were determined. Changes in signaling and immune gene expression were assessed by RNA sequencing and phosphoproteomic analyses of cystoscopic biopsies obtained before and during treatment, and at progression. The vemurafenib MTD was 37.5 mg/kg twice daily. Anorexia was the most common adverse event. At the MTD, partial remission occurred in 9 of 24 dogs (38%), with a median progression-free interval of 181 days (range, 53-608 days). In 18% of the dogs, new cutaneous squamous cell carcinoma and papillomas occurred, a known pharmacodynamic effect of vemurafenib in humans. Upregulation of genes in the classical and alternative MAPK-related pathways occurred in subsets of dogs at cancer progression. The most consistent transcriptomic changes were the increase in patterns of T lymphocyte infiltration during the first month of vemurafenib, and of immune failure accompanying cancer progression. In conclusion, the safety, antitumor activity, and cutaneous pharmacodynamic effects of vemurafenib, and the development of drug resistance in dogs closely mimic those reported in humans. This suggests BRAF-mutated canine InvUC offers an important complementary animal model to improve BRAF-targeted therapies in humans.


Asunto(s)
Carcinoma de Células Transicionales/tratamiento farmacológico , Proteínas Proto-Oncogénicas B-raf/efectos de los fármacos , Vemurafenib/uso terapéutico , Adolescente , Animales , Carcinoma de Células Transicionales/patología , Niño , Modelos Animales de Enfermedad , Perros , Humanos , Mutación , Vemurafenib/farmacología
7.
J Clin Pharmacol ; 47(1): 64-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17192503

RESUMEN

A bimodal extended-release formulation of d-methylphenidate (d-MPH) has been developed to enable fast onset of action and once-daily administration in patients with attention deficit hyperactivity disorder. The authors studied the dose proportionality of extended-release d-MPH pharmacokinetics. Twenty-five healthy adult volunteers received 5, 10, 20, 30, and 40 mg d-MPH in a crossover study with 7 days between doses. All doses were well tolerated. Dose proportionality was shown for all dose-dependent pharmacokinetic parameters. Geometric means (%gCV) for the first Cmax peak, Cmax0-4, were 3.25 (29.0%), 6.05 (27.1%), 12.6 (31.9%), 18.5 (31.9%), and 25.2 ng/mL (29.3%) for d-MPH 5, 10, 20, 30, and 40 mg, respectively. Geometric means (%gCV) for Cmax4-10 were 3.18 (27.5%), 5.84 (27.7%), 12.5 (31.7%), 17.7 (31.6%), and 23.6 ng/mL (29.0%), respectively. Geometric means for AUC(0-infinity) were 24.3 (30.7%), 45.9 (30.2%), 96.4 (35.5%), 144 (33.3%), and 195 ng x h/mL (30.9%), respectively. The pharmacokinetics of once-daily extended-release d-MPH are proportional to the dose.


Asunto(s)
Dopaminérgicos/farmacocinética , Metilfenidato/farmacocinética , Administración Oral , Adulto , Área Bajo la Curva , Cromatografía Líquida de Alta Presión , Estudios Cruzados , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/farmacocinética , Dopaminérgicos/administración & dosificación , Dopaminérgicos/sangre , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Femenino , Humanos , Masculino , Metilfenidato/administración & dosificación , Metilfenidato/sangre , Estereoisomerismo , Espectrometría de Masas en Tándem , Factores de Tiempo
8.
J Med Chem ; 59(13): 6086-100, 2016 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-27299419

RESUMEN

Spinal muscular atrophy (SMA) is the leading genetic cause of infant and toddler mortality, and there is currently no approved therapy available. SMA is caused by mutation or deletion of the survival motor neuron 1 (SMN1) gene. These mutations or deletions result in low levels of functional SMN protein. SMN2, a paralogous gene to SMN1, undergoes alternative splicing and exclusion of exon 7, producing an unstable, truncated SMNΔ7 protein. Herein, we report the identification of a pyridopyrimidinone series of small molecules that modify the alternative splicing of SMN2, increasing the production of full-length SMN2 mRNA. Upon oral administration of our small molecules, the levels of full-length SMN protein were restored in two mouse models of SMA. In-depth lead optimization in the pyridopyrimidinone series culminated in the selection of compound 3 (RG7800), the first small molecule SMN2 splicing modifier to enter human clinical trials.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Atrofia Muscular Espinal/tratamiento farmacológico , Pirimidinonas/química , Pirimidinonas/farmacología , ARN Mensajero/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Animales , Exones/efectos de los fármacos , Humanos , Ratones , Atrofia Muscular Espinal/genética , Pirimidinonas/farmacocinética , Pirimidinonas/uso terapéutico
9.
J Pharm Biomed Anal ; 114: 296-304, 2015 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-26093509

RESUMEN

Targeted immunocytokines (TICs) display potent activity in selective tumor suppression. This class of multi domain biotherapeutics (MDBs) is composed of the three major domains Fab, Fc, and a cytokine which may induce a complex polyclonal anti-drug antibody (ADA) response. However, classical ADA assays usually are not suitable to specify ADAs and to identify the immunogenic domains of a TIC. The purpose of the present study was to establish epitope characterization of ADA responses in order to specify immunogenic responses against a TIC and their direct impact on the pharmacokinetic profile, safety, and efficacy. Based on standard ADA screening and confirmation assays, respectively, domain detection assays (DDAs) and domain competition assays (DCAs) were established and compared by the use of 12 ADA-positive samples obtained from a cynomolgus monkey study in early development. Both domain-specific assays were sensitive enough to preserve the positive screening assay result and revealed an overall accordance for the evaluation of domain-specific ADA responses. About half of the samples displayed one ADA specificity, either for the Fab or for the cytokine (Cy) domain, and the remaining samples showed a combination of Fab-specific and Cy-specific ADA fractions. Fc-specific ADAs occurred in only one sample. In-depth comparison of DCAs and DDAs showed that both assays appeared to be appropriate to assess multi-specific ADA responses as well as minor ADA fractions. An advantage of DCAs is typically a fast and easy assay establishment, whereas, DDAs in some cases may be superior to assess low abundant ADAs in multi-specific responses. Our results reveal that both approaches benefit from thorough reagent development as an essential precondition for reliable epitope characterization of ADA responses.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Citocinas/inmunología , Epítopos/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Formación de Anticuerpos/inmunología , Unión Competitiva , Ensayo de Inmunoadsorción Enzimática/métodos , Epítopos/química , Femenino , Humanos , Inmunoglobulina G/inmunología , Macaca fascicularis , Masculino , Estructura Terciaria de Proteína , Reproducibilidad de los Resultados
10.
J Clin Pharmacol ; 44(8): 866-72, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15286090

RESUMEN

The pharmacokinetics of a meloxicam suspension were studied in 18 children with juvenile rheumatoid arthritis. Children received a single 0.25-mg/kg dose up to a maximum of 15 mg. Pharmacokinetic parameters after the first dose were calculated by noncompartmental methods. Geometric mean (percent coefficient of variation for geometric mean [gCV]) C(max), AUC(0- infinity ), apparent clearance, apparent volume of distribution, and elimination half-life values were 1.24 microg/mL (47% gCV), 25.6 microg x h/mL (81% gCV), 0.17 mL/min/kg (83% gCV), 0.19 L/kg (63% gCV), and 13.4 hours (54% gCV) in the younger group and 1.89 microg/mL (25% gCV), 35.8 microg x h/mL (21% gCV), 0.12 mL/min/kg (23% gCV), 0.13 L/kg (22% gCV), and 12.7 hours (21% gCV) for the older group, respectively. Area under the curve, volume of distribution, and clearance tended to be higher in the younger group, whereas elimination half-lives were similar. A post hoc comparison to pharmacokinetic data in adults revealed no relevant differences. Thus, a common body weight-normalized dose is considered appropriate for children older than 2 years.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Artritis Juvenil/tratamiento farmacológico , Inhibidores de la Ciclooxigenasa/farmacocinética , Tiazinas/farmacocinética , Tiazoles/farmacocinética , Administración Oral , Adolescente , Factores de Edad , Antiinflamatorios no Esteroideos/sangre , Antiinflamatorios no Esteroideos/uso terapéutico , Área Bajo la Curva , Niño , Preescolar , Inhibidores de la Ciclooxigenasa/sangre , Inhibidores de la Ciclooxigenasa/uso terapéutico , Femenino , Alemania , Semivida , Humanos , Masculino , Meloxicam , México , Tiazinas/sangre , Tiazinas/uso terapéutico , Tiazoles/sangre , Tiazoles/uso terapéutico
11.
Drug Discov Today ; 19(10): 1623-31, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24929223

RESUMEN

The pharmacokinetics (PK), metabolism and biodistribution of polyethylene glycol (PEG) in PEGylated proteins are important to understand the increased cellular vacuolation reported in various tissues in animals. The tissue distribution profile of PEGylated proteins and 'metabolic' PEG is guided largely by absolute PEG load, PEG molecular weight and, where applicable, receptor-mediated uptake via the protein moiety. High molecular weight PEGs show slow renal clearance, and consequently have a greater potential to accumulate within cells. The intracellular nonbiodegradable PEG can accumulate within the lysosome ultimately causing distension and vacuolation observed by standard histological examinations. Improved bioanalytical methodologies will contribute to the identification of specific PK parameters including distribution behavior to support development of PEGylated proteins as therapeutics.


Asunto(s)
Polietilenglicoles/farmacocinética , Proteínas/farmacocinética , Animales , Humanos , Polietilenglicoles/química , Proteínas/química , Distribución Tisular
12.
Thromb Res ; 131(5): 401-10, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23522853

RESUMEN

BACKGROUND AND OBJECTIVE: Elevated levels of platelet-leukocyte aggregates (PLAs) have been reported in several cardiovascular diseases and suggested to contribute to disease pathology. Our aim was to characterize the effects of inclacumab, a novel human anti-P-selectin antibody, on the interactions between leukocytes and platelets in preclinical and clinical studies. EXPERIMENTAL APPROACHES: Dual-label flow cytometry was used to detect the effect of inclacumab on agonist-induced platelet-leukocyte/platelet-monocyte aggregates in cynomolgus monkeys and humans, following ex vivo and in vivo administration. Platelet-dependent leukocyte activation and leukocyte adhesion to a platelet monolayer were also investigated after ex vivo administration of inclacumab to human blood. RESULTS: Treatment of cynomolgus monkeys with inclacumab profoundly inhibited thrombin receptor-activating peptide (TRAP) or adenosine diphosphate (ADP)-induced PLAs with an IC50 (<2 µg/mL) similar to the in vitro spiking experiments. Maximal inhibition of PLAs persisted for ≥28 days following single dose of inclacumab. In human blood, inclacumab was about 2-fold more potent in inhibiting TRAP-induced PLAs (IC50: 0.7 µg/mL) compared to monkeys. PLA formation was suppressed independently of the inducing platelet agonist. Inclacumab also inhibited the activation of the leukocyte integrin Mac-1 and leukocyte adhesion to a platelet monolayer under flow conditions. In clinical studies, inclacumab inhibited TRAP-induced PLA formation in a dose-dependent manner following single and multiple dose administration to healthy volunteers. It also reduced elevated circulating PLA levels in patients with peripheral arterial disease. CONCLUSION: By inhibiting platelet-leukocyte interactions, demonstrated in multiple preclinical and clinical studies, inclacumab may provide an effective treatment for cardiovascular diseases.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Plaquetas/citología , Plaquetas/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Leucocitos/citología , Leucocitos/efectos de los fármacos , Selectina-P/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Animales , Anticuerpos Monoclonales/inmunología , Plaquetas/inmunología , Comunicación Celular/inmunología , Femenino , Humanos , Leucocitos/inmunología , Macaca fascicularis , Masculino , Persona de Mediana Edad , Selectina-P/inmunología , Activación Plaquetaria , Resultado del Tratamiento , Adulto Joven
13.
Science ; 325(5939): 490-3, 2009 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-19541954

RESUMEN

Most antianxiety drugs (anxiolytics) work by modulating neurotransmitters in the brain. Benzodiazepines are fast and effective anxiolytic drugs; however, their long-term use is limited by the development of tolerance and withdrawal symptoms. Ligands of the translocator protein [18 kilodaltons (kD)] may promote the synthesis of endogenous neurosteroids, which also exert anxiolytic effects in animal models. Here, we found that the translocator protein (18 kD) ligand XBD173 enhanced gamma-aminobutyric acid-mediated neurotransmission and counteracted induced panic attacks in rodents in the absence of sedation and tolerance development. XBD173 also exerted antipanic activity in humans and, in contrast to benzodiazepines, did not cause sedation or withdrawal symptoms. Thus, translocator protein (18 kD) ligands are promising candidates for fast-acting anxiolytic drugs with less severe side effects than benzodiazepines.


Asunto(s)
Ansiolíticos/metabolismo , Purinas/uso terapéutico , Receptores de GABA/metabolismo , Adulto , Alprazolam/farmacología , Animales , Ansiolíticos/efectos adversos , Benzodiazepinas/efectos adversos , Línea Celular , Tolerancia a Medicamentos , Humanos , Isoquinolinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neurotransmisores/metabolismo , Trastorno de Pánico/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/metabolismo , Síndrome de Abstinencia a Sustancias/prevención & control , Tetragastrina , Ácido gamma-Aminobutírico/metabolismo
14.
J Rheumatol ; 29(5): 1079-83, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12022326

RESUMEN

OBJECTIVE: Use of meloxicam as a selective COX-2 inhibitor for treatment of adult rheumatic diseases decreases the frequency of gastrointestinal (GI) side effects in comparison with nonselective COX inhibitors. Up to 50% of children with juvenile rheumatoid arthritis (JRA) also develop GI side effects through nonselective COX inhibitors. In this 12 week Phase I/II study, with an additional open extension lasting up to 52 weeks, the safety, efficacy, and pharmacokinetics of meloxicam in JRA were investigated. METHODS: Meloxicam suspension 0.25 mg/kg once daily was given to 36 patients with JRA who required a nonsteroidal antiinflammatory drug. Safety evaluation and periodic measurement of efficacy were carried out using the Pediatric Rheumatology International Trials Organisation (PRINTO) criteria. Eighteen patients underwent pharmacokinetic (PK) evaluation. RESULTS: Thirty-one patients completed the study. Four were dropped due to administrative reasons. One patient, who found the drug ineffective, discontinued participation. A response was seen according to PRINTO outcome criteria in 44% of the patients at Week 4, 62% at Week 12, and 74% at Week 52. Drug related adverse events were observed in 5 patients. PK evaluation showed that the maximum plasma concentration Cmax of -34% and AUC(0-infinity) of -28% tended to be lower in younger children (2-6 years) versus older children. Plasma elimination half-life (13 h) was similar in all patients. CONCLUSION: Meloxicam suspension 0.25 mg/kg once daily seems to be effective and safe for treating active JRA over a period of 52 weeks.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Artritis Juvenil/tratamiento farmacológico , Tiazinas/administración & dosificación , Tiazoles/administración & dosificación , Adolescente , Antiinflamatorios no Esteroideos/efectos adversos , Antiinflamatorios no Esteroideos/farmacocinética , Niño , Preescolar , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/administración & dosificación , Quimioterapia Combinada , Femenino , Humanos , Isoenzimas/antagonistas & inhibidores , Masculino , Meloxicam , Proteínas de la Membrana , Satisfacción del Paciente , Prostaglandina-Endoperóxido Sintasas , Tiazinas/efectos adversos , Tiazinas/farmacocinética , Tiazoles/efectos adversos , Tiazoles/farmacocinética , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA