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

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Clin Psychopharmacol ; 38(5): 435-441, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30015676

RESUMO

BACKGROUND: Aripiprazole lauroxil (AL), a long-acting injectable antipsychotic for the treatment of schizophrenia, requires 21 days of oral aripiprazole supplementation upon initiation. We report findings from a phase 1 study investigating a nanocrystalline milled dispersion of AL (ALNCD) as a potential 1-day initiation regimen. The 1-day initiation regimen is designed to enable rapid achievement of plasma aripiprazole concentrations that are comparable with the 21-day oral initiation regimen. Here, a 6-month pharmacokinetic study compared 2 different initiation regimens for starting AL. METHODS: Patients were randomized 1:1:1:1 to receive 1 of 4 treatments consisting of the 1-day (single ALNCD injection + one 30-mg dose of oral aripiprazole on day 1 only) or the 21-day (15-mg daily dose of oral aripiprazole for 21 days) initiation regimen, each combined with a starting AL dose of either 441 mg or 882 mg. RESULTS: In total, 133/161 patients completed the study. The pharmacokinetic profile of the 1-day initiation regimen was comparable to the 21-day initiation regimen; both achieved aripiprazole concentrations in the therapeutic range within 4 days and remained in a comparable concentration range during treatment initiation. Common adverse events (≥5.0%) were injection-site pain, headache, increased weight, insomnia, dyspepsia, and anxiety. Nine akathisia events occurred (4 events in 4 patients and 5 events in 2 patients in the 1-day and 21-day initiation regimen groups, respectively). CONCLUSIONS: The 1-day initiation regimen resulted in plasma aripiprazole concentrations consistent with the 21-day initiation regimen. Therefore, a single dose of ALNCD with a single 30-mg oral dose of aripiprazole provides an alternative initiation regimen for starting AL.


Assuntos
Antipsicóticos/administração & dosagem , Antipsicóticos/farmacocinética , Aripiprazol/administração & dosagem , Aripiprazol/farmacocinética , Esquizofrenia/sangue , Esquizofrenia/tratamento farmacológico , Administração Oral , Adulto , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/farmacocinética , Método Duplo-Cego , Esquema de Medicação , Feminino , Humanos , Injeções Intramusculares , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
2.
J Clin Psychopharmacol ; 37(3): 289-295, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28350572

RESUMO

BACKGROUND: Aripiprazole lauroxil is an extended-release prodrug of aripiprazole for intramuscular injection, approved for schizophrenia treatment. We developed a population pharmacokinetic (PopPK) model to characterize aripiprazole lauroxil PK and evaluate dosing scenarios likely to be encountered in clinical practice. METHODS: Data from 616 patients with schizophrenia, collected from 5 clinical studies, were used to construct the PopPK model. The model was subsequently used to evaluate various dose levels and frequency and the impact of dosing delay on aripiprazole concentrations. FINDINGS: The results of the model indicate that aripiprazole is released into the systemic circulation after 5 to 6 days, and release continues for an additional 36 days. The slow increase in aripiprazole concentration after injection necessitates the coadministration of oral aripiprazole for 21 days with the first injection. Based on the PopPK model simulations, a dosing interval of 882 mg every 6 weeks results in aripiprazole concentrations that fall within the concentration range associated with the efficacious aripiprazole lauroxil dose range (441-882 mg dosed monthly). A 662-mg monthly dose also resulted in aripiprazole concentrations within the efficacious dose range. Aripiprazole lauroxil administration results in prolonged exposure, such that dose delays of 2 to 4 weeks, depending on the dose regimen, do not require oral aripiprazole supplementation upon resumption of dosing. CONCLUSIONS: This PopPK model and model-based simulations were effective means for evaluating aripiprazole lauroxil dosing regimens and management of missed doses. Such analyses play an important role in determining the use of this long-acting antipsychotic in clinical practice.


Assuntos
Antipsicóticos/farmacocinética , Aripiprazol/farmacocinética , Esquizofrenia/tratamento farmacológico , Antipsicóticos/administração & dosagem , Aripiprazol/administração & dosagem , Ensaios Clínicos como Assunto , Preparações de Ação Retardada , Humanos , Injeções Intramusculares , Modelos Teóricos
3.
J Cardiovasc Pharmacol ; 64(2): 164-71, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24691275

RESUMO

Mipomersen is a second-generation antisense oligonucleotide indicated as an adjunct therapy for homozygous familial hypercholesterolemia (HoFH). Warfarin is commonly prescribed for a variety of cardiac disorders in homozygous familial hypercholesterolemia population, and concurrent use of warfarin and mipomersen is likely. This open-label, single-sequence 2-period phase 1 study in healthy subjects evaluated the potential drug-drug interactions between mipomersen and warfarin. The subjects received a single oral 25 mg dose of warfarin alone on day 1, and after a 7-day washout period, received 200 mg mipomersen alone subcutaneously every other day on days 8-12, and received both concurrently on day 14. Coadministration of mipomersen did not change the pharmacodynamics (international normalized ratio, prothrombin time, and activated partial thromboplastin time) and pharmacokinetics (PK) of warfarin. There were no clinically significant changes in the PK of mipomersen with concurrent administration of warfarin. There were no events indicative of an increase in bleeding tendency when warfarin was coadministered with mipomersen, and the adverse event profile of mipomersen did not appear to be altered in combination with warfarin, as compared with that of the respective reference treatment. The combination of these 2 medications appeared to be safe and well tolerated. These results suggest that the dosage adjustment of warfarin or mipomersen is not expected to be necessary with coadministration.


Assuntos
Oligodesoxirribonucleotídeos Antissenso/farmacologia , Oligodesoxirribonucleotídeos Antissenso/farmacocinética , Oligonucleotídeos/farmacologia , Oligonucleotídeos/farmacocinética , Varfarina/farmacologia , Varfarina/farmacocinética , Adolescente , Adulto , Interações Medicamentosas , Feminino , Voluntários Saudáveis , Humanos , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Coeficiente Internacional Normatizado , Masculino , Pessoa de Meia-Idade , Oligodesoxirribonucleotídeos Antissenso/administração & dosagem , Oligodesoxirribonucleotídeos Antissenso/efeitos adversos , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos/efeitos adversos , Tempo de Tromboplastina Parcial , Tempo de Protrombina , Varfarina/administração & dosagem , Varfarina/efeitos adversos , Adulto Jovem
4.
Int J Clin Pharmacol Ther ; 52(4): 314-20, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24548981

RESUMO

OBJECTIVES: To characterize the safety, tolerability, pharmacokinetics (PK) and dose proportionality of mipomersen after single subcutaneous (SC) administration to Japanese healthy subjects; and to compare the PK profiles of Japanese and Western subjects. METHODS: 20 healthy first-generation Japanese male subjects were enrolled into one of three treatment cohorts (50, 100 and 200 mg SC) in a dose-escalation design. Within each cohort, subjects were randomized in a 4 : 1 ratio to receive mipomersen or placebo. RESULTS: Mipomersen was absorbed rapidly after SC administration; median tmax varied between 2 and 3 hours. After reaching peak levels, plasma concentrations of mipomersen decayed multiphasically with an initial distribution t1/2 in several hours and a terminal t1/2 of 261 - 393 hours. Mean Cmax increased in a dose-linear manner while all mean AUC from time 0 to different cut points increased slightly more than dose proportionally. Although mean terminal t1/2 varied in the dose range tested, it did not show dose-dependence. The PK profiles of mipomersen in Japanese subjects are similar to those observed in Western subjects. A single SC dose of 50 mg, 100 mg and 200 mg mipomersen was well tolerated by male Japanese subjects. CONCLUSION: Single SC doses of 50 - 200 mg were safe and well tolerated when administered to Japanese subjects. Comparison of PK between Japanese and Western subjects does not support any need for dose adjustment in Japanese population in future clinical development.


Assuntos
Oligonucleotídeos/farmacocinética , Adulto , Área Sob a Curva , Povo Asiático , Método Duplo-Cego , Voluntários Saudáveis , Humanos , Masculino , Oligonucleotídeos/efeitos adversos , Estudos Prospectivos
5.
Kidney Int ; 79(11): 1236-43, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21368745

RESUMO

Primary focal segmental glomerulosclerosis (FSGS) is a disease with poor prognosis and high unmet therapeutic need. Here, we evaluated the safety and pharmacokinetics of single-dose infusions of fresolimumab, a human monoclonal antibody that inactivates all forms of transforming growth factor-ß (TGF-ß), in a phase I open-label, dose-ranging study. Patients with biopsy-confirmed, treatment-resistant, primary FSGS with a minimum estimated glomerular filtration rate (eGFR) of 25 ml/min per 1.73 m(2), and a urine protein to creatinine ratio over 1.8 mg/mg were eligible. All 16 patients completed the study in which each received one of four single-dose levels of fresolimumab (up to 4 mg/kg) and was followed for 112 days. Fresolimumab was well tolerated with pustular rash the only adverse event in two patients. One patient was diagnosed with a histologically confirmed primitive neuroectodermal tumor 2 years after fresolimumab treatment. Consistent with treatment-resistant FSGS, there was a slight decline in eGFR (median decline baseline to final of 5.85 ml/min per 1.73 m(2)). Proteinuria fluctuated during the study with the median decline from baseline to final in urine protein to creatinine ratio of 1.2 mg/mg with all three Black patients having a mean decline of 3.6 mg/mg. The half-life of fresolimumab was ∼14 days, and the mean dose-normalized Cmax and area under the curve were independent of dose. Thus, single-dose fresolimumab was well tolerated in patients with primary resistant FSGS. Additional evaluation in a larger dose-ranging study is necessary.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Glomerulosclerose Segmentar e Focal/tratamento farmacológico , Rim/efeitos dos fármacos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Adulto , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Biomarcadores/urina , Biópsia , Creatinina/urina , Relação Dose-Resposta a Droga , Europa (Continente) , Feminino , Taxa de Filtração Glomerular/efeitos dos fármacos , Glomerulosclerose Segmentar e Focal/imunologia , Glomerulosclerose Segmentar e Focal/patologia , Glomerulosclerose Segmentar e Focal/fisiopatologia , Humanos , Infusões Parenterais , Rim/imunologia , Rim/patologia , Rim/fisiopatologia , Masculino , Pessoa de Meia-Idade , Proteinúria/tratamento farmacológico , Proteinúria/imunologia , Fator de Crescimento Transformador beta/imunologia , Resultado do Tratamento , Estados Unidos , Adulto Jovem
6.
Clin Pharmacol Drug Dev ; 10(8): 824-839, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34107166

RESUMO

Napabucasin is an orally administered reactive oxygen species generator that is bioactivated by the intracellular antioxidant nicotinamide adenine dinucleotide phosphate:quinone oxidoreductase 1. Napabucasin induces cell death in cancer cells, including cancer stem cells. This phase 1 study (NCT03411122) evaluated napabucasin drug-drug interaction potential for 7 cytochrome P450 (CYP) enzymes and the breast cancer resistance protein transporter/organic anion transporter 3. Healthy volunteers who tolerated napabucasin during period 1 received probe drugs during period 2, and in period 3 received napabucasin (240 mg twice daily; days 1-11) plus a phenotyping cocktail containing omeprazole (CYP2C19), caffeine (CYP1A2), flurbiprofen (CYP2C9), bupropion (CYP2B6), dextromethorphan (CYP2D6), midazolam (CYP3A) (all oral; day 6), intravenous midazolam (day 7), repaglinide (CYP2C8; day 8), and rosuvastatin (breast cancer resistance protein/organic anion transporter 3; day 9). Drug-drug interaction potential was evaluated in 17 of 30 enrolled volunteers. Napabucasin coadministration increased the area under the plasma concentration-time curve from time 0 extrapolated to infinity (geometric mean ratio [90% confidence interval]) of caffeine (124% [109.0%-141.4%]), intravenous midazolam (118% [94.4%-147.3%]), repaglinide (127% [104.7%-153.3%]), and rosuvastatin (213% [42.5%-1068.3%]) and decreased the area under the plasma concentration-time curve from time 0 extrapolated to infinity of dextromethorphan (71% [47.1%-108.3%]), bupropion (79% [64.6%-97.0%]), and hydroxybupropion (45% [15.7%-129.6%]). No serious adverse events/deaths were reported. Generally, napabucasin is not expected to induce/inhibit drug clearance to a clinically meaningful degree.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Benzofuranos/administração & dosagem , Sistema Enzimático do Citocromo P-450/metabolismo , Naftoquinonas/administração & dosagem , Proteínas de Neoplasias/metabolismo , Administração Oral , Adulto , Benzofuranos/farmacocinética , Bupropiona/administração & dosagem , Bupropiona/farmacocinética , Cafeína/administração & dosagem , Cafeína/farmacocinética , Dextrometorfano/administração & dosagem , Dextrometorfano/farmacocinética , Interações Medicamentosas , Feminino , Flurbiprofeno/administração & dosagem , Flurbiprofeno/farmacocinética , Regulação da Expressão Gênica/efeitos dos fármacos , Meia-Vida , Voluntários Saudáveis , Humanos , Masculino , Midazolam/administração & dosagem , Midazolam/farmacocinética , Naftoquinonas/farmacocinética , Omeprazol/administração & dosagem , Omeprazol/farmacocinética , Rosuvastatina Cálcica/administração & dosagem , Rosuvastatina Cálcica/farmacocinética , Adulto Jovem
7.
Pharmacol Res Perspect ; 9(1): e00722, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33576192

RESUMO

This phase 1, open-label study assessed14 C-napabucasin absorption, metabolism, and excretion, napabucasin pharmacokinetics, and napabucasin metabolites (primary objectives); safety/tolerability were also evaluated. Eight healthy males (18-45 years) received a single oral 240-mg napabucasin dose containing ~100 µCi14 C-napabucasin. Napabucasin was absorbed and metabolized to dihydro-napabucasin (M1; an active metabolite [12.57-fold less activity than napabucasin]), the sole major circulating metabolite (median time to peak concentration: 2.75 and 2.25 h, respectively). M1 plasma concentration versus time profiles generally mirrored napabucasin; similar arithmetic mean half-lives (7.14 and 7.92 h, respectively) suggest M1 formation was rate limiting. Napabucasin systemic exposure (per Cmax and AUC) was higher than M1. The total radioactivity (TRA) whole blood:plasma ratio (AUClast : 0.376; Cmax : 0.525) indicated circulating drug-related compounds were essentially confined to plasma. Mean TRA recovery was 81.1% (feces, 57.2%; urine, 23.8%; expired air, negligible). Unlabeled napabucasin and M1 recovered in urine accounted for 13.9% and 11.0% of the dose (sum similar to urine TRA recovered); apparent renal clearance was 8.24 and 7.98 L/h. No uniquely human or disproportionate metabolite was quantified. Secondary glucuronide and sulfate conjugates were common urinary metabolites, suggesting napabucasin was mainly cleared by reductive metabolism. All subjects experienced mild treatment-emergent adverse events (TEAEs), the majority related to napabucasin. The most commonly reported TEAEs were gastrointestinal disorders. There were no clinically significant laboratory, vital sign, electrocardiogram, or physical examination changes. Napabucasin was absorbed, metabolized to M1 as the sole major circulating metabolite, and primarily excreted via feces. A single oral 240-mg dose was generally well tolerated.


Assuntos
Antineoplásicos/farmacocinética , Benzofuranos/farmacocinética , Naftoquinonas/farmacocinética , Administração Oral , Adulto , Antineoplásicos/efeitos adversos , Antineoplásicos/sangue , Antineoplásicos/urina , Benzofuranos/efeitos adversos , Benzofuranos/sangue , Benzofuranos/urina , Radioisótopos de Carbono , Fezes/química , Humanos , Masculino , Naftoquinonas/efeitos adversos , Naftoquinonas/sangue , Naftoquinonas/urina , Adulto Jovem
8.
Biol Blood Marrow Transplant ; 16(1): 95-101, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19748593

RESUMO

Plerixafor is a selective antagonist of CXCR4 used for mobilization of hematopoietic stem cells (HSCs) for autologous stem cell transplantation (SCT) in patients with multiple myeloma (MM) and non-Hodgkin lymphoma (NHL). This Phase 1 open-label study in healthy subjects was conducted to evaluate the pharmacokinetic characteristics of plerixafor in subjects with renal impairment. All subjects received a single 0.24 mg/kg subcutaneous dose of plerixafor. Subjects were stratified into 4 cohorts based on creatinine clearance determined from a 24-hour urine collection: control (>90 mL/min), mild renal impairment (51-80 mL/min), moderate renal impairment (31-50 mL/min), and severe renal impairment (<31 mL/min, not requiring dialysis). Eleven female subjects (48%) and 12 male subjects (52%), ranging in age from 35 to 73 years, were enrolled. Plerixafor clearance was reduced in subjects with renal impairment and was positively correlated with creatinine clearance. The mean area under the concentration- versus-time curve from time 0 to 24 hours postdose of plerixafor in subjects with mild, moderate, and severe renal impairment was 7%, 32%, and 39% higher, respectively, than that in subjects with normal renal function. Renal impairment had no effect on maximal plasma concentrations. The safety profile was similar among subjects with renal impairment and controls. No renal impairment-related trends in the incidence of adverse events were apparent. A plerixaflor dose reduction to 160 microg/kg in patients with a creatinine clearance value

Assuntos
Compostos Heterocíclicos/farmacocinética , Receptores CXCR4/antagonistas & inibidores , Insuficiência Renal/metabolismo , Adulto , Idoso , Benzilaminas , Estudos de Coortes , Creatinina/metabolismo , Creatinina/urina , Ciclamos , Feminino , Mobilização de Células-Tronco Hematopoéticas , Compostos Heterocíclicos/efeitos adversos , Compostos Heterocíclicos/sangue , Compostos Heterocíclicos/urina , Humanos , Testes de Função Renal , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Insuficiência Renal/sangue , Insuficiência Renal/fisiopatologia , Insuficiência Renal/urina
9.
Ther Adv Psychopharmacol ; 9: 2045125319859964, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31308935

RESUMO

BACKGROUND: Aripiprazole lauroxil (AL), a long-acting injectable antipsychotic for the treatment of schizophrenia in adults, can be started with either 21 days of daily oral aripiprazole supplementation or a 1-day initiation regimen consisting of a single injection of a NanoCrystal® Dispersion formulation of AL (ALNCD) and a single dose of 30 mg oral aripiprazole. This phase I study assessed the pharmacokinetics and safety of deltoid versus gluteal ALNCD injections. METHODS: Patients with schizophrenia or schizoaffective disorder (N = 47) were randomized 1:1 to receive a single intramuscular dose of ALNCD in the deltoid or gluteal muscle. Plasma samples were collected over 85 days to measure ALNCD concentration by injection site. Relative aripiprazole bioavailability for deltoid versus gluteal injection was assessed based on area under the curve (AUC∞ and AUClast) and maximum concentration (Cmax) values. Adverse events were monitored throughout the study. RESULTS: Plasma aripiprazole concentrations after a single ALNCD injection were comparable between deltoid and gluteal administration. Mean maximum plasma aripiprazole concentrations were 196.1 ng/ml (deltoid) and 175.0 ng/ml (gluteal). Exposure to aripiprazole was similar, with mean AUC∞ values of 6591 day × ng/ml for deltoid and 6437 day × ng/ml for gluteal. Aripiprazole bioavailability was not significantly different between injection sites. ALNCD administration in the deltoid or gluteal muscle was well tolerated, with similar safety profiles at both sites. CONCLUSION: ALNCD demonstrated similar exposure and safety profiles between the two administration sites, suggesting that ALNCD can be given in either the gluteal or the deltoid muscles as a component of the 1-day initiation regimen for AL.

10.
Eur J Drug Metab Pharmacokinet ; 43(4): 461-469, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29943125

RESUMO

BACKGROUND AND OBJECTIVES: Aripiprazole lauroxil (AL), a long-acting injectable antipsychotic for the treatment of schizophrenia, requires 21 days of oral aripiprazole supplementation upon initiation (21-day initiation regimen). An alternative 1-day initiation regimen utilizing a nano-crystalline milled dispersion of AL (ALNCD) plus a single 30 mg oral aripiprazole dose achieved aripiprazole concentrations associated with therapeutic doses of aripiprazole in the same time frame as the 21-day initiation regimen when starting AL (441 or 882 mg). A population pharmacokinetic (PopPK) model was developed to describe aripiprazole pharmacokinetics following administration of ALNCD, AL and oral aripiprazole, and evaluate dosing scenarios likely to be encountered in clinical practice. METHODS: In total, 12,768 plasma aripiprazole concentrations from 343 patients (from 4 clinical studies) were included in the PopPK analysis and used to construct the model. RESULTS: Concomitant administration of the 1-day initiation regimen with all approved AL dosing regimens (441, 662, or 882 mg monthly, 882 mg every 6 weeks, or 1064 mg every 2 months) is predicted to achieve aripiprazole concentrations associated with therapeutic doses of AL using the 21-day initiation regimen within 4 days, maintaining these concentrations until the next AL dose. Administration of the first AL injection 10 days after the 1-day initiation regimen resulted in median aripiprazole concentrations just before the second dose of AL ≥ 77% of that when coadministered on the same day. Coadministration of AL with a single ALNCD injection was predicted to be effective in rapidly re-establishing concentrations associated with therapeutic doses of AL following dose delay. CONCLUSIONS: Model-based simulations demonstrate that the 1-day initiation regimen is suitable for starting treatment with all AL doses, allowing a window of ≤ 10 days between initiation and AL administration. ALNCD may also be used to re-establish concentrations associated with therapeutic doses of AL in conjunction with a delayed AL dose.


Assuntos
Aripiprazol/farmacocinética , Simulação por Computador , Modelos Biológicos , Administração Oral , Antipsicóticos/sangue , Antipsicóticos/farmacocinética , Aripiprazol/administração & dosagem , Aripiprazol/sangue , Aripiprazol/química , Preparações de Ação Retardada/farmacocinética , Esquema de Medicação , Humanos , Injeções Intramusculares , Nanopartículas/química
11.
CNS Drugs ; 31(7): 617-624, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28597226

RESUMO

BACKGROUND: Aripiprazole lauroxil (AL) is a long-acting injectable medication approved for the treatment of schizophrenia. Current AL regimens are 441 mg, 662 mg, and 882 mg administered monthly (every 4 weeks [q4wk]), or 882 mg administered every 6 weeks (q6wk). OBJECTIVE: We examined the feasibility of a 2-month (every 8 weeks [q8wk]) dosing interval of AL in a phase I open-label pharmacokinetic study investigating AL 1064 mg administered q8wk for 24 weeks, followed by 20 weeks of safety and pharmacokinetic measurements (ClinicalTrials.gov ID: NCT02320032). Second, a population pharmacokinetic model (referred to as the 2MPopPK model) was generated using data collected from the present trial, as well as data obtained from earlier studies. METHODS: The phase I study included patients with schizophrenia or schizoaffective disorder maintained on an oral antipsychotic (n = 140) who were assigned to one of three groups: AL 441 mg q4wk, AL 882 mg q6wk, or AL 1064 mg q8wk, with a total of seven, five, or four injections administered, respectively. No oral aripiprazole lead-in supplementation was administered and patients continued on maintenance oral antipsychotics. Pharmacokinetic samples were collected at various time points during the 24-week study period and the 20-week follow-up period. Plasma concentrations obtained from the phase I study were analyzed using non-compartmental methods. Additionally, the data were combined with data collected from prior studies to develop the 2MPopPK model. RESULTS: Following the final injection of AL in the phase I study, maximum aripiprazole concentrations were achieved 24.4-35.2 days after the last dose and persisted for the duration of the study. The mean C avg,ss values were 125.8 ng/ml, 131.1 ng/ml, and 140.7 ng/ml for the 441 mg q4wk, 882 mg q6wk, and 1064 mg q8wk doses, respectively. The mean elimination half-life of aripiprazole following the last dose was 53.9 days for the 1064 mg dose, 55.1 days for the 882 mg dose, and 57.2 days for the 441 mg dose. The 2MPopPK dataset included 14,524 aripiprazole concentrations from 700 patients with schizophrenia. The duration of absorption of aripiprazole was estimated as 43 days (95% confidence interval [CI] 42-45 days), which was preceded by a 3.2-day lag time (95% CI 3.0-3.5 days) for a total duration of input into the systemic circulation of 46 days following intramuscular administration of AL. Multiple-dose simulations showed that the 1064 mg q8wk regimen provides aripiprazole concentrations within the concentration range associated with 441 mg and 882 mg q4wk doses previously demonstrated to be efficacious in a phase III study. CONCLUSION: These data from the phase I study and the 2MPopPK model support the suitability of using the AL 1064 mg dose as a 2-month (q8wk) dose interval option for the treatment of schizophrenia.


Assuntos
Antipsicóticos/administração & dosagem , Antipsicóticos/farmacocinética , Aripiprazol/administração & dosagem , Aripiprazol/farmacocinética , Transtornos Psicóticos/tratamento farmacológico , Esquizofrenia/tratamento farmacológico , Administração Oral , Adulto , Antipsicóticos/efeitos adversos , Aripiprazol/efeitos adversos , Esquema de Medicação , Quimioterapia Combinada , Estudos de Viabilidade , Feminino , Seguimentos , Humanos , Injeções , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Transtornos Psicóticos/sangue , Esquizofrenia/sangue
12.
Placenta ; 24(2-3): 149-54, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12566241

RESUMO

BACKGROUND: Significant interindividual variability exists following maternal alcohol consumption; not all children born to alcoholic women manifest the symptoms associated with foetal alcohol spectrum disorder (FASD). OBJECTIVE: To investigate the potential role of the placenta as a source of variability by determining if interindividual variability exists in the binding of acetaldehyde to human placenta. METHODS: Acetaldehyde was added to ten different human placental homogenates and subjected to equilibrium dialysis. Homogenates of placentae obtained from guinea pigs chronically exposed to ethanol throughout gestation were also dialysed in the presence of acetaldehyde to look for alterations in binding after chronic alcohol exposure. Nonlinear least-squares regression analysis was used to characterize the binding system involved. RESULTS: It was found that the amount of acetaldehyde bound to human placentae varied by as much as 3-fold among placentae. The binding profile of acetaldehyde was characterized as a two site binding system (Ka(1)=9.8 x 10(5)+/-0.7 x 10(5)l/mol, N(1)=1.1 x 10(-8)+/-0.7 x 10(-8)mol/g tissue; Ka(2)=1.6 x 10(4)+/-0.9 x 10(4)l/mol, N(2)=1.7 x 10(-7)+/-0.4 x 10(-7)mol/g tissue). Chronic alcohol exposure had no effect on the degree of acetaldehyde binding. CONCLUSION: This previously unidentified source of variability may partially explain why some foetuses are adversely affected by prenatal alcohol exposure while others are not.


Assuntos
Acetaldeído/metabolismo , Placenta/metabolismo , Adulto , Animais , Sítios de Ligação , Diálise , Etanol/farmacologia , Feminino , Cobaias , Humanos , Técnicas In Vitro , Trabalho de Parto , Troca Materno-Fetal/fisiologia , Placenta/efeitos dos fármacos , Gravidez , Prenhez
14.
J Lab Clin Med ; 145(1): 47-54, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15668661

RESUMO

Exposure to alcohol in the embryonic mouse can lead to structural and neurophysiologic changes. The cause of these changes is poorly understood, but they are likely the result of numerous mechanisms. Here we investigate ethanol-induced alterations in gene expression in the fetal brain. Using complementary-DNA microarrays, we identified 25 genes that were down-regulated by prenatal ethanol exposure on days 7 and 9 of gestation. None were found to be up-regulated. Of those that were repressed, 6 (Timp4, Bmp15, Rnf25, Akt1, Tulp4, Dexras1) have been identified, and they are discussed here in the context of the developing fetus. The identified genes have been shown to be involved in cell proliferation, differentiation, and apoptosis, and they contribute to tissue growth and remodeling, as well as neuronal growth and survival. Microarray studies may be useful in the identification of a genetic marker for fetal alcohol syndrome, the discovery of novel pathways that may be involved in its origin, or both.


Assuntos
Depressores do Sistema Nervoso Central/toxicidade , Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/genética , Desenvolvimento Fetal/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Encéfalo/patologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Feminino , Transtornos do Espectro Alcoólico Fetal/etiologia , Transtornos do Espectro Alcoólico Fetal/patologia , Desenvolvimento Fetal/genética , Perfilação da Expressão Gênica , Marcadores Genéticos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA