Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Clin Pharmacol Drug Dev ; 12(8): 832-838, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37145975

RESUMEN

Zanubrutinib is a second-generation Bruton tyrosine kinase inhibitor that is primarily metabolized by CYP3A enzymes. Previous drug-drug interaction (DDI) studies have demonstrated that co-administration of zanubrutinib with rifampin, a strong CYP3A inducer, reduces zanubrutinib plasma concentrations, potentially impacting activity. The impact of the co-administration of zanubrutinib with less potent CYP3A inducers is unclear. This phase 1, open-label, fixed-sequence DDI study evaluated the pharmacokinetics, safety, and tolerability of zanubrutinib when co-administered with steady-state rifabutin, a known CYP3A inducer less potent than rifampin, in 13 healthy male volunteers (NCT04470908). Co-administration of zanubrutinib with rifabutin resulted in a less than 2-fold reduction of zanubrutinib exposures. Overall, zanubrutinib was well tolerated. The results of this study provide useful information for the evaluation of the DDI between rifabutin and zanubrutinib. In conjunction with safety and efficacy data from other clinical studies, these results will be taken into consideration to determine the appropriate dose recommendation of zanubrutinib when co-administered with CYP3A inducers.


Asunto(s)
Inductores del Citocromo P-450 CYP3A , Rifampin , Humanos , Masculino , Inductores del Citocromo P-450 CYP3A/efectos adversos , Inductores del Citocromo P-450 CYP3A/farmacocinética , Rifabutina/efectos adversos , Voluntarios Sanos , Citocromo P-450 CYP3A/metabolismo , Interacciones Farmacológicas
2.
Leuk Lymphoma ; 64(2): 329-338, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36480811

RESUMEN

BTK inhibitor exposure increases significantly when coadministered with CYP3A inhibitors, which may lead to dose-related toxicities. This study explored the pharmacokinetics, efficacy, and safety of zanubrutinib when coadministered with moderate or strong CYP3A inhibitors in 26 patients with relapsed or refractory B-cell malignancies. Coadministration of zanubrutinib (80 mg BID) with moderate CYP3A inhibitors fluconazole and diltiazem or zanubrutinib (80 mg QD) with strong CYP3A inhibitor voriconazole resulted in comparable exposures to zanubrutinib (320 mg QD) with AUC0-24h geometric least squares mean ratios approaching 1 (0.94, 0.81, and 0.83, for fluconazole, diltiazem, and voriconazole, respectively). The most common treatment-emergent adverse events were contusion (26.9%), back pain (19.2%), constipation and neutropenia (15.4% each), and rash, diarrhea, and fall (11.5% each). This study supports current United States Prescribing Information dose recommendations for the coadministration of reduced-dose zanubrutinib with moderate or strong CYP3A inhibitors and confirms the favorable efficacy and safety profile of zanubrutinib.


Asunto(s)
Inhibidores del Citocromo P-450 CYP3A , Neoplasias , Humanos , Inhibidores del Citocromo P-450 CYP3A/efectos adversos , Fluconazol/farmacología , Voriconazol , Diltiazem , Interacciones Farmacológicas
3.
Pharmacol Res Perspect ; 9(6): e00870, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34664792

RESUMEN

Zanubrutinib is a highly selective, potent, orally available, targeted covalent inhibitor (TCI) of Bruton's tyrosine kinase (BTK). This work investigated the in vitro drug metabolism and transport of zanubrutinib, and its potential for clinical drug-drug interactions (DDIs). Phenotyping studies indicated cytochrome P450 (CYP) 3A are the major CYP isoform responsible for zanubrutinib metabolism, which was confirmed by a clinical DDI study with itraconazole and rifampin. Zanubrutinib showed mild reversible inhibition with half maximal inhibitory concentration (IC50 ) of 4.03, 5.69, and 7.80 µM for CYP2C8, CYP2C9, and CYP2C19, respectively. Data in human hepatocytes disclosed induction potential for CYP3A4, CYP2B6, and CYP2C enzymes. Transport assays demonstrated that zanubrutinib is not a substrate of human breast cancer resistance protein (BCRP), organic anion transporting polypeptide (OATP)1B1/1B3, organic cation transporter (OCT)2, or organic anion transporter (OAT)1/3 but is a potential substrate of the efflux transporter P-glycoprotein (P-gp). Additionally, zanubrutinib is neither an inhibitor of P-gp at concentrations up to 10.0 µM nor an inhibitor of BCRP, OATP1B1, OATP1B3, OAT1, and OAT3 at concentrations up to 5.0 µM. The in vitro results with CYPs and transporters were correlated with the available clinical DDIs using basic models and mechanistic static models. Zanubrutinib is not likely to be involved in transporter-mediated DDIs. CYP3A inhibitors and inducers may impact systemic exposure of zanubrutinib. Dose adjustments may be warranted depending on the potency of CYP3A modulators.


Asunto(s)
Sistema Enzimático del Citocromo P-450/efectos de los fármacos , Proteínas de Transporte de Membrana/efectos de los fármacos , Piperidinas/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Perros , Interacciones Farmacológicas , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Concentración 50 Inhibidora , Proteínas de Transporte de Membrana/metabolismo , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Piperidinas/farmacocinética , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacocinética , Pirazoles/farmacología , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas
4.
Expert Rev Clin Pharmacol ; 14(11): 1329-1344, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34491123

RESUMEN

Introduction: Bruton's tyrosine kinase (BTK) inhibitors have revolutionized the treatment of B-cell lymphomas. Zanubrutinib was designed to achieve improved therapeutic concentrations and minimize off-target activities putatively accounting, in part, for the adverse effects seen with other BTK inhibitors.Areas covered: This drug profile covers zanubrutinib clinical pharmacology and the translation of pharmacokinetics (PK) and pharmacodynamics (PD) to clinical efficacy and safety profiles, by highlighting key differences between zanubrutinib and other BTK inhibitors. We discuss PK, sustained BTK occupancy, and potential factors affecting PK of zanubrutinib, including food effects, hepatic impairment, and drug-drug interactions. These data, along with exposure-response analyses, were used to support the recommended dose of 320 mg, either once daily or as 160 mg twice daily. Translation of PK/PD attributes into clinical effects was demonstrated in a randomized, phase 3 head-to-head study comparing it with ibrutinib in patients with Waldenström macroglobulinemia.Expert opinion: Among the approved BTK inhibitors, zanubrutinib is less prone to PK modulation by intrinsic and extrinsic factors, leading to more consistent, sustained therapeutic exposures and improved dosing convenience. Zanubrutinib PK/PD has translated into durable responses and improved safety, representing an important new treatment option for patients who benefit from BTK therapy.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Piperidinas/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Adenina/administración & dosificación , Adenina/análogos & derivados , Adenina/farmacocinética , Adenina/farmacología , Animales , Interacciones Farmacológicas , Humanos , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/enzimología , Piperidinas/farmacocinética , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacocinética , Pirazoles/farmacología , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto , Macroglobulinemia de Waldenström/tratamiento farmacológico , Macroglobulinemia de Waldenström/enzimología
5.
Leuk Lymphoma ; 62(11): 2612-2624, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34159878

RESUMEN

This report summarizes a totality-of-evidence approach supporting recommendation of a 320-mg total daily dose, either as 160-mg twice daily (BID) or 320-mg once daily (QD) for zanubrutinib in patients with mantle cell lymphoma. Data were derived from a phase 2 study in patients receiving 160-mg BID and a phase 1/2 study with similar response rates observed with 160-mg BID or 320-mg QD. Given the limited number of patients in the QD dose group, population pharmacokinetics and exposure-response analyses were employed to bridge the two regimens. The analyses showed that similar plasma exposure and BTK inhibition were achieved, and differences in trough concentration and maximum plasma concentration between the two regimens are unlikely to have a meaningful impact on efficacy and safety endpoints. The totality of data, including pharmacokinetic, pharmacodynamic, safety, efficacy, and exposure-response analyses, provided support for the recommended 320-mg total daily dose for the approved indication.


Asunto(s)
Linfoma de Células del Manto , Adulto , Humanos , Linfoma de Células del Manto/tratamiento farmacológico , Piperidinas , Pirazoles , Pirimidinas
6.
CPT Pharmacometrics Syst Pharmacol ; 10(5): 441-454, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33687157

RESUMEN

A physiologically based pharmacokinetic (PBPK) model was developed to evaluate and predict (1) the effect of concomitant cytochrome P450 3A (CYP3A) inhibitors or inducers on the exposures of zanubrutinib, (2) the effect of zanubrutinib on the exposures of CYP3A4, CYP2C8, and CYP2B6 substrates, and (3) the impact of gastric pH changes on the pharmacokinetics of zanubrutinib. The model was developed based on physicochemical and in vitro parameters, as well as clinical data, including pharmacokinetic data in patients with B-cell malignancies and in healthy volunteers from two clinical drug-drug interaction (DDI) studies of zanubrutinib as a victim of CYP modulators (itraconazole, rifampicin) or a perpetrator (midazolam). This PBPK model was successfully validated to describe the observed plasma concentrations and clinical DDIs of zanubrutinib. Model predictions were generally within 1.5-fold of the observed clinical data. The PBPK model was used to predict untested clinical scenarios; these simulations indicated that strong, moderate, and mild CYP3A inhibitors may increase zanubrutinib exposures by approximately four-fold, two- to three-fold, and <1.5-fold, respectively. Strong and moderate CYP3A inducers may decrease zanubrutinib exposures by two- to three-fold or greater. The PBPK simulations showed that clinically relevant concentrations of zanubrutinib, as a DDI perpetrator, would have no or limited impact on the enzyme activity of CYP2B6 and CYP2C8. Simulations indicated that zanubrutinib exposures are not impacted by acid-reducing agents. Development of a PBPK model for zanubrutinib as a DDI victim and perpetrator in parallel can increase confidence in PBPK models supporting zanubrutinib label dose recommendations.


Asunto(s)
Antineoplásicos/farmacocinética , Simulación por Computador , Inductores del Citocromo P-450 CYP3A/farmacocinética , Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Interacciones Farmacológicas , Piperidinas/farmacocinética , Pirazoles/farmacocinética , Pirimidinas/farmacocinética , Humanos
7.
Clin Transl Sci ; 14(2): 764-772, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33306268

RESUMEN

Zanubrutinib is a potent, second-generation Bruton's tyrosine kinase inhibitor that is currently being investigated in patients with B-cell malignancies and recently received accelerated approval in the United States for treatment of relapsed/refractory mantle cell lymphoma. The objective of this analysis was to develop a population pharmacokinetic (PK) model to characterize the PKs of zanubrutinib and identify the potential impact of intrinsic and extrinsic covariates on zanubrutinib PK. Data across nine clinical studies of patients with B-cell malignancies and data of healthy volunteers (HVs) were included in this analysis, at total daily doses ranging from 20 to 320 mg. In total, 4,925 zanubrutinib plasma samples from 632 subjects were analyzed using nonlinear mixed-effects modeling. Zanubrutinib PKs were adequately described by a two-compartment model with sequential zero-order then first-order absorption, and first-order elimination. A time-dependent residual error model was implemented in order to better capture the observed maximum concentration variability in subjects. Baseline alanine aminotransferase and health status (HVs or patients with B-cell malignancies) were identified as statistically significant covariates on the PKs of zanubrutinib. These factors are unlikely to be clinically meaningful based on a sensitivity analysis. No statistically significant differences in the PKs of zanubrutinib were observed based on age, sex, race (Asian, white, and other), body weight, mild or moderate renal impairment (creatinine clearance ≥ 30 mL/minute as estimated by Cockcroft-Gault), baseline aspartate aminotransferase, bilirubin, tumor type, or use of acid-reducing agents (including proton pump inhibitors). These results support that no dose adjustment is considered necessary based on the aforementioned factors.


Asunto(s)
Leucemia de Células B/tratamiento farmacológico , Linfoma de Células B/tratamiento farmacológico , Piperidinas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazoles/farmacocinética , Pirimidinas/farmacocinética , Macroglobulinemia de Waldenström/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Variación Biológica Poblacional , Estudios de Casos y Controles , Ensayos Clínicos como Asunto , Relación Dosis-Respuesta a Droga , Femenino , Voluntarios Sanos , Humanos , Leucemia de Células B/sangre , Linfoma de Células B/sangre , Masculino , Persona de Mediana Edad , Modelos Biológicos , Piperidinas/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Macroglobulinemia de Waldenström/sangre , Adulto Joven
8.
Br J Clin Pharmacol ; 87(7): 2926-2936, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33336408

RESUMEN

AIM: This study aims to assess the potential effects of zanubrutinib on the activity of cytochrome P450 (CYP) enzymes and drug transporter proteins using a cocktail probe approach. METHODS: Patients received single oral doses of probe drugs alone and after at least 8 days of treatment with zanubrutinib 160 mg twice daily in a single-sequence study in 18 healthy male volunteers. Simultaneous doses of 10 mg warfarin (CYP2C9) and 2 mg midazolam (CYP3A) were administered on Day 1 and Day 14, 0.25 mg digoxin (P-glycoprotein [P-gp]) and 10 mg rosuvastatin (breast cancer resistance protein [BCRP]) on Day 3 and Day 16, and 20 mg omeprazole (CYP2C19) on Day 5 and Day 18. Pharmacokinetic (PK) parameters were estimated from samples obtained up to 12 h post dose for zanubrutinib; 24 h for digoxin, omeprazole and midazolam; 48 h for rosuvastatin; and 144 h for warfarin. RESULTS: The ratios (%) of geometric least squares means (90% confidence intervals) for the area under the concentration-time curve from time zero to the last quantifiable concentration in the presence/absence of zanubrutinib were 99.80% (97.41-102.2%) for S-warfarin; 52.52% (48.49-56.88%) for midazolam; 111.3% (103.8-119.3%) for digoxin; 89.45% (78.73-101.6%) for rosuvastatin; and 63.52% (57.40-70.30%) for omeprazole. Similar effects were observed for maximum plasma concentrations. CONCLUSIONS: Zanubrutinib 320 mg total daily dose had minimal or no effect on the activity of CYP2C9, BCRP and P-gp, but decreased the systemic exposure of CYP3A and CYP2C19 substrates (mean reduction <50%).


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Citocromo P-450 CYP3A , Subfamilia B de Transportador de Casetes de Unión a ATP , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Cafeína , Citocromo P-450 CYP2C19/genética , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP3A/metabolismo , Interacciones Farmacológicas , Humanos , Masculino , Proteínas de Neoplasias , Piperidinas , Pirazoles , Pirimidinas
9.
Clin Transl Sci ; 13(5): 923-931, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32144955

RESUMEN

This thorough QT (TQT) study evaluated the effect of zanubrutinib on electrocardiogram (ECG) parameters by using concentration-QTc (C-QTc) analysis as the primary analysis for this study. Part A of the study determined the safety and tolerability of a single supratherapeutic dose of zanubrutinib (480 mg) in healthy volunteers. Part B was a randomized, blinded, placebo-controlled and positive-controlled, four-way crossover, TQT study of single therapeutic (160 mg) and supratherapeutic (480 mg) doses of zanubrutinib, placebo, and open-label moxifloxacin 400 mg. Thirty-two participants received at least 1 dose of zanubrutinib, and 26 participants completed all 4 periods. Zanubrutinib did not have any effect on heart rate or cardiac conduction (pulse rate, QRS interval, or T-wave morphology) and was generally well-tolerated. Using C-QTc analysis, the predicted placebo-corrected change-from-baseline QT interval using Fridericia's formula (ΔΔQTcF) was -3.4 msec (90% confidence interval: -4.9 to -1.9 msec) at peak concentrations of the 480 mg dose. A QT effect (ΔΔQTcF) exceeding 10 msec could be excluded within the observed concentration range at 160 and 480 mg doses. Assay sensitivity was established by moxifloxacin with 90% lower bound exceeding 5 msec. Implementing a C-QTc analysis prospectively in this TQT study resulted in a substantially smaller sample size to maintain a similar study power as shown in the traditional time-point analysis. A single 160-mg or 480-mg zanubrutinib dose did not prolong the QTc interval or have any other clinically relevant effects on ECG parameters.


Asunto(s)
Síndrome de QT Prolongado/diagnóstico , Piperidinas/efectos adversos , Inhibidores de Proteínas Quinasas/efectos adversos , Pirazoles/efectos adversos , Pirimidinas/efectos adversos , Adolescente , Adulto , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Electrocardiografía , Femenino , Voluntarios Sanos , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Síndrome de QT Prolongado/inducido químicamente , Masculino , Persona de Mediana Edad , Piperidinas/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Adulto Joven
10.
Leuk Lymphoma ; 61(6): 1355-1363, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32031037

RESUMEN

The pharmacokinetics and safety of single-dose zanubrutinib (80 mg) were assessed in subjects with mild, moderate, and severe hepatic impairment (n = 6 each, Child-Pugh class A, B, and C) relative to healthy controls (n = 11). Zanubrutinib median Tmax was 1.25-2.25 h in all groups. Compared to control group, mean zanubrutinib exposure (AUC0-inf) in the mild and moderate hepatic impairment groups was increased by 1.1- and 1.2-fold, which is within the range of PK variability for zanubrutinib. The total and unbound AUC of zanubrutinib were 1.60- and 2.9-fold higher in subjects with severe hepatic impairment compared to healthy controls. Terminal half-life was comparable between subjects with hepatic impairment and matched healthy controls. Zanubrutinib was generally well-tolerated when administered as a single, 80-mg dose to subjects in this study. Results of this study will be used, in conjunction with clinical safety and efficacy data, to develop dose recommendations for patients with hepatic impairment.


Asunto(s)
Hepatopatías , Pirimidinas , Área Bajo la Curva , Humanos , Hepatopatías/complicaciones , Hepatopatías/diagnóstico , Piperidinas , Pirazoles
11.
Pharm Res ; 31(3): 809-18, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24065594

RESUMEN

PURPOSE: This study was to characterize the pharmacokinetics (PK) and pharmacodynamics (PD) of a chimeric protein, CD4-anchoring bi-functional fusion inhibitor (CD4-BFFI), in monkeys and assess the feasibility for HIV-1 treatment in humans. METHODS: The serum concentrations of CD4-BFFI and CD4 receptors were determined and modeled using a target-mediated drug disposition (TMDD) model following intravenous administration of 1 or 10 mg/kg in monkeys. In vitro CD4 internalization was examined in human peripheral blood mononuclear cells. RESULTS: Noncompartmental analysis showed a decrease in clearance (1.35 to 0.563 mL/h/kg) and an increase in half-lives (35 to 50 h) with increasing doses. Dose-dependent CD4 occupancy was observed. The TMDD model reasonably captured the PK/PD profiles and suggested greater degradation rate constant for the free CD4 than the bound CD4. In vitro assay showed CD4-BFFI did not reduce the internalization of cell surface CD4. The simulated serum concentrations of CD4-BFFI were 20-fold above its in vitro IC50 for HIV-1 at 3 mg/kg weekly or biweekly following subcutaneous administration in humans. CONCLUSIONS: The TMDD modeling and in vitro CD4 internalization study indicate that CD4-BFFI does not induce CD4 internalization and CD4-BFFI short half-life is likely due to normal CD4 internalization. The simulated human PK supports CD4-BFFI as a promising anti-HIV-1 agent.


Asunto(s)
Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/farmacocinética , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/farmacocinética , Antígenos CD4/inmunología , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/farmacocinética , Administración Intravenosa , Animales , Fármacos Anti-VIH/sangre , Fármacos Anti-VIH/inmunología , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/inmunología , Femenino , Haplorrinos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Masculino , Modelos Biológicos , Proteínas Recombinantes de Fusión/sangre , Proteínas Recombinantes de Fusión/inmunología
12.
J Clin Pharmacol ; 53(11): 1112-20, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23913679

RESUMEN

Results of pharmacometric analyses influence high-level decisions such as clinical trial design, drug approval, and labeling. Key challenges for timely delivery of pharmacometric analyses are the data assembly process and tracking and documenting the modeling process and results. Since clinical efficacy and safety data typically reside in the biostatistics computing area, an integrated computing platform for pharmacometric and biostatistical analyses would be ideal. A case study is presented integrating a pharmacometric modeling platform into an existing statistical computing environment (SCE). The feasibility and specific configurations of running common PK/PD programs such as NONMEM and R inside of the SCE are provided. The case study provides an example of an integrated repository that facilitates efficient data assembly for pharmacometrics analyses. The proposed platform encourages a good pharmacometrics working practice to maintain transparency, traceability, and reproducibility of PK/PD models and associated data in supporting drug development and regulatory decisions.


Asunto(s)
Bioestadística/métodos , Farmacología Clínica/métodos , Programas Informáticos , Aminoglicósidos/sangre , Aminoglicósidos/farmacocinética , Antibacterianos/sangre , Antibacterianos/farmacocinética , Simulación por Computador , Infección Hospitalaria/sangre , Infección Hospitalaria/tratamiento farmacológico , Humanos , Lipoglucopéptidos , Modelos Biológicos , Neumonía Bacteriana/sangre , Neumonía Bacteriana/tratamiento farmacológico
13.
Arch Toxicol ; 81(2): 101-11, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16858609

RESUMEN

Chemical exposure via breast milk is one of the great concerns in public health. Previously, we demonstrated that most body burden of PCB 153 can be transferred from the mother to the pups in mice during lactational period. Here we present a physiologically based pharmacokinetic (PBPK) model to describe the lactational transfer of PCB 153 with or without PCB 126 in mice. The model incorporated physiological changes on the volume and the blood flow into mammary tissues, and considered mechanistic information on the movement of PCB 153 from adipose tissue to the mammary gland during lactational period. The mechanistic consideration includes fat volume changes, binding of PCB 153 to very low density lipoprotein (VLDL) and increased uptake of VLDL in mammary tissues. Model parameters depicting physiological changes were obtained from research articles dealing with chemical transfer during lactational period in rodents. Chemical-specific parameters were derived from previous PBPK models focusing on the PCB disposition in rodents. The developed model adequately described the lactational transfer of PCB 153 with or without PCB 126 in mice. Our model will provide a useful mechanistic tool to estimate the disposition of PCBs in diverse experimental designs regarding PCB effects during developmental period and to improve quantitative risk assessment of PCBs in the developing organisms.


Asunto(s)
Contaminantes Ambientales/farmacocinética , Lactancia/metabolismo , Bifenilos Policlorados/farmacocinética , Animales , Animales Lactantes , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Lipoproteínas VLDL/metabolismo , Glándulas Mamarias Animales/metabolismo , Ratones , Modelos Biológicos , Medición de Riesgo
14.
Toxicol Sci ; 73(2): 301-14, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12700395

RESUMEN

A combination of experimental and simulation approaches was used to analyze clonal growth of glutathione-S-transferase pi (GST-P) enzyme-altered foci during liver carcinogenesis in an initiation-promotion regimen for 1,4-dichlorobenzene (DCB), 1,2,4,5-tetrachlorobenzene (TECB), pentachlorobenzene (PECB), and hexachlorobenzene (HCB). Male Fisher 344 rats, eight weeks of age, were initiated with a single dose (200 mg/kg, ip) of diethylnitrosamine (DEN). Two weeks later, daily dosing of 0.1 mol/kg chlorobenzene was maintained for six weeks. Partial hepatectomy was performed three weeks after initiation. Liver weight, normal hepatocyte division rates, and the number and volume of GST-P positive foci were obtained at 23, 26, 28, 47, and 56 days after initiation. A clonal growth stochastic model separating the initiated cell population into two distinct subtypes (referred to as A and B cells) was successfully used to describe the foci development data for the four chlorobenzenes. The B cells are initiated cells that display a selective growth advantage under conditions that inhibit the growth of initiated A cells or normal hepatocytes. The simulation exercise for the four chlorobenzenes indicates a positive correlation between the estimated net growth rate of B cells during the 2-week regeneration period following partial hepatectomy and final foci volume at the end of the bioassay. This observation is consistent with the sensitivity analysis of model parameters. While TECB, PECB, and HCB all significantly increased foci volume, only HCB increased normal hepatocyte proliferation. Together, these results indicate that examining effects of chemicals on regenerative responses following partial hepatectomy may be a means for understanding the carcinogenicity potential of chlorobenzene compounds.


Asunto(s)
Carcinógenos/toxicidad , Clorobencenos/toxicidad , Hiperplasia Nodular Focal/inducido químicamente , Neoplasias Hepáticas/inducido químicamente , Lesiones Precancerosas/inducido químicamente , Administración Oral , Animales , Pruebas de Carcinogenicidad , Carcinógenos/administración & dosificación , Clorobencenos/administración & dosificación , Células Clonales , Simulación por Computador , Dietilnitrosamina/toxicidad , Quimioterapia Combinada , Hiperplasia Nodular Focal/enzimología , Hiperplasia Nodular Focal/patología , Glutatión Transferasa/metabolismo , Hepatectomía , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Masculino , Lesiones Precancerosas/patología , Ratas , Ratas Endogámicas F344
15.
Environ Health Perspect ; 110 Suppl 5: 859-64, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12426147

RESUMEN

Methylmercury (MeHg) has been an environmental concern to public health and regulatory agencies for over 50 years because of its toxicity to the human nervous system. Its association with nervous system toxicity in adults and infants near Minamata Bay, Japan, in the 1950s initiated environmental health research inquiries that continue to this day. Observations of greater neurotoxicity with gestational compared with adult exposure suggest a unique susceptibility of the developing nervous system to MeHg. Despite extensive research conducted over the last half century, determination of definitive molecular mechanisms underlying the observed neurotoxic effects of MeHg have not been identified. This paper summarizes results of a series of experiments conducted to examine the effects of MeHg on neuroepithelial cell proliferation, a hypothesized mode of action for its selective effects on neurogenesis. Observed effects of MeHg on cell cycle entry and progression were associated with alterations in a variety of cell cycle regulatory molecules, including p21 signaling pathways. We place these studies in the context of other cellular responses involved in signal transduction, including oxidative stress, altered protein phosphorylation, and altered intracellular calcium homeostasis. Although existing information suggests that no single mechanism underlies the diverse array of effects associated with MeHg-induced developmental neurotoxicity, we demonstrate characteristic effects of MeHg on cell signaling that contribute to observed effects on cell proliferation. Experimentally derived cell cycle kinetic and cytotoxicity data allowed development of a biologically based dose-response model of MeHg-induced alterations in neurodevelopment, which can form the basis for information synthesis and hypothesis testing and for use in assessing risks from environmental exposures.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Compuestos de Metilmercurio/efectos adversos , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/crecimiento & desarrollo , Animales , Calcio/metabolismo , Niño , Protección a la Infancia , Desarrollo Embrionario y Fetal , Homeostasis , Humanos , Cinética , Sistema Nervioso/embriología , Estrés Oxidativo , Fosforilación , Ratas , Medición de Riesgo , Transducción de Señal
16.
Toxicol Appl Pharmacol ; 178(2): 117-25, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11814332

RESUMEN

Methylmercury (MeHg) is an environmentally prevalent organometal that is particularly toxic to the developing central nervous system (CNS). Prenatal MeHg exposure is associated with reduced brain size and weight and a reduced number of neurons, which have been associated with impaired cell proliferation. We evaluate the role of p21, a cell cycle protein involved in the G1- and G2-phase checkpoint control, in the cell cycle inhibition induced by MeHg. Primary mouse embryonic fibroblasts (MEFs) of different p21 genotypes (wild-type, heterozygous, and null) were isolated at day 14 of gestation and treated at passages 4-6 with either 0, 2, 4, or 6 microM MeHg or 50 nM colchicine for 24 h. Changes in cell cycle distribution after continuous toxicant treatment were analyzed by DNA content-based flow cytometry using DAPI. MeHg induced an increase in the proportion of cells in G2/M at 2 and 4 microM MeHg (p < or = 0.05) irrespective of p21 genotype. Effects of MeHg on cell cycle progression were subsequently evaluated using BrdU-Hoechst flow cytometric analysis. Inhibition of cell cycle progression was observed in all p21 genotypes after continuous exposure to MeHg for 24 and 48 h. p21 null (-/-) cells reached the second-round G1 at a higher fraction compared to the wild type (+/+) and heterozygous (+/-) cells (p < or = 0.05). These data support previous observations that MeHg inhibits cell cycle progression through delayed G2/M transition. Whereas the G2/M accumulation induced by MeHg was independent of p21 status, a greater proportion of p21(-/-) cells were able to complete one round of cell division in the presence of MeHg compared to p21(+/-) or p21(+/+) cells. These data suggest a role for p21 in retarding cell cycle progression, but not mitotic inhibition, following exposure to MeHg.


Asunto(s)
Ciclinas/fisiología , Fase G2/efectos de los fármacos , Compuestos de Metilmercurio/toxicidad , Animales , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Fibroblastos , Citometría de Flujo , Genotipo , Ratones , Ratones Transgénicos
17.
Toxicol Sci ; 65(1): 26-34, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11752682

RESUMEN

Polychlorinated biphenyls (PCBs) are ubiquitous environmental contaminants that can induce neurological defects in infants and children via placental and lactational transfer. To investigate the lactational transfer of PCBs and compare pharmacokinetic interactions among nonpregnant, lactating mice and suckling pups, quantitative time-course measurements of PCB accumulation in tissues were performed. On postnatal day 1, nonpregnant and lactating C57BL/6 mice were exposed to PCB 153 (2,2',4,4',5,5'-hexachlorobiphenyl, 20 mg/kg) alone or a mixture of PCB 153 (20 mg/kg) and PCB 126 (3,3',4,4',5-pentachlorobiphenyl, 0.2 mg/kg) by oral gavage. At 1, 3, 6, and 13 days after treatment, PCB 153 and PCB 126 were determined in nonpregnant and maternal tissues as well as in neonatal tissues by gas chromatography (GC). Coadministration of PCB 153 and PCB 126 increased PCB 153 retention in the liver and decreased PCB 153 accumulation in the fat of nonpregnant mice. Lactational transfer was confirmed to be an efficient elimination mechanism for the lactating mice but a major source of exposure in the pups. However, little or no significant pharmacokinetic interactions were observed in lactating mice and suckling pups. To describe pharmacokinetic interactions between PCB 153 and PCB 126, a physiologically based pharmacokinetic model for PCB 153 disposition was developed. The effects of PCB 126 on the fat content in liver and a diffusion permeation constant in fat were incorporated into the physiologically based pharmacokinetic (PBPK) model. This model successfully describes PCB 153 disposition altered by PCB 126 in nonpregnant mice.


Asunto(s)
Antagonistas de Estrógenos/farmacocinética , Lactancia/metabolismo , Bifenilos Policlorados/farmacocinética , Tejido Adiposo/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Interacciones Farmacológicas , Femenino , Hígado/metabolismo , Exposición Materna , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Sensibilidad Química Múltiple , Embarazo , Factores de Tiempo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA