Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
CPT Pharmacometrics Syst Pharmacol ; 6(4): 249-257, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28109128

RESUMO

Benralizumab is a humanized, afucosylated, anti-interleukin-5 receptor α, immunoglobulin G (IgG) 1 κ monoclonal antibody. We developed a population pharmacokinetic (PK)/pharmacodynamic (PD) model for benralizumab by analyzing PK and blood eosinophil count data from two healthy volunteer studies (N = 48) and four studies in patients with asthma (N = 152). Benralizumab PK was dose-proportional and adequately described by a two-compartment model with first-order elimination from the central compartment and first-order absorption from the subcutaneous dosing site. The estimated systemic clearance and volume of distribution were typical for human IgG. Body weight and high-titer antidrug antibodies were identified as relevant covariates influencing the PK of benralizumab. Depletion of blood eosinophil counts was depicted by a modified transit model in which benralizumab induced depletion of eosinophils in each age compartment. Stochastic simulations supported an every-8-week dosing schedule of benralizumab for a phase IIb study in patients with uncontrolled asthma.


Assuntos
Antiasmáticos/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Asma/tratamento farmacológico , Adulto , Antiasmáticos/farmacocinética , Anticorpos Monoclonais Humanizados/farmacocinética , Asma/metabolismo , Peso Corporal , Relação Dose-Resposta a Droga , Eosinófilos/efeitos dos fármacos , Eosinófilos/metabolismo , Feminino , Voluntários Saudáveis , Humanos , Imunoglobulina G/metabolismo , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Processos Estocásticos , Adulto Jovem
3.
Clin Pharmacol Ther ; 93(6): 483-92, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23511714

RESUMO

A type I interferon (IFN) gene signature shared by systemic lupus erythematous (SLE) and systemic sclerosis (SSc) was used to evaluate an anti-type I IFN-α receptor (IFN-αR) monoclonal antibody, MEDI-546, in a phase I trial in SSc. MEDI-546 suppressed IFN signature in blood and skin of SSc patients in a dose-dependent manner. To bridge clinical indications to SLE, we developed a model incorporating (i) pharmacokinetics (PK) and pharmacodynamics (PD) in SSc patients, (ii) internalization kinetics of MEDI-546/IFN-αR complex, and (iii) the different IFN signatures between SSc and SLE. Simulations predicted that i.v. administration of MEDI-546 at 300- or 1,000-mg monthly doses could suppress IFN signature in blood to levels of healthy subjects in 53 and 68% of SLE patients, respectively. An innovative approach utilizing a novel biomarker characterized the PD of MEDI-546 by modeling and simulation and allowed rapid progression of MEDI-546 from a phase I study in SSc to a randomized, multiple-dose phase II trial.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Simulação por Computador , Interferon-alfa/metabolismo , Farmacogenética , Receptor de Interferon alfa e beta/imunologia , Receptor de Interferon alfa e beta/metabolismo , Escleroderma Sistêmico/tratamento farmacológico , Adulto , Idoso , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Biomarcadores/sangue , Biomarcadores/metabolismo , Ensaios Clínicos Fase II como Assunto , Relação Dose-Resposta a Droga , Feminino , Humanos , Interferon-alfa/genética , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Escleroderma Sistêmico/sangue , Escleroderma Sistêmico/metabolismo , Pele/metabolismo
4.
Br J Pharmacol ; 168(1): 200-11, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22913645

RESUMO

BACKGROUND AND PURPOSE: For antibody therapies against receptor targets, in vivo outcomes can be difficult to predict because of target-mediated clearance or antigen 'sink' effects. The purpose of this work was to engineer an antibody to the GM-CSF receptor α (GM-CSFRα) with pharmacological properties optimized for chronic, s.c. treatment of rheumatoid arthritis (RA) patients. EXPERIMENTAL APPROACH: We used an in silico model of receptor occupancy to guide the target affinity and a combinatorial phage display approach for affinity maturation. Mechanism of action and internalization assays were performed on the optimized antibody in vitro before refining the modelling predictions of the eventual dosing in man. Finally, in vivo pharmacology studies in cynomolgus monkeys were carried out to inform the predictions and support future clinical development. KEY RESULTS: Antibody potency was improved 8600-fold, and the target affinity was reached. The refined model predicted pharmacodynamic effects at doses as low as 1 mg kg(-1) and a study in cynomolgus monkeys confirmed in vivo efficacy at 1 mg kg(-1) dosing. CONCLUSIONS AND IMPLICATIONS: This rational approach to antibody drug discovery enabled the isolation of a potent molecule compatible with chronic, s.c. self-administration by RA patients. We believe this general approach enables the development of optimal biopharmaceuticals.


Assuntos
Anticorpos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/imunologia , Animais , Artrite Reumatoide/imunologia , Técnicas de Visualização da Superfície Celular , Feminino , Humanos , Imunoglobulina G/metabolismo , Concentração Inibidora 50 , Macaca fascicularis , Masculino , Modelos Biológicos , Ligação Proteica , Engenharia de Proteínas , Proteínas Recombinantes
5.
Int J Toxicol ; 27(4): 303-12, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18821393

RESUMO

Robust assessments of the nonclinical safety profile of biopharmaceuticals are best developed on a scientifically justified, case-by-case basis, with consideration of the therapeutic molecule, molecular target, and differences/similarities between nonclinical species and humans (ICH S6). Significant experience has been gained in the 10 years ensuing since publication of the ICH S6 guidance. In a PhRMA-FDA-sponsored workshop, "Nonclinical Aspects of Biopharmaceutical Development," industry and US regulatory representatives engaged in exploration of current scientific and regulatory issues relating to the nonclinical development of biopharmaceuticals in order to share scientific learning and experience and to work towards establishing consistency in application of general principles and approaches. The proceedings and discussions of this workshop confirm general alignment of strategy and tactics in development of biopharmaceuticals with regard to such areas as species selection, selection of high doses in toxicology studies, selection of clinical doses, the conduct of developmental and reproductive toxicity (DART) studies, and assessment of carcinogenic potential. However, several important aspects, including, for example, appropriate use of homologues, nonhuman primates, and/or in vitro models in the assessment of risk for potential developmental and carcinogenic effects, were identified as requiring further scientific exploration and discussion.


Assuntos
Fatores Biológicos , Química Farmacêutica , Animais , Humanos , Estados Unidos , United States Food and Drug Administration
6.
Bone Marrow Transplant ; 32(4): 399-404, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12900776

RESUMO

Daily administration of filgrastim decreases the duration of severe neutropenia in the clinical setting. A sustained-duration form of filgrastim, pegfilgrastim, significantly reduces scheduling protocols to a single injection per chemotherapy cycle while maintaining therapeutic efficiency. We examined the ability of a single injection of pegfilgrastim to significantly improve neutrophil recovery following autologous bone marrow transplantation (AuBMT) in rhesus macaques. On day 1, postmyeloablation (920 cGy x-irradiation) and AuBMT, animals received either 0.1% autologous serum for 18 consecutive days (n=13), or single doses of pegfilgrastim via the subcutaneous (s.c.) or intravenous (i.v.) route (300 or 100 micro g/kg), or a single dose of filgrastim at 300 micro g/kg via the s.c. or i.v. route, or filgrastim at 10 micro g/kg via the s.c. route (n=4) on a daily basis (range=days 12-17). Pharmacokinetic parameters and neutrophil recovery were assessed. A single dose of pegfilgrastim via the i.v. or s.c. route was as effective as daily filgrastim administration, resulting in significant improvement of neutrophil recovery after myeloablation and ABuMT. Effective pegfilgrastim plasma concentrations were maintained in neutropenic animals until after the onset of hematopoietic recovery. Enhanced pharmacokinetics in AuBMT cohorts are consistent with self-regulating, neutrophil-mediated clearance.


Assuntos
Transplante de Medula Óssea/métodos , Fator Estimulador de Colônias de Granulócitos/análogos & derivados , Fator Estimulador de Colônias de Granulócitos/farmacologia , Fator Estimulador de Colônias de Granulócitos/farmacocinética , Neutrófilos/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Células da Medula Óssea , Estudos de Coortes , Citocinas/metabolismo , Filgrastim , Macaca mulatta , Masculino , Neutropenia , Neutrófilos/efeitos dos fármacos , Polietilenoglicóis , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Condicionamento Pré-Transplante , Transplante Autólogo
7.
Epilepsia ; 43(7): 691-6, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12102670

RESUMO

PURPOSE: To evaluate the potential pharmacokinetic interactions between topiramate (TPM) and phenytoin (PHT) in patients with epilepsy by studying their pharmacokinetics (PK) after monotherapy and concomitant TPM/PHT treatment. METHODS: Twelve patients with epilepsy stabilized on PHT monotherapy were enrolled in this study, with 10 and seven patients completing the phases with 400 and 800 mg TPM daily doses, respectively. TPM was added at escalating doses, and after stabilization at the highest tolerated TPM dose, PHT doses were tapered. Serial blood and urine samples were collected for PK analysis during the monotherapy phase or the lowest PHT dose after taper and the concomitant TPM/PHT phase. Potential metabolic interaction between PHT and TPM also was studied in vitro in human liver microsomal preparations. RESULTS: In nine of the 12 patients, PHT plasma concentrations remained stable, with a mean (+/-SD) area under the curve (AUC) ratio (combination therapy/monotherapy) of 1.13 +/- 0.17 (range, 0.89-1.23). Three patients had AUC ratios of 1.25, 1.39, and 1.55, respectively, and with the addition of TPM (800, 400, and 400 mg daily, respectively), their peak PHT plasma concentrations increased from 15 to 21 mg/L, 28 to 36 mg/L, and 27 to 41 mg/L, respectively. Human liver microsomal studies with S-mephenytoin showed that TPM partially inhibited CYP2C19 at very high concentrations of 300 microM (11% inhibition) and 900 microM (29% inhibition). Such high plasma concentrations would correspond to doses in humans that are 5 to 15 times higher than the recommended dose (200-400 mg). TPM clearance was approximately twofold higher during concomitant TPM/PHT therapy CONCLUSIONS: This study provides evidence that the addition of TPM to PHT generally does not cause clinically significant PK interaction. PHT induces the metabolism of TPM, causing increased TPM clearance, which may require TPM dose adjustments when PHT therapy is added or is discontinued. TPM may affect PHT concentrations in a few patients because of inhibition by TPM of the CYP2C19-mediated minor metabolic pathway of PHT.


Assuntos
Anticonvulsivantes/farmacocinética , Hidrocarboneto de Aril Hidroxilases , Epilepsia/tratamento farmacológico , Frutose/farmacocinética , Fenitoína/farmacocinética , Adolescente , Adulto , Anticonvulsivantes/uso terapêutico , Citocromo P-450 CYP2C19 , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Interações Medicamentosas , Quimioterapia Combinada , Epilepsia/metabolismo , Feminino , Frutose/análogos & derivados , Frutose/uso terapêutico , Humanos , Masculino , Pessoa de Meia-Idade , Oxigenases de Função Mista/efeitos dos fármacos , Oxigenases de Função Mista/metabolismo , Fenitoína/uso terapêutico , Topiramato
8.
J Pharmacokinet Pharmacodyn ; 28(4): 321-42, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11677930

RESUMO

The pharmacokinetic-pharmacodynamic (PK-PD) relationship of the granulopoietic effects of Filgrastim in healthy volunteers was characterized via a population approach. Healthy male volunteers were enrolled into a four-way crossover clinical trial. Subjects received four single doses of Filgrastim (375 and 750 micrograms i.v. and s.c.) with an intervening washout period of 7 days. Serum concentrations of Filgrastim were determined using an enzyme-linked immunosorbent assay. Absolute neutrophil count (ANC) was determined. Data analysis was performed using mixed-effects modeling as implemented in the NONMEM software package. The final PKPD model incorporates a two-compartment PK model with bisegmental absorption from the s.c. site, first-order and saturable elimination pathways, and an indirect PD model. A sigmoidal Emax model for the stimulation of ANC input rate (kin) was superior to the conventional Emax model (mean +/- SE: Emax = 12.7 +/- 1.7; EC50 = 4.72 +/- 0.72 ng/ml; Hill = 1.34 +/- 0.19). In addition, a time-variant scaling factor for ANC observations was introduced to account for the early transient depression of ANC after Filgrastim administration. The absolute bioavailability of subcutaneously administered Filgrastim was estimated to be 0.619 +/- 0.058 and 0.717 +/- 0.028 for 375 micrograms and 750 micrograms s.c. doses, respectively. The time profiles of concentration and ANC, as well as the concentration approximately ANC relationship of Filgrastim in healthy volunteers were well described by the developed population PK-PD model.


Assuntos
Fator Estimulador de Colônias de Granulócitos/farmacocinética , Adolescente , Adulto , Disponibilidade Biológica , Filgrastim , Fator Estimulador de Colônias de Granulócitos/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Proteínas Recombinantes
9.
Blood ; 98(7): 2052-8, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11567989

RESUMO

ABX-CBL, an immunoglobulin M murine monoclonal antibody, recognizes CD147 and initiates cell killing through complement-mediated lysis. In a dose-finding trial, 27 patients with steroid-refractory acute graft-versus-host disease (GVHD) received ABX-CBL at 0.01 (presumed no effect dose), 0.1, 0.2, or 0.3 mg/kg per day, and an additional 32 patients were given ABX-CBL at 0.2 or 0.15 mg/kg per day. All patients had undergone allogeneic transplantation for malignant or nonmalignant disorders and received GVHD prophylaxis, generally with methotrexate- and cyclosporine-containing regimens. None responded to methylprednisolone, given for a minimum of 3 days. ABX-CBL was started 20 to 236 (median, 47) days after transplantation; it was given for 7 consecutive days and was followed by 2 infusions per week for 2 more weeks. Among 51 patients evaluable for efficacy, 26 (51%) responded, including 13 with complete responses (CR) and 13 with partial responses (PR). CR lasting 14 days or longer or PR lasting 7 days or longer occurred in 21 (41%; 8 CR, 13 PR) patients, including 19 of 43 (44%) patients who received 0.1 to 0.3 mg/kg ABX-CBL and 2 of 8 (25%) patients given 0.01 mg/kg per day. Myalgias at doses 0.2 mg/kg or greater were dose limiting and resolved without sequelae. Causes of death included organ failure, progressive GVHD, and infection. No death was attributed to ABX-CBL. At 6 months after the initiation of ABX-CBL therapy, 26 (44%) patients were surviving. These results are encouraging. Further studies on the use of ABX-CBL in the management of GVHD are warranted.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Antígenos CD , Antígenos de Neoplasias , Antígenos de Superfície , Antineoplásicos/administração & dosagem , Proteínas Aviárias , Proteínas Sanguíneas , Doença Enxerto-Hospedeiro/tratamento farmacológico , Glicoproteínas de Membrana/imunologia , Doença Aguda , Adolescente , Adulto , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/toxicidade , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidade , Basigina , Criança , Pré-Escolar , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Meia-Vida , Humanos , Lactente , Subpopulações de Linfócitos , Pessoa de Meia-Idade , Esteroides/uso terapêutico , Análise de Sobrevida , Equivalência Terapêutica
11.
Am J Hematol ; 68(3): 210-4, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11754405

RESUMO

We report a patient with cyclic thrombocytopenia and antiplatelet antibodies, a variant of chronic immune thrombocytopenic purpura (ITP), with a several year history of periodic fluctuation of the platelet count, megakaryocytic hyperplasia and high-titer anti-GPIb-specific antiplatelet antibodies. The patient was resistant to multiple forms of therapy but has responded to the thrombopoietic growth factor, pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF). This case suggests that some patients with classic ITP may respond to thrombopoietic growth factors.


Assuntos
Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/uso terapêutico , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Trombocitopenia/tratamento farmacológico , Trombopoetina/administração & dosagem , Trombopoetina/uso terapêutico , Idoso , Autoanticorpos/sangue , Feminino , Humanos , Contagem de Plaquetas , Complexo Glicoproteico GPIb-IX de Plaquetas/imunologia , Púrpura Trombocitopênica Idiopática/etiologia , Púrpura Trombocitopênica Idiopática/imunologia , Trombocitopenia/etiologia , Trombocitopenia/imunologia , Trombopoetina/sangue , Trombopoetina/deficiência
12.
J Clin Oncol ; 18(13): 2522-8, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10893282

RESUMO

PURPOSE: To explore the use of SD/01 (a polyethylene glycol-conjugated filgrastim shown in preclinical studies to have a prolonged half-life) in patients with chemotherapy-induced neutropenia. PATIENTS AND METHODS: Thirteen patients with non-small-cell lung cancer were randomized to receive daily filgrastim (5 microg/kg/d) or a single injection of SD/01 (30, 100, or 300 microg/kg) 2 weeks before chemotherapy and again 24 hours after administration of carboplatin and paclitaxel. Pharmacodynamic, pharmacokinetic, and safety analyses were performed. RESULTS: Peak serum concentrations of SD/01 and the duration of increased serum concentrations were dependent on the SD/01 dose. SD/01 concentrations remained increased longer in patients with chemotherapy-induced neutropenia. Prechemotherapy median absolute neutrophil counts (ANCs) in patients receiving SD/01 were increased in a dose-dependent fashion, with the duration of this effect also being dose dependent. After chemotherapy, median ANC nadirs were similar in the filgrastim cohort and the cohort receiving SD/01 30 microg/kg, with higher nadirs seen in the cohorts receiving SD/01 100 or 300 microg/kg. Dose-limiting toxicities were not noted. CD34(+) cells were mobilized in all cohorts. CONCLUSION: A single dose of SD/01 increases the serum concentration of SD/01 for several days in a dose-dependent fashion and is not associated with significant toxicity. The effects of SD/01 on ANC and CD34(+) cell mobilization are comparable or greater than those achieved with daily filgrastim. The self-regulation of this molecule provides a potential therapeutic advantage in a variety of clinical settings associated with neutropenia.


Assuntos
Antineoplásicos/efeitos adversos , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Neutropenia/tratamento farmacológico , Polietilenoglicóis/administração & dosagem , Idoso , Antígenos CD34/análise , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Relação Dose-Resposta a Droga , Feminino , Filgrastim , Fator Estimulador de Colônias de Granulócitos/efeitos adversos , Fator Estimulador de Colônias de Granulócitos/farmacocinética , Mobilização de Células-Tronco Hematopoéticas , Hemoglobinas/análise , Humanos , Contagem de Leucócitos/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Neutropenia/sangue , Neutropenia/induzido quimicamente , Neutrófilos , Projetos Piloto , Contagem de Plaquetas/efeitos dos fármacos , Polietilenoglicóis/efeitos adversos , Polietilenoglicóis/farmacocinética , Proteínas Recombinantes
13.
Growth Factors ; 18(3): 215-26, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11334057

RESUMO

Phase I studies with pegylated megakaryocyte growth and development factor (PEG-rHuMGDF), a c-Mpl ligand that stimulates megakaryopoiesis, have demonstrated that PEG-rHuMGDF is biologically active alone and causes a dose-related enhancement of platelet recovery when administered after chemotherapy. Here we report the dose-ranging pharmacokinetics of PEG-rHuMGDF. Pre-injection blood samples were drawn daily for pharmacokinetic studies on 43 patients. An ELISA, established using PEG-rHuMGDF as the standard, was able to quantitate Mpl ligand at concentrations > 0.02 ng/mL. Over the dose range 0.03 to 5.0 microg/kg/day, subcutaneous administration produced linear increases in steady-state serum levels. Maximum levels of PEG-rHuMGDF attained after 5.0 microg/kg/day were 5.88 to 10.9 ng/mL. After discontinuation of PEG-rHuMGDF, concentrations of Mpl ligand returned to baseline within 5 days. The pharmacokinetics were best described by a one-compartment model with first-order absorption, an absorption delay, and non linear clearance over the first 48 hours. The mean terminal half-life was 33.3 + 16.7 hours, and the average apparent at steady state was 27.7 + 14.0 mL/h/kg; both were independent of administered dose. The apparent clearance of PEG-rHuMGDF was not predicted by platelet count. Administration of chemotherapy and Filgrastim did not alter the pharmacokinetics of PEG-rHuMGDF.


Assuntos
Polietilenoglicóis/farmacocinética , Proteínas Recombinantes/farmacocinética , Trombopoetina/farmacocinética , Antineoplásicos/administração & dosagem , Relação Dose-Resposta a Droga , Esquema de Medicação , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Injeções Subcutâneas , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Contagem de Plaquetas , Polietilenoglicóis/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/sangue , Trombopoetina/administração & dosagem , Trombopoetina/sangue
14.
Pharm Res ; 17(12): 1531-6, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11303964

RESUMO

PURPOSE: To determine the enzyme kinetics (EK) and identify the human cytochrome(s) P450 (CYP) involved in the deethylation of phenacetin to acetaminophen using a population-based method. METHODS: A sparse data set was generated from incubations containing human liver microsomes (n = 19) with phenacetin. Estimates of the EK parameters were obtained by fitting the concentration-velocity data to Michaelis-Menten models by using nonlinear mixed effects modeling. Relationships between the EK parameters and the CYP activities determined for these liver microsomes were examined. RESULTS: A two-enzyme kinetic model with a saturated, low KM enzyme and an unsaturated, high KM enzyme capable of forming acetaminophen best fit the data. The population estimates of the EK parameters were Vmax1, 911 pmol/min/mg protein; KM1, 11.3 microM; and Cl(int2), 0.4 microl/min/mg. The coefficients of variation for interliver variability in Vmax1 and residual error of the model were 39% and 15%, respectively. When the selective catalytic activities were examined as potential covariates, 7-ethoxyresorufin O-deethylation (CYP1A2) activity was found to be associated with the low KM enzyme, however, the high KM enzyme(s) could not be identified. CONCLUSIONS: The population approach characterized the EK parameters and identified the low KM enzyme responsible for phenacetin O-deethylation as CYP1A2. Population modeling of EK provides valuable information on inter- and intraliver variability in CYP dependent activities.


Assuntos
Analgésicos não Narcóticos/metabolismo , Fenacetina/metabolismo , Algoritmos , Biotransformação , Sistema Enzimático do Citocromo P-450/metabolismo , Remoção de Radical Alquila , Humanos , Técnicas In Vitro , Microssomos Hepáticos/metabolismo , Modelos Biológicos , População
15.
Blood ; 95(8): 2514-22, 2000 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10753829

RESUMO

The effects of thrombopoietic stimulation on megakaryocytopoiesis, platelet production, and platelet viability and function were examined in normal volunteers randomized to receive single bolus subcutaneous injections of 3 microg/kg pegylated recombinant megakaryocyte growth and development factor (PEG-rHuMGDF) or placebo in a 3:1 ratio. PEG-rHuMGDF transiently doubled circulating platelet counts, from 237 +/- 41 x 10(3)/microL to 522 +/- 90 x 10(3)/microL (P <.0001), peaking on day 12. Baseline and day-12 samples showed no differences in responsiveness of platelets to adenosine diphosphate or thrombin receptor agonist peptide (P >.4 in all cases); expression of platelet ligand-induced binding sites or annexin V binding sites (P >.6 in both cases); or density of platelet TPO-receptors (P >.5). Platelet counts normalized by day 28. The life span of autologous (111)In-labeled platelets increased from 205 +/- 18 hours (baseline) to 226 +/- 22 hours (P <.01) on day 8. Platelet life span decreased from 226 +/- 22 hours (day 8) to 178 +/- 53 hours (P <.05) on day 18. The theoretical basis for senescent changes in mean platelet life span was illustrated by biomathematical modeling. Platelet turnover increased from 43.9 +/- 11.9 x 10(3) platelets/microL/d (baseline) to 101 +/- 27.6 x 10(3) platelets/microL/d (P =.0009), and marrow megakaryocyte mass expanded from 37.4 +/- 18.5 fL/kg to 62 +/- 17 x 10(10) fL/kg (P =. 015). Although PEG-rHuMGDF initially increased megakaryocyte volume and ploidy, subsequently ploidy showed a transient reciprocal decrease when the platelet counts exceeded placebo values. In healthy human volunteers PEG-rHuMGDF transiently increases megakaryocytopoiesis 2-fold. Additionally, peripheral platelets expand correspondingly and exhibit normal function and viability during the ensuing 10 days. The induced perturbation in steady state thrombopoiesis resolves by 4 weeks. (Blood. 2000;95:2514-2522)


Assuntos
Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Polietilenoglicóis/farmacologia , Trombopoetina/farmacologia , Plaquetas/citologia , Diferenciação Celular/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Humanos , Ativação Plaquetária/efeitos dos fármacos , Contagem de Plaquetas/efeitos dos fármacos , Proteínas Recombinantes/farmacologia
16.
Exp Hematol ; 27(1): 117-30, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9923450

RESUMO

A single injection of > or =10 microg/kg PEG-rHuMGDF in mice causes a dose-dependent increase in circulating platelets beginning on day 3 and peaking on days 5-6. The mean platelet volume and platelet distribution width at doses > or =100 microg/kg initially increase in a dose-dependent fashion and later decrease. However, the mean platelet volume does not change when platelets are incubated with PEG-rHuMGDF in vitro. The number of marrow megakaryocytes increases in a dose-dependent fashion as early as day 1 and peaks on day 3. Marrow megakaryocyte colony-forming units (CFU-Meg) do not increase on days 1-3 at a dose of 100 microg/kg (a dose that increases platelet numbers two- to threefold and may be clinically relevant), but the relative frequency of high ploidy megakaryocytes and the proportion of large marrow megakaryocytes (29-50 microm in diameter) increases. After a dose of 1,000 microg/kg the percentage of megakaryocytes in mitosis peaks at 24-48 hours and the percentage of megakaryocytes incorporating BrdU is maximal at 48 hours, the relatively delayed peak of BrdU incorporation most likely representing endomitosis. The relative frequency of type II and III megakaryocytes peaks on days 3 and 4, respectively. Pharmacokinetic analysis of PEG-rHuMGDF shows peak serum concentrations at 2-4 hours and a terminal half-life of 11.4+/-2.5 hours. A single injection of PEG-rHuMGDF ameliorates carboplatin-induced megakaryocytopenia and thrombocytopenia in a dose-response dependent fashion. In conclusion, a single injection of PEG-rHuMGDF increases megakaryocyte and platelet production in normal and myelo-suppressed mice.


Assuntos
Polietilenoglicóis/farmacologia , Polietilenoglicóis/uso terapêutico , Trombocitopenia/fisiopatologia , Trombopoetina/farmacologia , Trombopoetina/uso terapêutico , Acetilcolinesterase/metabolismo , Animais , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Medula Óssea/química , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Carboplatina/farmacologia , Contagem de Células/efeitos dos fármacos , Membrana Celular/ultraestrutura , Tamanho Celular/efeitos dos fármacos , Corantes , DNA/análise , DNA/metabolismo , Relação Dose-Resposta a Droga , Fêmur/citologia , Humanos , Injeções , Fígado/citologia , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Mitose/efeitos dos fármacos , Contagem de Plaquetas/efeitos dos fármacos , Ploidias , Polietilenoglicóis/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Reticulina/análise , Baço/citologia , Trombocitopenia/tratamento farmacológico , Trombopoetina/metabolismo , Fatores de Tempo
19.
J Pharmacol Exp Ther ; 276(2): 658-66, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8632334

RESUMO

The formation kinetics of 2-hydroxymethyl olanzapine (2-OH olanzapine), 4'-N-oxide olanzapine (N-O olanzapine) and 4'-N-desmethyl olanzapine (NdM olanzapine) were analyzed in vitro. Biphasic kinetics were observed for formation of 2-OH and NdM olanzapine. The high-affinity enzyme responsible for 2-OH olanzapine formation by two human liver samples exhibited an intrinsic clearance (CLint) of 0.2 microliter/min/mg. NdM olanzapine formation by two human liver samples exhibited a CLint of 1.0 microliter/min/mg for the high affinity enzyme. The formation of N-O olanzapine was linear up to 300 microM olanzapine, yielding a CLint of 0.32 to 1.70 microliters/min/mg. The formation of 7-hydroxy olanzapine (7-OH olanzapine) exhibited an apparent Km of 24.2 microM. The rates of 2-OH olanzapine formation correlated with CYP2D6 levels and activity, and it was formed to the greatest extent by cDNA-expressed CYP2D6. N-O olanzapine formation correlated with human liver flavin-containing monooxygenase (FMO3) levels and activity. NdM olanzapine and 7-OH olanzapine formation correlated with CYP1A2 catalytic activities and they were formed to the greatest extent by expressed CYP1A2. These results suggest that CYP1A2 catalyzes NdM olanzapine and 7-OH olanzapine formation, CYP2D6 catalyzes 2-OH olanzapine formation and FMO3 catalyzes N-O olanzapine formation.


Assuntos
Antipsicóticos/metabolismo , Sistema Enzimático do Citocromo P-450/fisiologia , Pirenzepina/análogos & derivados , Benzodiazepinas , Citocromo P-450 CYP1A2 , Citocromo P-450 CYP2D6 , Humanos , Oxigenases de Função Mista/fisiologia , Olanzapina , Oxirredução , Oxirredutases/fisiologia , Pirenzepina/metabolismo
20.
J Pharmacol Exp Ther ; 275(3): 1131-5, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8531073

RESUMO

The ability of fluoxetine, norfluoxetine, sertraline and desmethyl sertraline to inhibit the CYP3A subfamily of cytochromes P450 was examined in vitro, using the formation of 1'-hydroxy midazolam as a probe for CYP3A catalytic activity. The inhibition observed with these four compounds was modeled using competitive, noncompetitive, uncompetitive and mixed competitive/noncompetitive relationships by nonlinear regression analysis. The best fit model of the inhibition of CYP3A-mediated 1'-hydroxy midazolam formation by all four compounds examined was determined to be mixed inhibition. The calculated Ki values were 65.7 +/- 12.0 microM for fluoxetine, 19.1 +/- 1.9 microM for norfluoxetine, 64.4 +/- 11.6 microM for sertraline and 48.1 +/- 11.6 microM for desmethyl sertraline. Steady-state plasma levels of fluoxetine and norfluoxetine can approach a concentration of 1 microM (approximately 350 ng/ml of plasma). Assuming an inhibitor concentration of 1 microM and a concentration of the substrate substantially below its Km (at least 10-fold lower), the results reported predict that fluoxetine and norfluoxetine together would inhibit CYP3A catalytic activity by less than 7% (less than 0.7% if the unbound plasma concentration of fluoxetine is considered). By using the same assumptions and concentrations for sertraline and desmethyl sertraline, these agents together would be predicted to inhibit the metabolic clearance of a coadministered CYP3A metabolized drug by less than 4%. The observations reported here suggest that fluoxetine and sertraline would have little effect on CYP3A-mediated clearance of coadministered drugs.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Midazolam/análogos & derivados , Oxigenases de Função Mista/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , 1-Naftilamina/análogos & derivados , 1-Naftilamina/farmacologia , Catálise , Citocromo P-450 CYP2E1 , Fluoxetina/análogos & derivados , Fluoxetina/farmacologia , Humanos , Microssomos Hepáticos/enzimologia , Midazolam/metabolismo , Sertralina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...