Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 382(1): 31-43, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35489782

RESUMO

Erythropoietin (EPO) and thrombopoietin (TPO) have long been known to promote erythropoiesis and megakaryopoiesis, respectively. However, the fate-changing role of EPO and TPO on megakaryocyte-erythroid progenitors (MEPs) to develop along the erythroid versus megakaryocyte lineage remains unclear. We have previously shown that EPO may have a fate-changing role because EPO treatment could induce progenitor cells depletion and result in EPO resistance. Therefore, we hypothesize that a combination of romiplostim, a TPO receptor agonist that could stimulate the expansion of progenitors, with EPO can treat EPO resistance. Using rats with anemia due to chronic kidney disease, we demonstrated that romiplostim synergized with EPO to promote red blood cells production whereas EPO inhibited platelet production in a dose-dependent manner to reduce the risk of thrombosis. Corroborating findings from in vivo, in vitro experiments demonstrated that romiplostim expanded hematopoietic stem cells and stimulated megakaryopoiesis whereas EPO drove the progenitors toward an erythroid fate. We further developed a novel pharmacokinetic-pharmacodynamic model to quantify the effects of EPO and romiplostim on megakaryopoiesis and erythropoiesis simultaneously. The modeling results demonstrated that EPO increased the differentiation rate of MEPs into burst-forming unit-erythroid cells up to 22-fold, indicating that the slight increase of MEPs induced by romiplostim could be further amplified and recruited by EPO to promote erythropoiesis. The data herein support that romiplostim in combination with EPO can treat EPO resistance. SIGNIFICANCE STATEMENT: This study clarified that erythropoietin (EPO) drives the fate of megakaryocyte-erythroid progenitors (MEPs) toward the erythroid lineage, thus reducing their megakaryocyte (MK) lineage commitment, whereas romiplostim, a thrombopoietin receptor agonist, stimulates megakaryopoiesis through the MK-committed progenitor and MEP bifurcation pathways simultaneously. These findings support an innovative combination of romiplostim and EPO to treat EPO-resistant anemia because the combination therapy further promotes erythropoiesis compared to EPO monotherapy and inhibits platelet production compared to romiplostim monotherapy.


Assuntos
Eritropoetina , Trombopoetina , Animais , Células Precursoras Eritroides , Eritropoetina/farmacologia , Células-Tronco Hematopoéticas , Ratos , Receptores Fc , Proteínas Recombinantes de Fusão , Trombopoetina/farmacologia
2.
J Pharmacokinet Pharmacodyn ; 49(5): 493-510, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36040645

RESUMO

Therapeutic responses of most drugs are initiated by the rate and degree of binding to their receptors or targets. The law of mass action describes the rate of drug-receptor complex association (kon) and dissociation (koff) where the ratio koff/kon is the equilibrium dissociation constant (Kd). Drugs with slow reversible binding (SRB) often demonstrate delayed onset and prolonged pharmacodynamic effects. This report reviews evidence for drugs with SRB features, describes previous pharmacokinetic/pharmacodynamic (PK/PD) modeling efforts of several such drugs, provides a tutorial on the mathematics and properties of SRB models, demonstrates applications of SRB models to additional compounds, and compares PK/PD fittings of SRB with other mechanistic models. We identified and summarized 52 drugs with in vitro-confirmed SRB from a PubMed literature search. Simulations with a SRB model and observed PK/PD profiles showed delayed and prolonged responses and that increasing doses/kon or decreasing koff led to greater expected maximum effects and a longer duration of effects. Recession slopes for return of responses to baseline after single doses were nearly linear with an inflection point that approaches a limiting value at larger doses. The SRB model newly captured literature data for the antihypertensive effects of candesartan and antiallergic effects of noberastine. Their PD profiles could also be fitted with indirect response and biophase models with minimal differences. The applicability of SRB models is probably commonplace, but underappreciated, owing to the need for in vitro confirmation of binding kinetics and the similarity of PK/PD profiles to models with other mechanistic determinants.


Assuntos
Antialérgicos , Anti-Hipertensivos , Cinética , Modelos Biológicos
3.
J Pharmacokinet Pharmacodyn ; 48(5): 687-710, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34100188

RESUMO

Recombinant human erythropoietin (rHuEPO) is effective in managing chronic kidney disease and chemotherapy-induced anemia. However, hyporesponsiveness to rHuEPO treatment was reported in about 10% of the patients. A decreased response in rats receiving a single or multiple doses of rHuEPO was also observed. In this study, we aimed to develop a quantitative systems pharmacology (QSP) model to examine hyporesponsiveness to rHuEPO in rats. Pharmacokinetic (PK) and pharmacodynamic (PD) data after a single intravenous dose of rHuEPO (100 IU/kg) was obtained from a previous study (Yan et al. in Pharm Res, 30:1026-1036, 2013) including rHuEPO plasma concentrations, erythroid precursors counts in femur bone marrow and spleen, reticulocytes (RETs), red blood cells (RBCs), and hemoglobin (HGB) in circulation. Parameter values were obtained from literature or calibrated with experimental data. Global sensitivity analysis and model-based simulations were performed to assess parameter sensitivity and hyporesponsiveness. The final QSP model adequately characterizes time courses of rHuEPO PK and nine PD endpoints in both control and treatment groups simultaneously. The model indicates that negative feedback regulation, neocytolysis, and depletion of erythroid precursors are major factors leading to hyporesponsiveness to rHuEPO treatment in rats.


Assuntos
Eritropoetina/farmacologia , Anemia/induzido quimicamente , Anemia/tratamento farmacológico , Anemia/metabolismo , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Eritropoetina/farmacocinética , Estudos de Avaliação como Assunto , Hemoglobinas/metabolismo , Humanos , Masculino , Ratos , Ratos Wistar , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/metabolismo , Reticulócitos/efeitos dos fármacos , Reticulócitos/metabolismo , Baço/efeitos dos fármacos , Baço/metabolismo
4.
J Pharmacokinet Pharmacodyn ; 48(6): 763-802, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34302262

RESUMO

Delay differential equations (DDEs) are commonly used in pharmacometric models to describe delays present in pharmacokinetic and pharmacodynamic data analysis. Several DDE solvers have been implemented in NONMEM 7.5 for the first time. Two of them are based on algorithms already applied elsewhere, while others are extensions of existing ordinary differential equations (ODEs) solvers. The purpose of this tutorial is to introduce basic concepts underlying DDE based models and to show how they can be developed using NONMEM. The examples include previously published DDE models such as logistic growth, tumor growth inhibition, indirect response with precursor pool, rheumatoid arthritis, and erythropoiesis-stimulating agents. We evaluated the accuracy of NONMEM DDE solvers, their ability to handle stiff problems, and their performance in parameter estimation using both first-order conditional estimation (FOCE) and the expectation-maximization (EM) method. NONMEM control streams and excerpts from datasets are provided for all discussed examples. All DDE solvers provide accurate and precise solutions with the number of significant digits controlled by the error tolerance parameters. For estimation of population parameters, the EM method is more stable than FOCE regardless of the DDE solver.


Assuntos
Algoritmos , Modelos Biológicos , Simulação por Computador
5.
J Pharmacokinet Pharmacodyn ; 48(3): 411-438, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33954911

RESUMO

Population pharmacokinetic/pharmacodynamic (PK/PD) analysis was performed for extensive data for differing dosage forms and routes for dexamethasone (DEX) and betamethasone (BET) in 48 healthy nonpregnant Indian women in a partial and complex cross-over design. Single doses of 6 mg dexamethasone phosphate (DEX-P), betamethasone phosphate (BET-P), or 1:1 mixture of betamethasone phosphate and acetate (BET-PA) were administered orally (PO) or intramuscularly (IM) where each woman enrolled in a two-period cross-over study. Plasma concentrations collected over 96 h were described with a two-compartment model with differing PO and IM first-order absorption inputs. Overall, BET exhibited slower clearance, similar volume of distribution, faster absorption, and longer persistence than DEX with BET acetate producing extremely slow absorption but full bioavailability of BET. Six biomarkers were assessed over a 24-h baseline period with four showing circadian rhythms with complex baselines. These baselines and the strong responses seen after drug dosing were fitted with various indirect response models using the Laplace estimation methods in NONMEM 7.4. Both the PK and six biomarker responses were well-described with modest variability likely due to the homogeneous ages, weights, and ethnicities of the women. The drugs either inhibited or stimulated the influx processes with some models requiring joint inclusion of drug effects on circadian cortisol suppression. The biomarkers and order of sensitivity (lowest IC50/SC50 to highest) were: cortisol, T-helper cells, basophils, glucose, neutrophils, and T-cytotoxic cells. DEX sensitivities were generally greater than BET with corresponding mean ratios for these biomarkers of 2.86, 1.27, 1.72, 1.27, 2.69, and 1.06. Overall, the longer PK (e.g. half-life) of BET, but lesser PD activity (e.g. higher IC50), produces single-dose response profiles that appear quite similar, except for the extended effects from BET-PA. This comprehensive population modeling effort provides the first detailed comparison of the PK profiles and six biomarker responses of five commonly used dosage forms of DEX and BET in healthy women.


Assuntos
Betametasona/farmacocinética , Cronofarmacocinética , Dexametasona/farmacocinética , Modelos Biológicos , Administração Oral , Adulto , Betametasona/administração & dosagem , Biomarcadores , Ritmo Circadiano/fisiologia , Estudos Cross-Over , Dexametasona/administração & dosagem , Relação Dose-Resposta a Droga , Feminino , Meia-Vida , Voluntários Saudáveis , Humanos , Índia , Concentração Inibidora 50 , Injeções Intramusculares , Adulto Jovem
6.
J Pharmacokinet Pharmacodyn ; 48(2): 261-272, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33389521

RESUMO

Population analysis of pharmacokinetic data for five differing dosage forms and routes for dexamethasone and betamethasone in 48 healthy nonpregnant Indian women was performed that accounted for a partial and complex cross-over design. Single doses of 6 mg dexamethasone phosphate (DEX-P), betamethasone phosphate (BET-P), or 1:1 mixture of betamethasone phosphate and acetate (BET-PA) were administered orally (PO) or intramuscularly (IM). Plasma concentrations collected for two periods over 96 h were described with a two-compartment model with differing PO and IM first-order absorption inputs. Clearances and volumes were divided by the IM bioavailability [Formula: see text]. The homogeneous ages, body weights, and ethnicity of the women obviated covariate analysis. Parameter estimates were obtained by the Laplace estimation method implemented in NONMEM 7.4. Typical values for dexamethasone were clearance ([Formula: see text] of 9.29 L/h, steady-state volume ([Formula: see text] of 56.4 L, IM absorption constant [Formula: see text] of 0.460 1/h and oral absorption constant ([Formula: see text] of 0.936 1/h. Betamethasone parameters were CL/FIM of 5.95 L/h, [Formula: see text] of 72.4 L, [Formula: see text] of 0.971 1/h, and [Formula: see text] of 1.21 1/h. The PO to IM F values were close to 1.0 for both drugs. The terminal half-lives averaged about 7.5 h for DEX, 17 h for BET, and 78 h for BET from BET-PA with the latter reflecting very slow release of BET from the acetate ester. Overall, BET exhibited slower clearance, larger volume of distribution, faster absorption, and longer persistence than DEX. These data may be useful in considering exposures when substituting one form of corticosteroid for another.


Assuntos
Corticosteroides , Betametasona , Dexametasona , Adulto , Feminino , Humanos , Adulto Jovem , Administração Oral , Corticosteroides/administração & dosagem , Corticosteroides/farmacocinética , Betametasona/administração & dosagem , Betametasona/farmacocinética , Disponibilidade Biológica , Variação Biológica da População , Estudos Cross-Over , Dexametasona/administração & dosagem , Dexametasona/farmacocinética , Substituição de Medicamentos , Meia-Vida , Voluntários Saudáveis , Índia , Injeções Intramusculares
7.
J Pharmacokinet Pharmacodyn ; 46(1): 53-63, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30617672

RESUMO

In many models of pharmacodynamic systems with delays, a delay of an input is introduced by means of the convolution with the gamma distribution. An approximation of the convolution integral of bound functions based on a system of ordinary differential equations that utilizes properties of the binomial series has been introduced. The approximation converges uniformly on every compact time interval and an estimate of the approximation error has been found [Formula: see text] where [Formula: see text] is the number of differential equations and [Formula: see text] is the shape parameter of the gamma distribution. The accuracy of approximation has been tested on a set of input functions for which the convolution is known explicitly. For tested functions, [Formula: see text] has resulted in an accurate approximation, if [Formula: see text]. However, if [Formula: see text] the error of approximation decreases slowly with increasing [Formula: see text], and [Formula: see text] might be necessary to achieve acceptable accuracy. Finally, the approximation was applied to estimate parameters for the distributed delay model of chemotherapy-induced myelosuppression from previously published WBC count data in rats treated with 5-fluorouracil.


Assuntos
Modelos Biológicos , Algoritmos , Animais , Fluoruracila/farmacologia , Células Precursoras de Granulócitos/efeitos dos fármacos , Humanos , Ratos
8.
J Pharmacokinet Pharmacodyn ; 46(1): 89-101, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30694437

RESUMO

Rhythmicity in baseline responses over a 24-h period for an indirect pharmacological effect R(t) can arise from either a periodic time-dependent input rate [Formula: see text] or a periodic time-dependent loss constant [Formula: see text]. If either [Formula: see text] or [Formula: see text] follows some nonstationary biological rhythm (e.g., circadian), then the response R(t) also displays a periodic behavior. Indirect response models assuming time-dependent input rates [Formula: see text] have been utilized to capture drug effects on various physiological responses such as hormone suppression, immune cell trafficking, and gene expression in tissues. This paradigm was extended to consider responses with circadian-controlled loss [Formula: see text] mechanisms. Theoretical equations describing this model are presented and simulations were performed to examine expected response behaviors. The model was able to capture the chronobiology and pharmacodynamics of applicable drug responses, including the uricosuric effects of lesinurad in humans, suppression of the beta amyloid (Aß) peptide by a gamma-secretase inhibitor in mouse brain, and the modulation of extracellular dopamine by a dopamine transporter inhibitor in rat brain. This type of model has a mechanistic basis and shows utility for capturing drug responses displaying nonstationary baselines controlled by removal mechanism(s).


Assuntos
Relógios Circadianos/efeitos dos fármacos , Preparações Farmacêuticas/administração & dosagem , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Dopamina/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Modelos Biológicos , Ratos
9.
J Pharmacokinet Pharmacodyn ; 45(2): 329-337, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29435950

RESUMO

The distributed delay model has been introduced that replaces the transit compartments in the classic model of chemotherapy-induced myelosuppression with a convolution integral. The maturation of granulocyte precursors in the bone marrow is described by the gamma probability density function with the shape parameter (ν). If ν is a positive integer, the distributed delay model coincides with the classic model with ν transit compartments. The purpose of this work was to evaluate performance of the distributed delay model with particular focus on model deterministic identifiability in the presence of the shape parameter. The classic model served as a reference for comparison. Previously published white blood cell (WBC) count data in rats receiving bolus doses of 5-fluorouracil were fitted by both models. The negative two log-likelihood objective function (-2LL) and running times were used as major markers of performance. Local sensitivity analysis was done to evaluate the impact of ν on the pharmacodynamics response WBC. The ν estimate was 1.46 with 16.1% CV% compared to ν = 3 for the classic model. The difference of 6.78 in - 2LL between classic model and the distributed delay model implied that the latter performed significantly better than former according to the log-likelihood ratio test (P = 0.009), although the overall performance was modestly better. The running times were 1 s and 66.2 min, respectively. The long running time of the distributed delay model was attributed to computationally intensive evaluation of the convolution integral. The sensitivity analysis revealed that ν strongly influences the WBC response by controlling cell proliferation and elimination of WBCs from the circulation. In conclusion, the distributed delay model was deterministically identifiable from typical cytotoxic data. Its performance was modestly better than the classic model with significantly longer running time.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/farmacocinética , Medula Óssea/efeitos dos fármacos , Distribuição Tecidual/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Fluoruracila/farmacocinética , Fluoruracila/farmacologia , Humanos , Contagem de Leucócitos/métodos , Modelos Biológicos , Ratos , Sensibilidade e Especificidade
10.
J Pharmacokinet Pharmacodyn ; 45(2): 277-284, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29290034

RESUMO

Dexmedetomidine (DEX) is a fairly new alfa2-agonist which has been increasingly used in recent years for analgosedation, mostly because it offers a unique ability of providing both moderate level of sedation and analgesia without respiratory depression. Despite of many papers published, there are still only a few concerning the PK of the drug given as long-term infusion in ICU patients. The aim of this work was to characterize the population pharmacokinetics of dexmedetomidine and to investigate the potential benefits of individualization of drug dosing based on patient characteristics in the heterogeneous group of medical and surgical patients staying in intensive care unit. This study was performed in the group of 17 males and 10 females patients with a median age of 59.5 years and median body weight of 75 kg. Blood samples for dexmedetomidine assay were collected from arterial catheter, during and after discontinuation of a standard infusion, that ranged from 24 to 102 h. The following covariates were examined to influence dexmedetomidine PK: age, sex, body weight, patients' health status described by Sequential Organ Failure Assessment Score (SOFA), inotropes usage, and infusion duration. The dexmedetomidine PK was best described by a two-compartment model. The typical values of PK parameters were estimated as 27 L for the volume of the central compartment, 87.6 L for the volume of the peripheral compartment, 38.5 L/h (9.2 mL/min/kg for a 70 kg patient) for systemic clearance and 46.4 L/h for the distribution clearance. Those values are consistent with literature findings. We were unable to show any significant relationship between collected covariates and dexmedetomidine PK. This study does not provide sufficient evidence to support the individualization of dexmedetomidine dosing based on age, sex, body weight, SOFA, and infusion duration.


Assuntos
Dexmedetomidina/farmacocinética , Hipnóticos e Sedativos/farmacocinética , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Infusões Intravenosas/métodos , Unidades de Terapia Intensiva , Masculino , Taxa de Depuração Metabólica/fisiologia , Pessoa de Meia-Idade , Modelos Biológicos , Dinâmica não Linear , Adulto Jovem
11.
J Pharmacokinet Pharmacodyn ; 44(4): 305-316, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28357630

RESUMO

Age-structured cell population model was introduced to describe cell survival. The impact of the environment on the cell population is represented by drug plasma concentration. A key model variable is the hazard of cell removal that is a subject to the environment effect. The model is capable of describing cohort and random labeling cell survival data. In addition, it accounts for cell loss due to labeling of cell sample, but it lacks ability to describe the effect of label elution on the survival data. The model was applied to red blood cell (RBC) survival data in two groups of Wistar rats obtained by two techniques: cohort labeling using 14C-glycine (N = 4) and random labeling using biotin (N = 8). The Weibull probability density function was selected for the RBC lifespan distribution. The data were simultaneously fitted by the mixed effects model implemented in Monolix 4.3.3. The estimated typical values of RBC lifespan and age were 53.7 and 27.8 days, respectively. A noticeable effect of biotinylation on RBC survival was observed that resulted in a significant difference between the means of individual RBC lifespan for two groups. The model provides a mechanistic framework flexible enough to account for various experimental designs to generate the cell survival data. Despite model qualification using animal data, the model has the same potential to be applied to cell survival data analysis in humans.


Assuntos
Senescência Celular/fisiologia , Eritrócitos/fisiologia , Modelos Biológicos , Animais , Biotina/análise , Sobrevivência Celular/fisiologia , Eritrócitos/química , Masculino , Ratos , Ratos Wistar , Coloração e Rotulagem/métodos
12.
Pharm Res ; 33(8): 1833-49, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27059922

RESUMO

PURPOSE: To evaluate and model the pharmacokinetic and pharmacodynamic behavior in rats of FG-3019, a human monoclonal antibody targeting connective tissue growth factor (CTGF). METHODS: FG-3019, human CTGF (rhCTGF), or the N-terminal domain of rhCTGF were administered intravenously to rats and concentrations of these proteins as well as endogenous CTGF were determined by immunoassays. FG-3019, or (125)I-labeled FG-3019, and human CTGF (rhCTGF) were co-administered to assess the impact of CTGF on the elimination rate and tissue localization of FG-3019, which was further characterized by immunohistochemical analysis. A PK/PD model for target-mediated elimination of FG-3019 was developed to fit the kinetic data. RESULTS: FG-3019 exhibited non-linear pharmacokinetics in rats. Circulating concentrations of the N-terminal half of CTGF increased after dosing with FG-3019, reached maximal levels after 1-5 days, and returned toward baseline levels as FG-3019 cleared from the circulation, whereas the concentration of intact CTGF was unaffected by administration of FG-3019. Co-administration of rhCTGF dramatically enhanced the rate of FG-3019 elimination, redistributing the majority of (125)I-labeled FG-3019 from the blood to the liver, kidney, spleen and adrenal gland. FG-3019 co-administered with CTGF was found along the sinusoids of the liver and adrenal glands, the capillaries of the kidney glomeruli and in the spleen. A pharmacokinetic model for target-mediated elimination of FG-3019 was used to fit the time courses of FG-3019 and endogenous CTGF plasma concentrations, as well as time courses of rhCTGF and rhCTGF N-fragment after intravenous administration of these species. CONCLUSIONS: FG-3019 is subject to target mediated elimination in rats.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Fator de Crescimento do Tecido Conjuntivo/administração & dosagem , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Administração Intravenosa , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Relação Dose-Resposta a Droga , Interações Medicamentosas/fisiologia , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia
14.
J Pharmacokinet Pharmacodyn ; 43(5): 513-27, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27612462

RESUMO

We aimed to develop a cell-level pharmacodynamics-mediated drug disposition (PDMDD) model to analyze in vivo systems where the PD response to a drug has an appreciable effect on the pharmacokinetics (PK). An existing cellular level model of PD stimulation was combined with the standard target-mediated drug disposition (TMDD) model and the resulting model structure was parametrically identifiable from typical in vivo PK and PD data. The PD model of the cell population was controlled by the production rate k in and elimination rate k out which could be stimulated or inhibited by the number of bound receptors on a single cell. Simulations were performed to assess the impact of single and repeated dosing on the total drug clearance. The clinical utility of the cell-level PDMDD model was demonstrated by fitting published data on the stimulatory effects of filgrastim on absolute neutrophil counts in healthy subjects. We postulated repeated dosing as a means of detecting and quantifying PDMDD as a single dose might not be sufficient to elicit the cellular response capable of altering the receptor pool to visibly affect drug disposition. In the absence of any PD effect, the model reduces down to the standard TMDD model. The applications of this model can be readily extended to include chemotherapy-induced cytopenias affecting clearance of endogenous hematopoietic growth factors, different monoclonal antibodies and immunogenicity effects on PK.


Assuntos
Filgrastim/farmacocinética , Fármacos Hematológicos/farmacocinética , Modelos Biológicos , Neutrófilos/efeitos dos fármacos , Receptores de Droga/metabolismo , Transporte Biológico , Simulação por Computador , Relação Dose-Resposta a Droga , Filgrastim/administração & dosagem , Fármacos Hematológicos/administração & dosagem , Fármacos Hematológicos/sangue , Humanos , Taxa de Depuração Metabólica , Neutrófilos/citologia , Neutrófilos/metabolismo , Dinâmica não Linear , Ligação Proteica , Distribuição Tecidual
15.
J Pharmacokinet Pharmacodyn ; 42(5): 573-89, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26377617

RESUMO

The purpose of this work is to review basic pharmacodynamic (PD) models describing drug effects on cell populations and expand them to age-structured models using the theory of physiologically structured populations. The plasma drug concentrations are interpreted as the environment affecting the cell production and mortality rates. An explicit solution to model equations provides the age density distribution that serves to establish a relationship between the cell lifespan distribution and the hazard of cell removal. Given the lifespan distributions, the age distributions for most commonly applied PD models of cell responses including basic cell turnover, transit compartments, and basic lifespan models have been derived both for the baseline conditions and drug treatment. The steady-state age distribution for basic indirect response models is exponential, and it is uniform for the basic lifespan model. As an example of more complex cell population, the age distribution of human red blood cells has been simulated based on a recent model of red blood cell survival. The age distribution for cells in the transit compartment model is the sum of the gamma functions. Means and variances of age distributions for all discussed models were calculated. A brief discussion of numerical challenges and possible future model developments is presented.


Assuntos
Eritrócitos/efeitos dos fármacos , Modelos Biológicos , Preparações Farmacêuticas/administração & dosagem , Algoritmos , Sobrevivência Celular/efeitos dos fármacos , Simulação por Computador , Humanos , Modelos Estatísticos
16.
J Pharmacokinet Pharmacodyn ; 42(6): 709-20, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26341875

RESUMO

A mechanistic model describing the effects of chemotherapy and radiation on platelet counts and endogenous thrombopoietin (eTPO) in mice was developed. Thrombocytopenia was induced in mice by injection of carboplatin followed by the whole body irradiation on days 0, 28, and 56, with platelet and eTPO samples collected over 84 days. The pharmacodynamic model consisted of a series of aging compartments representing proliferating megakaryocyte precursors, megakaryocytes, and platelets with possible eTPO clearance through internalization. The cytotoxic effects of treatment were described by the kinetics of the effect (K-PD) model, and stimulation of platelet production by eTPO was considered to be driven by receptor occupancy. The proposed PD model adequately described the platelet counts and eTPO concentrations in mice by accounting for nadirs and peaks of platelet count, and rebounds in eTPO time course profiles. The estimates of model parameters were in good agreement with their physiological values reported in literature for mice with platelet lifespan of 4.3 days and 185 cMpl receptors per platelet. The predicted duration of the treatment effect was 0.82 h (approximately 5 carboplatin half-lives in mice). The data was not informative about the eTPO stimulatory effect as the nominal precursor production rate was sufficient to account for platelet response to treatment. The model quantified the inverse relationship between eTPO levels and platelet counts and offered an explanation of the tolerance effect observed in the eTPO data. The simulated rebound in free receptors levels correlated with rebounds in eTPO levels. The model suggests that the duration of the toxic effects is determined by the turnover of the proliferating cells in the bone marrow. This indicates that the lifespan of the target cells (megakaryocyte precursors, megakaryocytes and platelets) is a key determinant in the duration of both drug exposure and toxicity due to treatment. The model can be extended to account for pharmacokinetics of exogenous drugs and be applied to analysis of human data.


Assuntos
Antineoplásicos/toxicidade , Plaquetas/efeitos dos fármacos , Plaquetas/efeitos da radiação , Carboplatina/toxicidade , Quimiorradioterapia/efeitos adversos , Modelos Biológicos , Modelos Estatísticos , Lesões por Radiação/induzido quimicamente , Trombocitopenia/induzido quimicamente , Irradiação Corporal Total/efeitos adversos , Animais , Antineoplásicos/administração & dosagem , Biomarcadores/sangue , Plaquetas/metabolismo , Plaquetas/patologia , Carboplatina/administração & dosagem , Simulação por Computador , Esquema de Medicação , Feminino , Humanos , Camundongos , Contagem de Plaquetas , Doses de Radiação , Lesões por Radiação/sangue , Receptores de Trombopoetina/sangue , Medição de Risco , Trombocitopenia/sangue , Trombopoese/efeitos dos fármacos , Trombopoese/efeitos da radiação , Trombopoetina/sangue , Fatores de Tempo
17.
J Pharmacokinet Pharmacodyn ; 41(4): 291-318, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25142056

RESUMO

In pharmacokinetics/pharmacodynamics (PKPD) the measured response is often delayed relative to drug administration, individuals in a population have a certain lifespan until they maturate or the change of biomarkers does not immediately affects the primary endpoint. The classical approach in PKPD is to apply transit compartment models (TCM) based on ordinary differential equations to handle such delays. However, an alternative approach to deal with delays are delay differential equations (DDE). DDEs feature additional flexibility and properties, realize more complex dynamics and can complementary be used together with TCMs. We introduce several delay based PKPD models and investigate mathematical properties of general DDE based models, which serve as subunits in order to build larger PKPD models. Finally, we review current PKPD software with respect to the implementation of DDEs for PKPD analysis.


Assuntos
Matemática , Farmacocinética , Algoritmos , Simulação por Computador , Humanos , Absorção Intestinal , Modelos Estatísticos , Modelos Teóricos
18.
Biopharm Drug Dispos ; 35(6): 330-40, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24888906

RESUMO

A technique has recently been proposed for obtaining the reticulocyte (RET) age distribution from the flow cytometric reticulocyte count. It allows for a quantitative characterization of reticulocyte dynamics. In this work this technique was applied to characterize the blood, bone marrow and spleen reticulocytes in homeostatic and erythropoietically stimulated rats in order to determine the reticulocyte maturation times in the bone marrow and blood; and to confirm the presence of ineffective erythropoiesis (neocytolysis). The latter was done by comparing the reticulocyte removal rate from blood with bilirubin formation after erythropoiesis stimulation. A single subcutaneous dose (4050 IU/kg) of recombinant human erythropoietin (rHuEPO) was administered to rats, then their reticulocytes were stained with thiazole orange and the distribution of the fluorescent signal measured using flow cytometry. The obtained signal distribution of the reticulocytes was transformed to the age distribution and a set of basic parameters reflecting reticulocyte dynamics was determined. Bilirubin concentrations were measured to directly assess the presence of reticulocyte irreversible removal. The bilirubin formation was found to be considerably modulated by rHuEPO and corresponded well to the determined reticulocyte removal rate. The initial increase and subsequent decrease of the reticulocyte maturation time in blood was quantitated and directly linked with RET mobilization from the bone marrow. A substantial number (60%) of reticulocytes is sequestrated during homeostasis in rats. This number increases and then decreases after rHuEPO administration, as also reflected by bilirubin formation. Flow cytometry seems to be an excellent method for studying RET dynamics and the presence of young RBC neocytolysis.


Assuntos
Eritropoese/efeitos dos fármacos , Eritropoetina/farmacologia , Reticulócitos/citologia , Reticulócitos/efeitos dos fármacos , Animais , Bilirrubina/metabolismo , Células da Medula Óssea/citologia , Epoetina alfa , Contagem de Eritrócitos , Citometria de Fluxo , Hemoglobinas/metabolismo , Homeostase , Masculino , Ratos Wistar , Proteínas Recombinantes/farmacologia , Reticulócitos/fisiologia , Baço/citologia
19.
J Pharm Sci ; 113(1): 257-267, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37926235

RESUMO

OBJECTIVES: Cell trafficking encompasses movement of the immune system cells (e.g., granulocytes, lymphocytes) between the blood and the extravascular tissues (e.g., lymph nodes). Corticosteroids are known to suppress cell trafficking. The age-structured cell population models introduce the transit time as a structure that allows one to quantify the distribution of times the immune cells spend in the blood and the extravascular tissues. The objective of this work is to develop an age-structured cell population model describing drug effects on cell trafficking and to implement the model in pharmacometric software to enable parameter estimation and simulations. METHODS: We adopted the well-known McKendrick age-structured population model to describe the age distributions in two cell populations: blood cells and cells in the extravascular space. The hazard of cell recirculation from the extravascular tissues was age dependent and described by the Weibull function with the shape ν and scale ß parameters. The drug effect on cell trafficking was modeled as the product of the Emax function of the drug plasma concentration and the Weibull hazard. The model was implemented in NONMEM 7.5.1. The model was applied to the basophil data in 34 healthy subjects who received a single intramuscular or oral dose of 6 mg dexamethasone (DEX). A recently published pharmacokinetic model was applied to describe DEX plasma concentration. Typical values of parameter estimates were further used to simulate the DEX effect of the basophil mean transit time in the extravascular tissues. RESULTS: Simulations of basophil time courses for varying ν demonstrated that the rebound in the blood count data following drug administration is only possible for ν >1. The estimates of model parameters were ν = 3.02, ß = 0.00863 1/h, and IC50 = 7.47 ng/mL. The calculated baseline mean transit times of basophils in the blood 7.2 h and extravascular tissues 104.9 h agree with the values reported in the literature. CONCLUSIONS: We introduced an age-structured population model to describe cell trafficking between the blood and extravascular tissues. The model was adopted to account for the inhibitory drug effect on the cell recirculation. We showed that the age structure is essential to explain the rebound observed in the blood count response to a single dose drug administration. The model was validated using the basophil responses to DEX treatment in healthy subjects.


Assuntos
Modelos Biológicos , Software , Humanos , Linfócitos , Relação Dose-Resposta a Droga
20.
Pharm Res ; 30(4): 1026-36, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23187865

RESUMO

PURPOSE: The purpose of this study is to demonstrate that the erythroid precursor depletion in bone marrow induced by recombinant human erythropoietin (rHuEPO) treatment may be another contributing factor to erythropoietin hyporesponsiveness. METHODS: Healthy Wistar rats were given single dose (SD) or multiple doses (MD) of rHuEPO (100 IU/kg). In MD study, animals were challenged with thrice-weekly over two weeks. Blood, bone marrow and spleen (for SD only) were collected. The erythropoietic responses in bone marrow and spleen were quantified using a flow cytometric immunophenotyping technique. A mathematical approach involving measuring reticulocyte age distribution was developed to evaluate the reticulocyte loss due to neocytolysis. RESULTS: A reduced level of erythropoietic responses below the baseline was observed for both MD and SD studies. In SD study, the reticulocyte decreased below the baseline after day 6. A depletion of the bone marrow erythroid precursor cells was observed. However, neocytolysis of reticulocyte only occurs from day 3-5 after rHuEPO injection. CONCLUSIONS: The findings demonstrate that EPO-induced erythroid precursor depletion in bone marrow is responsible for reduced reticulocyte response and may contribute to erythropoietin hyporesponsiveness. Therefore, this study provides further justification for reducing the doses of erythropoietin-stimulating agents in anemic patients demonstrating hyporesponsiveness.


Assuntos
Células Precursoras Eritroides/efeitos dos fármacos , Eritropoetina/farmacologia , Proteínas Recombinantes/farmacologia , Administração Intravenosa , Animais , Medula Óssea/efeitos dos fármacos , Eritropoese/efeitos dos fármacos , Eritropoetina/administração & dosagem , Eritropoetina/efeitos adversos , Eritropoetina/sangue , Humanos , Masculino , Ratos , Ratos Wistar , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/sangue , Reticulócitos/efeitos dos fármacos , Baço/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA