Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
MAbs ; 15(1): 2261509, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37823690

RESUMO

There are few treatments that slow neurodegeneration in Alzheimer's disease (AD), and while therapeutic antibodies are being investigated in clinical trials for AD treatment, their access to the central nervous system is restricted by the blood-brain barrier. This study investigates a bispecific modular fusion protein composed of gantenerumab, a fully human monoclonal anti- amyloid-beta (Aß) antibody under investigation for AD treatment, with a human transferrin receptor 1-directed Brainshuttle™ module (trontinemab; RG6102, INN trontinemab). In vitro, trontinemab showed a similar binding affinity to fibrillar Aß40 and Aß plaques in human AD brain sections to gantenerumab. A single intravenous administration of trontinemab (10 mg/kg) or gantenerumab (20 mg/kg) to non-human primates (NHPs, Macaca fascicularis), was well tolerated in both groups. Immunohistochemistry indicated increased trontinemab uptake into the brain endothelial cell layer and parenchyma, and more homogeneous distribution, compared with gantenerumab. Brain and plasma pharmacokinetic (PK) parameters for trontinemab were estimated by nonlinear mixed-effects modeling with correction for tissue residual blood, indicating a 4-18-fold increase in brain exposure. A previously developed clinical PK/pharmacodynamic model of gantenerumab was adapted to include a brain compartment as a driver of plaque removal and linked to the allometrically scaled above model from NHP. The new brain exposure-based model was used to predict trontinemab dosing regimens for effective amyloid reduction. Simulations from these models were used to inform dosing of trontinemab in the first-in-human clinical trial.


Assuntos
Doença de Alzheimer , Anticorpos Monoclonais , Animais , Humanos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/uso terapêutico , Anticorpos Monoclonais/farmacologia , Encéfalo/metabolismo , Primatas/metabolismo
2.
CPT Pharmacometrics Syst Pharmacol ; 12(2): 139-143, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36418887

RESUMO

Immunogenicity against therapeutic proteins frequently causes attrition owing to its potential impact on pharmacokinetics, pharmacodynamics, efficacy, and safety. Predicting immunogenicity is complex because of its multifactorial drivers, including compound properties, subject characteristics, and treatment parameters. To integrate these, the Immunogenicity Simulator was developed using published, predominantly late-stage trial data from 15 therapeutic proteins. This single-blinded evaluation with subject-level data from 10 further monoclonals assesses the Immunogenicity Simulator's credibility for application during the drug development process.


Assuntos
Desenvolvimento de Medicamentos , Farmacologia em Rede , Humanos , Proteínas/imunologia , Proteínas/uso terapêutico
3.
Alzheimers Dement ; 19(6): 2287-2297, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36454709

RESUMO

INTRODUCTION: A quantitative model of Alzheimer's disease (AD) based on the amyloid/tau/neurodegeneration biomarker framework (Q-ATN model) was developed to sequentially link amyloid positron emission tomography (PET), tau PET, medial temporal cortical thickness, and clinical outcome (Clinical Dementia Rating - Sum of Boxes; CDR-SB). METHODS: Published data and biologically plausible mechanisms were used to construct, calibrate, and validate the model. Clinical trial simulations were performed for different anti-amyloid antibodies, including a 5-year simulation of subcutaneous gantenerumab treatment. RESULTS: The simulated time-course of biomarkers and CDR-SB was consistent with natural history studies and described the effects of several anti-amyloid antibodies observed in trials with positive and negative (or non-significant) outcomes. The 5-year simulation predicts that the beneficial effects of continued anti-amyloid treatment should increase markedly over time. DISCUSSION: The Q-ATN model offers a novel approach for linking amyloid PET to CDR-SB, and provides theoretical support for the potential clinical benefit of anti-amyloid therapy. HIGHLIGHTS: A semi-mechanistic model was developed to link amyloid/tau/neurodegeneration biomarkers to clinical outcome (Q-ATN model). The Q-ATN model describes the disease progression seen in natural history studies. Model simulations agree well with mean data from the aducanumab EMERGE study. A 5-year simulation of gantenerumab predicts greater benefit with longer treatment.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/tratamento farmacológico , Amiloide , Tomografia por Emissão de Pósitrons , Biomarcadores , Proteínas Amiloidogênicas , Peptídeos beta-Amiloides , Proteínas tau
4.
Alzheimers Dement (N Y) ; 8(1): e12306, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35676943

RESUMO

Introduction: Amyloid-related imaging abnormalities with edema/effusion (ARIA-E) are commonly observed with anti-amyloid therapies in Alzheimer's disease. We developed a semi-mechanistic, in silico model to understand the time course of ARIA-E and its dose dependency. Methods: Dynamic and statistical analyses of data from 112 individuals that experienced ARIA-E in the open-label extension of SCarlet RoAD (a study of gantenerumab in participants with prodromal Alzheimer's disease) and Marguerite RoAD (as study of Gantenerumab in participants with mild Alzheimer's disease) studies were used for model building. Gantenerumab pharmacokinetics, local amyloid removal, disturbance and repair of the vascular wall, and ARIA-E magnitude were represented in the novel vascular wall disturbance (VWD) model of ARIA-E. Results: The modeled individual-level profiles provided a good representation of the observed pharmacokinetics and time course of ARIA-E magnitude. ARIA-E dynamics were shown to depend on the interplay between drug-mediated amyloid removal and intrinsic vascular repair processes. Discussion: Upon further refinement and validation, the VWD model could inform strategies for dosing and ARIA monitoring in individuals with an ARIA-E history.

5.
Int J Mol Sci ; 23(2)2022 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-35054865

RESUMO

The prediction of monoclonal antibody (mAb) disposition within solid tumors for individual patients is difficult due to inter-patient variability in tumor physiology. Improved a priori prediction of mAb pharmacokinetics in tumors may facilitate the development of patient-specific dosing protocols and facilitate improved selection of patients for treatment with anti-cancer mAb. Here, we report the use of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), with tumor penetration of the contrast agent gadobutrol used as a surrogate, to improve physiologically based pharmacokinetic model (PBPK) predictions of cetuximab pharmacokinetics in epidermal growth factor receptor (EGFR) positive xenografts. In the initial investigations, mice bearing Panc-1, NCI-N87, and LS174T xenografts underwent DCE-MRI imaging with the contrast agent gadobutrol, followed by intravenous dosing of an 125Iodine-labeled, non-binding mAb (8C2). Tumor concentrations of 8C2 were determined following the euthanasia of mice (3 h-6 days after 8C2 dosing). Potential predictor relationships between DCE-MRI kinetic parameters and 8C2 PBPK parameters were evaluated through covariate modeling. The addition of the DCE-MRI parameter Ktrans alone or Ktrans in combination with the DCE-MRI parameter Vp on the PBPK parameters for tumor blood flow (QTU) and tumor vasculature permeability (σTUV) led to the most significant improvement in the characterization of 8C2 pharmacokinetics in individual tumors. To test the utility of the DCE-MRI covariates on a priori prediction of the disposition of mAb with high-affinity tumor binding, a second group of tumor-bearing mice underwent DCE-MRI imaging with gadobutrol, followed by the administration of 125Iodine-labeled cetuximab (a high-affinity anti-EGFR mAb). The MRI-PBPK covariate relationships, which were established with the untargeted antibody 8C2, were implemented into the PBPK model with considerations for EGFR expression and cetuximab-EGFR interaction to predict the disposition of cetuximab in individual tumors (a priori). The incorporation of the Ktrans MRI parameter as a covariate on the PBPK parameters QTU and σTUV decreased the PBPK model prediction error for cetuximab tumor pharmacokinetics from 223.71 to 65.02%. DCE-MRI may be a useful clinical tool in improving the prediction of antibody pharmacokinetics in solid tumors. Further studies are warranted to evaluate the utility of the DCE-MRI approach to additional mAbs and additional drug modalities.


Assuntos
Anticorpos Monoclonais/imunologia , Meios de Contraste/química , Imageamento por Ressonância Magnética , Neoplasias/imunologia , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Linhagem Celular Tumoral , Cetuximab/imunologia , Cetuximab/farmacocinética , Humanos , Masculino , Camundongos , Modelos Biológicos , Neoplasias/sangue , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico
6.
Pharmaceutics ; 13(12)2021 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-34959386

RESUMO

The goal of this study is to investigate the pharmacokinetics in plasma and tumour interstitial fluid of two T-cell bispecifics (TCBs) with different binding affinities to the tumour target and to assess the subsequent cytokine release in a tumour-bearing humanised mouse model. Pharmacokinetics (PK) as well as cytokine data were collected in humanised mice after iv injection of cibisatamab and CEACAM5-TCB which are binding with different binding affinities to the tumour antigen carcinoembryonic antigen (CEA). The PK data were modelled and coupled to a previously published physiologically based PK model. Corresponding cytokine release profiles were compared to in vitro data. The PK model provided a good fit to the data and precise estimation of key PK parameters. High tumour interstitial concentrations were observed for both TCBs, influenced by their respective target binding affinities. In conclusion, we developed a tailored experimental method to measure PK and cytokine release in plasma and at the site of drug action, namely in the tumour. Integrating those data into a mathematical model enabled to investigate the impact of target affinity on tumour accumulation and can have implications for the PKPD assessment of the therapeutic antibodies.

8.
AAPS J ; 22(3): 63, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32246215

RESUMO

Minipigs have been proposed as animal model to study the subcutaneous (SC) absorption of monoclonal antibodies (mAb), because they are more translatable to humans than other species. However, the minipig SC tissue structure differs markedly depending on its location. This study explored different SC administration sites for mAb SC administration, to explore which site translates best to humans. The study assessed the SC absorption of tocilizumab (Actemra®) following administration at several injection sites: Inguinal area, flank, caudal to the ear, and interscapular area, in comparison with an IV group. After SC administration, tocilizumab absorption was most rapid from the inguinal administration site, and slowest after administration behind the ear, with absorption from the other sites in between. Tocilizumab bioavailability was 98.6, 88.3, 74.1, and 86.3% after administration in inguinal area, flank, behind the ear, and interscapular area, as determined by non-compartmental analysis. Fitting of a single first-order absorption rate constant by compartmental analysis was dissatisfactory. A combined fitting of all data was done assuming two different kinds of SC depots, one undergoing fast absorption, the other undergoing a slower absorption. The split between these absorption depots differed across administration sites, with absorption from "fast depot" in inguinal area > flank > interscapular area > behind the ear. Comparisons with clinical data show that tocilizumab PK after SC administration behind the ear translates best to humans, considering both bioavailability and rate of absorption. Whether this translation from minipigs to humans is prototypic for other mAb remains to be confirmed.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Modelos Animais , Absorção Cutânea , Porco Miniatura , Animais , Anticorpos Monoclonais Humanizados/farmacocinética , Humanos , Injeções Subcutâneas , Masculino , Modelos Biológicos , Suínos
9.
J Pharm Sci ; 108(11): 3729-3736, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31351865

RESUMO

The pharmacokinetics (PK) of the anti-CD20 monoclonal antibody obinutuzumab was assessed after single intravenous dosing to cynomolgus monkeys. In addition, the pharmacokinetic-pharmacodynamic (PKPD) relationship for B-cell depletion was characterized. The PKPD model was used to estimate the B-cell repopulation during the recovery phase of chronic toxicology studies, thereby supporting the study design, in particular planning the recovery phase duration. Marked immunogenicity against obinutuzumab was observed approximately 10 days after single dose, leading to an up to ∼30-fold increase in obinutuzumab clearance in the affected monkeys. Despite this accelerated clearance, the PK could be characterized, either by disregarding the clearance in noncompartmental PK analysis or by capturing it explicitly as an additional time-dependent clearance process in compartmental modeling. This latter step was crucial to model the PKPD of B-cells as an indirect response to obinutuzumab exposure, showing that-without immune response-the limiting factor is obinutuzumab elimination with concentrations below 0.02 µg/mL required for initiation of B-cell recovery. Overall, the results demonstrate that despite a marked anti-drug antibody response in the nonclinical animal species, the PK and PKPD of obinutuzumab could be characterized successfully by appropriately addressing the immune-modulated clearance pathway in data analysis and modeling.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais/farmacocinética , Antígenos CD20/metabolismo , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Macaca fascicularis
10.
MAbs ; 10(5): 803-813, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29621428

RESUMO

The neonatal Fc receptor (FcRn) has been demonstrated to contribute to a high bioavailability of monoclonal antibodies (mAbs). In this study, we explored the cellular sites of FcRn-mediated protection after subcutaneous (SC) and intravenous (IV) administration. SC absorption and IV disposition kinetics of a mAb were studied in hFcRn transgenic (Tg) bone marrow chimeric mice in which hFcRn was restricted to radioresistant cells or hematopoietic cells. SC bioavailabilities close to 90% were observed in hFcRn Tg mice and chimeric mice with hFcRn expression in hematopoietic cells, whereas SC bioavailabilities were markedly lower when FcRn was missing in hematopoietic cells. Our study demonstrates: 1) FcRn in radiosensitive hematopoietic cells is required for high SC bioavailability, indicating first-pass catabolism after SC administration by hematopoietic cells; 2) FcRn-mediated transcytosis or recycling by radioresistent cells is not required for high SC bioavailability; and 3) after IV administration hematopoietic and radioresistent cells contribute about equally to clearance of the mAb. A pharmacokinetic model was devised to describe a mixed elimination via radioresistent and hematopoietic cells from vascular and extravascular compartments, respectively. Overall, the study indicates a relevant role of hematopoietic cells for first-pass clearance of mAbs after SC administration and confirms their role in the overall clearance of mAbs.


Assuntos
Anticorpos Monoclonais/farmacocinética , Células-Tronco Hematopoéticas/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Receptores Fc/metabolismo , Administração Intravenosa , Animais , Anticorpos Monoclonais/administração & dosagem , Disponibilidade Biológica , Transplante de Células-Tronco Hematopoéticas , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Injeções Subcutâneas , Taxa de Depuração Metabólica , Camundongos , Camundongos Transgênicos , Receptores Fc/genética
11.
Clin Cancer Res ; 24(14): 3325-3333, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29463551

RESUMO

Purpose: Optimal dosing is critical for immunocytokine-based cancer immunotherapy to maximize efficacy and minimize toxicity. Cergutuzumab amunaleukin (CEA-IL2v) is a novel CEA-targeted immunocytokine. We set out to develop a mathematical model to predict intratumoral CEA-IL2v concentrations following various systemic dosing intensities.Experimental Design: Sequential measurements of CEA-IL2v plasma concentrations in 74 patients with solid tumors were applied in a series of differential equations to devise a model that also incorporates the peripheral concentrations of IL2 receptor-positive cell populations (i.e., CD8+, CD4+, NK, and B cells), which affect tumor bioavailability of CEA-IL2v. Imaging data from a subset of 14 patients were subsequently utilized to additionally predict antibody uptake in tumor tissues.Results: We created a pharmacokinetic/pharmacodynamic mathematical model that incorporates the expansion of IL2R-positive target cells at multiple dose levels and different schedules of CEA-IL2v. Model-based prediction of drug levels correlated with the concentration of IL2R-positive cells in the peripheral blood of patients. The pharmacokinetic model was further refined and extended by adding a model of antibody uptake, which is based on drug dose and the biological properties of the tumor. In silico predictions of our model correlated with imaging data and demonstrated that a dose-dense schedule comprising escalating doses and shortened intervals of drug administration can improve intratumoral drug uptake and overcome consumption of CEA-IL2v by the expanding population of IL2R-positive cells.Conclusions: The model presented here allows simulation of individualized treatment plans for optimal dosing and scheduling of immunocytokines for anticancer immunotherapy. Clin Cancer Res; 24(14); 3325-33. ©2018 AACRSee related commentary by Ruiz-Cerdá et al., p. 3236.


Assuntos
Citocinas/administração & dosagem , Fatores Imunológicos/administração & dosagem , Modelos Teóricos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Antineoplásicos Imunológicos/administração & dosagem , Antineoplásicos Imunológicos/efeitos adversos , Antineoplásicos Imunológicos/farmacocinética , Biomarcadores , Citocinas/efeitos adversos , Citocinas/farmacocinética , Humanos , Fatores Imunológicos/efeitos adversos , Fatores Imunológicos/farmacocinética , Imunoterapia , Interleucina-2/administração & dosagem , Interleucina-2/efeitos adversos , Interleucina-2/farmacocinética , Modelos Biológicos , Imagem Molecular , Neoplasias/diagnóstico , Neoplasias/mortalidade , Prognóstico , Receptores de Interleucina-2/metabolismo , Resultado do Tratamento
12.
MAbs ; 9(6): 1007-1015, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28613103

RESUMO

Monoclonal antibodies are an important therapeutic entity, and knowledge of antibody pharmacokinetics has steadily increased over the years. Despite this effort, little is known about the extent of IgG antibody degradation in different tissues of the body. While studies have been published identifying sites of degradation with the use of residualizing and non-residualizing radiolabels, quantitative tissue clearances have not yet been derived. Here, we show that in physiologically-based pharmacokinetic (PBPK) models we can combine mouse data of Indium-111 and Iodine-125 labeled antibodies with prior physiologic knowledge to determine tissue-specific intrinsic clearances. Unspecific total tissue clearance (mL/day) in the mouse was estimated to be: liver = 4.75; brain = 0.02; gut = 0.40; heart = 0.07; kidney = 0.97; lung = 0.20; muscle = 3.02; skin = 3.89; spleen = 0.45; rest of body = 2.16. The highest catabolic activity (per g tissue) was in spleen for an FcRn wild-type antibody, but shifts to the liver for an antibody with reduced FcRn affinity. In the model developed, this shift can be explained by the liver having a greater FcRn-mediated protection capacity than the spleen. The quantification of tissue intrinsic clearances and FcRn salvage capacity increases our understanding of quantitative processes that drive the therapeutic responses of antibodies. This knowledge is critical, for instance to estimate the non-specific cellular uptake and degradation of antibodies used for targeted delivery of payloads.

13.
Clin Cancer Res ; 22(17): 4417-27, 2016 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-27117182

RESUMO

PURPOSE: CEA TCB (RG7802, RO6958688) is a novel T-cell bispecific antibody, engaging CD3ε upon binding to carcinoembryonic antigen (CEA) on tumor cells. Containing an engineered Fc region, conferring an extended blood half-life while preventing side effects due to activation of innate effector cells, CEA TCB potently induces tumor lysis in mouse tumors. Here we aimed to characterize the pharmacokinetic profile, the biodistribution, and the mode of action of CEA TCB by combining in vitro and in vivo fluorescence imaging readouts. EXPERIMENTAL DESIGN: CEA-expressing tumor cells (LS174T) and human peripheral blood mononuclear cells (PBMC) were cocultured in vitro or cografted into immunocompromised mice. Fluorescence reflectance imaging and intravital 2-photon (2P) microscopy were employed to analyze in vivo tumor targeting while in vitro confocal and intravital time-lapse imaging were used to assess the mode of action of CEA TCB. RESULTS: Fluorescence reflectance imaging revealed increased ratios of extravascular to vascular fluorescence signals in tumors after treatment with CEA TCB compared with control antibody, suggesting specific targeting, which was confirmed by intravital microscopy. Confocal and intravital 2P microscopy showed CEA TCB to accelerate T-cell-dependent tumor cell lysis by inducing a local increase of effector to tumor cell ratios and stable crosslinking of multiple T cells to individual tumor cells. CONCLUSIONS: Using optical imaging, we demonstrate specific tumor targeting and characterize the mode of CEA TCB-mediated target cell lysis in a mouse tumor model, which supports further clinical evaluation of CEA TCB. Clin Cancer Res; 22(17); 4417-27. ©2016 AACRSee related commentary by Teijeira et al., p. 4277.


Assuntos
Anticorpos Biespecíficos/imunologia , Antígeno Carcinoembrionário/imunologia , Citotoxicidade Imunológica , Imagem Molecular , Neoplasias/diagnóstico por imagem , Neoplasias/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Biespecíficos/metabolismo , Anticorpos Biespecíficos/farmacologia , Especificidade de Anticorpos/imunologia , Antineoplásicos Imunológicos/metabolismo , Antineoplásicos Imunológicos/farmacologia , Biomarcadores , Comunicação Celular/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Microscopia Confocal , Imagem Molecular/métodos , Neoplasias/metabolismo , Neoplasias/terapia , Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Fatores de Tempo , Distribuição Tecidual
14.
Mol Pharm ; 13(2): 586-98, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26674605

RESUMO

Drug absorption is a complex process involving dissolution and precipitation, along with other kinetic processes. The purpose of this work was to (1) establish an in vitro methodology to study dissolution and precipitation in early stages of drug development where low compound consumption and high throughput are necessary, (2) develop a mathematical model for a mechanistic explanation of generated in vitro dissolution and precipitation data, and (3) extrapolate in vitro data to in vivo situations using physiologically based models to predict oral drug absorption. Small-scale pH-shift studies were performed in biorelevant media to monitor the precipitation of a set of poorly soluble weak bases. After developing a dissolution-precipitation model from this data, it was integrated into a simplified, physiologically based absorption model to predict clinical pharmacokinetic profiles. The model helped explain the consequences of supersaturation behavior of compounds. The predicted human pharmacokinetic profiles closely aligned with the observed clinical data. In summary, we describe a novel approach combining experimental dissolution/precipitation methodology with a mechanistic model for the prediction of human drug absorption kinetics. The approach unifies the dissolution and precipitation theories and enables accurate predictions of in vivo oral absorption by means of physiologically based modeling.


Assuntos
Cloridrato de Erlotinib/farmacocinética , Absorção Gastrointestinal/efeitos dos fármacos , Modelos Biológicos , Permeabilidade/efeitos dos fármacos , Administração Oral , Simulação por Computador , Cloridrato de Erlotinib/administração & dosagem , Humanos , Cinética , Distribuição Tecidual
15.
Pharm Res ; 31(10): 2696-707, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24792825

RESUMO

PURPOSE: Onset and rate of gastric emptying are important determinants of drug absorption after oral dosing. Therefore, robust estimates of these parameters are needed in physiologically based absorption models to predict reliably plasma concentration time profiles. For human and some other laboratory animals, reasonable parameterization of gastric emptying has been established. However gastric emptying is less well characterized in minipigs, a large animal model rapidly gaining importance in pharmaceutical research. METHODS: A pharmacokinetic crossover study using different dosage forms of paracetamol (intravenous and oral solution, capsule and tablet) was conducted in four male and four female Göttingen minipigs after an overnight fast. Deconvolution analysis was performed to determine the absorption kinetics. Estimated lag times and first order gastric emptying parameters were incorporated in a previously published PBPK model of the minipig and simulations verified. Postmortem assessments of minipig stomachs were made after different fasting protocols. RESULTS: Fraction of dose absorbed vs. time profiles showed high interindividual variability, comparable to human fed state absorption. Mean gastric transit times were determined to be 0.63 h, 1.36 h, and 0.73 h for solution, capsules, and tablets, respectively. Postmortem assessment confirmed that minipig stomachs were not empty after an overnight fast. CONCLUSIONS: Gastric transit times in overnight fasted minipigs are longer than those observed in humans. This is most likely caused by delayed and incomplete food emptying and further work is needed to develop feasible and effective fasting protocols for minipigs.


Assuntos
Absorção Fisiológica/fisiologia , Acetaminofen/farmacocinética , Analgésicos não Narcóticos/farmacocinética , Esvaziamento Gástrico/fisiologia , Mucosa Gástrica/metabolismo , Modelos Biológicos , Acetaminofen/administração & dosagem , Acetaminofen/sangue , Administração Oral , Analgésicos não Narcóticos/sangue , Animais , Área Sob a Curva , Disponibilidade Biológica , Estudos Cross-Over , Jejum , Feminino , Conteúdo Gastrointestinal , Injeções Intravenosas , Masculino , Suínos , Porco Miniatura
16.
MAbs ; 5(5): 776-86, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23924796

RESUMO

CD81 is an essential receptor for hepatitis C virus (HCV). K21 is a novel high affinity anti-CD81 antibody with potent broad spectrum anti-HCV activity in vitro. The pharmacokinetics (PK), pharmacodynamics and liver distribution of K21 were characterized in cynomolgus monkeys after intravenous (i.v.) administration of K21. Characteristic target-mediated drug disposition (TMDD) was shown based on the PK profile of K21 and a semi-mechanistic TMDD model was used to analyze the data. From the TMDD model, the estimated size of the total target pool at baseline (V(c) • R(base)) is 16 nmol/kg and the estimated apparent Michaelis-Menten constant (KM) is 4.01 nM. A simulation using estimated TMDD parameters indicated that the number of free receptors remains below 1% for at least 3 h after an i.v. bolus of 7 mg/kg. Experimentally, the availability of free CD81 on peripheral lymphocytes was measured by immunostaining with anti-CD81 antibody JS81. After K21 administration, a dose- and time-dependent reduction in free CD81 on peripheral lymphocytes was observed. Fewer than 3% of B cells could bind JS81 3 h after a 7 mg/kg dose. High concentrations of K21 were found in liver homogenates, and the liver/serum ratio of K21 increased time-dependently and reached ~160 at 168 h post-administration. The presence of K21 bound to hepatocytes was confirmed by immunohistochemistry. The fast serum clearance of K21 and accumulation in the liver are consistent with TMDD. The TMDD-driven liver accumulation of the anti-CD81 antibody K21 supports the further investigation of K21 as a therapeutic inhibitor of HCV entry.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Fígado/metabolismo , Modelos Biológicos , Tetraspanina 28/antagonistas & inibidores , Administração Intravenosa , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/imunologia , Afinidade de Anticorpos/imunologia , Área Sob a Curva , Células CHO , Células Cultivadas , Simulação por Computador , Cricetinae , Cricetulus , Sistemas de Liberação de Medicamentos/métodos , Hepatócitos/imunologia , Hepatócitos/metabolismo , Humanos , Cinética , Fígado/citologia , Fígado/imunologia , Linfócitos/imunologia , Linfócitos/metabolismo , Macaca fascicularis , Taxa de Depuração Metabólica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Microscopia Confocal , Tetraspanina 28/imunologia , Tetraspanina 28/metabolismo , Fatores de Tempo
17.
Birth Defects Res B Dev Reprod Toxicol ; 98(2): 170-82, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23529913

RESUMO

Interleukin-6 (IL-6) is involved in the pathogenesis of multiple disorders, including juvenile autoimmune diseases. IL-6 participates in a broad spectrum of physiological events, and the IL-6 receptor (IL-6R) is widely distributed across multiple organs. The interrelationship of development phases in juveniles together with organs involved in IL-6 signaling called for evaluations of anti-IL-6R antibody induced effects in a juvenile mouse model to assess the safety of such an approach in human juvenile arthritis. Here we show that naive mice in which IL-6 signals have been transiently blocked during the juvenile period develop normally. The fatal immunogenic reactions recorded earlier by repeated administration of the chosen rat anti-mouse IL-6R antibody, MR16-1, to mice were avoided successfully by application of a high loading dose followed by lower maintenance doses, with the support of modeling data. The high loading-dose regimen enabled us to conduct assessments without any major interference due to immunogenicity. Transient and complete inhibition of IL-6 signals from postnatal days 22 to 79 in mice exhibited no biologically important changes in sexual maturation or development of immune and skeletal systems. Although tendencies toward reductions of peripheral blood T-cell counts were observed, normal levels of antigen-specific IgG/IgM antibody productions indicating sufficient immunological functions were confirmed. Our results demonstrate that blockage of IL-6R by the neutralizing antibody does not affect juvenile development. This may be in part due to the generation or existence of compensatory pathways in the whole body system.


Assuntos
Anticorpos Anti-Idiotípicos/farmacologia , Anticorpos Neutralizantes/farmacologia , Osso e Ossos/efeitos dos fármacos , Sistema Imunitário/efeitos dos fármacos , Receptores de Interleucina-6/antagonistas & inibidores , Reprodução/efeitos dos fármacos , Animais , Anticorpos Monoclonais/farmacologia , Doenças Autoimunes/imunologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Sistema Imunitário/metabolismo , Interleucina-6/imunologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Receptores de Interleucina-6/imunologia
18.
J Pharmacokinet Pharmacodyn ; 39(3): 227-37, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22481485

RESUMO

Inhibition of the enzyme(s) that produce the Amyloid beta (Aß) peptide, namely BACE and γ-secretase, is considered an attractive target for Alzheimer's disease therapy. However, the optimal pharmacokinetic-pharmacodynamic modelling method to describe the changes in Aß levels after drug treatment is unclear. In this study, turnover models were employed to describe Aß levels following treatment with the γ-secretase inhibitor RO5036450, in the wild type rat. Initially, Aß level changes in the brain, cerebral spinal fluid (CSF) and plasma were modeled as separate biological compartments, which allowed the estimation of a compound IC50 and Aß turnover. While the data were well described, the model did not take into consideration that the CSF pool of Aß most likely originates from the brain via the CSF drainage pathway. Therefore, a separate model was carried out, with the assumption that CSF Aß levels originated from the brain. The optimal model that described the data involved two brain Aß 40 sub-compartments, one with a rapid turnover, from which CSF Aß 40 is derived, and a second quasi-static pool of ~20%. Importantly, the estimated in vivo brain IC50 was in a good range of the in vitro IC50 (ratio, 1.4). In conclusion, the PK/PD models presented here are well suited for describing the temporal changes in Aß levels that occur after treatment with an Aß lowering drug, and identifying physiological parameters.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Modelos Biológicos , Inibidores de Proteases/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/metabolismo , Masculino , Ratos , Ratos Wistar , Resultado do Tratamento
19.
MAbs ; 4(2): 243-55, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22453096

RESUMO

Subcutaneous (SC) delivery is a common route of administration for therapeutic monoclonal antibodies (mAbs) with pharmacokinetic (PK)/pharmacodynamic (PD) properties requiring long-term or frequent drug administration. An ideal in vivo preclinical model for predicting human PK following SC administration may be one in which the skin and overall physiological characteristics are similar to that of humans. In this study, the PK properties of a series of therapeutic mAbs following intravenous (IV) and SC administration in Göttingen minipigs were compared with data obtained previously from humans. The present studies demonstrated: (1) minipig is predictive of human linear clearance; (2) the SC bioavailabilities in minipigs are weakly correlated with those in human; (3) minipig mAb SC absorption rates are generally higher than those in human and (4) the SC bioavailability appears to correlate with systemic clearance in minipigs. Given the important role of the neonatal Fc-receptor (FcRn) in the PK of mAbs, the in vitro binding affinities of these IgGs against porcine, human and cynomolgus monkey FcRn were tested. The result showed comparable FcRn binding affinities across species. Further, mAbs with higher isoelectric point tended to have faster systemic clearance and lower SC bioavailability in both minipig and human. Taken together, these data lend increased support for the use of the minipig as an alternative predictive model for human IV and SC PK of mAbs.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/farmacocinética , Modelos Imunológicos , Administração Intravenosa , Animais , Anticorpos Monoclonais/imunologia , Feminino , Humanos , Injeções Subcutâneas , Masculino , Suínos , Porco Miniatura
20.
J Pharmacokinet Pharmacodyn ; 38(5): 581-93, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21779940

RESUMO

The volume of distribution at steady state (Vss) of therapeutic proteins is usually assessed by non-compartmental or compartmental pharmacokinetic (PK) analysis wherein errors may arise due to the elimination of therapeutic proteins from peripheral tissues that are not in rapid equilibrium with the sampling compartment (usually blood). Here we explored another potential source of error in the estimation of Vss that is linked to the heterogeneity of therapeutic proteins which may consist of components (e.g. glycosylation variants) with different elimination rates. PK simulations were performed with such hypothetical binary protein mixtures where elimination was assumed to be exclusively from the central compartment. The simulations demonstrated that binary mixtures containing a rapid-elimination component can give rise to pronounced bi-phasic concentration-time profiles. Apparent Vss observed with both non-compartmental and 2-compartmental PK analysis, increased with increasing fraction as well as with increasing elimination rate k ( 10 ) of the rapid-elimination component. Simulation results were complemented by PK analysis of an in vivo study in cynomolgus monkeys with different lots of lenercept, a tumor necrosis factor receptor-immunoglobulin G1 fusion protein, with different heterogeneities. The comparative Vss data for the three lenercept lots with different amounts of rapidly cleared components were consistent with the outcome of our simulations. Both lots with a higher fraction of rapidly cleared components had a statistically significant higher Vss as compared to the reference lot. Overall our study demonstrates that Vss of a therapeutic protein may be overestimated in proteins with differently eliminated components.


Assuntos
Software , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Simulação por Computador , Composição de Medicamentos , Glicosilação , Humanos , Cadeias gama de Imunoglobulina/química , Cadeias gama de Imunoglobulina/uso terapêutico , Macaca fascicularis , Taxa de Depuração Metabólica , Polissacarídeos/farmacocinética , Estrutura Quaternária de Proteína , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/uso terapêutico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA