Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Thromb Haemost ; 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38863155

RESUMO

BACKGROUND: Fidanacogene elaparvovec, an adeno-associated virus-based gene therapy vector expressing the high-activity factor IX (FIX) variant FIX-R338L, is in development for hemophilia B. One-stage clotting (OS) assays and chromogenic substrate (CS) assays are commonly used to measure FIX-R338L variant activity. Data from ongoing trials suggest FIX activity varies between different OS and CS assays. MATERIAL AND METHODS: To better understand FIX-R338L activity in clinical samples, an international multisite field study was conducted across a central laboratory and 18 local laboratories, using standard protocols, reagents, and instrumentation, with individual participant samples from a phase 1/2a study of fidanacogene elaparvovec. RESULTS: Unlike the wild-type FIX control, FIX-R338L activity was higher with the OS silica-based assay versus OS ellagic acid-based and CS assays. Variation in FIX activity was greater at the lowest activity levels. Activated FIX (FIXa) in plasma could result in higher OS assay activity or increased thrombin generation, which could overestimate FIX activity. However, FIXa was not detected in the participant samples, indicating that it was not contributing to the OS assay differences. Since individuals on gene therapy may receive exogenous replacement FIX products, replacement products were spiked into patient plasma samples to target a therapeutic concentration. Exogenous FIX was additive to endogenous FIX-R338L, with no interference from FIX-R338L. CONCLUSION: These results demonstrate FIX-R338L activity can be measured with OS and CS assays in clinical laboratories and provide insight into assay variability when measuring FIX with endogenously produced FIX-R338L. The findings may help establish best practices for measuring FIX-R338L activity (Clinicaltrials.gov identifier: NCT02484092).

2.
CPT Pharmacometrics Syst Pharmacol ; 12(9): 1358-1370, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37470295

RESUMO

Tanezumab is a monoclonal antibody against nerve growth factor (NGF). We investigated tanezumab pharmacokinetic (PK)-NGF relationships and predicted the extent of systemic free NGF suppression with target-mediated drug disposition (TMDD) modeling using data from three pivotal phase III interventional studies (NCT02697773, NCT02709486, and NCT02528188) in patients with osteoarthritis. Patients received tanezumab 2.5 mg or 5 mg every 8 weeks (q8w) subcutaneously. A TMDD model using a previously established population PK model was used to describe plasma tanezumab and serum total NGF concentration data, and simulations were performed to predict "unobserved" free NGF versus time profiles and dose-response relationships for free NGF. A total of 2992 patients had available data for plasma tanezumab or serum total NGF concentrations and were included in the analysis; 706 of these had data for both tanezumab and total NGF concentrations. The model generally performed well to predict observed total NGF concentrations up to ~24 weeks after each dose. Simulations suggested free NGF concentration would be suppressed by ~75% (median) near the peak of tanezumab concentration and by less than 5% (median) around the trough tanezumab concentration with a tanezumab 2.5 mg q8w regimen. Free NGF concentration was predicted to return to baseline level at ~8 weeks (95% prediction interval: 5-16 weeks) after the last tanezumab dose. This model adequately described plasma tanezumab and serum total NGF concentrations following s.c. administration of tanezumab 2.5 or 5 mg q8w, allowed prediction of relative change in systemic free NGF following s.c. administration of tanezumab.


Assuntos
Anticorpos Monoclonais , Fator de Crescimento Neural , Humanos , Anticorpos Monoclonais Humanizados , Resultado do Tratamento
3.
Clin Drug Investig ; 43(7): 551-563, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37460782

RESUMO

BACKGROUND: Tanezumab, a humanized anti-nerve growth factor antibody, was developed for the treatment of pain associated with osteoarthritis. Due to its mechanism of action, peripheral nerve safety was assessed in all clinical studies. OBJECTIVES: To summarize the neurological safety of intravenous (IV) and subcutaneous (SC) tanezumab versus placebo in patients with osteoarthritis. METHODS: Data were pooled from 3389 patients across seven studies that investigated IV administration, and from 1840 patients across three studies that investigated SC administration. The treatment period of each study ranged from 16 to 24 weeks, and follow-up periods ranged from 8 to 24 weeks. Neurological safety evaluations focused on adverse events (AEs) of abnormal peripheral sensation (APS), neurologic examinations, and consultations. RESULTS: Across datasets, the incidence of AEs of APS was higher in tanezumab groups versus placebo. Paresthesia and hypoesthesia were the most frequently reported AEs in tanezumab-treated patients, compared with placebo. In both datasets, most AEs were of mild severity, resolved, and rarely resulted in discontinuation. In all treatment groups in both IV and SC studies, over 90% of patients had no new or worsened neurological examination abnormalities at the last study visit. Across datasets, mononeuropathy was diagnosed more frequently in tanezumab groups compared with placebo. Polyneuropathy was diagnosed in ≤ 0.9% of patients in tanezumab and placebo groups. CONCLUSIONS: Tanezumab IV or SC had an increased incidence of AEs of APS, such as paresthesia and hypoesthesia, and diagnoses of mononeuropathy compared with placebo. However, tanezumab was not associated with generalized peripheral neuropathy. GOV IDENTIFIERS: NCT00733902, NCT00744471, NCT00830063, NCT00863304, NCT00863772, NCT01089725, NCT00985621, NCT02697773, and NCT02709486.


Assuntos
Anticorpos Monoclonais Humanizados , Fator de Crescimento Neural , Osteoartrite do Joelho , Parestesia , Humanos , Hipestesia/complicações , Osteoartrite do Joelho/tratamento farmacológico , Dor/tratamento farmacológico , Parestesia/complicações , Nervos Periféricos , Resultado do Tratamento , Ensaios Clínicos Fase III como Assunto , Ensaios Clínicos Controlados Aleatórios como Assunto , Anticorpos Monoclonais Humanizados/uso terapêutico , Fator de Crescimento Neural/antagonistas & inibidores
5.
Br J Clin Pharmacol ; 88(7): 3321-3334, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35112378

RESUMO

AIMS: Describe population pharmacokinetics of intravenous (IV) and subcutaneous (SC) tanezumab across Phase 2b/3 studies of osteoarthritis and chronic low back pain. METHODS: Data from 10 studies of IV or SC tanezumab (2.5-20 mg every 8 wk for up to 56 wk) were included in a multistep analysis. In Step 1, a 2-compartment model with linear and nonlinear elimination (based on prior analysis of pre-2015 IV osteoarthritis studies) was expanded to include other pre-2015 studies. In Step 2, post-2015 SC studies were combined into the model. Steps 3 and 4 evaluated impact of baseline nerve growth factor (NGF) and treatment-emergent anti-drug antibodies (TE ADA). RESULTS: SC bioavailability was estimated at 62-76%. The key disposition parameters CL, Vc , Vp and KM were estimated to be 0.133 L d-1 , 2.6 L, 1.77 L and 31.2 µg L-1 , respectively. Plasma tanezumab concentration was predicted to reach Cmax at 8.9-11.2 days following single and multiple SC administration in typical patients within the dose range of SC Phase 3 studies (2.5-10 mg every 8 wk). Exposure of a typical patient was similar between IV and SC for the second part of the dosing interval (wk 4-8). Covariates selected on the absorption parameters were weight, age, sex and injection site. Baseline NGF had minimal effect on maximum elimination capacity and TE ADA status was associated with slightly higher tanezumab clearance (6-7%). CONCLUSION: Our model adequately described plasma tanezumab concentration vs. time following IV or SC administration. Weight was the most influential covariate with respect to absorption of tanezumab in comparison to patient population (osteoarthritis and chronic low back pain) or other demographics. There was no clinically relevant effect of baseline NGF or TE ADA on tanezumab PK.


Assuntos
Dor Lombar , Osteoartrite , Administração Intravenosa , Anticorpos Monoclonais Humanizados/uso terapêutico , Humanos , Dor Lombar/tratamento farmacológico , Fator de Crescimento Neural/uso terapêutico , Osteoartrite/tratamento farmacológico , Resultado do Tratamento
6.
AAPS J ; 22(2): 19, 2020 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-31900604

RESUMO

After tier 1 and 2 cut points for anti-drug antibody (ADA) assays are derived during pre-study assay validation in a population, there is a need to verify the continued appropriateness of the previously derived cut points during sample analysis in the same or different populations, per FDA guidance (US HHS, FDA, CDER, CBER, 2019). Proper sample size-dependent criteria with statistical underpinning were derived and presented in this technical note to aid in assessing the appropriateness of tier 1 and tier 2 cut points, respectively.


Assuntos
Anticorpos/análise , Testes Imunológicos/normas , Proteínas/imunologia , Projetos de Pesquisa , Interpretação Estatística de Dados , Reações Falso-Positivas , Humanos , Valor Preditivo dos Testes , Proteínas/uso terapêutico , Reprodutibilidade dos Testes , Tamanho da Amostra
7.
J Pharmacokinet Pharmacodyn ; 46(6): 617-626, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31667657

RESUMO

Cardiac safety assessment is a key regulatory requirement for almost all new drugs. Until recently, one evaluation aspect was via a specifically designated, expensive, and resource intensive thorough QTc study, and a by-time-point analysis using an intersection-union test (IUT). ICH E14 Q&A (R3) (http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Efficacy/E14/E14_Q_As_R3__Step4.pdf) allows for analysis of the PK-QTc relationship using early Phase I data to assess QTc liability. In this paper, we compared the cardiac risk assessment based on the early Phase I analysis with that from a thorough QTc study across eleven drug candidate programs, and demonstrate that the conclusions are largely the same. The early Phase I analysis is based upon a linear mixed effect model with known covariance structure (Dosne et al. in Stat Med 36(24):3844-3857, 2017). The treatment effect was evaluated at the supratherapeutic Cmax as observed in the thorough QTc study using a non-parametric bootstrap analysis to generate 90% confidence intervals for the treatment effect, and implementation of the standardized methodology in R and SAS software yielded consistent results. The risk assessment based on the concentration-response analysis on the early Phase I data was concordant with that based on the standard analysis of the thorough QTc study for nine out of the eleven drug candidates. This retrospective analysis is consistent with and supportive of the conclusion of a previous prospective analysis by Darpo et al. (Clin Pharmacol Ther 97(4):326-335, 2015) to evaluate whether C-QTc analysis can detect QTc effects in a small study with healthy subjects.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Eletrocardiografia/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Coração/efeitos dos fármacos , Preparações Farmacêuticas/administração & dosagem , Ensaios Clínicos Fase I como Assunto , Estudos Cross-Over , Relação Dose-Resposta a Droga , Humanos , Estudos Prospectivos , Estudos Retrospectivos , Medição de Risco/métodos
8.
CPT Pharmacometrics Syst Pharmacol ; 7(9): 581-592, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30043511

RESUMO

A pharmacokinetic (PK) model was developed for nusinersen, an antisense oligonucleotide (ASO) that is the first approved treatment for spinal muscular atrophy (SMA). The model was built with data from 92 nonhuman primates (NHPs) following intrathecal doses (0.3-7 mg) and characterized the PK in cerebrospinal fluid (CSF), plasma, total spinal cord, brain, and pons. The estimated volumes were 13.6, 937, 4.5, 53.8, and 2.11 mL, respectively. Global sensitivity analysis demonstrated that the CSF-to-plasma drug distribution rate (0.09 hour-1 ) is a major determinant of the maximum nusinersen concentration in central nervous system (CNS) tissues. Physiological age-based and body weight-based allometric scaling was implemented with exponent values of -0.08 and 1 for the rate constants and the volume of distribution, respectively. Simulations of the scaled model were in agreement with clinical observations from 52 pediatric phase I PK profiles. The developed model can be used to guide the design of clinical trials with ASOs.


Assuntos
Modelos Biológicos , Atrofia Muscular Espinal/tratamento farmacológico , Oligonucleotídeos/farmacocinética , Oligonucleotídeos/uso terapêutico , Animais , Feminino , Macaca fascicularis , Masculino
9.
Clin Pharmacol Ther ; 104(4): 699-708, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29271001

RESUMO

Type 2 diabetes mellitus (T2DM) is a chronic, progressive disease characterized by persistently elevated blood glucose concentration (hyperglycemia). We developed a mechanistic drug-disease modeling platform based on data from more than 4,000 T2DM subjects in seven phase II/III clinical trials. The model integrates longitudinal changes in clinically relevant biomarkers of glycemic control with information on baseline disease state, demographics, disease progression, and different therapeutic interventions, either when given alone or as add-on combination therapy. The model was able to simultaneously characterize changes in fasting plasma glucose, fasting serum insulin, and glycated hemoglobin A1c following administration of sulfonylurea, metformin, and thiazolidinedione as well as disease progression in clinical trials ranging from 16-104 weeks of treatment. The mechanistic components of this generalized mechanism-based platform, based on knowledge of pharmacology, insulin-glucose homeostatic feedback, and diabetes pathophysiology, allows its application to be further expanded to other antidiabetic drug classes and combination therapies.


Assuntos
Glicemia/efeitos dos fármacos , Ensaios Clínicos como Assunto/métodos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Determinação de Ponto Final , Hipoglicemiantes/uso terapêutico , Modelos Biológicos , Projetos de Pesquisa , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/diagnóstico , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Feminino , Hemoglobinas Glicadas/metabolismo , Humanos , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/farmacocinética , Insulina/sangue , Masculino , Metformina/uso terapêutico , Pessoa de Meia-Idade , Compostos de Sulfonilureia/uso terapêutico , Tiazolidinedionas/uso terapêutico , Fatores de Tempo , Resultado do Tratamento
10.
Clin Pharmacokinet ; 55(7): 769-788, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26798033

RESUMO

Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease, which affects millions of people worldwide. The disease is characterized by chronically elevated blood glucose concentrations (hyperglycaemia), which result in comorbidities and multi-organ dysfunction. This is due to a gradual loss of glycaemic control as a result of increasing insulin resistance, as well as decreasing ß-cell function. The objective of T2DM drug interventions is, therefore, to reduce fasting and postprandial blood glucose concentrations to normal, healthy levels without hypoglycaemia. Several classes of novel antihyperglycaemic drugs with various mechanisms of action have been developed over the past decades or are currently under clinical development. The development of these drugs is routinely supported by the application of pharmacokinetic/pharmacodynamic modelling and simulation approaches. They integrate information on the drug's pharmacokinetics, clinically relevant biomarker information and disease progression into a single, unifying approach, which can be used to inform clinical study design, dose selection and drug labelling. The objective of this review is to provide a comprehensive overview of the quantitative approaches that have been reported since the 2008 review by Landersdorfer and Jusko in an increasing order of complexity, starting with glucose homeostasis models. Each of the presented approaches is discussed with respect to its strengths and limitations, and respective knowledge gaps are highlighted as potential opportunities for future drug-disease model development in the area of T2DM.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Modelos Biológicos , Biomarcadores , Glicemia/efeitos dos fármacos , Inibidores da Dipeptidil Peptidase IV/farmacologia , Peptídeo 1 Semelhante ao Glucagon/agonistas , Glucoquinase/biossíntese , Hemoglobinas Glicadas , Humanos , Hiperglicemia/prevenção & controle , Hipoglicemiantes/farmacocinética , Incretinas/biossíntese , Insulina/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores de Glucagon/antagonistas & inibidores , Inibidores do Transportador 2 de Sódio-Glicose
11.
Eur J Pharm Sci ; 66: 157-62, 2015 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-25459532

RESUMO

Hemophilia A is a bleeding disorder caused by the deficiency of an important coagulation factor; Factor VIII (FVIII). Replacement therapy using exogenously administered recombinant FVIII is the most commonly used method of treatment. However, approximately 30% of Hemophilia A patients develop neutralizing antibodies (Nabs) against the recombinant protein. Nabs abolish FVIII activity and drastically influence efficacy of the protein. The immunogenic epitopes of FVIII reside predominantly in the C2 domain of FVIII. However, the C2 domain also contains a lipid binding region. O-Phospho-L-Serine (OPLS) which is the head-group moiety of phosphatidylserine, interacts with the lipid binding region of FVIII. Previous studies have shown that FVIII complexed with OPLS lowered Nab development against FVIII following subcutaneous administration. In dendritic cell-T-cell co-culture studies, OPLS treatment increased the secretion of immunosuppressive cytokines (Transforming Growth Factor-ß and Interleukin-10), and simultaneously decreased pro-inflammatory IL-17 cytokine. Here, we investigated FVIII immune response and pharmacokinetics upon intravenous administration of FVIII-OPLS complex. We studied the effect of FVIII-OPLS complex on the interaction between a professional antigen presenting cell; dendritic cell and T-cell, and T-cell clonal expansion. Pharmacokinetics parameters were estimated following intravenous administration of FVIII and FVIII-OPLS. The results suggest that OPLS lowers FVIII immune response following intravenous administration. OPLS also hinders FVIII-specific T-cell clonal proliferation and preserves FVIII PK profile. Thus, the ease of protein-lipid complexation, preservation of FVIII activity and in vivo behavior, and improved in vitro FVIII stability, makes OPLS an attractive excipient in the preparation of next generation or biosimilar FVIII products with improved safety profile.


Assuntos
Fator VIII/química , Fator VIII/imunologia , Hemofilia A/terapia , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Administração Intravenosa , Animais , Área Sob a Curva , Fator VIII/administração & dosagem , Meia-Vida , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfosserina/administração & dosagem , Subpopulações de Linfócitos T
12.
J Biol Chem ; 288(24): 17051-6, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23649621

RESUMO

Administration of recombinant factor VIII (FVIII), an important co-factor in blood clotting cascade, elicits unwanted anti-FVIII antibodies in hemophilia A (HA) patients. Previously, FVIII associated with phosphatidylserine (PS) showed significant reduction in the anti-FVIII antibody response in HA mice. The reduction in the immune response to FVIII-PS could be due either to a failure of the immune system to recognize the antigen (i.e. immunological ignorance) or to an active induction of an antigen-specific nonresponsiveness (i.e. immunological tolerance). If it were a result of tolerance, one would predict that pre-exposure to FVIII-PS would render the mice hypo-responsive to a subsequent FVIII challenge. Here, we have demonstrated that naive HA mice that were pretreated with FVIII-PS showed a significantly reduced FVIII immune response to further challenge with native FVIII and that this decreased responsiveness could be adoptively transferred to other mice. An increase in number of FoxP3-expressing CD4(+) regulatory T-cells (Treg) was observed for the FVIII-PS-immunized group as compared with animals that received FVIII alone, suggesting the involvement of Treg in PS-mediated hypo-responsiveness. The PS-mediated reduction in antibody response was reversed by the co-administration of function-blocking anti-TGF-ß antibody with FVIII-PS. The decreased response to FVIII induced by FVIII-PS was determined to be antigen-specific because the immune response to another non-cross-reactive antigen (ovalbumin) was not altered. These results are consistent with the notion that FVIII-PS is tolerogenic and suggest that immunization with this tolerogenic form of the protein could be a useful treatment option to minimize immunogenicity of FVIII and other protein-based therapeutics.


Assuntos
Fator VIII/imunologia , Hemofilia A/terapia , Fosfatidilserinas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Transferência Adotiva , Animais , Anticorpos Neutralizantes/sangue , Fator VIII/administração & dosagem , Fatores de Transcrição Forkhead/metabolismo , Hemofilia A/imunologia , Humanos , Tolerância Imunológica , Camundongos , Camundongos Knockout , Fosfatidilserinas/administração & dosagem , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante
13.
J Pharm Sci ; 101(1): 48-55, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21953409

RESUMO

Factor VIII (FVIII) is an important coagulation cofactor and its deficiency causes Hemophilia A, a bleeding disorder. Replacement therapy using recombinant FVIII is currently the first line of therapy for Hemophilia A, but the development of neutralizing antibody is a major clinical complication for this therapy. Recently, it has been shown that FVIII associated with phosphatidylinositol (PI)-containing lipidic nanoparticles reduced development of neutralizing antibodies in Hemophilia A mice (Peng A, Straubinger RM, Balu-Iyer SV. 2010. AAPS J 12(3):473-481). Here, we investigated the underlying mechanism of this reduction in antibody response in culturing conditions. In vitro, PI interfered with the processing of FVIII by cultured dendritic cells (DC), resulting in a reduction in the upregulation of phenotypic costimulatory signal CD40. Furthermore, PI increased secretion of regulatory cytokines Transforming Growth Factor ß1 and Interleukin 10 (IL-10) but reduced the secretion of proinflammatory cytokines IL-6 and IL-17. The data suggest that PI reduces immunogenicity of FVIII by modulating DC maturation and inducing secretion of regulatory cytokines.


Assuntos
Antígenos CD40/metabolismo , Células Dendríticas/imunologia , Fator VIII/imunologia , Fator VIII/farmacologia , Fosfatidilinositóis/imunologia , Fosfatidilinositóis/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD40/imunologia , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Fator VIII/metabolismo , Humanos , Interleucinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator de Crescimento Transformador beta1/imunologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
14.
Methods Mol Biol ; 716: 267-80, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21318912

RESUMO

Immunogenicity against therapeutic proteins is a clinical problem in the successful treatment of many diseases and, as such, immunogenicity risk assessment in preclinical setting would be useful to improve safety and efficacy of protein-based therapeutics in the product development stages. Here, we attempted a mechanism-based in vitro study as screening tool to capture clinically observed antibody-based immune response against two representative therapeutic proteins: recombinant human Erythropoietin-alpha (rHuEPO)and recombinant Factor VIII (rFVIII). Flow-cytometry was used to determine the maturation level of dendritic cells (DCs), a primary initiator of T-cell responses. Studies to capture T-lymphocyte proliferation upon challenge with free rFVIII were performed and secretion of immunomodulatory cytokines was analyzed by ELISA assay. These in vitro techniques could be used as risk assessment tool to determine the immunogenic potential of formulations of recombinant proteins in preclinical setting.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Eritropoetina/imunologia , Fator VIII/imunologia , Proteínas Recombinantes/imunologia , Animais , Proliferação de Células , Células Cultivadas , Citocinas/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Citometria de Fluxo/métodos , Humanos , Camundongos , Medição de Risco , Linfócitos T/citologia , Linfócitos T/imunologia
15.
Clin Immunol ; 138(2): 135-45, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21094627

RESUMO

A major clinical complication in the treatment of Hemophilia A using exogenously administered recombinant Factor VIII (FVIII) is the development of neutralizing antibodies. It has been shown previously that FVIII complexed with phosphatidylserine (PS) reduces the development of total and neutralizing antibody titers in hemophilic mice. The effect of complexation of FVIII with PS upon dendritic cell (DC) uptake, maturation and processing, T-cell proliferation and cytokine secretion profiles was investigated. Flow cytometric studies of DC showed that PS inhibited the up-regulation of cell surface co-stimulatory markers (CD86 and CD40). PS reduced T-cell proliferation and significantly increased levels of TGF-ß and IL-10 but reduced secretion of IL-6 and IL-17 compared to controls. The data suggest that PS reduces immunogenicity of FVIII by regulating dendritic cell maturation and subsequent T-lymphocyte activity through modulation of cytokine secretion. A possible mechanism for PS-mediated induction of FVIII tolerance is discussed.


Assuntos
Anticorpos Neutralizantes/imunologia , Células Dendríticas/efeitos dos fármacos , Fator VIII/imunologia , Hemofilia A/imunologia , Tolerância Imunológica/efeitos dos fármacos , Fosfatidilserinas/farmacologia , Proteínas Recombinantes/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Antígeno B7-2/imunologia , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Humanos , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Proteínas Recombinantes/uso terapêutico , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
16.
J Pharm Sci ; 98(12): 4480-4, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19499565

RESUMO

Factor VIII is a multi-domain glycoprotein and is an essential cofactor in the blood coagulation cascade. Its deficiency or dysfunction causes Hemophilia A, a bleeding disorder. Replacement using exogenous recombinant Factor VIII (FVIII) is the first line of therapy for Hemophilia A. Immunogenicity, the development of binding (total) and neutralizing (inhibitory) antibody against administered protein is a clinical complication of the therapy. There are several product related factors such as presence of aggregates, route and frequency of administration and glycosylation have been shown to contribute to immunogenicity. The effect of route of administration of FVIII on antibody development in Hemophilia A is not completely understood. Here we investigated the effect of route of administration (s.c. or i.v.) on immunogenicity in Hemophilia A mice. The total and inhibitory titers were determined using ELISA and modified Bethesda Assay respectively. The results indicated that s.c. is more immunogenic compared to i.v. route in terms of total antibody titer development (binding antibodies) but no significant differences in inhibitory titer levels could be established.


Assuntos
Fator VIII/administração & dosagem , Fator VIII/imunologia , Hemofilia A/metabolismo , Animais , Anticorpos/análise , Anticorpos Bloqueadores/farmacologia , Células CHO , Cricetinae , Cricetulus , Ensaio de Imunoadsorção Enzimática , Fator VIII/farmacologia , Glicosilação , Hemofilia A/tratamento farmacológico , Humanos , Injeções Intravenosas , Injeções Subcutâneas , Camundongos
17.
J Pharm Sci ; 97(9): 3753-64, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18300296

RESUMO

The replacement therapy using recombinant human FVIII (rFVIII) is the first line of therapy for hemophilia A. Approximately 15-30% of the patients develop inhibitory antibodies. Recently, we reported that liposomes composed of phosphatidylserine (PS) could reduce the immunogenicity of rFVIII. However, PS containing liposomal-rFVIII is likely to reduce the systemic exposure and efficacy of FVIII due to rapid uptake of the PS containing liposomes by the reticuloendothelial system (RES). Here, we investigated whether phosphatidylserine (PS) liposomes containing Polyethylene glycol (PEG) (PEGylated), could reduce the immunogenicity of rFVIII and reverse the reduction in systemic exposure of rFVIII. Animals given PEGylated liposomal-rFVIII had lower total and inhibitory anti-rFVIII antibody titers, compared to animals treated with rFVIII alone. The mean stimulation index of CD4+ T-cells from animals given PEGylated liposomal-rFVIII also was lower than for animals that were given rFVIII alone. Pharmacokinetic studies following intravenous dosing indicated that the systemic exposure (area under the activity curve, AUAC(0-24h)) of PEGylated liposomal-rFVIII was approximately 59 IU/mL x h and significantly higher than that of non-PEGylated liposomal-rFVIII (AUAC(0-24h) approximately 36 IU/mL x h). Based on these studies, we speculate that PEGylated PS-containing liposomal rFVIII may improve efficacy of rFVIII.


Assuntos
Modelos Animais de Doenças , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Polietilenoglicóis/química , Animais , Anticorpos/imunologia , Proliferação de Células , Fator VIII/química , Fator VIII/imunologia , Fator VIII/farmacocinética , Feminino , Lipossomos , Masculino , Camundongos , Micelas , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapêutico , Linfócitos T/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA