Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Clin Infect Dis ; 75(1): e459-e465, 2022 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34554235

RESUMO

BACKGROUND: After the failure of antibody therapies in treating hospitalized patients with coronavirus disease 2019 (COVID-19), we investigated the impact of viral replication on the pharmacokinetics and efficacy of a hyperimmune severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunoglobulin (CoVIG) product in treating SARS-CoV-2 infection using an adult Syrian hamster model. METHODS: The CoVIG was manufactured from plasma donors who had recovered from COVID-19. The dose used (400 mg/kg) was based on the dose given in clinical trials to hospitalized patients with COVID-19. Hamsters were given a single dose of CoVIG 2 days after challenge with the SARS-CoV-2 virus (isolate NY/PV08410/2020), followed by sampling of blood, nasal, tracheal, and lung tissues at different time points. The blood samples were assayed for anti-SARS-CoV-2 spike binding and used to calculate pharmacokinetic (PK) parameters. Nasal wash, tracheal, and lung tissue samples were assayed for viral replication by polymerase chain reaction (subgenomic messenger RNA). RESULTS: CoVIG-treated hamsters showed a reduction in viral replication in the lower respiratory tract, but minimal reduction in the upper respiratory tract, after challenge with SARS-CoV-2. Challenge resulted in altered PK parameters proportionate to viral replication, resulting in decreased area under the curve, accelerated clearance, and shorter half-life of CoVIG. CONCLUSIONS: These data indicate that in the presence of actively replicating SARS-CoV-2 virus, PK parameters are altered and should trigger an adjustment in CoVIG dosing.


Assuntos
Tratamento Farmacológico da COVID-19 , Adulto , Animais , Cricetinae , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Pulmão , Mesocricetus , SARS-CoV-2
2.
Pharm Res ; 39(8): 1669-1680, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35552984

RESUMO

Model-informed drug development (MIDD) is a powerful approach to support drug development and regulatory review. There is a rich history of MIDD applications at the U.S. Food and Drug Administration (FDA). MIDD applications span across the life cycle of the development of new drugs, generics, and biologic products. In new drug development, MIDD approaches are often applied to inform clinical trial design including dose selection/optimization, aid in the evaluation of critical regulatory review questions such as evidence of effectiveness, and development of policy. In the biopharmaceutics space, we see a trend for increasing role of computational modeling to inform formulation development and help strategize future in vivo studies or lifecycle plans in the post approval setting. As more information and knowledge becomes available pre-approval, quantitative mathematical models are becoming indispensable in supporting generic drug development and approval including complex generic drug products and are expected to help reduce overall time and cost. While the application of MIDD to inform the development of cell and gene therapy products is at an early stage, the potential for future application of MIDD include understanding and quantitative evaluation of information related to biological activity/pharmacodynamics, cell expansion/persistence, transgene expression, immune response, safety, and efficacy. With exciting innovations on the horizon, broader adoption of MIDD is poised to revolutionize drug development for greater patient and societal benefit.


Assuntos
Desenvolvimento de Medicamentos , Medicamentos Genéricos , Animais , Aprovação de Drogas , Humanos , Estágios do Ciclo de Vida , Preparações Farmacêuticas , Estados Unidos , United States Food and Drug Administration
3.
Haemophilia ; 26(5): 817-825, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32842165

RESUMO

INTRODUCTION: Emerging, systematic approaches for capturing patient input, such as preference elicitation, can provide valuable information for the benefit-risk assessment of medical products for treating bleeding disorders, such as haemophilia. AIM: This study aims to identify existing and develop new methods to capture, rank and summarize preference scores for clotting factor therapies. METHODS: Haemophilia patient preference data were compiled from studies identified through literature review and publicly available US FDA patient-focused drug development meeting documents. Text mining was performed to identify major themes across studies. A standardized preference score was estimated and aggregated. RESULTS: Ten preference studies that employed qualitative (n = 3), and quantitative methods (n = 7) met the inclusion criteria. Text mining of qualitative and quantitative studies revealed similar themes as the standardized preference attribute importance. We found that seven quantitative studies employed discrete choice experiments (DCE)/conjoint analysis (CA) and examined a range of 5-12 attributes. For DCE/CA studies published prior to 2014 (n = 4), safety attributes (inhibitor and viral safety) were among the most important attributes, accounting for ~46% of the total utility measured. DCE/CA studies published after 2014 (n = 3) focused on frequency of infusion and reduction of bleeding risk, accounting for ~67% of the total utility. Interestingly, two studies that used different preference elicitation approaches (DCE and a monadic conjoint approach) both ranked infusion frequency as the most important attribute. CONCLUSIONS: Although there are few published patient preference studies for haemophilia, the results of this study can be viewed in the larger context of enhancing scientific methods of incorporating patient input in medical product development.


Assuntos
Fatores de Coagulação Sanguínea/uso terapêutico , Hemofilia A/sangue , Fatores de Coagulação Sanguínea/farmacologia , Feminino , Humanos , Masculino
4.
Haemophilia ; 25(4): e240-e246, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31131515

RESUMO

INTRODUCTION: Clinical pharmacology studies are one of the major types of regulatory data submitted for review of therapeutic proteins regulated by the Center for Biologics Evaluation and Research (CBER). AIM: The primary objective of the current study is to provide an overview of the role of clinical pharmacology including pharmacokinetics (PK), pharmacodynamics (PD) and exposure-response analysis at CBER. Furthermore, we aim to provide a baseline estimate for the use of quantitative clinical pharmacology studies prior to implementation of FDA's model-informed drug development (MIDD) pilot programme. METHODS: We survey original Biologics License Applications (BLAs) for plasma-derived and related recombinant therapeutic protein products approved by CBER/FDA (2008-2017). RESULTS: There were 37 original BLAs that met our inclusion criteria, and 34 of these products (92%) contained human PK data as part of the biological licensing. The products were broadly classified as coagulation factors (54%), IgG and related proteins (24%), and other therapeutic proteins (22%). Coagulation factor VIII and IX products constitute 32% of the BLAs and indicated for treatment of haemophilia A and B, respectively. Twelve products (35%) used model-based approaches (population PK/PD and exposure-response). Over the past 5 years (2013 to 2017), there is a trend for increased application of MIDD approaches as compared to the previous cohort years (2008 to 2012). CONCLUSION: In conclusion, clinical pharmacology has played a major role in regulatory review of plasma-derived products, and we expect that the application of quantitative methods will further evolve for these products under the FDA MIDD programme.


Assuntos
Proteínas Sanguíneas/farmacologia , Descoberta de Drogas , Proteínas Recombinantes/farmacologia , Adulto , Proteínas Sanguíneas/farmacocinética , Criança , Feminino , Humanos , Masculino , Modelos Teóricos , Projetos Piloto , Proteínas Recombinantes/farmacocinética
5.
Regul Toxicol Pharmacol ; 108: 104436, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31381939

RESUMO

BACKGROUND: In recent years, there has been great interest from academia, industry and government scientists for an increased understanding of the mode of action of vaccine adjuvants to characterize the safety and efficacy of vaccines. In this context, pharmacokinetic (PK) and biodistribution studies are useful for quantifying the concentration of vaccine adjuvants in mechanistically or toxicologically relevant target tissues. METHODS: In this study, we conducted a comparative analysis of the PK and biodistribution profile of radiolabeled squalene for up to 336 h (14 days) after intramuscular injection of mice with adjuvanted H5N1 influenza vaccines. The evaluated adjuvants included an experimental-grade squalene-in-water (SQ/W) emulsion (AddaVax®) and an adjuvant system (AS03®) that contained squalene and α-tocopherol in the oil phase of the emulsion. RESULTS: The half-life of the initial exponential decay from quadriceps muscle was 1.5 h for AS03 versus 12.9 h for AddaVax. At early time points (1-6 h), there was about a 10-fold higher concentration of labeled squalene in draining lymph nodes following AS03 injection compared to AddaVax. The area-under-concentration curve up to 336 h (AUC0-336hr) and peak concentration of squalene in spleen (immune organ) was about 1.7-fold higher following injection of AS03 than AddaVax. The peak systemic tissue concentration of squalene from the two adjuvants, with or without antigen, remained below 1% of injected dose for toxicologically relevant target tissues, such as spinal cord, brain, and kidney. The pharmacokinetics of AS03 was unaffected by the presence of H5N1 antigen. CONCLUSIONS: This study demonstrates a rapid decline of AS03 from the quadriceps muscles of mice as compared to conventional SQ/W emulsion adjuvant, with an increased transfer to mechanistically relevant tissues such as local lymph nodes. Systemic tissue exposure to potential toxicological target tissues was very low.


Assuntos
Adjuvantes Imunológicos/farmacocinética , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/farmacocinética , Polissorbatos/farmacocinética , Esqualeno/farmacocinética , alfa-Tocoferol/farmacocinética , Animais , Antígenos/imunologia , Combinação de Medicamentos , Emulsões , Feminino , Injeções Intramusculares , Linfonodos/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Músculo Quadríceps/metabolismo , Distribuição Tecidual
6.
Ther Drug Monit ; 40(5): 635-641, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30086085

RESUMO

BACKGROUND: In population pharmacokinetic modeling, bodyweight is often incorporated as an important covariate using fixed (0.75) or single-exponent model. In recent years, several variations of allometric models have been suggested for the prediction of drug clearance across a wide age range. The objective of this study is to develop and evaluate single-exponent, bodyweight-dependent allometric exponent (BDE), age-dependent exponent (ADE), and segmented regression models for predicting clearance and maintenance dose of theophylline. METHODS: The BDE model was described by the following equation: (Equation is included in full-text article.), where L × BW defines the BDE for clearance. The coefficient and the exponents L and M were estimated. The ADE model consisted of several empirical exponents based on age and ranged from 0.75 (children >5 years and adults) to 1.2 (premature neonates). Data for model development and validation were based on 52 subjects each. RESULTS: All structural and statistical parameters were estimated with acceptable precision for single-exponent and BDE models (<30%); however, the BDE model was superior in describing theophylline clearance across a wide age range for the training data. The segmented regression model on log-transformed data also adequately described theophylline clearance. When models were evaluated with validation data, a single-exponent model overpredicted clearance and dosing rate in premature neonates and adults with a mean prediction error of ≥50%. For premature neonates and adults, mean clearance and dosing rate were predicted within a 30% prediction error using the BDE, ADE, and segmented models. CONCLUSIONS: This study demonstrates that the BDE, ADE, and segmented models performed better than a single-exponent model for predicting clearance and dose of theophylline across a wide age range.


Assuntos
Envelhecimento , Peso Corporal , Cálculos da Dosagem de Medicamento , Modelos Biológicos , Teofilina/administração & dosagem , Teofilina/farmacocinética , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino
7.
Regul Toxicol Pharmacol ; 97: 57-62, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29894734

RESUMO

The objectives of this study were to develop and evaluate allometric methods for predicting tissue-to-plasma partition coefficients (Kp) in mice from experimentally determined in-vivo volume of distribution at steady state (Vss) for monoclonal antibodies (mAbs). The Vss was allometrically predicted (using a fixed exponent 1.0 or 0.9) in a given tissue of the mice. The Kp was predicted using Vss and tissue specific physiological parameters. In total, Kp values were predicted for 20 mAbs, 121 tissues, and 665 tissue concentrations. The predicted Kp values and tissue concentrations were compared with the experimental results as well as an empirically predicted antibody biodistribution coefficient (ABC). Comparison of the predicted Kp values by the two proposed methods with experimentally determined Kp values indicated that 64-75% of the predicted Kp values were within two-fold prediction error. For 665 tissue concentrations, 63%, 74%, and 48% tissue concentration ratio were within 0.5-2 fold prediction error by exponent 1.0, exponent 0.9, and ABC, respectively. The proposed allometric methods are better than ABC method for the prediction of tissue Kp values and tissue concentrations. The proposed methods can reasonably predict tissue concentrations of mAbs using plasma concentration gathered at early stage of biologics development.


Assuntos
Anticorpos Monoclonais/sangue , Animais , Camundongos , Distribuição Tecidual
8.
Regul Toxicol Pharmacol ; 81: 113-119, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27498239

RESUMO

Squalene is a component of oil-in-water emulsion adjuvants developed for potential use in some influenza vaccines. The biodistribution of the squalene-containing emulsion adjuvant (AddaVax™) alone and as part of complete H5N1 vaccine was quantified in mechanistically and toxicologically relevant target tissues up to 336 h (14 days) following injection into quadriceps muscle. At 1 h, about 55% of the intramuscularly injected dose of squalene was detected in the local quadriceps muscles and this decreased to 26% at 48 h. Twenty-four hours after the injection, approximately 5%, 1%, and 0.6% of the injected dose was detected in inguinal fat, draining lymph nodes, and sciatic nerve, respectively. The peak concentration for kidney, brain, spinal cord, bone marrow, and spleen was each less than 1% of the injected dose, and H5N1 antigen did not significantly alter the biodistribution of squalene to these tissues. The area-under-blood-concentration curve (AUC) and peak blood concentration (Cmax) of squalene were slightly higher (20-25%) in the presence of H5N1 antigen. A population pharmacokinetic model-based statistical analysis identified body weight and H5N1 antigen as covariates influencing the clearance of squalene. The results contribute to the body of knowledge informing benefit-risk analyses of squalene-containing emulsion vaccine adjuvants.


Assuntos
Adjuvantes Imunológicos/farmacocinética , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/farmacocinética , Polissorbatos/farmacocinética , Esqualeno/farmacocinética , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/sangue , Adjuvantes Imunológicos/toxicidade , Animais , Área Sob a Curva , Simulação por Computador , Emulsões , Feminino , Meia-Vida , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/sangue , Vacinas contra Influenza/toxicidade , Injeções Intramusculares , Masculino , Taxa de Depuração Metabólica , Camundongos Endogâmicos BALB C , Modelos Biológicos , Dinâmica não Linear , Polissorbatos/administração & dosagem , Polissorbatos/toxicidade , Medição de Risco , Esqualeno/administração & dosagem , Esqualeno/sangue , Esqualeno/toxicidade , Distribuição Tecidual , Toxicocinética
9.
Regul Toxicol Pharmacol ; 71(3): 353-64, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25683773

RESUMO

Alpha (α)-tocopherol is a component of a new generation of squalene-containing oil-in-water (SQ/W) emulsion adjuvants that have been licensed for use in certain influenza vaccines. Since regulatory pharmacokinetic studies are not routinely required for influenza vaccines, the in vivo fate of this vaccine constituent is largely unknown. In this study, we constructed a physiologically based pharmacokinetic (PBPK) model for emulsified α-tocopherol in human adults and infants. An independent sheep PBPK model was also developed to inform the local preferential lymphatic transfer and for the purpose of model evaluation. The PBPK model predicts that α-tocopherol will be removed from the injection site within 24h and rapidly transfer predominantly into draining lymph nodes. A much lower concentration of α-tocopherol was estimated to peak in plasma within 8h. Any systemically absorbed α-tocopherol was predicted to accumulate slowly in adipose tissue, but not in other tissues. Model evaluation and uncertainty analyses indicated acceptable fit, with the fraction of dose taken up into the lymphatics as most influential on plasma concentration. In summary, this study estimates the in vivo fate of α-tocopherol in adjuvanted influenza vaccine, may be relevant in explaining its immunodynamics in humans, and informs current regulatory risk-benefit analyses.


Assuntos
Adjuvantes Imunológicos/farmacocinética , Vacinas contra Influenza/química , Modelos Biológicos , Polissorbatos/farmacocinética , Esqualeno/farmacocinética , alfa-Tocoferol/farmacocinética , Tecido Adiposo/metabolismo , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Adjuvantes Imunológicos/sangue , Adjuvantes Imunológicos/química , Adulto , Animais , Química Farmacêutica , Simulação por Computador , Combinação de Medicamentos , Emulsões , Humanos , Lactente , Injeções Intramusculares , Sistema Linfático/metabolismo , Modelos Animais , Polissorbatos/administração & dosagem , Polissorbatos/efeitos adversos , Polissorbatos/química , Medição de Risco , Ovinos , Esqualeno/administração & dosagem , Esqualeno/efeitos adversos , Esqualeno/sangue , Esqualeno/química , alfa-Tocoferol/administração & dosagem , alfa-Tocoferol/efeitos adversos , alfa-Tocoferol/sangue , alfa-Tocoferol/química
10.
Glia ; 62(12): 1982-91, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25042766

RESUMO

Microglia are rapidly activated in the central nervous system (CNS) in response to a variety of injuries, including inflammation, trauma, and stroke. In addition to modulation of the innate immune response, a key function of microglia is the phagocytosis of dying cells and cellular debris, which can facilitate recovery. Despite emerging evidence that axonal debris can pose a barrier to regeneration of new axons in the CNS, little is known of the cellular and molecular mechanisms that underlie clearance of degenerating CNS axons. We utilize a custom micropatterned microfluidic system that enables robust microglial-axon co-culture to explore the role of Toll-like receptors (TLRs) in microglial phagocytosis of degenerating axons. We find that pharmacologic and genetic disruption of TLR4 blocks induction of the Type-1 interferon response and inhibits phagocytosis of axon debris in vitro. Moreover, TLR4-dependent microglial clearance of unmyelinated axon debris facilitates axon outgrowth. In vivo, microglial phagocytosis of CNS axons undergoing Wallerian degeneration in a dorsal root axotomy model is impaired in adult mice in which TLR4 has been deleted. Since purinergic receptors can influence TLR4-mediated signaling, we also explored a role for the microglia P2 receptors and found that the P2X7R contributes to microglial clearance of degenerating axons. Overall, we identify TLR4 as a key player in axonal debris clearance by microglia, thus creating a more permissive environment for axonal outgrowth. Our findings have significant implications for the development of protective and regenerative strategies for the many inflammatory, traumatic, and neurodegenerative conditions characterized by CNS axon degeneration.


Assuntos
Axônios/patologia , Microglia/metabolismo , Degeneração Neural/patologia , Fagocitose/genética , Receptor 4 Toll-Like/deficiência , Animais , Antígeno CD11b/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Técnicas de Cocultura , Citocinas/metabolismo , Embrião de Mamíferos , Hipocampo/citologia , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Técnicas Analíticas Microfluídicas , Degeneração Neural/genética , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2X7/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sulfonamidas/farmacologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
11.
Clin Cancer Res ; 30(11): 2309-2316, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38324398

RESUMO

In June 2022, the FDA extended the indication for lisocabtagene maraleucel (liso-cel) to include adults with large B-cell lymphoma (LBCL) who have refractory disease or relapse within 12 months of first-line chemoimmunotherapy (CIT), as well as transplant-ineligible adults with refractory disease or relapse after first-line CIT. Two clinical trials evaluating a single infusion of liso-cel preceded by lymphodepleting chemotherapy supported the second-line indications. TRANSFORM is a randomized, phase 3, open-label trial comparing liso-cel with standard second-line therapy, including planned autologous hematopoietic stem cell transplantation (HSCT), in 184 transplant-eligible patients. On interim analysis, event-free survival (EFS) by independent review committee (IRC) assessment was statistically significantly improved for the liso-cel arm, with a stratified hazard ratio of 0.34 [95% confidence interval (CI), 0.22-0.51; P < 0.0001]; the estimated median EFS was 10.1 months in the liso-cel arm versus 2.3 months in the control arm. PILOT is a single-arm phase 2 trial of second-line liso-cel in patients who were transplant-ineligible due to age or comorbidities but had adequate organ function for chimeric antigen receptor (CAR) T-cell therapy. Among 61 patients who received liso-cel (median age, 74 years), the IRC-assessed complete response rate was 54% (95% CI, 41-67). Among patients achieving complete response, the estimated 1-year rate of continued response was 68% (95% CI, 45-83). Of the 268 patients combined who received liso-cel as second-line therapy for LBCL, cytokine release syndrome occurred in 45% (Grade 3, 1.3%) and CAR T-cell-associated neurologic toxicities occurred in 27% (Grade 3, 7%), warranting a continued risk evaluation and mitigation strategy.


Assuntos
Aprovação de Drogas , Imunoterapia Adotiva , Linfoma Difuso de Grandes Células B , United States Food and Drug Administration , Humanos , Estados Unidos , Imunoterapia Adotiva/métodos , Imunoterapia Adotiva/efeitos adversos , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/terapia , Idoso , Masculino , Feminino , Pessoa de Meia-Idade , Adulto , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Produtos Biológicos/uso terapêutico
12.
J Neurosci ; 32(22): 7745-57, 2012 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-22649252

RESUMO

Following CNS injury, microglial phagocytosis of damaged endogenous tissue is thought to play an important role in recovery and regeneration. Previous work has focused on delineating mechanisms of clearance of neurons and myelin. Little, however, is known of the mechanisms underlying phagocytosis of axon debris. We have developed a novel microfluidic platform that enables coculture of microglia with bundles of CNS axons to investigate mechanisms of microglial phagocytosis of axons. Using this platform, we find that axon degeneration results in the induction of type-1 interferon genes within microglia. Pharmacologic and genetic disruption of Toll/interleukin-1 receptor domain-containing adapter inducing interferon-ß (TRIF), a Toll-like receptor adapter protein, blocks induction of the interferon response and inhibits microglial phagocytosis of axon debris in vitro. In vivo, microglial phagocytosis of axons following dorsal root axotomy is impaired in mice in which TRIF has been genetically deleted. Furthermore, we identify the p38 mitogen-activated protein kinase (MAPK) cascade as a signaling pathway downstream of TRIF following axon degeneration and find that inhibition of p38 MAPK by SB203580 (4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)-1H-imidazole) also blocked clearance of axon debris. Finally, we find that TRIF-dependent microglial clearance of unmyelinated axon debris facilitates axon outgrowth. Overall, we provide evidence that TRIF-mediated signaling plays an unexpected role in axonal debris clearance by microglia, thereby facilitating a more permissive environment for axonal outgrowth. Our study has significant implications for the development of novel regenerative and restorative strategies for the many traumatic, neuroinflammatory, and neurodegenerative conditions characterized by CNS axon degeneration.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Interferon beta/metabolismo , Microglia/fisiologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Fagocitose/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/química , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Trifosfato de Adenosina/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Axônios/patologia , Axotomia , Antígeno CD11b/metabolismo , Células Cultivadas , Quimiocina CXCL10/metabolismo , Técnicas de Cocultura , Modelos Animais de Doenças , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Gânglios Espinais/lesões , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hipocampo/citologia , Humanos , Imidazóis/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Técnicas Analíticas Microfluídicas , Degeneração Neural/induzido quimicamente , Neurônios/citologia , Neurônios/efeitos dos fármacos , Óxido Nítrico/farmacologia , Peptídeos/farmacologia , Fagocitose/genética , Piridinas/farmacologia , Compostos de Amônio Quaternário/toxicidade , Ratos , Ratos Sprague-Dawley , Receptores de Interleucina-1/metabolismo , Fatores de Tempo , Transfecção
13.
J Pharmacokinet Pharmacodyn ; 40(5): 545-56, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23912214

RESUMO

Squalene is used in the oil phase of certain emulsion vaccine adjuvants, but its fate as a vaccine component following intramuscular (IM) injection in humans is unknown. In this study, we constructed a physiologically-based pharmacokinetic (PBPK) model for intramuscularly injected squalene-in-water (SQ/W) emulsion, in order to make a quantitative estimation of the tissue distribution of squalene following a single IM injection in humans. The PBPK model incorporates relevant physicochemical properties of squalene; estimates of the time course of cracking of a SQ/W emulsion; anatomical and physiological parameters at the injection site and beyond; and local, preferential lymphatic transport. The model predicts that a single dose of SQ/W emulsion will be removed from human deltoid muscle within six days following IM injection. The major proportion of the injected squalene will be distributed to draining lymph nodes and adipose tissues. The model indicates slow decay from the latter compartment most likely due to partitioning into neutral lipids and a low rate of squalene biotransformation there. Parallel pharmacokinetic modeling for mouse muscle suggests that the kinetics of SQ/W emulsion correspond to the immunodynamic time course of a commercial squalene-containing adjuvant reported in that species. In conclusion, this study makes important pharmacokinetic predictions of the fate of a squalene-containing emulsion in humans. The results of this study may be relevant for understanding the immunodynamics of this new class of vaccine adjuvants and may be useful in future quantitative risk analyses that incorporate mode-of-action data.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/farmacocinética , Esqualeno/imunologia , Esqualeno/farmacocinética , Vacinas/imunologia , Vacinas/farmacocinética , Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Animais , Emulsões/administração & dosagem , Humanos , Injeções Intramusculares/métodos , Cinética , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos , Modelos Biológicos , Distribuição Tecidual/imunologia
14.
J Clin Pharmacol ; 63 Suppl 2: S110-S116, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37942903

RESUMO

The current dosing strategy of immune globulin products for the treatment of primary immunodeficiency diseases (PIDDs) in the USA is based on total body weight (BW). The aim of our study was to assess the relationship between dose and trough level, and to determine whether an alternative dosing strategy should be considered for patients who are overweight or obese. We analyzed data in a total of 533 patients from 11 studies. We modeled the relationship between trough level and dose per week using a linear mixed model. We used an over-dispersed Poisson model to model the relationship between infection and trough level. In these analyses, we then combined the study-specific treatment effects using a random-effect or fixed-effect model. The mean administered dose per week was 9.77, 14.00, or 18.17 g in patients who were normal weight, overweight, or obese, respectively. Compared with a patient of normal weight, a 1 g increase in dose per week in a patient who was overweight was associated with a smaller increase in the trough level, 0.08 g/L less (95%CI -0.14 to -0.03 g/L), and a 1 g increase in dose per week in a patient who was obese was associated with a much smaller increase in trough level, 0.01 g/L less (95% CI -0.07 to 0.06 g/L). Last, for a 1 unit (g/L) increase in trough level, the expected number of infections remained the same, with a multiplicative factor of 1.01 (95%CI 0.98-1.04). Overall, we found no compelling evidence to justify a reconsideration of the current dosing strategy based on total BW for patients with PIDDs who are overweight or obese.


Assuntos
Sobrepeso , Doenças da Imunodeficiência Primária , Humanos , Imunoglobulina G/uso terapêutico , Obesidade/complicações , Obesidade/tratamento farmacológico , Sobrepeso/complicações , Doenças da Imunodeficiência Primária/complicações , Doenças da Imunodeficiência Primária/tratamento farmacológico , Ensaios Clínicos como Assunto
15.
Int Immunopharmacol ; 117: 109914, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36841154

RESUMO

Pharmacokinetics (PK) studies are important to determine a safe and effective dose of both small and large molecule drugs. Intrinsic factors such as pregnancy can substantially alter the PK of a drug. Several PK studies have been published for small molecules administered during pregnancy, but such investigations are scarce for macromolecules including monoclonal and polyclonal antibodies. In this part 1 of 2 reviews, we first provide a general description of macromolecule drugs, the PK differences with small molecules, and current knowledge on their absorption, distribution, metabolism and elimination in non-pregnant subjects. We then review in detail the physiological changes during pregnancy. While some of the physiologic adaptions of pregnancy, for example increased plasma volume and cardiac output, are expected to impact PK of antibody therapeutics, the effects of others, such as increased GFR and altered immune responses are not fully understood. We conclude that further investigations are needed to fully elucidate how pregnancy can impact PK properties of macromolecules.


Assuntos
Anticorpos Monoclonais , Anticorpos , Farmacocinética , Feminino , Humanos , Gravidez , Anticorpos/farmacologia , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia
16.
Int Immunopharmacol ; 119: 109915, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36842918

RESUMO

In Part 1, we provided a general description of macromolecules, pharmacokinetics (PK) characteristics in non-pregnant subjects, and the physiological changes during pregnancy. Here we further elaborate on the impact of pregnancy on the PK of antibodies through illustrative case studies (immunoglobulins, infliximab, adalimumab and eculizumab). Using published data from nonclinical and clinical studies, we present measured or calculated PK parameters from pregnant subjects comparing with data from non-pregnant subjects, if available. Due to the paucity of PK data evaluating PK of antibodies during pregnancy, we also provide examples of PK studies for small molecules. Finally, we draw conclusions on the nature and direction of PK changes for both antibodies and small molecules as well as provide recommendations for areas that would benefit from further studies.


Assuntos
Modelos Biológicos , Farmacocinética , Gravidez , Feminino , Humanos , Adalimumab , Infliximab
17.
Cell Mol Neurobiol ; 32(6): 1021-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22373696

RESUMO

Botulinum neurotoxins (BoNTs) internalize into nerve terminals and block the release of neurotransmitters into the synapse. BoNTs are widely used as a therapeutic agent for treatment of movement disorders and recently gained more attention as a biological weapon. Consequently, there is strong interest to develop a cell-based assay platform to screen the toxicity and bioactivity of the BoNTs. In this study, we present an in vitro screening assay for BoNT/A based on differentiated human embryonal carcinoma stem (NT2) cells. The human NT2 cells fully differentiated into mature neurons that display immunoreactivity to cytoskeletal markers (ßIII-tubulin and MAP2) and presynaptic proteins (synapsin and synaptotagmin I). We showed that the human NT2 cells undergo a process of exo-endocytotic synaptic vesicle recycling upon depolarization with high K(+) buffer. By employing an antibody directed against light chain of BoNT/A, we detected internalized toxin as a punctate staining along the neurites of the NT2 neurons. Using well-established methods of synaptic vesicle exocytosis assay (luminal synaptotagmin I and FM1-43 imaging) we show that pre-incubation with BoNT/A resulted in a blockade of vesicle release from human NT2 neurons in a dose-dependent manner. Moreover, this blocking effect of BoNT/A was abolished by pre-adsorbing the toxin with neutralizing antibody. In a proof of principle, we demonstrate that our cell culture assay for vesicle release is sensitive to BoNT/A and the activity of BoNT/A can be blocked by specific neutralizing antibodies. Overall our data suggest that human NT2 neurons are suitable for large scale screening of botulinum bioactivity.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurotransmissores/metabolismo , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/metabolismo , Anticorpos Neutralizantes/farmacologia , Diferenciação Celular/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Humanos , Modelos Biológicos , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Teratocarcinoma/patologia
18.
Cell Mol Life Sci ; 68(12): 2089-99, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20957508

RESUMO

Neuronal migration is one of the most critical processes during early brain development. The gaseous messenger nitric oxide (NO) has been shown to modulate neuronal and glial migration in various experimental models. Here, we analyze a potential role for NO signaling in the migration of fetal human neural progenitor cells. Cells migrate out of cultured neurospheres and differentiate into both neuronal and glial cells. The neurosphere cultures express neuronal nitric oxide synthase and soluble guanylyl cyclase that produces cGMP upon activation with NO. By employing small bioactive enzyme activators and inhibitors in both gain and loss of function experiments, we show NO/cGMP signaling as a positive regulator of migration in neurosphere cultures of early developing human brain cells. Since NO signaling regulates cell movements from developing insects to mammalian nervous systems, this transduction pathway may have evolutionary conserved functions.


Assuntos
Movimento Celular , GMP Cíclico/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Técnicas de Cultura de Células , Diferenciação Celular , Humanos , Neuroglia/citologia , Neurônios/citologia
19.
Cell Mol Neurobiol ; 31(4): 635-43, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21331625

RESUMO

Human neurons derived from stem cells can be employed as in vitro models to predict the potential of neurochemicals affecting neurodevelopmental cellular processes including proliferation, migration, and differentiation. Here, we developed a model of differentiating human neurons from well characterized human embryonal carcinoma stem cells (NT2). NT2 cells were induced to differentiate into neuronal phenotypes after 2 weeks of treatment with retinoic acid in aggregate culture. Nestin positive progenitor cells migrate out of NT2 aggregates and differentiate into ßIII-tubulin expressing neuronal cells. Culturing the NT2 cells for an additional 7-14 days resulted in increased percentage of ßIII-tubulin expressing cells, elaborating a long neurite that positively stained for axonal marker (Tau) and presynaptic protein (synapsin). We then asked whether neurite outgrowth from NT2 cells is modulated by bioactive chemicals. Since the cAMP/PKA pathway has been widely investigated as a regulator of neurite outgrowth/regeneration in several experimental systems, we used chemical activators and inhibitors of cAMP/PKA pathway in the culture. The adenylyl cyclase activator, forskolin, and cell-permeable analog of cAMP, 8-Br-cAMP increased the percentage of neurite bearing cells and neurite extension. Application of the protein kinase A inhibitors, H-89 and Rp-cAMP, blocked neurite formation. Taken together, NT2 aggregates undergo migration, differentiation, and neurite elaboration and can be used as a model of differentiating human neurons to screen neurochemicals and to understand cellular mechanisms of human nerve cell development.


Assuntos
Diferenciação Celular , Células-Tronco de Carcinoma Embrionário/citologia , Neuritos/metabolismo , Axônios/efeitos dos fármacos , Axônios/metabolismo , Diferenciação Celular/efeitos dos fármacos , Colforsina/farmacologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células-Tronco de Carcinoma Embrionário/efeitos dos fármacos , Humanos , Neuritos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia
20.
J Clin Pharmacol ; 61 Suppl 1: S108-S116, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34185903

RESUMO

There is a growing interest in the use of physiologically based pharmacokinetic (PBPK) models as clinical pharmacology drug development tools. In PBPK modeling, not every organ or physiological parameter is required, leading to the development of a minimal PBPK (mPBPK) model, which is simple and efficient. The objective of this study was to streamline mPBPK modeling approaches and enable straightforward prediction of clearance of protein-based products in children. Four mPBPK models for scaling clearance from adult to children were developed and evaluated on Excel spreadsheets using (1) liver and kidneys; (2) liver, kidneys, and skin; (3) liver, kidneys, skin, and lymph; and (4) interstitial, lymph, and plasma volume. There were 35 therapeutic proteins with a total of 113 observations across different age groups (premature neonates to adolescents). For monoclonal and polyclonal antibodies, more than 90% of observations were within a 0.5- to 2-fold prediction error for all 4 methods. For nonantibodies, 79% to 100% of observations were within the 0.5- to 2-fold prediction error for the 4 different methods. Methods 1 and 4 provided the best results, >90% of the total observations were within the 0.5- to 2-fold prediction error for all 3 classes of protein-based products across a wide age range. The precision of clearance prediction was comparatively lower in children ≤2 years of age vs older children (>2 years of age) with methods 1 and 4 predicting 80% to 100% and 75% to 90% of observations within the 0.5- to 2-fold prediction error, respectively. The results of the study indicated that mPBPK models can be developed on spreadsheets, with acceptable performance for prediction of clearance.


Assuntos
Produtos Biológicos/farmacocinética , Vias de Eliminação de Fármacos/fisiologia , Taxa de Depuração Metabólica/fisiologia , Modelos Biológicos , Pediatria/métodos , Proteínas/farmacocinética , Adolescente , Fatores Etários , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Produtos Biológicos/administração & dosagem , Criança , Pré-Escolar , Humanos , Imunoglobulinas Intravenosas/administração & dosagem , Imunoglobulinas Intravenosas/farmacocinética , Lactente , Recém-Nascido , Proteínas/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA