Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
CPT Pharmacometrics Syst Pharmacol ; 13(4): 544-550, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38343040

RESUMO

Personalized dosing approaches play important roles in clinical practices to improve benefit: risk profiles. Whereas this is also important for drug development, especially in the context of drugs with narrow therapeutic windows, such approaches have not been fully evaluated during clinical development. Fazpilodemab (BFKB8488A) is an agonistic bispecific antibody which was being developed for the treatment of nonalcoholic steatohepatitis. The objective of this study was to characterize the exposure-response relationships of fazpilodemab with the purpose of guiding dose selection for a phase II study, as well as to evaluate various personalized dosing strategies to optimize the treatment benefit. Fazpilodemab exhibited clear exposure-response relationships for a pharmacodynamic (PD) biomarker and gastrointestinal adverse events (GIAEs), such as nausea and vomiting. Static exposure-response analysis, as well as longitudinal adverse event (AE) analysis using discrete-time Markov model, were performed to characterize the observations. Clinical trial simulations were performed based on the developed exposure-response models to evaluate probability of achieving target PD response and the frequency of GIAEs to inform phase II dose selection. Dynamic simulation of personalized dosing strategies demonstrated that the AE-based personalized dosing is the most effective approach for optimizing the benefit-risk profiles. The approach presented here can be a useful framework for quantifying the benefit of personalized dosing for drugs with narrow therapeutic windows.


Assuntos
Proteínas Klotho , Humanos
2.
Gut ; 72(8): 1451-1461, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36732049

RESUMO

BACKGROUND: The interleukin-22 cytokine (IL-22) has demonstrated efficacy in preclinical colitis models with non-immunosuppressive mechanism of action. Efmarodocokin alfa (UTTR1147A) is a fusion protein agonist that links IL-22 to the crystallisable fragment (Fc) of human IgG4 for improved pharmacokinetic characteristics, but with a mutation to minimise Fc effector functions. METHODS: This randomised, phase 1b study evaluated the safety, tolerability, pharmacokinetics and pharmacodynamics of repeat intravenous dosing of efmarodocokin alfa in healthy volunteers (HVs; n=32) and patients with ulcerative colitis (n=24) at 30-90 µg/kg doses given once every 2 weeks or monthly (every 4 weeks) for 12 weeks (6:2 active:placebo per cohort). RESULTS: The most common adverse events (AEs) were on-target, reversible, dermatological effects (dry skin, erythema and pruritus). Dose-limiting non-serious dermatological AEs (severe dry skin, erythema, exfoliation and discomfort) were seen at 90 µg/kg once every 2 weeks (HVs, n=2; patients, n=1). Pharmacokinetics were generally dose-proportional across the dose levels, but patients demonstrated lower drug exposures relative to HVs at the same dose. IL-22 serum biomarkers and IL-22-responsive genes in colon biopsies were induced with active treatment, and microbiota composition changed consistent with a reversal in baseline dysbiosis. As a phase 1b study, efficacy endpoints were exploratory only. Clinical response was observed in 7/18 active-treated and 1/6 placebo-treated patients; clinical remission was observed in 5/18 active-treated and 0/6 placebo-treated patients. CONCLUSION: Efmarodocokin alfa had an adequate safety and pharmacokinetic profile in HVs and patients. Biomarker data confirmed IL-22R pathway activation in the colonic epithelium. Results support further investigation of this non-immunosuppressive potential inflammatory bowel disease therapeutic. TRIAL REGISTRATION NUMBER: NCT02749630.


Assuntos
Colite Ulcerativa , Humanos , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/patologia , Voluntários Saudáveis , Administração Intravenosa , Biomarcadores
3.
Hepatology ; 78(3): 847-862, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35993161

RESUMO

BACKGROUND AND AIMS: BFKB8488A is a bispecific antibody targeting fibroblast growth factor receptor 1c and Klothoß. This phase 1b study assessed safety, tolerability, pharmacokinetics, immunogenicity, and pharmacodynamics of BFKB8488A in patients with type 2 diabetes mellitus (T2DM) or NAFLD. APPROACH AND RESULTS: Patients were randomized to receive multiple doses of BFKB8488A at various dose levels and dosing intervals (weekly, every 2 weeks, or every 4 weeks) or placebo for 12 weeks. The primary outcome was the safety of BFKB8488A. Overall, 153 patients (T2DM: 91; NAFLD: 62) were enrolled and received at least one dose of treatment. Of these, 102 patients (62.7%) reported at least one adverse event (BFKB8488A: 83 [68.6%]; placebo: 19 [59.4%]). BFKB8488A exhibited nonlinear pharmacokinetics, with greater than dose-proportional increases in exposure. The treatment-emergent antidrug antibody incidence was 22.7%. Overall, trends in exposure-dependent increases in high-density lipoprotein (HDL) and decreases in triglyceride levels were observed. Decreases in alanine aminotransferase and aspartate aminotransferase were 0.7% and 9.2% for medium exposure and 7.3% and 11.2% for high-exposure tertiles, compared with increases of 7.5% and 17% in the placebo group, respectively, at Day 85. In patients with NAFLD, the mean decrease from baseline liver fat was 13.0%, 34.5%, and 49.0% in the low-, medium-, and high-exposure tertiles, respectively, compared with 0.1% with placebo at Day 85. CONCLUSIONS: BFKB8488A was adequately tolerated in patients with T2DM or NAFLD, leading to triglyceride reduction, HDL improvements, and trends in improvement in markers of liver health for both populations and marked liver fat reduction in patients with NAFLD. ( ClinicalTrials.gov : NCT03060538).


Assuntos
Diabetes Mellitus Tipo 2 , Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/uso terapêutico , Método Duplo-Cego , Triglicerídeos , Lipídeos
4.
ACS Nano ; 16(9): 14456-14462, 2022 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-36074897

RESUMO

Optical contrast is the most common preliminary method to identify layer number of two-dimensional (2D) materials, but it is seldom used as a confirmatory technique. We explain the reason for variation of optical contrast between imaging systems, motivating system-independent measurement of optical contrast as a critical need. We describe a universal method to quantify the layer number using the RGB (red-green-blue) and RAW optical images. For RGB images, the slope of 2D flake (MoS2, WSe2, graphene) intensity vs substrate intensity is extracted from optical images with varying lamp power. The intensity slope identifies layer number and is system independent. For RAW images, intensity slopes and intensity ratios are completely system and intensity independent. Intensity slope (for RGB) and intensity ratio (for RAW) are thus universal parameters for identifying layer number. The RAW format is not present in all imaging systems, but it can confirm layer number using a single optical image, making it a rapid and system-independent universal method. A Fresnel-reflectance-based optical model provides an excellent match with experiments. Furthermore, we have created a MATLAB-based graphical user interface that can identify layer number rapidly. This technique is expected to accelerate the preparation of heterostructures and to fulfill a prolonged need for universal optical contrast method.

5.
J Allergy Clin Immunol ; 150(6): 1517-1524, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36041655

RESUMO

BACKGROUND: The binding of IL-33 to its receptor ST2 (alias of IL1RL1) leads to the release of inflammatory mediators and may play a role in the pathogenesis of atopic dermatitis. Astegolimab is a fully human, IgG2 mAb that binds to ST2 and inhibits IL-33 signaling. OBJECTIVES: This study sought to assess the efficacy, safety, and pharmacokinetics of astegolimab in patients with atopic dermatitis. METHODS: This was a randomized, placebo-controlled, phase 2 study in which adults with chronic atopic dermatitis were randomized 1:1 to receive astegolimab 490 mg every 4 weeks or placebo, for 16 weeks. The primary outcome was the percentage of change from baseline to week 16 of the Eczema Area and Severity Index score. RESULTS: A total of 65 patients were enrolled in the study (placebo, n = 32; astegolimab, n = 33). The adjusted mean percentage of change from baseline to week 16 in the Eczema Area and Severity Index score was -51.47% for astegolimab compared with -58.24% for placebo, with a nonsignificant treatment difference of 6.77% (95% CI: -16.57-30.11; P = .5624). No differences were observed between treatment groups for secondary efficacy outcomes and in exploratory biomarkers (blood eosinophils, serum IL-5, serum CCL13). With the use of loading dose, pharmacokinetic exposure was sufficient from week 1. Astegolimab was well-tolerated, with a safety profile consistent with that observed in previous clinical trials. CONCLUSIONS: In patients with atopic dermatitis, astegolimab did not show a significant difference compared to placebo for the primary or secondary outcomes.


Assuntos
Anticorpos Monoclonais Humanizados , Dermatite Atópica , Humanos , Dermatite Atópica/tratamento farmacológico , Interleucina-33 , Anticorpos Monoclonais Humanizados/uso terapêutico
6.
J Allergy Clin Immunol ; 148(3): 790-798, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33872652

RESUMO

BACKGROUND: The IL-33/ST2 pathway is linked with asthma susceptibility. Inhaled allergens, pollutants, and respiratory viruses, which trigger asthma exacerbations, induce release of IL-33, an epithelial-derived "alarmin." Astegolimab, a human IgG2 mAb, selectively inhibits the IL-33 receptor, ST2. Approved biologic therapies for severe asthma mainly benefit patients with elevated blood eosinophils (type 2-high), but limited options are available for patients with low blood eosinophils (type 2-low). Inhibiting IL-33 signaling may target pathogenic pathways in a wider spectrum of asthmatics. OBJECTIVES: This study evaluated astegolimab efficacy and safety in patients with severe asthma. METHODS: This double-blind, placebo-controlled, dose-ranging study (ZENYATTA [A Study to Assess the Efficacy and Safety of MSTT1041A in Participants With Uncontrolled Severe Asthma]) randomized 502 adults with severe asthma to subcutaneous placebo or 70-mg, 210-mg, or 490-mg doses of astegolimab every 4 weeks. The primary endpoint was the annualized asthma exacerbation rate (AER) at week 54. Enrollment caps ensured ∼30 patients who were eosinophil-high (≥300 cells/µL) and ∼95 patients who were eosinophil-low (<300 cells/µL) per arm. RESULTS: Overall, adjusted AER reductions relative to placebo were 43% (P = .005), 22% (P = .18), and 37% (P = .01) for 490-mg, 210-mg, and 70-mg doses of astegolimab, respectively. Adjusted AER reductions for patients who were eosinophil-low were comparable to reductions in the overall population: 54% (P = .002), 14% (P = .48), and 35% (P = .05) for 490-mg, 210-mg, and 70-mg doses of astegolimab. Adverse events were similar in astegolimab- and placebo-treated groups. CONCLUSIONS: Astegolimab reduced AER in a broad population of patients, including those who were eosinophil-low, with inadequately controlled, severe asthma. Astegolimab was safe and well tolerated.


Assuntos
Antiasmáticos/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Asma/tratamento farmacológico , Adulto , Antiasmáticos/efeitos adversos , Antiasmáticos/farmacocinética , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Asma/imunologia , Progressão da Doença , Método Duplo-Cego , Eosinófilos/imunologia , Feminino , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1/antagonistas & inibidores , Interleucina-33/antagonistas & inibidores , Contagem de Leucócitos , Masculino , Pessoa de Meia-Idade , Método Simples-Cego , Resultado do Tratamento
7.
Drug Saf ; 44(2): 133-165, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33141341

RESUMO

With the widespread development of new drugs to treat chronic liver diseases (CLDs), including viral hepatitis and nonalcoholic steatohepatitis (NASH), more patients are entering trials with abnormal baseline liver tests and with advanced liver injury, including cirrhosis. The current regulatory guidelines addressing the monitoring, diagnosis, and management of suspected drug-induced liver injury (DILI) during clinical trials primarily address individuals entering with normal baseline liver tests. Using the same laboratory criteria cited as signals of potential DILI in studies involving patients with no underlying liver disease and normal baseline liver tests may result in premature and unnecessary cessation of a study drug in a clinical trial population whose abnormal and fluctuating liver tests are actually due to their underlying CLD. This position paper focuses on defining best practices for the detection, monitoring, diagnosis, and management of suspected acute DILI during clinical trials in patients with CLD, including hepatitis C virus (HCV) and hepatitis B virus (HBV), both with and without cirrhosis and NASH with cirrhosis. This is one of several position papers developed by the IQ DILI Initiative, comprising members from 16 pharmaceutical companies in collaboration with DILI experts from academia and regulatory agencies. It is based on an extensive literature review and discussions between industry members and experts from outside industry to achieve consensus regarding the recommendations. Key conclusions and recommendations include (1) the importance of establishing laboratory criteria that signal potential DILI events and that fit the disease indication being studied in the clinical trial based on knowledge of the natural history of test fluctuations in that disease; (2) establishing a pretreatment value that is based on more than one screening determination, and revising that baseline during the trial if a new nadir is achieved during treatment; (3) basing rules for increased monitoring and for stopping drug for potential DILI on multiples of baseline liver test values and/or a threshold value rather than multiples of the upper limit of normal (ULN) for that test; (4) making use of more sensitive tests of liver function, including direct bilirubin (DB) or combined parameters such as aspartate transaminase:alanine transaminase (AST:ALT) ratio or model for end-stage liver disease (MELD) to signal potential DILI, especially in studies of patients with cirrhosis; and (5) being aware of potential confounders related to complications of the disease being studied that may masquerade as DILI events.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Consenso , Guias de Prática Clínica como Assunto , Adulto , Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Doença Hepática Induzida por Substâncias e Drogas/terapia , Ensaios Clínicos como Assunto , Hepatite B/complicações , Hepatite C/complicações , Hepatite Crônica/epidemiologia , Humanos , Cirrose Hepática/etiologia , Cirrose Hepática/virologia , Hepatopatia Gordurosa não Alcoólica/complicações
8.
Clin Pharmacol Ther ; 107(2): 333-346, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31314926

RESUMO

The diagnosis and management of drug-induced liver injury (DILI) remains a challenge in clinical trials in drug development. The qualification of emerging biomarkers capable of predicting DILI soon after the initiation of treatment, differentiating DILI from underlying liver disease, identifying the causal entity, and assigning appropriate treatment options after DILI is diagnosed are needed. Qualification efforts have been hindered by lack of properly stored and consented biospecimens that are linked to clinical data relevant to a specific context of use. Recommendations are made for biospecimen collection procedures, with the focus on clinical trials, and for specific emerging biomarkers to focus qualification efforts.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Desenvolvimento de Medicamentos/métodos , Desenvolvimento de Medicamentos/normas , Manejo de Espécimes/métodos , Manejo de Espécimes/normas , Biomarcadores , Doença Hepática Induzida por Substâncias e Drogas/sangue , Ensaios Clínicos como Assunto/normas , Humanos , Consentimento Livre e Esclarecido , Testes de Função Hepática , Fenótipo
9.
Expert Opin Drug Discov ; 13(2): 193-205, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29190166

RESUMO

INTRODUCTION: The progressive disease spectrum of non-alcoholic fatty liver disease (NAFLD), which includes non-alcoholic steatohepatitis (NASH), is a rapidly emerging public health crisis with no approved therapy. The diversity of various therapies under development highlights the lack of consensus around the most effective target, underscoring the need for better translatable preclinical models to study the complex progressive disease and effective therapies. Areas covered: This article reviews published literature of various mouse models of NASH used in preclinical studies, as well as complex organotypic in vitro and ex vivo liver models being developed. It discusses translational challenges associated with both kinds of models, and describes some of the studies that validate their application in NAFLD. Expert opinion: Animal models offer advantages of understanding drug distribution and effects in a whole body context, but are limited by important species differences. Human organotypic in vitro and ex vivo models with physiological relevance and translatability need to be used in a tiered manner with simpler screens. Leveraging newer technologies, like metabolomics, proteomics, and transcriptomics, and the future development of validated disease biomarkers will allow us to fully utilize the value of these models to understand disease and evaluate novel drugs in isolation or combination.


Assuntos
Desenho de Fármacos , Descoberta de Drogas/métodos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Animais , Modelos Animais de Doenças , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Camundongos , Modelos Biológicos , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Pesquisa Translacional Biomédica/métodos
10.
Expert Opin Drug Metab Toxicol ; 13(2): 193-204, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27759439

RESUMO

INTRODUCTION: Drug induced steatohepatitis (DISH), a form of drug induced liver injury (DILI) is characterized by intracellular accumulation of lipids in hepatocytes and subsequent inflammatory events, in some ways similar to the pathology seen with other metabolic, viral and genetic causes of non alcoholic fatty liver disease and steatohepatitis (NAFLD and NASH). Areas covered: This paper provides a comprehensive review of the main underlying mechanisms by which various drugs cause DISH, and outlines existing preclinical tools to predict it and study underlying pathways involved. The translational hurdles of these models are discussed, with the example of an organotypic liver system designed to address them. Finally, we describe the clinical assessment and management of DISH. Expert Opinion: The complexity of the interconnected mechanistic pathways underlying DISH makes it important that preclinical evaluation of drugs is done in a physiologically and metabolically relevant context. Advanced organotypic tissue models, coupled with translational functional biomarkers and next-generational pan-omic measurements, may offer the best shot at gathering mechanistic knowledge and potential of a drug causing steatohepatitis. Ultimately this information could also help predict, detect or guide the development of specific treatments for DISH, which is an unmet need as of today.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/etiologia , Fígado Gorduroso/induzido quimicamente , Hepatócitos/efeitos dos fármacos , Animais , Doença Hepática Induzida por Substâncias e Drogas/patologia , Avaliação Pré-Clínica de Medicamentos/métodos , Fígado Gorduroso/patologia , Hepatócitos/patologia , Humanos , Metabolismo dos Lipídeos , Modelos Biológicos
11.
ALTEX ; 34(2): 301-310, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27846345

RESUMO

Translating in vitro biological data into actionable information related to human health holds the potential to improve disease treatment and risk assessment of chemical exposures. While genomics has identified regulatory pathways at the cellular level, translation to the organism level requires a multiscale approach accounting for intra-cellular regulation, inter-cellular interaction, and tissue/organ-level effects. Tissue-level effects can now be probed in vitro thanks to recently developed systems of three-dimensional (3D), multicellular, "organotypic" cell cultures, which mimic functional responses of living tissue. However, there remains a knowledge gap regarding interactions across different biological scales, complicating accurate prediction of health outcomes from molecular/genomic data and tissue responses. Systems biology aims at mathematical modeling of complex, non-linear biological systems. We propose to apply a systems biology approach to achieve a computational representation of tissue-level physiological responses by integrating empirical data derived from organotypic culture systems with computational models of intracellular pathways to better predict human responses. Successful implementation of this integrated approach will provide a powerful tool for faster, more accurate and cost-effective screening of potential toxicants and therapeutics. On September 11, 2015, an interdisciplinary group of scientists, engineers, and clinicians gathered for a workshop in Research Triangle Park, North Carolina, to discuss this ambitious goal. Participants represented laboratory-based and computational modeling approaches to pharmacology and toxicology, as well as the pharmaceutical industry, government, non-profits, and academia. Discussions focused on identifying critical system perturbations to model, the computational tools required, and the experimental approaches best suited to generating key data.


Assuntos
Técnicas de Cultura de Células , Simulação por Computador , Biologia de Sistemas , Alternativas aos Testes com Animais , Animais , Técnicas de Cultura de Células/métodos , Substâncias Perigosas/toxicidade , Humanos , Dispositivos Lab-On-A-Chip , Medição de Risco
12.
JCI Insight ; 1(20): e90954, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27942596

RESUMO

A barrier to drug development for nonalcoholic steatohepatitis (NASH) is the absence of translational preclinical human-relevant systems. An in vitro liver model was engineered to incorporate hepatic sinusoidal flow, transport, and lipotoxic stress risk factors (glucose, insulin, free fatty acids) with cocultured primary human hepatocytes, hepatic stellate cells (HSCs), and macrophages. Transcriptomic, lipidomic, and functional endpoints were evaluated and compared with clinical data from NASH patient biopsies. The lipotoxic milieu promoted hepatocyte lipid accumulation (4-fold increase, P < 0.01) and a lipidomics signature similar to NASH biopsies. Hepatocyte glucose output increased with decreased insulin sensitivity. These changes were accompanied by increased inflammatory analyte secretion (e.g., IL-6, IL-8, alanine aminotransferase). Fibrogenic activation markers increased with lipotoxic conditions, including secreted TGF-ß (>5-fold increase, P < 0.05), extracellular matrix gene expression, and HSC activation. Significant pathway correlation existed between this in vitro model and human biopsies. Consistent with clinical trial data, 0.5 µM obeticholic acid in this model promoted a healthy lipidomic signature, reduced inflammatory and fibrotic secreted factors, but also increased ApoB secretion, suggesting a potential adverse effect on lipoprotein metabolism. Lipotoxic stress activates similar biological signatures observed in NASH patients in this system, which may be relevant for interrogating novel therapeutic approaches to treat NASH.


Assuntos
Técnicas de Cocultura , Células Estreladas do Fígado/citologia , Hepatócitos/citologia , Macrófagos/citologia , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Animais , Glucose/metabolismo , Humanos , Inflamação , Resistência à Insulina , Lipídeos/análise , Fígado , Metaboloma , Camundongos Endogâmicos C57BL , Modelos Biológicos , Transcriptoma
13.
Mol Genet Metab ; 117(3): 355-362, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26740382

RESUMO

BACKGROUND: Propionic acidemia (PA) is a disorder of intermediary metabolism with defects in the alpha or beta subunits of propionyl CoA carboxylase (PCCA and PCCB respectively) enzyme. We previously described a liver culture system that uses liver-derived hemodynamic blood flow and transport parameters to restore and maintain primary human hepatocyte biology and metabolism utilizing physiologically relevant milieu concentrations. METHODS: In this study, primary hepatocytes isolated from the explanted liver of an 8-year-old PA patient were cultured in the liver system for 10 days and evaluated for retention of differentiated polarized morphology. The expression of PCCA and PCCB was assessed at a gene and protein level relative to healthy donor controls. Ammonia and urea levels were measured in the presence and absence of amino acid supplements to assess the metabolic consequences of branched-chain amino acid metabolism in this disease. RESULTS: Primary hepatocytes from the PA patient maintained a differentiated polarized morphology (peripheral actin staining) over 10 days of culture in the system. We noted lower levels of PCCA and PCCB relative to normal healthy controls at the mRNA and protein level. Supplementation of branched-chain amino acids, isoleucine (5mM) and valine (5mM) in the medium, resulted in increased ammonia and decreased urea in the PA patient hepatocyte system, but no such response was seen in healthy hepatocytes or patient-derived fibroblasts. CONCLUSIONS: We demonstrate for the first time the successful culture of PA patient-derived primary hepatocytes in a differentiated state, that stably retain the PCCA and PCCB enzyme defects at a gene and protein level. Phenotypic response of the system to an increased load of branched-chain amino acids, not possible with fibroblasts, underscores the utility of this system in the better understanding of the molecular pathophysiology of PA and examining the effectiveness of potential therapeutic agents in the most relevant tissue.


Assuntos
Hepatócitos/citologia , Hepatócitos/metabolismo , Acidemia Propiônica/metabolismo , Actinas/análise , Aminoácidos de Cadeia Ramificada/metabolismo , Amônia/metabolismo , Carbono-Carbono Ligases/genética , Carbono-Carbono Ligases/metabolismo , Células Cultivadas , Criança , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Hemodinâmica , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Humanos , Isoleucina/farmacologia , Fígado/enzimologia , Fígado/metabolismo , Fígado/patologia , Metilmalonil-CoA Descarboxilase/genética , Metilmalonil-CoA Descarboxilase/metabolismo , Mutação , Ureia/metabolismo , Valina/farmacologia
14.
Chem Biol Interact ; 255: 31-44, 2016 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-26626330

RESUMO

Drug induced liver injury (DILI), a major cause of pre- and post-approval failure, is challenging to predict pre-clinically due to varied underlying direct and indirect mechanisms. Nevirapine, a non-nucleoside reverse transcriptase inhibitor (NNRTI) and Ritonavir, a protease inhibitor, are antiviral drugs that cause clinical DILI with different phenotypes via different mechanisms. Assessing DILI in vitro in hepatocyte cultures typically requires drug exposures significantly higher than clinical plasma Cmax concentrations, making clinical interpretations of mechanistic pathway changes challenging. We previously described a system that uses liver-derived hemodynamic blood flow and transport parameters to restore primary human hepatocyte biology, and drug responses at concentrations relevant to in vivo or clinical exposure levels. Using this system, primary hepatocytes from 5 human donors were exposed to concentrations approximating clinical therapeutic and supra-therapeutic levels of Nevirapine (11.3 and 175.0 µM) and Ritonavir (3.5 and 62.4 µM) for 48 h. Whole genome transcriptomics was performed by RNAseq along with functional assays for metabolic activity and function. We observed effects at both doses, but a greater number of genes were differentially expressed with higher probability at the toxic concentrations. At the toxic doses, both drugs showed direct cholestatic potential with Nevirapine increasing bile synthesis and Ritonavir inhibiting bile acid transport. Clear differences in antigen presentation were noted, with marked activation of MHC Class I by Nevirapine and suppression by Ritonavir. This suggests CD8+ T cell involvement for Nevirapine and possibly NK Killer cells for Ritonavir. Both compounds induced several drug metabolizing genes (including CYP2B6, CYP3A4 and UGT1A1), mediated by CAR activation in Nevirapine and PXR in Ritonavir. Unlike Ritonavir, Nevirapine did not increase fatty acid synthesis or activate the respiratory electron chain with simultaneous mitochondrial uncoupling supporting clinical reports of a lower propensity for steatosis. This in vitro study offers insights into the disparate direct and immune-mediated toxicity mechanisms underlying Nevirapine and Ritonavir toxicity in the clinic.


Assuntos
Fármacos Anti-HIV/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/genética , Hepatócitos/efeitos dos fármacos , Nevirapina/toxicidade , Ritonavir/toxicidade , Transcriptoma , Técnicas de Cultura de Células/métodos , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Avaliação Pré-Clínica de Medicamentos/métodos , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia
15.
Photochem Photobiol ; 90(1): 57-65, 2014 01.
Artigo em Inglês | MEDLINE | ID: mdl-24102233

RESUMO

Weak photon absorption and fast carrier kinetics in graphene restrict its applications in photosensitive reactions. Such restrictions/limitations can be overcome by covalent coupling of another photosensitive nanostructure to graphene, forming graphene-semiconductor nanocomposites. Herein, we report one-pot synthesis of RGO-Ag3 VO4 nanocomposites using various sacrificial agents like ethanol, methanol, propanol and ethylene glycol (EG) under visible light illumination. The Raman spectral analysis and (13) C MAS NMR suggest ethanol to be the best sacrificial agent among those studied. Thermal analysis studies, further, confirm the stability of the synthesized nanocomposite with ethanol as sacrificial agent. In view of this, the activity toward dye degradation was focused over the composites prepared via ethanol as sacrificial agent. It was observed and proved that cationic dyes could be degraded quantitatively and swiftly compared to anionic dyes (37.79%) in 1.5 h. This suggests that the surface of the nanocomposites is anionic as partial reduction takes place during synthesis process. In case of mixed dye degradation process, it was noticed that the presence of cationic dye doubles the degradation of anionic dye. The activity of these synthesized nanocomposites is more than five-fold toward the phototransformation of phenol and photodegradation of textile dyes under visible light illumination.

16.
Expert Opin Drug Metab Toxicol ; 8(8): 999-1014, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22632603

RESUMO

INTRODUCTION: The frequent failure of high-throughput screening cell-based tools to accurately predict in vivo responses, coupled with limitations of animal models in predicting human safety or drug efficacy, impairs the de-risking process for biotechnology/pharmaceutical companies as they make important decisions to enter human clinical trials. Organotypic systems strive to fill the gap between these screening and in vivo studies and provide a solution. AREAS COVERED: The authors examine the various approaches to recreate physiological response on the bench and trace the evolution of organotypic systems, while discussing intrinsic challenges and opportunities that lie ahead. Furthermore, they cite literature that is the foundation of several biotechnology research companies addressing this issue and discuss major government-funded initiatives to aid the development of these systems in an effort to fill this existing gap. EXPERT OPINION: Decisions from translational systems that bridge basic drug efficacy and toxicity with clinical outcome must be benchmarked against human-relevant endpoints and clinical data for early meaningful pre-clinical decisions. The use of human primary cells coupled with emerging technologies that allow precise control of the culture environment and analysis of meaningful endpoints paves the way for human organotypic systems as a major initiative in de-risking the drug discovery and development process.


Assuntos
Técnicas de Cultura de Células , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Inativação Metabólica , Preparações Farmacêuticas/metabolismo , Linhagem Celular , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Hemodinâmica , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos
17.
Chem Res Toxicol ; 23(3): 557-67, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20131800

RESUMO

The hydroperoxide of linoleic acid (13-HPODE) degrades to 9,12-dioxo-10(E)-dodecenoic acid (DODE), which readily modifies proteins. This study identified the major proteins in MCF7 cells modified by DODE. To reduce false positives, three methods were used to identify DODE-modified proteins. First, cells were treated with a synthetically biotinylated 13-HPODE (13-HPODE-biotin). Modified proteins were enriched by neutravidin affinity and identified by two-dimensional liquid chromatography--tandem mass spectrometry (2D LC-MS/MS). Second, cells were treated with native 13-HPODE. Protein carbonyls were biotinylated with an aldehyde reactive probe, and modified proteins were enriched by neutravidin affinity and identified by 2D LC-MS/MS. Third, using a newly developed DODE antibody, DODE-modified proteins were located by 2D sodium dodecyl sulfate--polyacrylamide gel electrophoresis and Western blot and identified by in-gel digestion and LC-MS/MS. Analysis of the proteins characterized by all three methods revealed a significant overlap and identified 32 primary proteins modified by DODE in MCF7 cells. These results demonstrated the feasibility for the cellular formation of DODE protein-carbonyl adducts that may be future indicators of oxidative stress.


Assuntos
Ácidos Graxos Monoinsaturados/metabolismo , Peroxidação de Lipídeos , Proteínas/metabolismo , Linhagem Celular Tumoral , Cromatografia Líquida , Citocromos c/metabolismo , Humanos , Ácidos Linoleicos/metabolismo , Peróxidos Lipídicos/metabolismo , Proteínas/análise , Espectrometria de Massas em Tandem
18.
Lab Chip ; 10(1): 51-8, 2010 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-20024050

RESUMO

In vitro models that capture the complexity of in vivo tissue and organ behaviors in a scalable and easy-to-use format are desirable for drug discovery. To address this, we have developed a bioreactor that fosters maintenance of 3D tissue cultures under constant perfusion and we have integrated multiple bioreactors into an array in a multiwell plate format. All bioreactors are fluidically isolated from each other. Each bioreactor in the array contains a scaffold that supports formation of hundreds of 3D microscale tissue units. The tissue units are perfused with cell culture medium circulated within the bioreactor by integrated pneumatic diaphragm micropumps. Electronic controls for the pumps are kept outside the incubator and connected to the perfused multiwell by pneumatic lines. The docking design and open-well bioreactor layout make handling perfused multiwell plates similar to using standard multiwell tissue culture plates. A model of oxygen consumption and transport in the circulating culture medium was used to predict appropriate operating parameters for primary liver cultures. Oxygen concentrations at key locations in the system were then measured as a function of flow rate and time after initiation of culture to determine oxygen consumption rates. After seven days of culture, tissue formed from cells seeded in the perfused multiwell reactor remained functionally viable as assessed by immunostaining for hepatocyte and liver sinusoidal endothelial cell (LSEC) phenotypic markers.


Assuntos
Descoberta de Drogas , Dispositivos Lab-On-A-Chip , Fígado/citologia , Técnicas Analíticas Microfluídicas/métodos , Engenharia Tecidual/métodos , Animais , Reatores Biológicos , Sobrevivência Celular , Técnicas de Cocultura , Desenho de Equipamento , Proteínas de Fluorescência Verde , Fígado/metabolismo , Técnicas Analíticas Microfluídicas/instrumentação , Modelos Biológicos , Técnicas de Cultura de Órgãos , Consumo de Oxigênio/fisiologia , Perfusão , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Engenharia Tecidual/instrumentação
19.
Expert Opin Drug Metab Toxicol ; 5(10): 1159-74, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19637986

RESUMO

Assessment of drug-liver interactions is an integral part of predicting the safety profile of new drugs. Existing model systems range from in vitro cell culture models to FDA-mandated animal tests. Data from these models often fail, however, to predict human liver toxicity, resulting in costly failures of clinical trials. In vitro screens based on cultured hepatocytes are now commonly used in early stages of development, but many toxic responses in vivo seem to be mediated by a complex interplay among several different cell types. We discuss some of the evolving trends in liver cell culture systems applied to drug safety assessment and describe an experimental model that captures complex liver physiology through incorporation of heterotypic cell-cell interactions, 3D architecture and perfused flow. We demonstrate how heterotypic interactions in this system can be manipulated to recreate an inflammatory environment and apply the model to test compounds that potentially exhibit idiosyncratic drug toxicity. Finally, we provide a perspective on how the range of existing and emerging in vitro liver culture approaches, from simple to complex, might serve needs across the range of stages in drug discovery and development, including applications in molecular therapeutics.


Assuntos
Fígado/metabolismo , Engenharia Tecidual/métodos , Testes de Toxicidade/métodos , Animais , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas , Desenho de Fármacos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Humanos , Fígado/efeitos dos fármacos
20.
Adv Cancer Res ; 97: 225-46, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17419948

RESUMO

Metastatic seeding leads to most of the morbidity from carcinomas. However, little is known of this key event as current methods to study the cellular behaviors utilize nonrepresentative in vitro models or follow indirect subsequent developments in vivo. Therefore, we developed a system to visualize over a multiday to multiweek period the interactions between tumor cells and target organ parenchyma. We employ an ex vivo microscale perfusion culture system that provides a tissue-relevant environment to assess metastatic seeding behavior. The bioreactor recreates many features of the fluid flow, scale, and biological functionality of a hepatic parenchyma, a common site of metastatic spread for a wide range of carcinomas. As a test of this model, prostate and breast carcinoma cells were introduced. Tumor cell invasion and expansion could be observed by two-photon microscopy of red fluorescent protein (RFP)- and CellTracker-labeled carcinoma cells against a green fluorescent protein (GFP)-labeled hepatic tissue bed over a 14-day period. Tumors visible to the naked eye could be formed by day 25, without evident necrosis in the >0.3-mm tumor mass. These tumor cells failed to grow in the absence of the supporting three-dimensional (3D) hepatic microtissue, suggesting paracrine or stromal support function for the liver structure in tumor progression. Initial ultrastructural studies suggest that early during the tumor-parenchyma interactions, there are extensive interactions between and accommodations of the cancer and host cells, suggesting that the tumor-related epithelial-mesenchymal transition (EMT) reverts, at least transiently, to promote metastatic seeding. In sum, our 3D ex vivo organotypic liver tissue system presents a critical vehicle to examine tumor-host interactions during cancer metastasis and/or invasion. It also circumvents current limitations in assays to assess early events in metastasis, and provides new approaches to study molecular events during tumor progression.


Assuntos
Reatores Biológicos , Neoplasias Hepáticas/secundário , Metástase Neoplásica/patologia , Técnicas de Cultura de Órgãos/métodos , Animais , Neoplasias da Mama/patologia , Carcinoma/secundário , Linhagem Celular Tumoral/patologia , Técnicas de Cocultura , Células Endoteliais/citologia , Desenho de Equipamento , Feminino , Proteínas de Fluorescência Verde , Hepatócitos/citologia , Humanos , Circulação Hepática , Masculino , Microscopia de Fluorescência , Invasividade Neoplásica , Metástase Neoplásica/fisiopatologia , Técnicas de Cultura de Órgãos/instrumentação , Perfusão/métodos , Neoplasias da Próstata/patologia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Células Estromais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...