Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 360(1): 226-238, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27821712

RESUMEN

Bruton's tyrosine kinase (BTK) is a member of the Tec family of cytoplasmic tyrosine kinases involved in B-cell and myeloid cell signaling. Small molecule inhibitors of BTK are being investigated for treatment of several hematologic cancers and autoimmune diseases. GDC-0853 ((S)-2-(3'-(hydroxymethyl)-1-methyl-5-((5-(2-methyl-4-(oxetan-3-yl)piperazin-1-yl)pyridin-2-yl)amino)-6-oxo-1,6-dihydro-[3,4'-bipyridin]-2'-yl)-7,7-dimethyl-3,4,7,8-tetrahydro-2H-cyclopenta[4,5]pyrrolo[1,2-a]pyrazin-1(6H)-one) is a selective and reversible oral small-molecule BTK inhibitor in development for the treatment of rheumatoid arthritis and systemic lupus erythematosus. In Sprague-Dawley (SD) rats, administration of GDC-0853 and other structurally diverse BTK inhibitors for 7 days or longer caused pancreatic lesions consisting of multifocal islet-centered hemorrhage, inflammation, fibrosis, and pigment-laden macrophages with adjacent lobular exocrine acinar cell atrophy, degeneration, and inflammation. Similar findings were not observed in mice or dogs at much higher exposures. Hemorrhage in the peri-islet vasculature emerged between four and seven daily doses of GDC-0853 and was histologically similar to spontaneously occurring changes in aging SD rats. This suggests that GDC-0853 could exacerbate a background finding in younger animals. Glucose homeostasis was dysregulated following a glucose challenge; however, this occurred only after 28 days of administration and was not directly associated with onset or severity of pancreatic lesions. There were no changes in other common serum biomarkers assessing endocrine and exocrine pancreatic function. Additionally, these lesions were not readily detectable via Doppler ultrasound, computed tomography, or magnetic resonance imaging. Our results indicate that pancreatic lesions in rats are likely a class effect of BTK inhibitors, which may exacerbate an islet-centered pathology that is unlikely to be relevant to humans.


Asunto(s)
Páncreas/efectos de los fármacos , Piperazinas/toxicidad , Inhibidores de Proteínas Quinasas/toxicidad , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Piridonas/toxicidad , Pirroles/toxicidad , Agammaglobulinemia Tirosina Quinasa , Animales , Perros , Relación Dosis-Respuesta a Droga , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Humanos , Masculino , Ratones , Páncreas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Ratas , Especificidad de la Especie
2.
Toxicol Appl Pharmacol ; 334: 100-109, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28893587

RESUMEN

The contribution of animal testing in drug development has been widely debated and challenged. An industry-wide nonclinical to clinical translational database was created to determine how safety assessments in animal models translate to First-In-Human clinical risk. The blinded database was composed of 182 molecules and contained animal toxicology data coupled with clinical observations from phase I human studies. Animal and clinical data were categorized by organ system and correlations determined. The 2×2 contingency table (true positive, false positive, true negative, false negative) was used for statistical analysis. Sensitivity was 48% with a 43% positive predictive value (PPV). The nonhuman primate had the strongest performance in predicting adverse effects, especially for gastrointestinal and nervous system categories. When the same target organ was identified in both the rodent and nonrodent, the PPV increased. Specificity was 84% with an 86% negative predictive value (NPV). The beagle dog had the strongest performance in predicting an absence of clinical adverse effects. If no target organ toxicity was observed in either test species, the NPV increased. While nonclinical studies can demonstrate great value in the PPV for certain species and organ categories, the NPV was the stronger predictive performance measure across test species and target organs indicating that an absence of toxicity in animal studies strongly predicts a similar outcome in the clinic. These results support the current regulatory paradigm of animal testing in supporting safe entry to clinical trials and provide context for emerging alternate models.


Asunto(s)
Bases de Datos Factuales , Investigación Biomédica Traslacional , Animales , Evaluación Preclínica de Medicamentos , Industria Farmacéutica , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Modelos Animales , Medición de Riesgo
3.
Chem Res Toxicol ; 29(4): 452-72, 2016 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-26625186

RESUMEN

Discovery toxicology focuses on the identification of the most promising drug candidates through the development and implementation of lead optimization strategies and hypothesis-driven investigation of issues that enable rational and informed decision-making. The major goals are to [a] identify and progress the drug candidate with the best overall drug safety profile for a therapeutic area, [b] remove the most toxic drugs from the portfolio prior to entry into humans to reduce clinical attrition due to toxicity, and [c] establish a well-characterized hazard and translational risk profile to enable clinical trial designs. This is accomplished through a framework that balances the multiple considerations to identify a drug candidate with the overall best drug characteristics and provides a cogent understanding of mechanisms of toxicity. The framework components include establishing a target candidate profile for each program that defines the qualities of a successful candidate based on the intended therapeutic area, including the risk tolerance for liabilities; evaluating potential liabilities that may result from engaging the therapeutic target (pharmacology-mediated or on-target) and that are chemical structure-mediated (off-target); and characterizing identified liabilities. Lead optimization and investigation relies upon the integrated use of a variety of technologies and models (in silico, in vitro, and in vivo) that have achieved a sufficient level of qualification or validation to provide confidence in their use. We describe the strategic applications of various nonclinical models (established and new) for a holistic and integrated risk assessment that is used for rational decision-making. While this review focuses on strategies for small molecules, the overall concepts, approaches, and technologies are generally applicable to biotherapeutics.


Asunto(s)
Descubrimiento de Drogas/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Animales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Humanos , Preparaciones Farmacéuticas/química , Farmacología/métodos , Medición de Riesgo/métodos , Pruebas de Toxicidad/métodos
4.
Toxicol Appl Pharmacol ; 266(1): 86-94, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23142475

RESUMEN

Several toxicities are clearly driven by free drug concentrations in plasma, such as toxicities related to on-target exaggerated pharmacology or off-target pharmacological activity associated with receptors, enzymes or ion channels. However, there are examples in which organ toxicities appear to correlate better with total drug concentrations in the target tissues, rather than with free drug concentrations in plasma. Here we present a case study in which a small molecule Met inhibitor, GEN-203, with significant liver and bone marrow toxicity in preclinical species was modified with the intention of increasing the safety margin. GEN-203 is a lipophilic weak base as demonstrated by its physicochemical and structural properties: high LogD (distribution coefficient) (4.3) and high measured pKa (7.45) due to the basic amine (N-ethyl-3-fluoro-4-aminopiperidine). The physicochemical properties of GEN-203 were hypothesized to drive the high distribution of this compound to tissues as evidenced by a moderately-high volume of distribution (Vd>3l/kg) in mouse and subsequent toxicities of the compound. Specifically, the basicity of GEN-203 was decreased through addition of a second fluorine in the 3-position of the aminopiperidine to yield GEN-890 (N-ethyl-3,3-difluoro-4-aminopiperidine), which decreased the volume of distribution of the compound in mouse (Vd=1.0l/kg), decreased its tissue drug concentrations and led to decreased toxicity in mice. This strategy suggests that when toxicity is driven by tissue drug concentrations, optimization of the physicochemical parameters that drive tissue distribution can result in decreased drug concentrations in tissues, resulting in lower toxicity and improved safety margins.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/toxicidad , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Femenino , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Masculino , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas c-met/metabolismo , Distribución Aleatoria , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
6.
Vet Immunol Immunopathol ; 126(3-4): 377-81, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18771806

RESUMEN

CD137 plays an important role as a co-stimulatory molecule in activated T cells. Agonistic CD137 specific antibodies have been investigated as therapeutic agents to promote tumor-specific immune responses by direct activation of T cells. As part of the pre-clinical pharmacological evaluation of cynomolgus monkeys, monkey CD137 was cloned and characterized. The deduced amino acid sequence encoded a full-length gene of 254 amino acids 95% identical to human CD137. Sequence variants identified in monkey CD137 include four splice variants lacking the transmembrane domain. These variants were detectable in human including two previously unreported variants. Two missense single nucleotide polymorphisms were detected present in 42 and 50% of 36 monkeys tested. In both monkey and human, mRNA expression of full-length CD137 and splice variants were significantly increased in peripheral blood mononuclear cells (PBMCs) upon stimulation by anti-CD3 antibodies. Recombinant monkey CD137 protein was bound with high affinity by an agonistic anti-human CD137 antibody but not by an anti-mouse CD137 antibody. In summary, compared to human, monkey CD137 showed distinct extracellular domain amino acid sequence and sequence polymorphisms. Thus, antibodies directed against epitopes in this extracellular domain could have differences in pharmacologic activity between cynomolgus monkeys and human or across individual cynomolgus monkeys.


Asunto(s)
Macaca fascicularis/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos/metabolismo , Secuencia de Bases , Clonación Molecular , Cartilla de ADN/genética , Humanos , Datos de Secuencia Molecular , Mutación Missense/genética , Polimorfismo de Nucleótido Simple , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Alineación de Secuencia , Análisis de Secuencia de ADN/veterinaria
7.
Clin Cancer Res ; 22(11): 2618-22, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27250932

RESUMEN

Multidisciplinary approaches that incorporate nonclinical pharmacologic and toxicologic characterization of small-molecule oncology drugs into clinical development programs may facilitate improved benefit-risk profiles and clinical toxicity management in patients. The performance of the current nonclinical safety-testing scheme was discussed, highlighting current strengths and areas for improvement. While current nonclinical testing appears to predict the clinical outcome where the prevalence of specific adverse effects are high, nonclinical testing becomes less reliable for predicting clinical adverse effects that occur infrequently, as with some kinase inhibitors. Although adverse effects associated with kinase inhibitors can often be predicted on the basis of target biology, drugs can be promiscuous and inhibit targets with poorly defined function and associated risks. Improvements in adverse effect databases and better characterization of the biologic activities of drug targets may enable better use of computational modeling approaches in predicting adverse effects with kinase inhibitors. Assessing safety of a lead candidate in parallel with other drug properties enables incorporation of a molecule's best features during chemical design, eliminates the worst molecules early, and permits timely investigation/characterization of toxicity mechanisms for identified liabilities. A safety lead optimization and candidate identification strategy that reduces intrinsic toxicity and metabolic risk and enhances selectivity can deliver selective kinase inhibitors that demonstrate on-target adverse effects identified nonclinically. Integrating clinical and nonclinical data during drug development can facilitate better identification and management of oncology drugs. Follow-up nonclinical studies may be used to better understand the risks in a given patient population and minimize or manage these risks more appropriately. Clin Cancer Res; 22(11); 2618-22. ©2016 AACR SEE ALL ARTICLES IN THIS CCR FOCUS SECTION, "NEW APPROACHES FOR OPTIMIZING DOSING OF ANTICANCER AGENTS".


Asunto(s)
Antineoplásicos/toxicidad , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/toxicidad , Animales , Antineoplásicos/uso terapéutico , Ensayos Clínicos como Asunto , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Inhibidores de Proteínas Quinasas/uso terapéutico
8.
Biomed Res Int ; 2016: 9737920, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27689095

RESUMEN

Drug-induced liver injury (DILI) is a major cause of late-stage clinical drug attrition, market withdrawal, black-box warnings, and acute liver failure. Consequently, it has been an area of focus for toxicologists and clinicians for several decades. In spite of considerable efforts, limited improvements in DILI prediction have been made and efforts to improve existing preclinical models or develop new test systems remain a high priority. While prediction of intrinsic DILI has improved, identifying compounds with a risk for idiosyncratic DILI (iDILI) remains extremely challenging because of the lack of a clear mechanistic understanding and the multifactorial pathogenesis of idiosyncratic drug reactions. Well-defined clinical diagnostic criteria and risk factors are also missing. This paper summarizes key data interpretation challenges, practical considerations, model limitations, and the need for an integrated risk assessment. As demonstrated through selected initiatives to address other types of toxicities, opportunities exist however for improvement, especially through better concerted efforts at harmonization of current, emerging and novel in vitro systems or through the establishment of strategies for implementation of preclinical DILI models across the pharmaceutical industry. Perspectives on the incorporation of newer technologies and the value of precompetitive consortia to identify useful practices are also discussed.

9.
Toxicol Sci ; 151(2): 245-60, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26917699

RESUMEN

PRO304186, a humanized monoclonal antibody targeting soluble interleukin-17 A and F, was developed for autoimmune and inflammatory disease indications. When administered to cynomolgus monkeys PRO304186 induced unexpected adverse effects characterized by clinical signs of hematemesis, hematochezia, and moribundity. Pathology findings included hemorrhage throughout the gastrointestinal tract without any evidence of vascular wall damage or inflammatory cellular infiltration. Mechanistic investigation of these effects revealed mild elevations of serum MCP-1 and IL-12/23 but without a classical proinflammatory profile in PRO304186-treated animals. In vitro studies demonstrated off-target effects on vascular endothelial cells including activation of nitric oxide synthase leading to production of nitric oxide (NO) accompanied by increased mitochondrial membrane depolarization, glutathione depletion, and increased paracellular permeability. Additionally, endothelial cell-PRO304186-conditioned medium reduced myosin light chain phosphorylation in vascular smooth muscle cells. Furthermore, an ex vivo study utilizing segments from cynomolgus aorta and femoral artery confirmed PRO304186-induced endothelium-dependent smooth muscle relaxation and vasodilation mediated via NO. Finally, a single dose of PRO304186 in cynomolgus monkeys induced a rapid and pronounced increase in NO in the portal circulation that preceded a milder elevation of NO in the systemic circulation and corresponded temporally with systemic hypotension; findings consistent with NO-mediated vasodilation leading to hypotension. These changes were associated with non-inflammatory, localized hemorrhage in the gastrointestinal tract consistent with hemodynamic vascular injury associated with intense local vasodilation. Together, these data demonstrate that PRO304186-associated toxicity in monkeys was due to an off-target effect on endothelium that involved regional NO release resulting in severe systemic vasodilation, hypotension, and hemorrhage.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Arterias/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Hemorragia Gastrointestinal/inducido químicamente , Hipotensión/inducido químicamente , Óxido Nítrico/metabolismo , Vasodilatación/efectos de los fármacos , Animales , Anticuerpos Monoclonales Humanizados/metabolismo , Arterias/metabolismo , Arterias/fisiopatología , Células Cultivadas , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Femenino , Hemorragia Gastrointestinal/metabolismo , Hemorragia Gastrointestinal/fisiopatología , Hematemesis/inducido químicamente , Hematemesis/metabolismo , Hematemesis/fisiopatología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hipotensión/metabolismo , Hipotensión/fisiopatología , Interleucina-17/antagonistas & inhibidores , Interleucina-17/inmunología , Interleucina-17/metabolismo , Macaca fascicularis , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Factores de Tiempo
10.
Curr Top Med Chem ; 5(10): 929-39, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16178738

RESUMEN

Inhibitors of p38 MAP kinases show promise for the treatment of inflammatory and immunological disorders and some cancers. There is a substantial body of experimental evidence across several organ systems suggesting that p38 also mediates developmental, differentiation and proliferation processes. As a consequence of the wide-ranging regulatory role of p38 kinase in diverse cellular processes, the possibility of adverse events resulting from undesired pharmacological activity is a major concern for the p38 inhibitor drug class. Taking into consideration the limitations of experimental modeling systems, together the data may indicate that profound inhibition of p38 has the potential to impact these processes during fetal or neonatal development. The difficulty comes in extrapolating these findings to predict potential adverse effects under conditions of partial inhibition of p38 activity, and in an adult population in which these processes are typically only recapitulated during repair or adaptive responses. As such, the goal of this review of the targets of p38 activity is to bring an awareness of the those organ systems that should be monitored for potential toxicity, as well as to present a potential mechanistic basis for such monitoring or for investigation of adverse effects that may develop with administration of a p38 inhibitor.


Asunto(s)
Inhibidores Enzimáticos/efectos adversos , Proteína Quinasa 11 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Animales , Coagulación Sanguínea/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Corazón/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Humanos , Sistema Inmunológico/efectos de los fármacos , Hígado/efectos de los fármacos , Ratones , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Sistema Nervioso/efectos de los fármacos , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Reproducción/efectos de los fármacos , Distribución Tisular/efectos de los fármacos
11.
Toxicol In Vitro ; 18(4): 533-41, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15130611

RESUMEN

Drug-induced idiosyncratic hepatotoxicity continues to be an important safety issue for the pharmaceutical industry. This toxicity is due, in part, to the limited predictive nature of current pre-clinical study systems. A hypothesis was formed that treatment of existing in vitro hepatocyte cultures with drugs clinically linked to idiosyncratic hepatotoxicity would result in the release of extracellular protein biomarkers indicative of liver toxicity. To test this hypothesis, a combination of proteomic and immunological techniques were used to first identify, and subsequently verify, components of the protein-laden conditioned culture media from immortalized human hepatocytes which overexpressed cytochrome p450 3A4. These cells were treated separately with seven individual compounds made up of a combination of thiazolidinedione and l-tyrosine PPARgamma agonists and HIV protease inhibitors, plus a vehicle control (dimethyl sulfoxide). For each drug class, clinically determined hepatotoxic and non-hepatotoxic compounds were compared. Two proteins, BMS-PTX-265 and BMS-PTX-837, were reproducibly and significantly increased in the conditioned media from cells treated with each of the toxic compounds as compared to media from cells treated with the non-toxic compounds (and vehicle). This result supported the hypothesis, and so a series of successive assays (western blots and enzyme linked immunosorbent assays) were used to measure the response of these two proteins as a function of an expanded set of 20 compounds. For all 20 drugs, elevations of BMS-PTX-265 correlated exactly with the known safety profile; whereas changes in BMS-PTX-837 correctly predicted the safety profile in 19 of 20 drugs (one false negative). In summary, the data supports both the pre-clinical in vitro method as a means to identify new biomarkers of liver toxicity, as well as the validity of the biomarkers themselves.


Asunto(s)
Biomarcadores/análisis , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Hepatocitos/fisiología , Hígado/efectos de los fármacos , Proteínas/análisis , Western Blotting , Cromatografía Líquida de Alta Presión , Medios de Cultivo/química , Evaluación Preclínica de Medicamentos/métodos , Ensayo de Inmunoadsorción Enzimática , Reacciones Falso Negativas , Predicción , Humanos , Hígado/patología , Proteómica
12.
J Vet Intern Med ; 16(1): 52-62, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11822805

RESUMEN

Dilated cardiomyopathy recently has been recognized in juvenile Portuguese Water Dogs. The purpose of this study was to evaluate unaffected and affected puppies by physical examination, electrocardiogram (ECG), echocardiogram, specific biochemical assays, and ultrastructure to document disease progression and to develop a method of early detection. Results of segregation analysis were consistent with autosomal recessive inheritance. Of 124 puppies evaluated clinically and echocardiographically, 10 were affected. No significant differences were found between unaffected and affected puppies for blood and myocardial carnitine or taurine concentrations, serum chemical variables, results of ophthalmological examinations, ECGs, or measurement of urine metabolites. Ultrastructural examination of myocardium from affected dogs revealed myofibrillar atrophy and small regions of myofibrillar degeneration, most prominently at the region of the intercalated discs. Only echocardiography allowed detection of affected puppies before clinical signs became evident. Echocardiography revealed a significant difference in the shortening fraction, E point to septal separation, and the end systolic and diastolic left ventricular internal diameters. Affected puppies were detected 1-4 weeks before the development of acute congestive heart failure.


Asunto(s)
Cardiomiopatía Dilatada/veterinaria , Enfermedades de los Perros/diagnóstico , Animales , Animales Recién Nacidos , Cruzamiento , Cardiomiopatía Dilatada/diagnóstico , Carnitina/análisis , Estudios de Casos y Controles , Progresión de la Enfermedad , Enfermedades de los Perros/diagnóstico por imagen , Enfermedades de los Perros/patología , Perros , Ecocardiografía/veterinaria , Electrocardiografía/veterinaria , Miocardio/ultraestructura , Examen Físico/veterinaria , Estudios Retrospectivos , Taurina/análisis
13.
J Pharm Sci ; 102(10): 3816-29, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23878104

RESUMEN

Having an understanding of drug tissue accumulation can be informative in the assessment of target organ toxicities; however, obtaining tissue drug levels from toxicology studies by bioanalytical methods is labor-intensive and infrequently performed. Additionally, there are no described methods for predicting tissue drug distribution for the experimental conditions in toxicology studies, which typically include non-steady-state conditions and very high exposures that may saturate several processes. The aim was the development of an algorithm to provide semiquantitative and quantitative estimates of tissue-to-plasma concentration ratios (Kp ) for several tissues from readily available parameters of pharmacokinetics (PK) such as volume of distribution (Vd ) and clearance of each drug, without performing tissue measurement in vivo. The computational approach is specific for the oral route of administration and non-steady-state conditions and was applied for a dataset of 29 Genentech small molecules such as neutral compounds as well as weak and strong organic bases. The maximum success rate in predicting Kp values within 2.5-fold error of observed Kp values was 82% at low doses (<100 mg/kg) in preclinical species. Prediction accuracy was relatively lower with saturation at high doses (≥100 mg/kg); however, an approach to perform low-to-high dose extrapolations of Kp values was presented and applied successfully in most cases. An approach for the interspecies scaling was also applied successfully. Finally, the proposed algorithm was used in a case study and successfully predicted differential tissue distribution of two small-molecule MET kinase inhibitors, which had different toxicity profiles in mice. This newly developed algorithm can be used to predict the partition coefficients Kp for small molecules in toxicology studies, which can be leveraged to optimize the PK drivers of tissue distribution in an attempt to decrease drug tissue level, and improve safety margins.


Asunto(s)
Preparaciones Farmacéuticas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacocinética , Algoritmos , Animales , Perros , Ratones , Modelos Biológicos , Ratas , Distribución Tisular
14.
Sci Transl Med ; 4(159): 159ps22, 2012 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-23136041

RESUMEN

Widespread sharing and analysis of clinical trial data and a U.S. government initiative to engineer nonclinical cell-based models that mimic human biological processes have the potential to improve predictions of drug-related adverse events.


Asunto(s)
Medición de Riesgo/normas , Ensayos Clínicos como Asunto , Conducta Cooperativa , Humanos , Dispositivos Laboratorio en un Chip/normas , Modelos Biológicos
15.
Toxicol Sci ; 125(1): 187-95, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21976371

RESUMEN

MEK, a kinase downstream of Ras and Raf oncogenes, constitutes a high priority target in oncology research. MEK small molecule inhibitors cause soft tissue mineralization in rats secondary to serum inorganic phosphorus (iP) elevation, but the molecular mechanism for this toxicity remains undetermined. We performed investigative studies with structurally distinct MEK inhibitors GEN-A and PD325901 (PD-901) in Sprague-Dawley rats. Our data support a mechanism that involves FGF-23 signal blockade in the rat kidney, causing transcriptional upregulation of 25-hydroxyvitamin D(3) 1-alpha-hydroxylase (Cyp27b1), the rate-limiting enzyme in vitamin D activation, and downregulation of 1,25-dihydroxyvitamin D(3) 24-hydroxylase (Cyp24a1), the enzyme that initiates the degradation of the active form of vitamin D. These transcriptional changes increase serum vitamin D levels, which in turn drive the increase in serum iP, leading to soft tissue mineralization in the rat.


Asunto(s)
Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Riñón/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Fósforo/sangre , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Animales , Calcio/sangre , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Homeostasis/efectos de los fármacos , Riñón/enzimología , Riñón/metabolismo , Masculino , Estructura Molecular , Hormona Paratiroidea/sangre , Inhibidores de Proteínas Quinasas/química , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Espectrometría de Masas en Tándem , Vitamina D/sangre
16.
Toxicol Pathol ; 35(5): 621-35, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17654404

RESUMEN

Toxicogenomics is considered a valuable tool for reducing pharmaceutical candidate attrition by facilitating earlier identification, prediction and understanding of toxicities. A retrospective evaluation of 3 years of routine transcriptional profiling in non-clinical safety studies was undertaken to assess the utility of toxicogenomics in drug safety assessment. Based on the analysis of studies with 33 compounds, marked global transcriptional changes (> 4% transcripts at p < 0.01) were shown to be a robust biomarker for dosages considered to be toxic . In general, there was an inconsistent correlation between transcription and histopathology, most likely due to differences in sensitivity to focal microscopic lesions, to secondary effects, and to events that precede structural tissue changes. For 60% of toxicities investigated with multiple time-point data, transcriptional changes were observed prior to changes in traditional study endpoints. Candidate transcriptional markers of pharmacologic effects were detected in 40% of targets profiled. Mechanistic classification of toxicity was obtained for 30% of targets. Furthermore, data comparison to compendia of transcriptional changes provided assessments of the specificity of transcriptional responses. Overall, our experience suggests that toxicogenomics has contributed to a greater understanding of mechanisms of toxicity and to reducing drug attrition by empiric analysis where safety assessment combines toxicogenomic and traditional evaluations.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Toxicogenética , Animales , Biomarcadores , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Humanos , Estudios Retrospectivos , Medición de Riesgo , Seguridad
17.
Toxicol Appl Pharmacol ; 211(2): 157-65, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16081117

RESUMEN

Oxidative stress plays an important role in acetaminophen (APAP)-induced hepatotoxicity. In addition to inducing direct cellular damage, oxidants can activate transcription factors including NF-kappaB, which regulate the production of inflammatory mediators implicated in hepatotoxicity. Here, we investigated the role of APAP-induced oxidative stress and NF-kappaB in inflammatory mediator production. Treatment of mice with APAP (300 mg/kg, i.p.) resulted in centrilobular hepatic necrosis and increased serum aminotransferase levels. This was correlated with depletion of hepatic glutathione and CuZn superoxide dismutase (SOD). APAP administration also increased expression of the proinflammatory mediators, interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNFalpha), macrophage chemotactic protein-1 (MCP-1), and KC/gro, and the anti-inflammatory cytokine, interleukin-10 (IL-10). Pretreatment of mice with the antioxidant, N-acetylcysteine (NAC) prevented APAP-induced depletion of glutathione and CuZnSOD, as well as hepatotoxicity. NAC also abrogated APAP-induced increases in TNFalpha, KC/gro, and IL-10, but augmented expression of the anti-inflammatory cytokines interleukin-4 (IL-4) and transforming growth factor-beta (TGFbeta). No effects were observed on IL-1beta or MCP-1 expression. To determine if NF-kappaB plays a role in regulating mediator production, we used transgenic mice with a targeted disruption of the gene for NF-kappaB p50. As observed with NAC pretreatment, the loss of NF-kappaB p50 was associated with decreased ability of APAP to upregulate TNFalpha, KC/gro, and IL-10 expression and increased expression of IL-4 and TGFbeta. However, in contrast to NAC pretreatment, the loss of p50 had no effect on APAP-induced hepatotoxicity. These data demonstrate that APAP-induced cytokine expression in the liver is influenced by oxidative stress and that this is dependent, in part, on NF-kappaB. However, NF-kappaB p50-dependent responses do not appear to play a major role in the pathogenesis of toxicity in this model.


Asunto(s)
Acetaminofén/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Mediadores de Inflamación/metabolismo , Subunidad p50 de NF-kappa B/fisiología , Acetaminofén/administración & dosificación , Acetilcisteína/farmacología , Alanina Transaminasa/sangre , Analgésicos no Narcóticos/administración & dosificación , Analgésicos no Narcóticos/toxicidad , Animales , Western Blotting , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/genética , Quimiocinas/genética , Quimiocinas/metabolismo , Expresión Génica/efectos de los fármacos , Glutatión/metabolismo , Humanos , Inyecciones Intraperitoneales , Interleucina-1/biosíntesis , Interleucina-1/genética , Interleucina-10/biosíntesis , Interleucina-10/genética , Interleucina-4/genética , Interleucina-4/metabolismo , Hígado/química , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteínas Inflamatorias de Macrófagos/genética , Proteínas Inflamatorias de Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Subunidad p50 de NF-kappa B/genética , Necrosis/inducido químicamente , Necrosis/patología , Estrés Oxidativo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/metabolismo , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
18.
Toxicol Pathol ; 33(1): 17-26, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15805052

RESUMEN

Hepatotoxicity remains a significant cause for drug failures during clinical trials. This is due, in part, to the idiosyncratic nature of toxicity in humans and inherent physiological differences between humans and preclinical species leading to limited correct prediction of adverse responses in humans. To address this issue, robust screening assays are being developed, which have heightened predictive capacity for human hepatotoxicity, and may be utilized throughout the discovery and development phases in conjunction with traditional in vivo methods, for decision making during drug selection and risk assessment. This manuscript describes an example application of in vitro-based strategies using human hepatocyte cultures in lead optimization screening in conjunction with ADME profiling, for evaluation of compound-associated CYP450 induction potential, and the identification of potentially useful biomarkers as predictors of hepatotoxicity for use in vitro, and in preclinical species and humans.


Asunto(s)
Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Hepatocitos/efectos de los fármacos , Animales , Biomarcadores , Células Cultivadas , Sistema Enzimático del Citocromo P-450/biosíntesis , Diseño de Fármacos , Evaluación de Medicamentos , Inducción Enzimática , Hepatocitos/enzimología , Hepatocitos/metabolismo , Humanos
19.
Methods ; 35(3): 291-302, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15722225

RESUMEN

Discovery of novel protein biomarkers is essential for successful drug discovery and development. These novel protein biomarkers may aid accelerated drug efficacy, response, or toxicity decision making based on their enhanced sensitivity and/or specificity. These biomarkers, if necessary, could eventually be converted into novel diagnostic marker assays. Proteomic platforms developed over the past few years have given us the ability to rapidly identify novel protein biomarkers in various biological matrices from cell cultures (lysates, supernatants) to human clinical samples (serum, plasma, and urine). In this article, we delineate an approach to biomarker discovery. This approach is divided into three steps, (i) identification of markers, (ii) prioritization of identified markers, and (iii) preliminary validation (qualification) of prioritized markers. Using drug-induced idiosyncratic hepatotoxicity as a case study, the article elaborates methods and techniques utilized during the three steps of biomarker discovery process. The first step involves identification of markers using multi-dimensional protein identification technology. The second step involves prioritization of a subset of marker candidates based on several criteria such as availability of reagent set for assay development and literature association to disease biology. The last step of biomarker discovery involves development of preliminary assays to confirm the bio-analytical measurements from the first step, as well as qualify the marker(s) in pre-clinical models, to initiate future marker validation and development.


Asunto(s)
Biomarcadores/química , Cromatografía Líquida de Alta Presión/métodos , Análisis por Matrices de Proteínas/métodos , Proteínas/aislamiento & purificación , Proteómica/métodos , Proteínas 14-3-3/química , Alanina Transaminasa/metabolismo , Animales , Aspartato Aminotransferasas/metabolismo , Western Blotting , Células Cultivadas , Biología Computacional , Perros , Ensayo de Inmunoadsorción Enzimática , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Inmunoensayo/métodos , Hígado/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/química , Espectrometría de Masas/métodos , Proteínas/química , Ratas , Sensibilidad y Especificidad , Tecnología Farmacéutica/métodos , Factores de Tiempo
20.
Hepatology ; 35(5): 1093-103, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11981759

RESUMEN

The biological effects of monocyte chemoattractant protein (MCP) 1 are mediated by binding to C-C chemokine receptor (CCR) 2. In the present studies, we used CCR2 knockout (CCR2-/-) mice to examine the role of MCP-1 in acetaminophen-induced macrophage accumulation in the liver, expression of inflammatory cytokines, and hepatotoxicity. We found that hepatic expression of CCR2 and MCP-1 was increased 10-fold and 20-fold, respectively, 12 to 72 hours after administration of acetaminophen to wild-type mice. Expression of these proteins was localized in centrilobular regions of the liver. Whereas MCP-1 was expressed by both hepatocytes and macrophages, CCR2 was identified in inflammatory macrophages. F4/80 is a marker of mature macrophages expressed in large quantities by Kupffer cells. In wild-type mice, a 75% decrease in F4/80-positive macrophages was observed 24 to 48 hours after administration of acetaminophen. In contrast, expression of macrosialin (CD68), a marker of activated macrophages, increased 2-fold 24 to 72 hours after administration of acetaminophen and was associated with inflammatory cells. Although there was a decrease in the overall severity of inflammation and in the number of macrosialin-positive macrophages 72 hours after administration of acetaminophen in CCR2-/- mice, the number of F4/80-positive cells did not change. Loss of CCR2 was also found to alter acetaminophen-induced expression of tumor necrosis factor alpha, monocyte chemoattractant protein 3, and KC/gro. However, the overall outcome of acetaminophen-induced hepatic injury was not affected. In conclusion, these data indicate that MCP-1 and CCR2 contribute to the recruitment of a subset of activated macrophages into the liver during acetaminophen-induced hepatotoxicity that may be important in resolution of tissue injury.


Asunto(s)
Acetaminofén/toxicidad , Analgésicos no Narcóticos/toxicidad , Movimiento Celular/inmunología , Hígado/inmunología , Macrófagos/citología , Receptores de Quimiocina/metabolismo , Animales , Quimiocina CCL2/metabolismo , Expresión Génica/inmunología , Interleucina-1/genética , Hígado/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Receptores CCR2 , Receptores de Quimiocina/genética , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA