Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
ILAR J ; 62(1-2): 7-16, 2021 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-34181728

RESUMEN

Animals have been closely observed by humans for at least 17 000 years to gain critical knowledge for human and later animal survival. Routine scientific observations of animals as human surrogates began in the late 19th century driven by increases in new compounds resulting from synthetic chemistry and requiring characterization for potential therapeutic utility and safety. Statistics collected by the United States Department of Agriculture's Animal and Plant Health Inspection Service and United Kingdom Home Office show that animal usage in biomedical research and teaching activities peaked after the mid-20th century and thereafter fell precipitously until the early 21st century, when annual increases (in the UK) were again observed, this time driven by expansion of genetically modified animal technologies. The statistics also show a dramatic transfer of research burden in the 20th and 21st centuries away from traditional larger and more publicly sensitive species (dogs, cats, non-human primates, etc) towards smaller, less publicly sensitive mice, rats, and fish. These data show that new technology can produce multi-faceted outcomes to reduce and/or to increase annual animal usage and to redistribute species burden in biomedical research. From these data, it is estimated that annual total vertebrate animal usage in biomedical research and teaching in the United States was 15 to 25 million per year during 2001-2018. Finally, whereas identification and incorporation of non-animal alternatives are products of, but not an integral component of, the animal research cycle, they replace further use of animals for specific research and product development purposes and create their own scientific research cycles, but are not necessarily a substitute for animals or humans for discovery, acquisition, and application of new (eg, previously unknown and/or unsuspected) knowledge critical to further advance human and veterinary medicine and global species survival.


Asunto(s)
Experimentación Animal , Investigación Biomédica , Animales , Perros , Ratones , Ratas , Estados Unidos
2.
Regul Toxicol Pharmacol ; 107: 104413, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31229519

RESUMEN

The draft ICH S5(R3) guideline includes an exposure-based endpoint as an option for selecting the high dose in developmental and reproductive toxicity (DART) studies. In 2016, IQ DruSafe conducted an anonymous survey to identify industry practices and experiences related to pharmacokinetic assessments in DART studies in order to facilitate a pragmatic data-driven approach to development of an acceptable multiple of the clinical exposure to be proposed for dose selection in the guideline. Questions in the survey were designed to explore pharmacokinetic differences in pregnant versus non-pregnant animals, and to assess exposure levels attained in the absence of maternal toxicity as well as DART outcomes in animal studies associated with those exposures. Small molecule and therapeutic proteins were analyzed separately. The key findings for small molecules were: a) differences in exposures between pregnant and non-pregnant animals were generally ≤3-fold, b) Cmax or AUC exposures ≥25-fold the clinical exposure were achieved in the absence of maternal toxicity for 31% and 23% of rat and rabbit developmental toxicity studies, respectively, and c) only 3.3% (5/153) and 1.6% (2/128) of the developmental toxicity studies were positive for malformations or embryofetal lethality in rats and rabbits, respectively, that were not observed until exposure margins were ≥25-fold.


Asunto(s)
Evaluación Preclínica de Medicamentos , Teratógenos/farmacocinética , Teratógenos/toxicidad , Pruebas de Toxicidad , Animales , Industria Farmacéutica , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Desarrollo Embrionario/efectos de los fármacos , Femenino , Desarrollo Fetal/efectos de los fármacos , Haplorrinos , Embarazo , Conejos , Ratas , Reproducción/efectos de los fármacos , Encuestas y Cuestionarios
3.
Regul Toxicol Pharmacol ; 105: 62-68, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30981719

RESUMEN

The draft Step 2 ICH S5(R3) guideline includes an exposure-based endpoint as an option for selecting the high-dose in reproductive and developmental toxicity studies. To help determine an appropriate exposure margin for embryofetal developmental toxicity testing, a retrospective analysis was undertaken to determine what threshold would have been sufficient to detect hazards to embryofetal development in rats and rabbits for 18 known and 4 presumed human teratogens. The analysis showed that using a high dose that provided at least a 6-fold exposure margin in the developmental toxicity studies would have been sufficient to detect the teratogenic hazard with relevance for humans for all these therapeutics. With regards to human risk assessment practices for developmental toxicity, the analysis showed that, after excluding lenalidomide and pomalidomide data in rats, all available AUC margins at the NOAEL for the induction of malformations or embryofetal lethality were <4-fold of the exposure at the MRHD for all 22 therapeutics. These data support the proposed general approach of increased level of concern for human risk when exposure margins of the NOAEL to the MRHD are <10-fold, reduced concern when the exposure margins are 10- to 25-fold, and minimal concern when the exposure margin is > 25-fold.


Asunto(s)
Embrión de Mamíferos/efectos de los fármacos , Medición de Riesgo/métodos , Teratógenos/toxicidad , Pruebas de Toxicidad/métodos , Animales , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Nivel sin Efectos Adversos Observados , Embarazo , Conejos , Ratas , Estudios Retrospectivos , Especificidad de la Especie
4.
Regul Toxicol Pharmacol ; 87 Suppl 3: S1-S15, 2017 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-28483710

RESUMEN

The transition from nonclinical to First-in-Human (FIH) testing is one of the most challenging steps in drug development. In response to serious outcomes in a recent Phase 1 trial (sponsored by Bial), IQ Consortium/DruSafe member companies reviewed their nonclinical approach to progress small molecules safely to FIH trials. As a common practice, safety evaluation begins with target selection and continues through iterative in silico and in vitro screening to identify molecules with increased probability of acceptable in vivo safety profiles. High attrition routinely occurs during this phase. In vivo exploratory and pivotal FIH-enabling toxicity studies are then conducted to identify molecules with a favorable benefit-risk profile for humans. The recent serious incident has reemphasized the importance of nonclinical testing plans that are customized to the target, the molecule, and the intended clinical plan. Despite the challenges and inherent risks of transitioning from nonclinical to clinical testing, Phase 1 studies have a remarkably good safety record. Given the rapid scientific evolution of safety evaluation, testing paradigms and regulatory guidance must evolve with emerging science. The authors posit that the practices described herein, together with science-based risk assessment and management, support safe FIH trials while advancing development of important new medicines.


Asunto(s)
Ensayos Clínicos Fase I como Asunto , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/efectos adversos , Humanos , Medición de Riesgo/métodos , Seguridad
5.
Regul Toxicol Pharmacol ; 82: 94-98, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27769827

RESUMEN

Chronic (>3 months) preclinical toxicology studies are conducted to support the safe conduct of clinical trials exceeding 3 months in duration. We have conducted a review of 32 chronic toxicology studies in non-rodents (22 studies in dogs and 10 in non-human primates) and 27 chronic toxicology studies in rats dosed with Merck compounds to determine the frequency at which additional target organ toxicities are observed in chronic toxicology studies as compared to subchronic studies of 3 months in duration. Our review shows that majority of the findings are observed in the subchronic studies since additional target organs were not observed in 24 chronic non rodent studies and in 21 chronic rodent studies. However, 6 studies in non rodents and 6 studies in rodents yielded new findings that were not seen in studies of 3-month or shorter duration. For 3 compounds the new safety findings did contribute to termination of clinical development plans. Although the incidence of compound termination associated with chronic toxicology study observations is low (∼10%), the observations made in these studies can be important for evaluating human safety risk.


Asunto(s)
Pruebas de Toxicidad Crónica/métodos , Pruebas de Toxicidad Subcrónica/métodos , Animales , Bases de Datos Factuales , Relación Dosis-Respuesta a Droga , Industria Farmacéutica , Humanos , Modelos Animales , Reproducibilidad de los Resultados , Medición de Riesgo , Especificidad de la Especie , Factores de Tiempo
6.
Hepatology ; 64(5): 1430-1441, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27474787

RESUMEN

In 2015, European and U.S. health agencies issued warning letters in response to 9 reported clinical cases of severe bradycardia/bradyarrhythmia in hepatitis C virus (HCV)-infected patients treated with sofosbuvir (SOF) in combination with other direct acting antivirals (DAAs) and the antiarrhythmic drug, amiodarone (AMIO). We utilized preclinical in vivo models to better understand this cardiac effect, the potential pharmacological mechanism(s), and to identify a clinically translatable model to assess the drug-drug interaction (DDI) cardiac risk of current and future HCV inhibitors. An anesthetized guinea pig model was used to elicit a SOF+AMIO-dependent bradycardia. Detailed cardiac electrophysiological studies in this species revealed SOF+AMIO-dependent selective nodal dysfunction, with initial, larger effects on the sinoatrial node. Further studies in conscious, rhesus monkeys revealed an emergent bradycardia and bradyarrhythmia in 3 of 4 monkeys administered SOF+AMIO, effects not observed with either agent alone. Morever, bradycardia and bradyarrhythmia were not observed in rhesus monkeys when intravenous infusion of MK-3682 was completed after AMIO pretreatment. CONCLUSIONS: These are the first preclinical in vivo experiments reported to replicate the severe clinical SOF+AMIO cardiac DDI and provide potential in vivo mechanism of action. As such, these data provide a preclinical risk assessment paradigm, including a clinically relevant nonhuman primate model, with which to better understand cardiovascular DDI risk for this therapeutic class. Furthermore, these studies suggest that not all HCV DAAs and, in particular, not all HCV nonstructural protein 5B inhibitors may exhibit this cardiac DDI with amiodarone. Given the selective in vivo cardiac electrophysiological effect, these data enable targeted cellular/molecular mechanistic studies to more precisely identify cell types, receptors, and/or ion channels responsible for the clinical DDI. (Hepatology 2016;64:1430-1441).


Asunto(s)
Amiodarona/farmacología , Antiarrítmicos/farmacología , Antivirales/farmacología , Corazón/efectos de los fármacos , Hepacivirus/efectos de los fármacos , Nucleótidos/antagonistas & inhibidores , Sofosbuvir/farmacología , Amiodarona/efectos adversos , Animales , Antiarrítmicos/efectos adversos , Antivirales/efectos adversos , Interacciones Farmacológicas , Cobayas , Corazón/fisiología , Macaca mulatta , Masculino , Sofosbuvir/efectos adversos
7.
J Pharmacol Toxicol Methods ; 81: 107-14, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27166580

RESUMEN

INTRODUCTION: There has been an increasing need to conduct investigative safety pharmacology studies to complement regulatory-required studies, particularly as it applies to a comprehensive assessment of cardiovascular (CV) risk. METHODS: We describe refined methodology using a combination of telemetry and direct signal acquisition to record concomitant peripheral hemodynamics, ECG, and left ventricular (LV) structure (LV chamber size and LV wall thickness) and function, including LV pressure-volume (PV) loops to determine load independent measures of contractility (end systolic elastance, Ees, and preload recruitable stroke work, PRSW) in conscious beagle dogs. Following baseline characterization, 28days of chronic rapid ventricular pacing (RVP) was performed and cardiac function monitored: both as a way to compare measures during development of dysfunction and to characterize feasibility of a model to assess CV safety in animals with underlying cardiac dysfunction. RESULTS: While ±dP/dT decreased within a few days of RVP and remained stable, more comprehensive cardiac function measurements, including Ees and PRSW, provided a more sensitive assessment confirming the value of such endpoints for a more clear functional assessment. After 28days of RVP, the inodilator pimobendan was administered to further demonstrate the ability to detect changes in cardiac function. Expectedly pimobendan caused a leftward shift in the PV loop, improved ejection fraction (EF) and significantly improved Ees and PRSW. DISCUSSION: In summary, the data show the feasibility and importance in measuring enhanced cardiac functional parameters in conscious normal beagle dogs and further describe a relatively stable cardiac dysfunction model that could be used as an investigative safety pharmacology risk assessment tool.


Asunto(s)
Pruebas de Función Cardíaca/métodos , Pruebas de Función Cardíaca/normas , Modelos Biológicos , Farmacología/métodos , Seguridad , Telemetría/métodos , Animales , Presión Sanguínea/efectos de los fármacos , Estimulación Cardíaca Artificial , Cardiotónicos/farmacología , Perros , Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Electrocardiografía/efectos de los fármacos , Electrodos Implantados , Hemodinámica/efectos de los fármacos , Masculino , Contracción Miocárdica/efectos de los fármacos , Piridazinas/farmacología , Medición de Riesgo , Función Ventricular Izquierda/efectos de los fármacos
8.
Birth Defects Res B Dev Reprod Toxicol ; 107(2): 108-19, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27062127

RESUMEN

This report discusses the principles of reproductive toxicity risk assessment for biopharmaceuticals blocking the PD-1/programmed cell death ligand 1 (PD-L1) pathway, which have been developed for the treatment of patients with advanced malignancies. The PD-1/PD-L1 pathway is a T-cell co-inhibitory pathway that normally maintains immune tolerance to self. Its role in pregnancy is to maintain immune tolerance to the fetal allograft. In cancer patients, this signaling pathway is hijacked by some neoplasms to avoid immune destruction. PD-1/PD-L1-blocking agents enhance functional activity of the target lymphocytes to eventually cause immune rejection of the tumor. A therapeutic blockade of PD-1/PD-L1 pathway that occurs at full target engagement provides a unique challenge to address the risk to pregnancy because disruption of the same pathway may also reduce or abrogate maternal immune tolerance to the fetal alloantigens inherited through the father. Typically, nonclinical reproductive and developmental toxicity (DART) studies in animals (rats and rabbits) with clinical drug candidates are conducted to identify potential risk in humans and to determine exposure margin for the effects on reproduction as part of the risk assessment. However, for biopharmaceuticals for which the desired mechanism of action cannot be separated from potential deleterious effects to the fetus and when the only relevant toxicology species is nonhuman primate (NHP), the risk to reproduction can be predicted by a mechanism-based assessment using data generated from murine surrogate models as supportive information without conducting DART in NHPs. Such an approach has been used in the evaluation of pregnancy risk of anti-PD-1 agent, pembrolizumab, and has been demonstrated as an important alternative to performing DART studies in NHPs.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Antígeno B7-H1/metabolismo , Reproducción/efectos de los fármacos , Animales , Antígeno B7-H1/antagonistas & inhibidores , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Femenino , Humanos , Inmunoterapia , Ratones , Neoplasias/tratamiento farmacológico , Placenta/efectos de los fármacos , Placenta/metabolismo , Embarazo , Medición de Riesgo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
10.
ILAR J ; 57(2): 101-108, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-28053064

RESUMEN

Human discovery of pharmacologically active substances is arguably the oldest of the biomedical sciences with origins >3500 years ago. Since ancient times, four major transformations have dramatically impacted pharmaceutical development, each driven by advances in scientific knowledge, technology, and/or regulation: (1) anesthesia, analgesia, and antisepsis; (2) medicinal chemistry; (3) regulatory toxicology; and (4) targeted drug discovery. Animal experimentation in pharmaceutical development is a modern phenomenon dating from the 20th century and enabling several of the four transformations. While each transformation resulted in more effective and/or safer pharmaceuticals, overall attrition, cycle time, cost, numbers of animals used, and low probability of success for new products remain concerns, and pharmaceutical development remains a very high risk business proposition. In this manuscript we review pharmaceutical development since ancient times, describe its coevolution with animal experimentation, and attempt to predict the characteristics of future transformations.


Asunto(s)
Experimentación Animal , Descubrimiento de Drogas , Animales , Humanos , Dolor
11.
Toxicol Pathol ; 43(5): 621-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26047967

RESUMEN

High-dose selection for 6-month carcinogenicity studies of pharmaceutical candidates in Tg.rasH2-transgenic mice currently primarily relies on (1) estimation of a maximum tolerated dose (MTD) from the results of a 1-month range-finding study, (2) determination of the maximum dose administrable to the animals (maximum feasible dose [MFD]), (3) demonstration of a plateau in systemic exposure, and (4) use of a limit dose of 1,500 mg/kg/day for products with human daily doses not exceeding 500 mg. Eleven 6-month Tg.rasH2 carcinogenicity studies and their corresponding 1-month range-finding studies conducted at Merck were reviewed. High doses were set by estimation of the MTD in 6, by plateau of exposure in 3, and by MFD in 2 cases. For 4 of 6 studies where MTD was used for high-dose selection, the 1-month study accurately predicted the 6-month study tolerability whereas in the remaining 2 studies the high doses showed poorer tolerability than expected. The use of 3 or more drug-treated dose levels proved useful to ensure that a study would successfully and unambiguously demonstrate that a drug candidate was adequately evaluated for carcinogenicity at a minimally toxic high dose level, especially when the high dose may be found to exceed the MTD.


Asunto(s)
Pruebas de Carcinogenicidad/métodos , Evaluación Preclínica de Medicamentos/métodos , Animales , Femenino , Masculino
12.
Biomark Med ; 5(4): 497-514, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21861671

RESUMEN

One promise of new translational safety biomarkers (TSBs) is their ability to demonstrate that toxicities in animal studies are monitorable at an early stage, such that human relevance of potential adverse effects of drugs can be safely and definitively evaluated in clinical trials. Another is that they would provide an earlier, more definitive and deeper insight to patient prognosis compared with conventional biomarkers. Recent experience with regulatory authorities indicates that resource demands for new TSB qualifications under the current framework are daunting and the rate of their expansion will be slow, particularly in light of mounting financial pressures on the pharmaceutical industry. Sponsors face a dilemma over engaging in safety biomarker qualification consortia. While it is clear new TSBs could be considered catalysts to drug development and that patient health, business and scientific benefits, described here using examples, should outweigh qualification costs, concerns exist that early ambiguities in biomarker interpretations at the introduction of such new TSBs might hinder drug development.


Asunto(s)
Biomarcadores Farmacológicos/metabolismo , Animales , Ensayos Clínicos como Asunto/economía , Ensayos Clínicos como Asunto/métodos , Evaluación Preclínica de Medicamentos/economía , Industria Farmacéutica/economía , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Regulación Gubernamental , Humanos , Preparaciones Farmacéuticas/economía , Ratas
13.
Toxicol Pathol ; 39(4): 716-44, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21666103

RESUMEN

Data collected from 182 marketed and nonmarketed pharmaceuticals demonstrate that there is little value gained in conducting a rat two-year carcinogenicity study for compounds that lack: (1) histopathologic risk factors for rat neoplasia in chronic toxicology studies, (2) evidence of hormonal perturbation, and (3) positive genetic toxicology results. Using a single positive result among these three criteria as a test for outcome in the two-year study, fifty-two of sixty-six rat tumorigens were correctly identified, yielding 79% test sensitivity. When all three criteria were negative, sixty-two of seventy-six pharmaceuticals (82%) were correctly predicted to be rat noncarcinogens. The fourteen rat false negatives had two-year study findings of questionable human relevance. Applying these criteria to eighty-six additional chemicals identified by the International Agency for Research on Cancer as likely human carcinogens and to drugs withdrawn from the market for carcinogenicity concerns confirmed their sensitivity for predicting rat carcinogenicity outcome. These analyses support a proposal to refine regulatory criteria for conducting a two-year rat study to be based on assessment of histopathologic findings from a rat six-month study, evidence of hormonal perturbation, genetic toxicology results, and the findings of a six-month transgenic mouse carcinogenicity study. This proposed decision paradigm has the potential to eliminate over 40% of rat two-year testing on new pharmaceuticals without compromise to patient safety.


Asunto(s)
Pruebas de Carcinogenicidad/métodos , Carcinógenos/toxicidad , Pruebas de Mutagenicidad/métodos , Animales , Pruebas de Carcinogenicidad/normas , Carcinógenos/normas , Bases de Datos Factuales , Árboles de Decisión , Modelos Animales de Enfermedad , Estudios de Evaluación como Asunto , Femenino , Guías como Asunto , Humanos , Inmunosupresores , Masculino , Ratones , Ratones Transgénicos , Pruebas de Mutagenicidad/normas , Neoplasias/inducido químicamente , Ratas , Ratas Endogámicas F344 , Factores de Riesgo , Estadística como Asunto , Pruebas de Toxicidad Crónica
14.
Toxicol Pathol ; 38(1): 51-61, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19893055

RESUMEN

International guidelines allow for use of a short-term cancer bioassay (twenty-six weeks) in transgenic mice as a substitute for one of the two required long-term rodent bioassays in the preclinical safety evaluation of pharmaceuticals. The two models that have gained the widest acceptance by sponsors and regulatory authorities are the CB6F1-RasH2 mouse hemizygous for a human H-ras transgene and the B6.129N5-Trp53 mouse heterozygous for a p53 null allele. The p53(+/-) model is of particular value for compounds with residual concern that genotoxic activity may contribute to tumorigenesis. The rasH2 model is an appropriate alternative without regard to evidence of genotoxic potential. Since results from a short-term bioassay can be obtained relatively early in drug development, there is the potential for more timely assessment of cancer risk for individuals in long-term clinical trials. Use of these models in preclinical safety evaluation also significantly reduces animal use, time, and manpower. Preliminary findings indicate that prediction of two-year rat bioassay outcomes based on data from chronic rat toxicity studies, together with early assessment of carcinogenic potential in short-term transgenic models, may have the potential to increase the timeliness and efficiency of strategies for the identification of human carcinogenic hazards.


Asunto(s)
Pruebas de Carcinogenicidad/métodos , Industria Farmacéutica , Ratones Transgénicos , Animales , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Ratones , Ratas , Proteína p53 Supresora de Tumor/fisiología
15.
Chem Res Toxicol ; 22(2): 257-62, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19170595

RESUMEN

Species differences in drug metabolism present two challenges that may confound the nonclinical safety assessment of candidate drugs. The first challenge is encountered when metabolites are formed uniquely or disproportionately in humans. Another challenge is understanding the human relevance of toxicities associated with metabolites formed uniquely or disproportionately in a nonclinical species. One potential approach to minimize the impact of metabolite related challenges is to consider genetically engineered mouse models that express human P450 enzymes. Human P450 expressing mouse models may have the ability to generate major human metabolites and eliminate or reduce the formation of mouse specific metabolites. Prior to determining the utility of any particular model, it is important to qualify by characterizing protein expression, establishing whether the model generates an in vivo metabolite profile more closely related to that of humans than the wild-type mouse, verifying genetic stability, and evaluating animal health. When compared to the current strategy for handling metabolite challenges (i.e., direct administration of metabolite), identifying an appropriate human P450 expressing model could provide a number of benefits. Such benefits include improved scientific relevance of the evaluation, decreased resource needs, and a possible reduction in the number of animals used. These benefits may ultimately improve the quality and speed by which promising new drug candidates are developed and delivered to patients.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Ratones Transgénicos , Modelos Animales , Preparaciones Farmacéuticas/metabolismo , Pruebas de Toxicidad/métodos , Administración Oral , Animales , Costos y Análisis de Costo , Sistema Enzimático del Citocromo P-450/genética , Perros , Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/metabolismo , Guías como Asunto , Humanos , Legislación de Medicamentos , Ratones , Ratones Noqueados , Ratas , Medición de Riesgo , Pruebas de Toxicidad/economía
16.
Methods Mol Biol ; 460: 239-61, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18449491

RESUMEN

Scientists in the pharmaceutical industry have ready access to samples from animal toxicology studies carefully designed to test the safety characteristics of a steady pipeline of agents advancing toward clinical testing. Applications of toxicogenomics to the evaluation of compounds could best be realized if this promising technology could be implemented in these studies fully anchored in the traditional study end points currently used to characterize phenotypic outcome and to support the safe conduct of clinical testing. Regulatory authorities worldwide have declared their support for toxicogenomics and related technological tools to positively impact drug development, and guidance has been published. However, applications of exploratory "omics" technologies to compounds undergoing safety testing remain inhibited due to two core data submission responsibility implications and ambiguities: (1) constraints arising from continual literature surveillance and data reanalysis burdens, under the shadow of looming subsequent reporting requirements to regulatory authorities as gene expression end points loosely linked to safety gain attention in the published literature, and (2) ambiguities in interpretation of validation stature remain between exploratory, probable valid, and known valid safety biomarkers. A proposal is offered to address these regulatory implementation barriers to open access for exploring this technology in prospective drug development animal toxicology studies.


Asunto(s)
Diseño de Fármacos , Genómica , Toxicología , Biomarcadores , Conducta Cooperativa , Sector Privado , Sector Público
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...