Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mutagenesis ; 31(3): 309-21, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26443851

RESUMEN

The anti-parasitic benzimidazole flubendazole has been used for many years to treat intestinal infections in humans and animals. Previous genotoxicity studies have shown that the compound is not a bacterial mutagen and a bone marrow micronucleus test, using a formulation that limited systemic absorption, was negative. The purpose of this study is to explore the genotoxicity of flubendazole and its main metabolites in in vitro micronucleus studies and to test a new oral formulation that improves systemic absorption in an in vivo micronucleus test. The isolated metabolites were also screened using the Ames test for bacterial mutagenicity. It was found that flubendazole, like other chemically related benzimidazoles used in anti-parasitic therapies, is a potent aneugen in vitro The hydrolysed metabolite of flubendazole is negative in these tests, but the reduced metabolite (R- and S-forms) shows both aneugenic and clastogenic activity. However, in vitro micronucleus tests of flubendazole in the presence of rat liver S9 gave almost identical signals for aneugenicity as they did in the absence of S9, suggesting that any clastogenicity from the reduced metabolite is not sufficient to change the overall profile. Like flubendazole itself, both metabolites are negative in the Ames test. Analysis of dose-response curves from the in vitro tests, using recently developed point of departure approaches, demonstrate that the aneugenic potency of flubendazole is very similar to related anti-parasitic benzimidazoles, including albendazole, which is used in mass drug administration programmes to combat endemic filarial diseases. The in vivo micronucleus test of the new formulation of flubendazole also showed evidence of induced aneugenicity. Analysis of the in vivo data allowed a reference dose for aneugenicity to be established which can be compared with therapeutic exposures of flubendazole when this has been established. Analysis of the plasma from the animals used in the in vivo micronucleus test showed that there is increased exposure to flubendazole compared with previously tested formulations, as well as significant formation of the non-genotoxic hydrolysed metabolite of flubendazole and small levels of the reduced metabolite. In conclusion, this study shows that flubendazole is a potent aneugen in vitro with similar potency to chemically related benzimidazoles currently used as anti-parasitic therapies. The reduced metabolite also has aneugenic properties as well as clastogenic properties. Treatment with a new formulation of flubendazole that allows increased systemic exposure, compared with previously used formulations, also results in detectable aneugenicity in vivo. Based on the lack of carcinogenicity of this class of benzimidazoles and the intended short-term dosing, it is unlikely that flubendazole treatment will pose a carcinogenic risk to patients.


Asunto(s)
Aneugénicos/toxicidad , Aberraciones Cromosómicas , Daño del ADN , Linfocitos/efectos de los fármacos , Mebendazol/análogos & derivados , Activación Metabólica , Aneugénicos/metabolismo , Animales , Antinematodos/metabolismo , Antinematodos/toxicidad , Células Cultivadas , Cromosomas Humanos/efectos de los fármacos , ADN/efectos de los fármacos , Humanos , Linfocitos/metabolismo , Masculino , Mebendazol/metabolismo , Mebendazol/toxicidad , Pruebas de Micronúcleos , Mutágenos/metabolismo , Mutágenos/toxicidad , Ratas
3.
Mutagenesis ; 27(5): 523-32, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22539226

RESUMEN

The parasitic disease human African trypanomiasis (HAT), also known as sleeping sickness, is a highly neglected fatal condition endemic in sub-Saharan Africa, which is poorly treated with medicines that are toxic, no longer effective or very difficult to administer. New, safe, effective and easy-to-use treatments are urgently needed. Many nitroimidazoles possess antibacterial and antiprotozoal activity and examples such as tinidazole are used to treat trichomoniasis and guardiasis, but concerns about toxicity including genotoxicity limit their usefulness. Fexinidazole, a 2-substituted 5-nitroimidazole rediscovered by the Drugs for Neglected Diseases initiative (DNDi) after extensive compound mining of public and pharmaceutical company databases, has the potential to become a short-course, safe and effective oral treatment, curing both acute and chronic HAT. This paper describes the genotoxicity profile of fexinidazole and its two active metabolites, the sulfoxide and sulfone derivatives. All the three compounds are mutagenic in the Salmonella/Ames test; however, mutagenicity is either attenuated or lost in Ames Salmonella strains that lack one or more nitroreductase(s). It is known that these enzymes can nitroreduce compounds with low redox potentials, whereas their mammalian cell counterparts cannot, under normal conditions. Fexinidazole and its metabolites have low redox potentials and all mammalian cell assays to detect genetic toxicity, conducted for this study either in vitro (micronucleus test in human lymphocytes) or in vivo (ex vivo unscheduled DNA synthesis in rats; bone marrow micronucleus test in mice), were negative. Thus, fexinidazole does not pose a genotoxic hazard to patients and represents a promising drug candidate for HAT. Fexinidazole is expected to enter Phase II clinical trials in 2012.


Asunto(s)
Mutágenos/toxicidad , Nitroimidazoles/toxicidad , Tripanocidas/toxicidad , Animales , Bacterias/efectos de los fármacos , Bacterias/enzimología , Células de la Médula Ósea/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Masculino , Ratones , Pruebas de Micronúcleos , Pruebas de Mutagenicidad , Mutágenos/metabolismo , Nitroimidazoles/metabolismo , Nitrorreductasas/metabolismo , Ratas , Tripanocidas/metabolismo , Tripanosomiasis Africana/tratamiento farmacológico
4.
AIDS Res Ther ; 6: 18, 2009 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-19660105

RESUMEN

Roche's protease inhibitor nelfinavir mesylate (Viracept) produced between March 2007-June 2007 was found to contain elevated levels of ethyl methanesulfonate (EMS), a known mutagen (alkylator) - leading to a global recall of the drug. EMS levels in a daily dose (2,500 mg Viracept/day) were predicted not to exceed a dose of approximately 2.75 mg/day (approximately 0.055 mg/kg/day based on 50 kg patient). As existing toxicology data on EMS did not permit an adequate patient risk assessment, a comprehensive animal toxicology evaluation of EMS was conducted. General toxicity of EMS was investigated in rats over 28 days. Two studies for DNA damage were performed in mice; chromosomal damage was assessed using a micronucleus assay and gene mutations were detected using the MutaMouse transgenic model. In addition, experiments designed to extrapolate animal exposure to humans were undertaken. A general toxicity study showed that the toxicity of EMS occurred only at doses >or= 60 mg/kg/day, which is far above that received by patients. Studies for chromosomal damage and mutations in mice demonstrated a clear threshold effect with EMS at 25 mg/kg/day, under chronic dosing conditions. Exposure analysis (Cmax) demonstrated that approximately 370-fold higher levels of EMS than that ingested by patients, are needed to saturate known, highly conserved, error-free, mammalian DNA repair mechanisms for alkylation. In summary, animal studies suggested that patients who took nelfinavir mesylate with elevated levels of EMS are at no increased risk for carcinogenicity or teratogenicity over their background risk, since mutations are prerequisites for such downstream events. These findings are potentially relevant to >40 marketed drugs that are mesylate salts.

5.
Regul Toxicol Pharmacol ; 55(1): 52-68, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19505521

RESUMEN

The Globally Harmonized System of Classification and Labeling of Chemicals (GHS) requires classification of chemicals on germ cell mutagenicity. The Japanese government has conducted GHS classification on about 1400 chemicals in a 2-year project (J-GHS) for implementing GHS domestically. Prior to the classification work, the technical guidance for classification of germ cell mutagens was prepared. This guidance introduces the concept of heritable mutagenicity, and presents detailed criteria for germ cell mutagens, test data to be used, and a practical decision tree for classification. These practical guidance and supporting explanations are useful for non-expert Classifiers (scientists applying the classification criteria). Several issues, however, were identified during the course of J-GHS and in re-evaluating the classification results. These include: (1) the information sources when available data are limited; (2) lack of understanding GHS classification criteria or insufficient review of the information by Classifiers; (3) varying opinions of experts on data quality and weight of evidence, and; (4) decision tree approaches, e.g., inadequacy for use in overall evaluation in some cases. Ideally, classification should be performed by Classifiers with high expertise using high quality information sources. Genetic toxicologists as experts should consider data quality and reliability, and give a critical review of all available information for support of classification. A weight of evidence approach is also required to assess mutagenic potential of chemicals. Critical points for suitable classification for GHS are discussed.


Asunto(s)
Contaminantes Ambientales/clasificación , Salud Global , Sustancias Peligrosas/clasificación , Mutágenos/clasificación , Toxicología/normas , Clasificación/métodos , Contaminantes Ambientales/toxicidad , Células Germinativas/efectos de los fármacos , Sustancias Peligrosas/toxicidad , Humanos , Cooperación Internacional , Japón , Mutágenos/toxicidad , Toxicología/métodos
6.
Artículo en Inglés | MEDLINE | ID: mdl-31708072

RESUMEN

As part of the 7th International Workshops on Genotoxicity Testing held in Tokyo, Japan in November 2017, a workgroup of experts reviewed and assessed the risk of aneugens for human health. The present manuscript is one of three manuscripts from the workgroup and reports on the unanimous consensus reached on the evidence for aneugens affecting germ cells, their mechanisms of action and role in hereditary diseases. There are 24 chemicals with strong or sufficient evidence for germ cell aneugenicity providing robust support for the ability of chemicals to induce germ cell aneuploidy. Interference with microtubule dynamics or inhibition of topoisomerase II function are clear characteristics of germ cell aneugens. Although there are mechanisms of chromosome segregation that are unique to germ cells, there is currently no evidence for germ cell-specific aneugens. However, the available data are heavily skewed toward chemicals that are aneugenic in somatic cells. Development of high-throughput screening assays in suitable animal models for exploring additional targets for aneuploidy induction, such as meiosis-specific proteins, and to prioritize chemicals for the potential to be germ cell aneugens is encouraged. Evidence in animal models support that: oocytes are more sensitive than spermatocytes and somatic cells to aneugens; exposure to aneugens leads to aneuploid conceptuses; and, the frequencies of aneuploidy are similar in germ cells and zygotes. Although aneuploidy in germ cells is a significant cause of infertility and pregnancy loss in humans, there is currently limited evidence that aneugens induce hereditary diseases in human populations because the great majority of aneuploid conceptuses die in utero. Overall, the present work underscores the importance of protecting the human population from exposure to chemicals that can induce aneuploidy in germ cells that, in contrast to carcinogenicity, is directly linked to an adverse outcome.


Asunto(s)
Aneugénicos/toxicidad , Aneuploidia , Carcinogénesis , Enfermedades Genéticas Congénitas/patología , Células Germinativas/efectos de los fármacos , Animales , Células Germinativas/patología , Humanos , Factores de Riesgo
7.
Artículo en Inglés | MEDLINE | ID: mdl-31699346

RESUMEN

An aneuploidy workgroup was established as part of the 7th International Workshops on Genotoxicity Testing. The workgroup conducted a review of the scientific literature on the biological mechanisms of aneuploidy in mammalian cells and methods used to detect chemical aneugens. In addition, the current regulatory framework was discussed, with the objective to arrive at consensus statements on the ramifications of exposure to chemical aneugens for human health risk assessment. As part of these efforts, the workgroup explored the use of adverse outcome pathways (AOPs) to document mechanisms of chemically induced aneuploidy in mammalian somatic cells. The group worked on two molecular initiating events (MIEs), tubulin binding and binding to the catalytic domain of aurora kinase B, which result in several adverse outcomes, including aneuploidy. The workgroup agreed that the AOP framework provides a useful approach to link evidence for MIEs with aneuploidy on a cellular level. The evidence linking chemically induced aneuploidy with carcinogenicity and hereditary disease was also reviewed and is presented in two companion papers. In addition, the group came to the consensus that the current regulatory test batteries, while not ideal, are sufficient for the identification of aneugens and human risk assessment. While it is obvious that there are many different MIEs that could lead to the induction of aneuploidy, the most commonly observed mechanisms involving chemical aneugens are related to tubulin binding and, to a lesser extent, inhibition of mitotic kinases. The comprehensive review presented here should help with the identification and risk management of aneugenic agents.


Asunto(s)
Rutas de Resultados Adversos , Aneuploidia , Enfermedades Genéticas Congénitas/inducido químicamente , Mitosis/efectos de los fármacos , Pruebas de Mutagenicidad/métodos , Mutágenos/toxicidad , Neoplasias/inducido químicamente , Animales , Aurora Quinasa B/antagonistas & inhibidores , Aurora Quinasa B/fisiología , Carcinógenos/toxicidad , Aberraciones Cromosómicas/inducido químicamente , Segregación Cromosómica/efectos de los fármacos , Cromosomas/efectos de los fármacos , Genes Reporteros , Enfermedades Genéticas Congénitas/genética , Células Germinativas/efectos de los fármacos , Células Germinativas/ultraestructura , Humanos , Ratones , Pruebas de Micronúcleos , Microtúbulos/efectos de los fármacos , Mitosis/fisiología , Pruebas de Mutagenicidad/normas , Mutágenos/análisis , Neoplasias/genética , No Disyunción Genética/efectos de los fármacos , Gestión de Riesgos/legislación & jurisprudencia , Moduladores de Tubulina/toxicidad
8.
Artículo en Inglés | MEDLINE | ID: mdl-31699349

RESUMEN

Aneuploidy is regarded as a hallmark of cancer, however, its role is complex with both pro- and anti-carcinogenic effects evident. In this IWGT review, we consider the role of aneuploidy in cancer biology; cancer risk associated with constitutive aneuploidy; rodent carcinogenesis with known chemical aneugens; and chemotherapy-related malignant neoplasms. Aneuploidy is seen at various stages in carcinogenesis. However, the relationship between induced aneuploidy occurring after exposure and clonal aneuploidy present in tumours is not clear. Recent evidence indicates that the induction of chromosomal instability (CIN), may be more important than aneuploidy per se, in the carcinogenic process. Down Syndrome, trisomy 21, is associated with altered hematopoiesis in utero which, in combination with subsequent mutations, results in an increased risk for acute megakaryoblastic and lymphoblastic leukemias. In contrast, there is reduced cancer risk for most solid tumours in Down Syndrome. Mouse models with high levels of aneuploidy are also associated with increased cancer risk for particular tumours with long latencies, but paradoxically other types of tumour often show decreased incidence. The aneugens reviewed that induce cancer in humans and animals all possess other carcinogenic properties, such as mutagenicity, clastogenicity, cytotoxicity, organ toxicities, hormonal and epigenetic changes which likely account for, or interact with aneuploidy, to cause carcinogenesis. Although the role that aneuploidy plays in carcinogenesis has not been fully established, in many cases, it may not play a primary causative role. Tubulin-disrupting aneugens that do not possess other properties linked to carcinogenesis, were not carcinogenic in rodents. Similarly, in humans, for the tubulin-disrupting aneugens colchicine and albendazole, there is no reported association with increased cancer risk. There is a need for further mechanistic studies on agents that induce aneuploidy, particularly by mechanisms other than tubulin disruption and to determine the role of aneuploidy in pre-neoplastic events and in early and late stage neoplasia.


Asunto(s)
Aneuploidia , Carcinogénesis/genética , Carcinógenos/toxicidad , Inestabilidad Cromosómica , Pruebas de Mutagenicidad/métodos , Neoplasias/inducido químicamente , Animales , Centrosoma , Trastornos de los Cromosomas/genética , Cromosomas/efectos de los fármacos , Síndrome de Down/complicaciones , Síndrome de Down/genética , Predisposición Genética a la Enfermedad , Humanos , Ratones , Modelos Animales , Pruebas de Mutagenicidad/normas , Mutágenos/toxicidad , Neoplasias/genética , Neoplasias Primarias Secundarias/inducido químicamente , Neoplasias Primarias Secundarias/genética , Huso Acromático/efectos de los fármacos , Moduladores de Tubulina/toxicidad
9.
Mutat Res ; 653(1-2): 23-33, 2008 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-18450500

RESUMEN

Sixteen coded compounds were blind-tested at 4 laboratories using the recently described GADD45a-GFP genotoxicity assay. The compounds were chosen to include non-genotoxic compounds as well as weak and strong genotoxins. None of the compounds required metabolic activation in order to exhibit genotoxic effects. The participating laboratories included 2 global pharmaceutical companies, a global consumer goods company and the Gentronix laboratory in Manchester. Each compound was tested 4 times on different days following a protocol previously described. The tests were carried out after a 3-day training period from the parent lab (Manchester). Following the exclusion of data from tests with positive control failures and data series with 'spikes', 92% of assays gave the correct result: non-genotoxins giving negative results and genotoxins giving positive results. There were no randomly distributed problems suggesting that differences between the results from different sites reflected the use of different instruments, procedural differences and operator experience. In naïve operator laboratories the quality of data improved with operator practice. It was concluded that simple clarification of the protocol would provide the level of reliability required for widespread use of the assay in hazard assessment.


Asunto(s)
Proteínas de Ciclo Celular/biosíntesis , Proteínas Fluorescentes Verdes/biosíntesis , Pruebas de Mutagenicidad , Mutágenos/análisis , Proteínas Nucleares/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Animales , Proteínas de Ciclo Celular/genética , Línea Celular , Proteínas Fluorescentes Verdes/genética , Humanos , Pruebas de Mutagenicidad/métodos , Pruebas de Mutagenicidad/normas , Proteínas Nucleares/genética , Distribución Aleatoria , Proteínas Recombinantes de Fusión/genética , Reproducibilidad de los Resultados
10.
Mutat Res ; 628(1): 31-55, 2007 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-17293159

RESUMEN

Workshop participants agreed that genotoxicity tests in mammalian cells in vitro produce a remarkably high and unacceptable occurrence of irrelevant positive results (e.g. when compared with rodent carcinogenicity). As reported in several recent reviews, the rate of irrelevant positives (i.e. low specificity) for some studies using in vitro methods (when compared to this "gold standard") means that an increased number of test articles are subjected to additional in vivo genotoxicity testing, in many cases before, e.g. the efficacy (in the case of pharmaceuticals) of the compound has been evaluated. If in vitro tests were more predictive for in vivo genotoxicity and carcinogenicity (i.e. fewer false positives) then there would be a significant reduction in the number of animals used. Beyond animal (or human) carcinogenicity as the "gold standard", it is acknowledged that genotoxicity tests provide much information about cellular behaviour, cell division processes and cellular fate to a (geno)toxic insult. Since the disease impact of these effects is seldom known, and a verification of relevant toxicity is normally also the subject of (sub)chronic animal studies, the prediction of in vivo relevant results from in vitro genotoxicity tests is also important for aspects that may not have a direct impact on carcinogenesis as the ultimate endpoint of concern. In order to address the high rate of in vitro false positive results, a 2-day workshop was held at the European Centre for the Validation of Alternative Methods (ECVAM), Ispra, Italy in April 2006. More than 20 genotoxicity experts from academia, government and industry were invited to review data from the currently available cell systems, to discuss whether there exist cells and test systems that have a reduced tendency to false positive results, to review potential modifications to existing protocols and cell systems that might result in improved specificity, and to review the performance of some new test systems that show promise of improved specificity without sacrificing sensitivity. It was concluded that better guidance on the likely mechanisms resulting in positive results that are not biologically relevant for human health, and how to obtain evidence for those mechanisms, is needed both for practitioners and regulatory reviewers. Participants discussed the fact that cell lines commonly used for genotoxicity testing have a number of deficiencies that may contribute to the high false positive rate. These include, amongst others, lack of normal metabolism leading to reliance on exogenous metabolic activation systems (e.g. Aroclor-induced rat S9), impaired p53 function and altered DNA repair capability. The high concentrations of test chemicals (i.e. 10 mM or 5000 microg/ml, unless precluded by solubility or excessive toxicity) and the high levels of cytotoxicity currently required in mammalian cell genotoxicity tests were discussed as further potential sources of false positive results. Even if the goal is to detect carcinogens with short in vitro tests under more or less acute conditions, it does not seem logical to exceed the capabilities of cellular metabolic turnover, activation and defence processes. The concept of "promiscuous activation" was discussed. For numerous mutagens, the decisive in vivo enzymes are missing in vitro. However, if the substrate concentration is increased sufficiently, some other enzymes (that are unimportant in vivo) may take over the activation-leading to the same or a different active metabolite. Since we often do not use the right enzyme systems for positive controls in vitro, we have to rely on their promiscuous activation, i.e. to use excessive concentrations to get an empirical correlation between genotoxicity and carcinogenicity. A thorough review of published and industry data is urgently needed to determine whether the currently required limit concentration of 10mM or 5000 microg/ml, and high levels of cytotoxicity, are necessary for the detection of in vivo genotoxins and DNA-reactive, mutagenic carcinogens. In addition, various measures of cytotoxicity are currently allowable under OECD test guidelines, but there are few comparative data on whether different measures would result in different maximum concentrations for testing. A detailed comparison of cytotoxicity assessment strategies is needed. An assessment of whether test endpoints can be selected that are not intrinsically associated with cytotoxicity, and therefore are less susceptible to artefacts produced by cytotoxicity, should also be undertaken. There was agreement amongst the workshop participants that cell systems which are p53 and DNA-repair proficient, and have defined Phase 1 and Phase 2 metabolism, covering a broad set of enzyme forms, and used within the context of appropriately set limits of concentration and cytotoxicity, offer the best hope for reduced false positives. Whilst there is some evidence that human lymphocytes are less susceptible to false positives than the current rodent cell lines, other cell systems based on HepG2, TK6 and MCL-5 cells, as well as 3D skin models based on primary human keratinocytes also show some promise. Other human cell lines such as HepaRG, and human stem cells (the target for carcinogenicity) have not been used for genotoxicity investigations and should be considered for evaluation. Genetic engineering is also a valuable tool to incorporate missing enzyme systems into target cells. A collaborative research programme is needed to identify, further develop and evaluate new cell systems with appropriate sensitivity but improved specificity. In order to review current data for selection of appropriate top concentrations, measures and levels of cytotoxicity, metabolism, and to be able to improve existing or validate new assay systems, the participants called for the establishment of an expert group to identify the in vivo genotoxins and DNA-reactive, mutagenic carcinogens that we expect our in vitro genotoxicity assays to detect as well as the non-genotoxins and non-carcinogens we expect them not to detect.


Asunto(s)
Pruebas de Mutagenicidad , Animales , Células Cultivadas , Reacciones Falso Positivas , Humanos , Modelos Biológicos , Juego de Reactivos para Diagnóstico , Técnicas de Cultivo de Tejidos
11.
Pharmacogenomics ; 6(2): 181-4, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15882136

RESUMEN

A workshop was held on October 26-27, 2004, in Bonn, Germany, to discuss the potential use of omic technologies for regulatory non-clinical safety testing of pharmaceuticals. The meeting was hosted by the European Federation of Pharmaceutical Industries and Associations (EFPIA). The workshop was held in conjunction with the 6th European preclinical assessors meeting, which was organized in Bonn by the German Federal Institute for Drugs and Medical Devices (BfArM) and the Safety Working Party (SWP) of the Committee for Medicinal Products for Human Use (CHMP). Approximately 100 scientists, roughly half from the European pharmaceutical industry and half from European regulatory authorities, attended the workshop. The authors of this report constitute the organizing committee members.


Asunto(s)
Industria Farmacéutica/normas , Guías como Asunto/normas , Tecnología Farmacéutica/normas , Biotecnología/métodos , Biotecnología/normas , Unión Europea , Genómica , Humanos , Proteómica , Medición de Riesgo/métodos , Medición de Riesgo/normas , Tecnología Farmacéutica/métodos
12.
Eur J Med Chem ; 46(5): 1524-35, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21353728

RESUMEN

Nitroimidazoles are a well-known class of antibacterial and antiprotozoal drugs but in spite of the widespread clinical and veterinary use of these drugs, this family has been stigmatized in part due to associated genotoxicity problems. Here we report the synthesis, the anti-trypanosomal activity and a structure-activity relationship (SAR) study of a series of about fifty 1-aryl-4-nitro-1H-imidazoles, with an emphasis on selected in vivo active molecules. Compounds 4-nitro-1-{4-(trifluoromethoxy)phenyl}-1H-imidazole and 1-(3,4-dichlorophenyl)-4-nitro-1H-imidazole are curative in mouse models of both acute and chronic African trypanosomiasis when given orally at doses of 25-50 mg/kg for 4 days for the acute infection, and 50-100 mg/kg (bid) for 5 days in the chronic model. While both compounds are bacterial mutagens, activity is lost in strains lacking bacterial specific nitro-reductases. Mammalian nitro-reductases do not reduce nitroaromatic compounds with low redox potentials with same avidity as their bacterial counterparts and these compounds were shown to be devoid of genotoxicity in mammalian cells. Both compounds are promising leads for the treatment of human African trypanosomiasis (HAT or sleeping sickness), including the fatal stage 2 of the disease, for which new treatments are urgently needed.


Asunto(s)
Antiprotozoarios/uso terapéutico , Nitroimidazoles/uso terapéutico , Tripanosomiasis Africana/tratamiento farmacológico , Animales , Antiprotozoarios/síntesis química , Antiprotozoarios/química , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Ratones , Estructura Molecular , Pruebas de Mutagenicidad , Nitroimidazoles/síntesis química , Nitroimidazoles/química , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
13.
PLoS Negl Trop Dis ; 4(12): e923, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21200426

RESUMEN

BACKGROUND: Human African trypanosomiasis (HAT), also known as sleeping sickness, is a fatal parasitic disease caused by trypanosomes. Current treatment options for HAT are scarce, toxic, no longer effective, or very difficult to administer, in particular for the advanced, fatal stage of the disease (stage 2, chronic HAT). New safe, effective and easy-to-use treatments are urgently needed. Here it is shown that fexinidazole, a 2-substituted 5-nitroimidazole rediscovered by the Drugs for Neglected Diseases initiative (DNDi) after extensive compound mining efforts of more than 700 new and existing nitroheterocycles, could be a short-course, safe and effective oral treatment curing both acute and chronic HAT and that could be implemented at the primary health care level. To complete the preclinical development and meet the regulatory requirements before initiating human trials, the anti-parasitic properties and the pharmacokinetic, metabolic and toxicological profile of fexinidazole have been assessed. METHODS AND FINDINGS: Standard in vitro and in vivo anti-parasitic activity assays were conducted to assess drug efficacy in experimental models for HAT. In parallel, a full range of preclinical pharmacology and safety studies, as required by international regulatory guidelines before initiating human studies, have been conducted. Fexinidazole is moderately active in vitro against African trypanosomes (IC50 against laboratory strains and recent clinical isolates ranged between 0.16 and 0.93 µg/mL) and oral administration of fexinidazole at doses of 100 mg/kg/day for 4 days or 200 mg/kg/day for 5 days cured mice with acute and chronic infection respectively, the latter being a model for the advanced and fatal stage of the disease when parasites have disseminated into the brain. In laboratory animals, fexinidazole is well absorbed after oral administration and readily distributes throughout the body, including the brain. The absolute bioavailability of oral fexinidazole was 41% in mice, 30% in rats, and 10% in dogs. Furthermore, fexinidazole is rapidly metabolised in vivo to at least two biologically active metabolites (a sulfoxide and a sulfone derivative) that likely account for a significant portion of the therapeutic effect. Key pharmacokinetic parameter after oral absorption in mice for fexinidazole and its sulfoxide and sulfone metabolites are a C(max) of 500, 14171 and 13651 ng/mL respectively, and an AUC0₋24 of 424, 45031 and 96286 h.ng/mL respectively. Essentially similar PK profiles were observed in rats and dogs. Toxicology studies (including safety pharmacology and 4-weeks repeated-dose toxicokinetics in rat and dog) have shown that fexinidazole is well tolerated. The No Observed Adverse Event Levels in the 4-weeks repeated dose toxicity studies in rats and dogs was 200 mg/kg/day in both species, with no issues of concern identified for doses up to 800 mg/kg/day. While fexinidazole, like many nitroheterocycles, is mutagenic in the Ames test due to bacterial specific metabolism, it is not genotoxic to mammalian cells in vitro or in vivo as assessed in an in vitro micronucleus test on human lymphocytes, an in vivo mouse bone marrow micronucleus test, and an ex vivo unscheduled DNA synthesis test in rats. CONCLUSIONS: The results of the preclinical pharmacological and safety studies indicate that fexinidazole is a safe and effective oral drug candidate with no untoward effects that would preclude evaluation in man. The drug has entered first-in-human phase I studies in September 2009. Fexinidazole is the first new clinical drug candidate with the potential for treating advanced-stage sleeping sickness in thirty years.


Asunto(s)
Antiprotozoarios/administración & dosificación , Nitroimidazoles/administración & dosificación , Tripanosomiasis Africana/tratamiento farmacológico , Administración Oral , Animales , Antiprotozoarios/efectos adversos , Antiprotozoarios/metabolismo , Antiprotozoarios/farmacocinética , Modelos Animales de Enfermedad , Perros , Femenino , Concentración 50 Inhibidora , Ratones , Nitroimidazoles/efectos adversos , Nitroimidazoles/metabolismo , Nitroimidazoles/farmacocinética , Pruebas de Sensibilidad Parasitaria , Ratas , Resultado del Tratamiento
15.
Drug Discov Today ; 14(7-8): 337-42, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19340928

RESUMEN

The drug development process is currently being hindered by non-optimal prediction of toxicity. Advances in molecular profiling approaches, such as transcriptomics, proteomics and metabolomics, offer the potential to provide a more comprehensive insight into toxicological effects than hitherto possible. These new technologies present their own challenges, however, particularly in relation to standardization and assessment. The focus of this article is on describing the current trends concerning the application of omic approaches in drug safety assessment, with specific emphasis on the role of public-private partnerships in advancing this emerging arena.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Perfilación de la Expresión Génica/tendencias , Asociación entre el Sector Público-Privado/organización & administración , Asociación entre el Sector Público-Privado/tendencias , Toxicogenética/tendencias , Perfilación de la Expresión Génica/métodos , Humanos , Metabolómica/métodos , Metabolómica/tendencias , Proteómica/métodos , Proteómica/tendencias , Tecnología Farmacéutica/métodos , Tecnología Farmacéutica/tendencias , Toxicogenética/métodos
16.
Toxicol Lett ; 190(3): 333-9, 2009 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-19866516

RESUMEN

This perspective first considers the potential impact of the Viracept-EMS case in the framework of the current understanding of the low-dose effects of DNA-reactive chemicals and the approaches used to estimate health risks from genotoxins occurring as impurities in pharmaceutical products or as contaminants in the environment or workplace. It also presents an outlook on the nature of additional research building upon the Viracept-EMS case to test assumptions underlying thresholded dose-response relationships and to establish biologically based risk assessment models in lieu of default models for DNA-reactive compounds.


Asunto(s)
Alquilantes/toxicidad , Contaminación de Medicamentos , Metanosulfonato de Etilo/toxicidad , Inhibidores de la Proteasa del VIH/química , Mutágenos/toxicidad , Mutación/efectos de los fármacos , Nelfinavir/química , Alquilantes/análisis , Animales , Relación Dosis-Respuesta a Droga , Exposición a Riesgos Ambientales , Metanosulfonato de Etilo/análisis , Humanos , Ratones , Pruebas de Mutagenicidad , Mutágenos/análisis , Nivel sin Efectos Adversos Observados , Vigilancia de Productos Comercializados , Ratas , Medición de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA