Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
ALTEX ; 40(3): 534-540, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36888967

RESUMO

Progress in developing new tools, assays, and approaches to assess human hazard and health risk provides an opportunity to re-evaluate the necessity of dog studies for the safety evaluation of agrochemicals. A workshop was held where partic­ipants discussed the strengths and limitations of past use of dogs for pesticide evaluations and registrations. Opportunities were identified to support alternative approaches to answer human safety questions without performing the required 90-day dog study. Development of a decision tree for determining when the dog study might not be necessary to inform pesticide safety and risk assessment was proposed. Such a process will require global regulatory authority participation to lead to its acceptance. The identification of unique effects in dogs that are not identified in rodents will need further evaluation and determination of their relevance to humans. The establishment of in vitro and in silico approaches that can provide critical data on relative species sensitivity and human relevance will be an important tool to advance the decision process. Promising novel tools including in vitro comparative metabolism studies, in silico models, and high-throughput assays able to identify metabolites and mechanisms of action leading to development of adverse outcome pathways will need further development. To replace or eliminate the 90-day dog study, a collaborative, multidisciplinary, international effort that transcends organi­zations and regulatory agencies will be needed in order to develop guidance on when the study would not be necessary for human safety and risk assessment.


Assuntos
Rotas de Resultados Adversos , Praguicidas , Animais , Cães , Humanos , Agroquímicos/toxicidade , Praguicidas/toxicidade , Medição de Risco , Simulação por Computador
2.
ILAR J ; 62(1-2): 7-16, 2021 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-34181728

RESUMO

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.


Assuntos
Experimentação Animal , Pesquisa Biomédica , Animais , Cães , Camundongos , Ratos , Estados Unidos
3.
ILAR J ; 62(3): 314-331, 2021 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-35512294

RESUMO

The organization and function of the institutional animal care and use committee (IACUC) is the key component of government regulation and oversight of necessary scientific research using live animals and of AAALAC - International accreditation of animal care and use programs in the United States. The regulations, roles, and responsibilities of IACUCs have evolved since their inception 35 years ago from a limited focus on animal welfare and specific animal procedures to embracing scientific quality, data reproducibility and translation, and animal welfare as inextricably interdependent and critical components of generation of new scientific knowledge and medical treatments. A current challenge for IACUCs is in evaluating whether benefits to be derived (eg, new knowledge or treatments) justify any unavoidable pain, stress, or injury associated with proposed research protocols, because the former are long-term and at best speculative outcomes, whereas the latter are immediate and tangible for the study animals. Scientific consensus is that research most likely to generate significant new knowledge and medical treatments is that conducted to high scientific, technical, and quality standards and reported with full transparency to facilitate reproducibility. As an alternative to current benefits evaluations included in risk benefit and harm benefit constructs, the authors propose that IACUCs assess the proposed research for scientific quality and alignment of study elements with the study purpose (e.g., Fit for Purpose [FfP]), including justifications for study design components, selection of primary endpoints and technologies, rationale for data and statistical analyses, and research communication plans. Fit for Purpose endpoints are objective, immediate, and impactful as are the potential risks for study animals, and at the same time they are the best predictors for achievement of longer-term benefits. We propose that IACUCs and any revision of The ILAR Guide consider FfP concepts in place of traditional benefits assessment to accelerate the generation of new knowledge and treatments benefiting medical and veterinary patients and the environment through better science and animal welfare rather than to continue to rely on speculative future outcomes.


Assuntos
Comitês de Cuidado Animal , Animais , Estados Unidos , Reprodutibilidade dos Testes , Bem-Estar do Animal
4.
ILAR J ; 62(3): 278-294, 2021 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-36515581

RESUMO

The Institute for Laboratory Animal Research (ILAR) was created within the National Academies of Sciences, Engineering, and Medicine (National Academies) in 1953 when biomedical research using animals was in its infancy in terms of quantity, quality, complexity, sophistication, and care. Over the intervening 69 years, ILAR has witnessed unprecedented growth, followed by unprecedented decline, and then regrowth in usage of specific species and models and an overall shift in experimental burden away from larger to smaller species (ie, mice, fish, and rats). ILAR has contributed much to the evolution of necessary research using animals and animal models for the benefit of humans, animals, and the environment and to the development and implementation of humane principles and standards for care and use of research animals. ILAR has served as a "neutral broker" seeking consensus, solutions, common ground, and pathways forward for all professional constituencies engaged in conduct of animal research. In 2022, ILAR will become the Board on Animal Health Sciences, Conservation, and Research (BAHSCR) within the Division on Earth and Life Studies of the National Academies and the ILAR Journal will pause publication with volume 62. This manuscript recounts the history and accomplishments of ILAR 1953-2022, emphasizing the past 2 decades. The manuscript draws upon ILAR's communications and previously published histories to document ILAR's leaders, reports, publications, conferences, workshops, and roundtables using text, tables, references, and extensive supplemental tables. The authors' intention is to provide the scientific community with a single source document for ILAR, and they apologize for any omissions and errors.


Assuntos
Experimentação Animal , Pesquisa Biomédica , Animais , Humanos , Camundongos , Ratos , Estados Unidos , Animais de Laboratório , Modelos Animais
5.
Toxicol Appl Pharmacol ; 336: 66-74, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29051111

RESUMO

Multiplex biomarker panel assays would enable early de-risking of discovery compound related kidney safety liabilities. The objective of this study was to evaluate the usefulness of the Myriad RBM Human KidneyMAP (Multi-Analyte Profile)® v.1.0 panel to detect experimental nephrotoxicity in Cynomolgus monkeys following a single intravenous administration of cisplatin (2.5mg/kg). Urine samples were collected at baseline on day -2; at approximately 4hr post-dose on day 1; and on days 4, 9, 15 and/or 20. Blood samples were collected at predose on day -2; at 4hr post-dose on day 1; and on days 2, 5, 10 and/or 21. Changes in toxicokinetic and biochemistry parameters in plasma, qualitative/quantitative urinalysis parameters, and urinary kidney safety biomarkers were assessed. Kidney tissues were collected on days 2, 5, 10 and 21 for routine microscopy. Cisplatin-induced tubular alterations were characterized by acute and progressive cortical tubular degeneration/necrosis, regeneration, tubular dilation and proteinaceous cast in the absence of statistically significant changes in traditional plasma biochemistry and urinalysis parameters. When normalized to urinary creatinine, cisplatin-induced significant increases in urinary levels of kidney injury molecule 1 (females on day 4), increases in calbindin D28k (males and females on day 4), decreases in Tamm-Horsfall glycoprotein (males on days 1, 4 and 9), and increases in clusterin (females and males on days 15 and 20, respectively), when compared to concurrent controls. This study revealed the usefulness of the Human KidneyMAP® multiplex panel when measuring changes in urine-based biomarkers to reliably detect cisplatin-induced acute/progressive cortical tubular injury in male and female Cynomolgus monkeys.


Assuntos
Antineoplásicos/toxicidade , Biomarcadores/urina , Cisplatino/toxicidade , Nefropatias/induzido quimicamente , Nefropatias/urina , Túbulos Renais/efeitos dos fármacos , Testes de Toxicidade/métodos , Animais , Biomarcadores/sangue , Nitrogênio da Ureia Sanguínea , Calbindinas/urina , Clusterina/urina , Creatinina/sangue , Feminino , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Humanos , Nefropatias/sangue , Nefropatias/patologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Macaca fascicularis , Masculino , Modelos Animais , Valor Preditivo dos Testes , Fatores Sexuais , Especificidade da Espécie , Fatores de Tempo , Toxicocinética , Urinálise , Uromodulina/urina
6.
ILAR J ; 57(2): 101-108, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-28053064

RESUMO

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.


Assuntos
Experimentação Animal , Descoberta de Drogas , Animais , Humanos , Dor
7.
Lab Anim (NY) ; 44(10): 403-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26398615

RESUMO

The longstanding concept of risk-benefit analysis is an established and familiar practice among animal research programs. It is generally preferred by researchers and statisticians and this term is used throughout the Guide for the Care and Use of Laboratory Animals. However, the term 'harm-benefit analysis' has recently come into use, particularly in the accreditation process for animal research programs. The Association for Assessment and Accreditation of Laboratory Animal Care has incorporated a new interpretation of the eighth edition of the Guide for the Care and Use of Laboratory Animals into their assessment and evaluation process, whereby they may require institutions to conduct a 'harm-benefit analysis'. However, whereas 'risk-benefit analysis' is specifically described in the Guide for the Care and Use of Laboratory Animals, harm-benefit analysis is not mentioned at all. The source of this harm-benefit language appears to be Article 38 of the European Directive 2010/63/EU. Here, the authors present a case for retaining the current language of risk-benefit analyses and not specifically introducing the language of harm-benefit analyses into ethical considerations of animal research activities, including protocol review procedures of Institutional Animal Care and Use Committees.


Assuntos
Criação de Animais Domésticos/normas , Animais de Laboratório , Medição de Risco/normas , Terminologia como Assunto , Animais
8.
Handb Exp Pharmacol ; 229: 3-45, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26091634

RESUMO

Professor Gerhard Zbinden recognized in the 1970s that the standards of the day for testing new candidate drugs in preclinical toxicity studies failed to identify acute pharmacodynamic adverse events that had the potential to harm participants in clinical trials. From his vision emerged the field of safety pharmacology, formally defined in the International Conference on Harmonization (ICH) S7A guidelines as "those studies that investigate the potential undesirable pharmacodynamic effects of a substance on physiological functions in relation to exposure in the therapeutic range and above." Initially, evaluations of small-molecule pharmacodynamic safety utilized efficacy models and were an ancillary responsibility of discovery scientists. However, over time, the relationship of these studies to overall safety was reflected by the regulatory agencies who, in directing the practice of safety pharmacology through guidance documents, prompted transition of responsibility to drug safety departments (e.g., toxicology). Events that have further shaped the field over the past 15 years include the ICH S7B guidance, evolution of molecular technologies leading to identification of new therapeutic targets with uncertain toxicities, introduction of data collection using more sophisticated and refined technologies, and utilization of transgenic animal models probing critical scientific questions regarding novel targets of toxicity. The collapse of the worldwide economy in the latter half of the first decade of the twenty-first century, continuing high rates of compound attrition during clinical development and post-approval and sharply increasing costs of drug development have led to significant strategy changes, contraction of the size of pharmaceutical organizations, and refocusing of therapeutic areas of investigation. With these changes has come movement away from dedicated internal safety pharmacology capability to utilization of capabilities within external contract research organizations. This movement has created the opportunity for the safety pharmacology discipline to come "full circle" and return to the drug discovery arena (target identification through clinical candidate selection) to contribute to the mitigation of the high rate of candidate drug failure through better compound selection decision making. Finally, the changing focus of science and losses in didactic training of scientists in whole animal physiology and pharmacology have revealed a serious gap in the future availability of qualified individuals to apply the principles of safety pharmacology in support of drug discovery and development. This is a significant deficiency that at present is only partially met with academic and professional society programs advancing a minimal level of training. In summary, with the exception that the future availability of suitably trained scientists is a critical need for the field that remains to be effectively addressed, the prospects for the future of safety pharmacology are hopeful and promising, and challenging for those individuals who want to assume this responsibility. What began in the early part of the new millennium as a relatively simple model of testing to assure the safety of Phase I clinical subjects and patients from acute deleterious effects on life-supporting organ systems has grown with experience and time to a science that mobilizes the principles of cellular and molecular biology and attempts to predict acute adverse events and those associated with long-term treatment. These challenges call for scientists with a broad range of in-depth scientific knowledge and an ability to adapt to a dynamic and forever changing industry. Identifying individuals who will serve today and training those who will serve in the future will fall to all of us who are committed to this important field of science.


Assuntos
Avaliação Pré-Clínica de Medicamentos/tendências , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Coleta de Dados , Humanos , Segurança
9.
Environ Health Perspect ; 123(8): 743-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25782181

RESUMO

BACKGROUND: Employers, courts, and the general public judge the credibility of professionals based on credentials such as academic degrees, publications, memberships in professional organizations, board certifications, and professional registrations. However, the relevance and merit of these credentials can be difficult to determine objectively. Board certification can be a reliable indicator of proficiency if the certifying organization demonstrates, through regularly scheduled independent review, that its processes meet established standards and when a certificate holder is required to periodically demonstrate command of a body of knowledge that is essential to current professional practice. OBJECTIVE: We report herein a current Standard of Knowledge in general toxicology compiled from the experience and opinions of 889 certified practicing professional toxicologists. DISCUSSION: An examination is the most commonly used instrument for testing a certification candidate's command of the body of knowledge. However, an examination-based certification is only creditable when the body of knowledge, to which a certification examination tests, is representative of the current knowledge, skills, and capabilities needed to effectively practice at the professional level. Thus, that body of knowledge must be the current "Standard of Knowledge" for the profession, compiled in a transparent fashion from current practitioners of the profession. CONCLUSION: This work was conducted toward ensuring the scientific integrity of the products produced by professional toxicologists.


Assuntos
Certificação , Prática Profissional/normas , Toxicologia/normas
10.
Toxicol Rep ; 1: 1202-1212, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-28962330

RESUMO

BACKGROUND: Ticagrelor is an orally available, direct acting and reversible P2Y12 receptor antagonist approved for treatment of acute coronary syndrome. The objectives of these studies were to (1) evaluate the Ticagrelor 2-year rat carcinogenicity bioassay data; (2) investigate potential mode of action (MOA) and (3) interpret human relevance. METHODS: The following studies were done (1) rat two-year carcinogenicity study in male and female rats, (2) in vitro and in vivo genotoxicity assays, (3) quantitative whole body autoradiography (QWBA; male and female rats), (4) in vitro pharmacological profiling for more than 300 assays, and (5) in vivo ovariectomized rat assay. RESULTS: The carcinogenicity study indicated Ticagrelor increased uterine tumor incidence while decreasing mammary and pituitary tumors/hyperplasia incidences in only high dose female rats. However, this altered tumor incidences were not P2Y12 target related since marketed non-reversible P2Y12 receptor antagonists were not associated with alter tumor incidences. MOA studies determined Ticagrelor exposure in the anterior pituitary and Ticagrelor was (1) non-genotoxic, (2) peripherally-restricted, (3) a dopamine transport (DAT) inhibitor with an IC50 lower than systemic free exposure in the rat carcinogenic study and more than a log higher than the free systemic exposure seen in clinical trials and (4) an inhibitor of estradiol-induced prolactin secretion. DISCUSSION: Similar to Ticagrelor, centrally active dopamine agonists induce the same altered tumor incidence patterns that according to literature do not translate into the clinical setting, with a MOA involving decreased prolactin secretion. The Ticagrelor MOA data and literature suggest that altered dopamine levels in the hypophyseal part of the hypothalamus-hypophyseal axis (by Ticagrelor) will result in similar altered tumor incidences in rat that do not translate into the clinical setting, based on qualitative species differences. In conclusion Ticagrelor increased uterine tumors in the rat carcinogenesis study by a MOA consistent with reduced dopamine inhibition of prolactin, which is not a patient safety risk.

11.
Regul Toxicol Pharmacol ; 66(1): 88-103, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23524271

RESUMO

Evaluation of the safety of new chemicals and pharmaceuticals requires the combination of information from various sources (e.g. in vitro, in silico and in vivo) to provide an assessment of risk to human health and the environment. The authors have identified opportunities to maximize the predictivity of this information to humans while reducing animal use in four key areas; (i) accelerating the uptake of in vitro methods; (ii) incorporating the latest science into safety pharmacology assessments; (iii) optimizing rodent study design in biological development and (iv) consolidating approaches in developmental and reproductive toxicology. Through providing a forum for open discussion of novel proposals, reviewing current research and obtaining expert opinion in each of the four areas, the authors have developed recommendations on good practice and future strategy.


Assuntos
Alternativas aos Testes com Animais/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Toxicologia/métodos , Animais , Humanos , Projetos de Pesquisa , Medição de Risco/métodos , Roedores , Especificidade da Espécie , Pesquisa Translacional Biomédica/métodos
12.
Toxicol Appl Pharmacol ; 268(3): 352-61, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23415679

RESUMO

Current diagnosis of drug-induced kidney injury (DIKI) primarily relies on detection of elevated plasma creatinine (Cr) or blood urea nitrogen (BUN) levels; however, both are indices of overall kidney function and changes are delayed with respect to onset of nephron injury. Our aim was to investigate whether early changes in new urinary DIKI biomarkers predict plasma Cr, BUN, renal hemodynamic and kidney morphological changes associated with kidney injury following a single dose of cisplatin (CDDP) using an integrated platform in rodent. Conscious surgically prepared male Han Wistar rats were given a single intraperitoneal dose of CDDP (15mg/kg). Glomerular filtration rate (GFR), effective renal plasma flow (ERPF), urinalysis, DIKI biomarkers, CDDP pharmacokinetics, blood pressures, heart rate, body temperature and electroencephalogram (EEG) were measured in the same vehicle- or CDDP-treated animals over 72h. Plasma chemistry (including Cr and BUN) and renal tissues were examined at study termination. Cisplatin caused progressive reductions of GFR, ERPF, heart rate and body temperature from day 1 (0-24h). DIKI biomarkers including alpha-glutathione S-transferase (α-GST) significantly increased as early as 6h post-dose, which preceded significant declines of GFR and ERPF (24h), increased plasma Cr and BUN (72h), and associated with renal acute tubular necrosis at 72h post-dose. The present study adds to the current understanding of CDDP action by demonstrating that early increases in urinary excretion of α-GST predict DIKI risk following acute exposure to CDDP in rats, before changes in traditional DIKI markers are evident.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Cisplatino/metabolismo , Cisplatino/toxicidade , Injúria Renal Aguda/patologia , Animais , Nitrogênio da Ureia Sanguínea , Taxa de Filtração Glomerular/efeitos dos fármacos , Taxa de Filtração Glomerular/fisiologia , Glutationa/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Masculino , Distribuição Aleatória , Ratos , Ratos Wistar
13.
Toxicology ; 303: 133-8, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23159986

RESUMO

Alpha 2u-globulin mediated hyaline droplet nephropathy (HDN) is a male rat specific lesion induced when a compound or metabolite binds to alpha 2u-globulin. The objective of this study was to investigate if the newer and more sensitive renal biomarkers would be altered with HDN as well as be able to distinguish between HDN and oxidative stress-induced kidney injury. Rats were dosed orally for 7 days to determine (1) if HDN (induced by 2-propanol or D-limonene) altered the newer renal biomarkers and not BUN or creatinine, (2) if renal biomarkers could distinguish between HDN and oxidative stress-induced kidney injury (induced by potassium bromate), (3) sensitivity of HDN-induced renal biomarker changes relative to D-limonene dose, and (4) reversibility of HDN and renal biomarkers, using vehicle or 300 mg/kg/day D-limonene with 7 days of dosing and necropsies scheduled over the period of Days 8-85. HDN-induced renal biomarker changes in male rats were potentially compound specific: (1) 2-propanol induced mild HDN without increased renal biomarkers, (2) potassium bromate induced moderate HDN with increased clusterin, and (3) D-limonene induced marked HDN with increased αGST, µGST and albumin. Administration of potassium bromate did not result in oxidative stress-induced kidney injury, based on histopathology and renal biomarkers creatinine and BUN. The compound D-limonene induced a dose dependent increase in HDN severity and renal biomarker changes without altering BUN, creatinine or NAG: (1) minimal induction of HDN and no altered biomarkers at 10 mg/kg/day, (2) mild induction of HDN with increased αGST and µGST at 50 mg/kg/day and (3) marked induction of HDN with increased αGST, µGST and albumin at 300 mg/kg/day. HDN induced by D-limonene was reversible, but with a variable renal biomarker pattern over time: Day 8 there was increased αGST, µGST and albumin; on Day 15 increased clusterin, albumin and Kim-1. In summary, HDN altered the newer and more sensitive renal biomarkers in a time and possibly compound dependent manner.


Assuntos
alfa-Globulinas/metabolismo , Hialina/metabolismo , Nefropatias/patologia , Estresse Oxidativo , 1-Propanol/administração & dosagem , 1-Propanol/toxicidade , Animais , Biomarcadores/metabolismo , Nitrogênio da Ureia Sanguínea , Bromatos/toxicidade , Creatinina/metabolismo , Cicloexenos/administração & dosagem , Cicloexenos/toxicidade , Relação Dose-Resposta a Droga , Feminino , Nefropatias/diagnóstico , Limoneno , Masculino , Ratos , Ratos Wistar , Índice de Gravidade de Doença , Terpenos/administração & dosagem , Terpenos/toxicidade , Fatores de Tempo
14.
Basic Clin Pharmacol Toxicol ; 111(6): 411-6, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22742711

RESUMO

Centrally acting dopamine agonists (e.g. bromocriptine) and dopamine transport inhibitors (e.g. GBR12909) are known to inhibit oestradiol-induced prolactin release. The capacity of peripherally restricted compounds to do likewise, however, is unknown. Here, the effects of the peripherally restricted dopamine receptor agonist carmoxirole on oestradiol-induced prolactin release were investigated. Dual-cannulated ovariectomized rats were used, so that a robust, reproducible response to exogenous oestrogen could be induced and sequential blood samples were taken with minimal stress. Carmoxirole (15 mg/kg) inhibited oestradiol-induced prolactin release, similar to bromocriptine and GBR12909. However, carmoxirole also induced a rapid, transient, oestradiol-independent release of prolactin. These data show that peripherally restricted dopamine receptor agonists are sufficient to inhibit oestradiol-induced prolactin release. Like centrally acting compounds, they may therefore be expected to affect the incidence of prolactin-dependent tumours in rat carcinogenesis studies without inducing central-mediated side effects.


Assuntos
Estradiol/metabolismo , Antagonistas de Estrogênios/farmacologia , Indóis/farmacologia , Prolactina/metabolismo , Piridinas/farmacologia , Animais , Bromocriptina/farmacologia , Agonistas de Dopamina/farmacologia , Feminino , Ovariectomia , Piperazinas/farmacologia , Prolactina/sangue , Ratos , Ratos Wistar , Estimulação Química
15.
Environ Toxicol ; 27(10): 573-82, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21384489

RESUMO

The conservation of common physiological systems across vertebrate classes suggests the potential for certain pharmaceuticals, which have been detected in surface waters, to produce biological effects in nontarget vertebrates such as fish. However, previous studies assessing the effects of such compounds in fish have not taken into account the potential for metabolism and elimination. This study aimed to assess if propranolol, a ß-adrenergic receptor antagonist or ß-blocker, could modulate EROD activity (indicative of CYP1A activity) in rainbow trout (Oncorhynchus mykiss) gills and liver. For this, an in vivo time course exposure with 1 mg/L was conducted. Additionally, using measured in vivo plasma concentrations, an in vitro exposure at human therapeutic levels was undertaken. This allowed comparison of in vitro and in vivo rates of EROD activity, thus investigating the applicability of cell preparations as surrogates for whole animal enzyme activity analysis. In vitro exposure of suspended liver and gill cells at concentrations similar to in vivo levels resulted in EROD activity in both tissues, but with significantly higher rates (up to six times in vivo levels). These results show that propranolol exposure elevated EROD activity in the liver and gill of rainbow trout, and that this is demonstrable both in vivo (albeit nonsignificantly in the liver) and in vitro, thus supporting the use of the latter as a surrogate of the former. These data also provide an insight into the potential role of the gill as a site of metabolism of pharmaceuticals in trout, suggesting that propranolol (and feasibly other pharmaceuticals) may undergo "first pass" metabolism in this organ.


Assuntos
Citocromo P-450 CYP1A1/metabolismo , Brânquias/enzimologia , Fígado/enzimologia , Oncorhynchus mykiss/metabolismo , Propranolol/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Células Cultivadas , Citocromo P-450 CYP1A1/efeitos dos fármacos , Feminino , Brânquias/citologia , Brânquias/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Fígado/efeitos dos fármacos , Propranolol/sangue
16.
J Pharmacol Toxicol Methods ; 64(1): 7-15, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21689769

RESUMO

In recognition of the tenth anniversary of the Safety Pharmacology Society (SPS), this review summarizes the significant events of the past 10years that have led to the birth, growth and evolution the SPS and presents a roadmap to the immediate-, intermediate- and long-term future of the SPS. The review discusses (i) the rationale for an optimal non-clinical Safety Pharmacology testing, (ii) the evolution of Safety Pharmacology over the last decade, (iii) its impact on drug discovery and development, (iv) the merits of adopting an integrated risk assessment approach, (v) the translation of non-clinical findings to humans and finally (vi) the future challenges and opportunities facing this discipline. Such challenges include the emergence of new molecular targets and new approaches to treat diseases, the rapid development of science and technologies, the growing regulatory concerns and associated number of guidance documents, and the need to train and educate the next generation of safety pharmacologist.


Assuntos
Avaliação Pré-Clínica de Medicamentos/tendências , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Animais , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Medição de Risco/métodos , Sociedades Farmacêuticas , Testes de Toxicidade/tendências
17.
Toxicol Pathol ; 39(4): 716-44, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21666103

RESUMO

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.


Assuntos
Testes de Carcinogenicidade/métodos , Carcinógenos/toxicidade , Testes de Mutagenicidade/métodos , Animais , Testes de Carcinogenicidade/normas , Carcinógenos/normas , Bases de Dados Factuais , Árvores de Decisões , Modelos Animais de Doenças , Estudos de Avaliação como Assunto , Feminino , Guias como Assunto , Humanos , Imunossupressores , Masculino , Camundongos , Camundongos Transgênicos , Testes de Mutagenicidade/normas , Neoplasias/induzido quimicamente , Ratos , Ratos Endogâmicos F344 , Fatores de Risco , Estatística como Assunto , Testes de Toxicidade Crônica
19.
Environ Toxicol Chem ; 29(12): 2831-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20836071

RESUMO

Two studies to examine the effect of waterborne clofibric acid (CA) on growth-rate and condition of rainbow trout were conducted using accepted regulatory tests (Organisation for Economic Co-operation and Development [OECD] 215). The first study (in 2005) showed significant reductions after 21 d of exposure (21-d growth lowest-observed-effect concentration [LOEC] = 0.1 µg/L, 21-d condition LOEC = 0.1 µg/L) that continued to 28 d. Growth rate was reduced by approximately 50% (from 5.27 to 2.67% per day), while the condition of the fish reduced in a concentration-dependant manner. Additionally, in a concentration-dependent manner, significant changes in relative liver size were observed, such that increasing concentrations of CA resulted in smaller livers after 28-d exposure. A no-observed-effect concentration (NOEC) was not achieved in the 2005 study. An expanded second study (in 2006) that included a robust bridge to the 2005 study, with four replicate tanks of eight individual fish per concentration, did not repeat the 2005 findings. In the 2006 study, no significant effect on growth rate, condition, or liver biometry was observed after 21 or 28 d (28-d growth NOEC = 10 µg/L, 28-d condition NOEC = 10 µg/L), contrary to the 2005 findings. We do not dismiss either of these findings and suggest both are relevant and stand for comparison. However, the larger 2006 study carries more statistical power and multiple-tank replication, so probably produced the more robust findings. Despite sufficient statistical power in each study, interpretation of these and similar studies should be conducted with caution, because much significance is placed on the role of limited numbers of individual and tank replicates and the influence of control animals.


Assuntos
Ácido Clofíbrico/toxicidade , Oncorhynchus mykiss/crescimento & desenvolvimento , Testes de Toxicidade Crônica/métodos , Poluentes Químicos da Água/toxicidade , Animais , Anticolesterolemiantes/sangue , Anticolesterolemiantes/toxicidade , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Ácido Clofíbrico/sangue , Relação Dose-Resposta a Droga , Feminino , Oncorhynchus mykiss/sangue , Tamanho do Órgão/efeitos dos fármacos , Poluentes Químicos da Água/sangue
20.
Integr Environ Assess Manag ; 6(1): 38-51, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19558206

RESUMO

Over recent years, human pharmaceuticals have been detected in the aquatic environment. This, combined with the fact that many are (by design) biologically active compounds, has raised concern about potential impacts in wildlife species. This concern was realized with two high-profile cases of unforeseen environmental impact (i.e., estrogens and diclofenac), which have led to a flurry of work addressing how best to predict such effects in the future. One area in which considerable research effort has been made, partially in response to regulatory requirements, has been on the potential use of preclinical and clinical pharmacological and toxicological data (generated during drug development from nonhuman mammals and humans) to predict possible effects in nontarget, environmentally relevant species: so-called read across. This approach is strengthened by the fact that many physiological systems are conserved between mammals and certain environmentally relevant species. Consequently, knowledge of how a pharmaceutical works (the "mode-of-action," or MoA) in nonclinical species and humans could assist in the selection of appropriate test species, study designs, and endpoints, in an approach referred to as "intelligent testing." Here we outline the data available from the human drug development process and suggest how this might be used to design a testing strategy best suited to the specific characteristics of the drug in question. In addition, we review published data that support this type of approach, discuss the potential pitfalls associated with read across, and identify knowledge gaps that require filling to ensure accuracy in the extrapolation of data from preclinical and clinical studies, for use in the environmental risk assessment of human pharmaceuticals.


Assuntos
Descoberta de Drogas , Monitoramento Ambiental/métodos , Medição de Risco/métodos , Humanos , Preparações Farmacêuticas/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...