Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Forensic Toxicol ; 42(2): 125-141, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38102417

RESUMEN

PURPOSE: There is very little information about the toxicological and pathological effects of synthetic cannabinoids, which have cannabis-like properties. This study was carried out to histopathologically, hematologically, and biochemically determine the toxic effects of acute and subacute exposure to a novel synthetic cannabinoid 1-(4-cyanobutyl)-N-(2-phenylpropan-2-yl)indazole-3-carboxamide in internal organs of adult male rats. METHODS: The cannabinoid was injected intraperitoneally at three doses (0.5, 1.0, and 2.0 mg/kg, body weight). The cannabinoid was administered to acute groups for 2 days and to subacute groups for 14 days. Observations were made for 14 days and various changes such as mortality, injury, and illness were recorded daily. Hematological and biochemical changes were evaluated and histopathological analyses in lung, liver, and kidney tissues were also performed. RESULTS: No mortality was observed. It was observed that there were fluctuations in hematological and serum biochemical parameters. Among the oxidative stress parameters, significant decreases in superoxide dismutase, catalase levels and significant increases in lipid peroxidation levels were determined. Serious pathological changes such as necrosis, vacuolation, congestion, and fibrosis were observed in the internal organs in a dose-dependent and time-dependent manner. It was also found that the synthetic cannabinoid triggered apoptosis in the organs. The results demonstrated that the most affected organ by the cannabinoid was the kidney. CONCLUSION: This study showed for the first time that CUMYL-4CN-BINACA adversely affects healthy male albino rats. It can be estimated that the abuse of the cannabinoid may harm human health in the same way.


Asunto(s)
Cannabinoides , Riñón , Peroxidación de Lípido , Hígado , Pulmón , Estrés Oxidativo , Animales , Masculino , Riñón/efectos de los fármacos , Riñón/patología , Riñón/metabolismo , Cannabinoides/toxicidad , Ratas , Hígado/efectos de los fármacos , Hígado/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Indazoles/toxicidad , Catalasa/metabolismo , Relación Dosis-Respuesta a Droga , Pruebas de Toxicidad Subaguda , Ratas Wistar , Pruebas de Toxicidad Aguda
2.
Front Endocrinol (Lausanne) ; 13: 951800, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36060935

RESUMEN

Tea (Camellia sinensis) has several reported health benefits, including that on bone health attributed to catechins of which the most abundant is epigallocatechin-3-gallate (EGCG). However, several preclinical and clinical studies raise safety concerns about EGCG in tea extract causing acute liver failure. Tea also contains kaempferol, albeit scanty, and it has hepatoprotective and osteogenic effects. Here, we utilized a novel extraction procedure of acid hydrolysis to enhance the osteogenic effect of tea extract while reducing its hepatotoxicity. The resultant extract (USKECSE) has a ~40-fold increase in kaempferol and a 2.5-fold reduction in EGCG content compared with the hydroethanolic extract (USCSE). In a female Sprague Dawley (SD) rat femur osteotomy model, USKECSE (100 mg/kg) but not USCSE promoted bone regeneration. In a rat postmenopausal osteoporosis model induced by bilateral ovariectomy (OVX), USKECSE through an osteogenic mechanism maintained bone mass, strength, and microarchitecture to the levels of ovary-intact rats with no hepatotoxic effect. After a single oral dose (100 mg/kg) of USKECSE to adult rats, kaempferol was detectable for 48 hours, suggesting its significant absorption and distribution in plasma. Peak kaempferol concentration in plasma (Cmax) was 483 ng/ml (2 µM), and at this concentration, kaempferol induces osteoblast differentiation. USKECSE had no genotoxicity, and its safety index assessed by preclinical toxicity studies, including safety pharmacology, was >20-fold. Taken together, we report a novel extraction process that enhanced the osteogenicity and concomitantly reduced hepatotoxicity of tea extract with significant kaempferol bioavailability and a favorable systemic safety profile. Based on these data, we propose assessing the USKECSE effect for postmenopausal osteoporosis treatment.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Osteoporosis Posmenopáusica , Osteoporosis , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Femenino , Humanos , Quempferoles/farmacología , Quempferoles/uso terapéutico , Osteoporosis/tratamiento farmacológico , Osteoporosis Posmenopáusica/tratamiento farmacológico , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Ratas , Ratas Sprague-Dawley ,
3.
Int J Toxicol ; 41(2): 143-162, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35230174

RESUMEN

Determining the adverse nature of findings from nonclinical safety studies often poses a challenge for the key stakeholders responsible for interpreting the results of definitive toxicity studies in support of pharmaceutical product development. Although there are instances in which responses to treatment clearly indicate intolerability or tissue injury associated with dysfunction; in practice, more often there is uncertainty in characterizing an effect of drug treatment as adverse or not. This is due to the inherent variability in responses of biological test systems to toxicological insults, leaving the ultimate analyses of adversity to individual interpretation and subjectivity. This article is a follow-up to the workshop entitled, "Adverse or Not Adverse?: Thinking process behind adversity determination during nonclinical drug development," conducted at the 58th Annual Meeting of the Society of Toxicology, March 2019 in Baltimore, MD. In this paper, we further discuss and incorporate the perspectives of authors representing different roles, such as Study Director, Study Pathologist, Pharmacology/Toxicology Reviewer (U.S. Food and Drug Administration), and Sponsor in the determination and use of adversity. We also present a practical stepwise approach as an aid in this assessment, and further apply these principles to discuss 10 case studies with different therapeutic modalities and unique challenges.


Asunto(s)
Desarrollo de Medicamentos , Evaluación Preclínica de Medicamentos/métodos , Nivel sin Efectos Adversos Observados , Preparaciones Farmacéuticas , Medición de Riesgo/métodos , Estados Unidos , United States Food and Drug Administration
4.
Regul Toxicol Pharmacol ; 123: 104939, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33961952

RESUMEN

In standard general toxicology studies in two species to support clinical development, cyclocreatine, a creatine analog for the treatment of creatine transporter deficiency, caused deaths, convulsions, and/or multi-organ pathology. The potential translatability of these findings to patients was evaluated by comparing toxicity of cyclocreatine in wild-type mice to creatine transporter-deficient mice, a model of the human disease. A biodistribution study indicated greater accumulation of cyclocreatine in the brains of wild-type mice, consistent with its ability to be transported by the creatine transporter. Subsequent toxicology studies confirmed greater sensitivity of wild-type mice to cyclocreatine-induced toxicity. Exposure at the no observed adverse effect level in creatine transporter-deficient (554 µg*hr/ml) mice exceeded exposure at the maximum tolerated dose in wild-type (248 µg*hr/ml) mice. When dosed at 300 mg/kg/day for 3 months, cyclocreatine-related mortality, convulsions, and multi-organ pathology were observed in wild-type mice whereas there were no adverse findings in creatine transporter-deficient mice. Brain vacuolation was common to both strains. Although transporter-deficient mice appeared to be more sensitive, the finding had no functional correlates in this strain. The results highlight the importance of considering models of disease for toxicology in cases where they may be relevant to assessing safety in the intended patient population.


Asunto(s)
Antineoplásicos/toxicidad , Creatinina/análogos & derivados , Modelos Animales de Enfermedad , Animales , Encéfalo , Encefalopatías Metabólicas Innatas , Creatina/deficiencia , Creatinina/toxicidad , Humanos , Proteínas de Transporte de Membrana , Discapacidad Intelectual Ligada al Cromosoma X , Ratones , Nivel sin Efectos Adversos Observados , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/deficiencia , Convulsiones , Distribución Tisular
5.
Front Pharmacol ; 12: 578796, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33867974

RESUMEN

Chinese materia medica (CMM) has been applied for the prevention and treatment of diseases for thousands of years. However, arrhythmia, myocardial ischemia, heart failure, and other cardiac adverse reactions during CMM application were gradually reported. CMM-induced cardiotoxicity has aroused widespread attention. Our review aimed to summarize the risk compounds, preclinical toxicity evaluation, and potential mechanisms of CMM-induced cardiotoxicity. All relevant articles published on the PubMed, Embase, and China National Knowledge Infrastructure (CNKI) databases for the latest twenty years were searched and manually extracted. The risk substances of CMM-induced cardiotoxicity are relatively complex. A single CMM usually contains various risk compounds, and the same risk substance may exist in various CMM. The active and risk substances in CMM may be transformed into each other under different conditions, such as drug dosage, medication methods, and body status. Generally, the risk compounds of CMM-induced cardiotoxicity can be classified into alkaloids, terpenoids, steroids, heavy metals, organic acids, toxic proteins, and peptides. Traditional evaluation methods of chemical drug-induced cardiotoxicity primarily include cardiac function monitoring, endomyocardial biopsy, myocardial zymogram, and biomarker determination. In the preclinical stage, CMM-induced cardiotoxicity should be systematically evaluated at the overall, tissue, cellular, and molecular levels, including cardiac function, histopathology, cytology, myocardial zymogram, and biomarkers. Thanks to the development of systematic biology, the higher specificity and sensitivity of biomarkers, such as genes, proteins, and metabolic small molecules, are gradually applied for evaluating CMM-induced cardiotoxicity. Previous studies on the mechanisms of CMM-induced cardiotoxicity focused on a single drug, monomer or components of CMM. The interaction among ion homeostasis (sodium, potassium, and calcium ions), oxidative damage, mitochondrial injury, apoptosis and autophagy, and metabolic disturbance is involved in CMM-induced cardiotoxicity. Clarification on the risk compounds, preclinical toxicity evaluation, and potential mechanisms of CMM-induced cardiotoxicity must be beneficial to guide new CMM development and post-marketed CMM reevaluation.

6.
Int J Toxicol ; 40(1): 4-14, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33131343

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) is a potent neuroprotective biologic in Parkinson's disease models. Adeno-associated viral vector serotype 2 (AAV2)-human GDNF safety was assessed in rats treated with a single intracerebral dose of vehicle, 6.8 × 108, 6.8 × 109, or 5.2 × 1010 vector genomes (vg)/dose followed by interim sacrifices on day 7, 31, 90, and 376. There were no treatment-related effects observed on food consumption, body weight, hematology, clinical chemistry, coagulation parameters, neurobehavioral parameters, organ weights, or serum GDNF and anti-GDNF antibody levels. Increased serum anti-AAV2 neutralizing antibody titers were observed in the 5.2 × 1010 vg/dose group. Histopathological lesions were observed at the injection site in the 6.8 × 109 vg/dose (day 7) and 5.2 × 1010 vg/dose groups (days 7 and 31) and consisted of gliosis, mononuclear perivascular cuffing, intranuclear inclusion bodies, and/or apoptosis on day 7 and mononuclear perivascular cuffing on day 31. GDNF immunostaining was observed in the injection site in all dose groups through day 376 indicating no detectable impacts of anti-AAV2 neutralizing antibody. There was no evidence of increased expression of calcitonin gene-related peptide or Swann cell hyperplasia in the cervical and lumbar spinal cord or medulla oblongata at the 5.2 × 1010 vg/dose level indicating lack of hyperplastic effects. In conclusion, no systemic toxicity was observed, and the local toxicity observed at the injection site appeared to be reversible demonstrating a promising safety profile of intracerebral AAV2-GDNF delivery. Furthermore, an intracerebral dose of 6.8 × 108 AAV2-GDNF vg/dose was considered to be a no observed adverse effect level in rats.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/administración & dosificación , Factor Neurotrófico Derivado de la Línea Celular Glial/toxicidad , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/toxicidad , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
7.
Ther Innov Regul Sci ; 55(2): 309-323, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32996106

RESUMEN

Pharmaceutical residues can harm the environment. Therefore any Marketing Authorisation Application (MAA) for a medicinal product for human use shall be accompanied by an "indication of any potential risks presented by the medicinal product for the environment" according to Article 8(3)(g) of the Directive 2001/83/EC. Since 2006 a guideline document from the European Medicines Agency (EMA, formerly EMEA) is available for this task, which is now called the "Environmental Risk Assessment" (ERA). Recently the EMA released a draft revision of an updated ERA guideline. The present paper provides a summary of the intended innovation. A number of options have been discontinued, but some of them cannot be easily recognised, even though they are potentially affecting the MAA and its costs in a relevant way. A new specifically tailored experimental testing course for antibiotics and a secondary poisoning assessment is introduced. The selection of required study types is altered and the environmental fate evaluation is adapted to the scientific progress. In the recent situation it is suggested that marketing authorisation applicants may reconsider the conduct of water/sediment studies for substances not requiring these according to the guideline revision. The new tailored testing approach for antibiotics avoids any vertebrate use, because it demands no fish study, therefore it may be applied henceforth. If the new prescriptions are not yet in force at the point in time of the MAA submission, appropriate waivers should be stated in the ERA. No further HPLC-study types and acute earthworm toxicity tests should be performed, since the guideline update draft does not accept them. In preclinical toxicity and pharmacokinetic studies, additional data gathering on metabolites should be considered in order to avoid unnecessary leaflet warnings. The cost for ERA may change significantly, with a tendency to increase for generic pharmaceutical MAA, but with a possible reduction for substances of low environmental concern. Earlier consideration of ERA in the drug development process is recommended.


Asunto(s)
Medición de Riesgo , Humanos
8.
Regul Toxicol Pharmacol ; 116: 104716, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32619635

RESUMEN

Pancreatic cancer is a leading cause of cancer-related deaths in the U.S. Ninety percent of patients with stage IV pancreatic cancer die within one year of diagnosis due to complications of metastasis. A metastatic potential of cancer cells has been shown to be closely associated with formation of perinucleolar compartment (PNC). Metarrestin, a first-in-class PNC inhibitor, was evaluated for its toxicity, toxicokinetics, and safety pharmacology in beagle dogs following every other day oral (capsule) administration for 28 days to support its introduction into clinical trials. The study consisted of four dose groups: vehicle; 0.25, 0.75 and 1.50 mg/kg/dose. Metarrestin reached its maximum concentration in blood at 3 h (overall median Tmax) across all doses with a mean t1/2 over 168 h of 55.5 h. Dose dependent increase in systemic exposure (Cmax and AUClast) with no sex difference was observed on days 1 and 27. Metarrestin accumulated from Day 1 to Day 27 at all dose levels and in both sexes by an overall factor of about 2.34. No mortality occurred during the dosing period; however, treatment-related clinical signs of toxicity consisting of hypoactivity, shaking/shivering, thinness, irritability, salivation, abnormal gait, tremors, ataxia and intermittent seizure-like activity were seen in both sexes at mid and high dose groups. Treatment-related effects on body weight and food consumption were seen at the mid and high dose levels. Safety pharmacology study showed no treatment-related effects on blood pressure, heart rate, corrected QT, PR, RR, or QRS intervals, or respiratory function parameters (respiratory rate, tidal volume, minute volume). There were no histopathological changes observed, with the exception of transient thymic atrophy which was considered to be non-adverse. Based primarily on clinical signs of toxicity, the No Observed Adverse Effect Level (NOAEL) in dogs was considered to be 0.25 mg/kg metarrestin after every other day dosing for 28 days with a mean of male and female Cmax = 82.5 ng/mL and AUClast = 2521 h*ng/mL, on Day 27.


Asunto(s)
Antineoplásicos , Pirimidinas , Pirroles , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Perros , Evaluación Preclínica de Medicamentos , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/fisiología , Femenino , Células HEK293 , Humanos , Masculino , Nivel sin Efectos Adversos Observados , Neoplasias Pancreáticas/tratamiento farmacológico , Pirimidinas/administración & dosificación , Pirimidinas/farmacocinética , Pirimidinas/toxicidad , Pirroles/administración & dosificación , Pirroles/farmacocinética , Pirroles/toxicidad
9.
Clin Ther ; 40(11): 1813-1822, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30458928

RESUMEN

PURPOSE: Advances in methods designed to evaluate preclinical toxicity have not kept up with progress in regenerative medicine. Preclinical toxicity studies of regenerative therapies must be designed logically and should be flexible to accurately reflect toxicity of products under development. The purpose of this review is to discuss requirements of preclinical toxicity studies of this type developed in Japan. METHODS: We conducted MEDLINE and PubMed literature searches to identify recent reports relevant to regenerative medicine. Information regarding approved drugs and public announcements, including existing guidelines and guidance in Japan, was collected from the website of Japan's Ministry of Health, Labor and Welfare (https://www.mhlw.go.jp/index.html) and the Pharmaceuticals and Medical Devices Agency (https://www.pmda.go.jp/). FINDINGS: Four cell therapy products have been developed and approved in Japan so far. The principal preclinical toxicity data submitted to regulatory authorities in the Pharmaceuticals and Medical Devices Agency in Japan are summarized here. The potential for tumor formation, a major concern in such clinical applications, is assessed in 3 ways: tumor-forming capacity of the original cell, quantitation of residual pluripotent stem cells in the product, and the possibility that a tumor will form at the product's engraftment site. Although gene therapy and oncolytic virus products are under development, these types of products are not yet approved in Japan. Guidelines relevant to the development of these products are now being created based on existing guidelines and considerations established by the International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use. IMPLICATIONS: Because of cell tropism and heterologous immunity, animal species or strains useful for preclinical studies of regenerative therapies are often restricted. Nonetheless, preclinical toxicity studies must be designed to predict results relevant to humans.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Terapia Genética/efectos adversos , Medicina Regenerativa/métodos , Animales , Humanos , Japón
10.
Food Chem Toxicol ; 121: 467-471, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30248479

RESUMEN

Certified LabDiet® 5K96 Advanced Protocol™ Verified Casein Diet 10 IF (5K96) is a commercial diet low in soy isoflavones developed for use in developmental and reproductive toxicity (DART) studies, especially those designed to detect endocrine disruptors. The objective of this study was to determine the incidences and severities of 5K96-associated renal lesions in control F0 and F1 cohorts of rats fed the 5K96 diet. Kidneys from control animals of four DART studies involving Sprague-Dawley rats fed the 5K96 diet, were evaluated microscopically. Mineralization and basophilic tubules were present in high incidence/severity in males and females compared to historical controls fed conventional diets. F1 cohorts were affected to a far greater degree than F0 cohorts, and females were affected more than males. Consideration of target tissue and mode of action should be given before automatically incorporating the 5K96 diet into DART study designs, and caution should be exercised when identifying and interpreting renal toxicity in the F1 cohorts of such studies.


Asunto(s)
Envejecimiento , Alimentación Animal/análisis , Glycine max/química , Isoflavonas/farmacología , Enfermedades Renales/etiología , Animales , Caseínas , Dieta/veterinaria , Femenino , Isoflavonas/química , Ciencia de los Animales de Laboratorio , Masculino , Embarazo , Fenómenos Fisiologicos de la Nutrición Prenatal , Ratas
11.
Toxicol Pathol ; 45(6): 764-773, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28891435

RESUMEN

A 90-day study in Göttingen minipigs was conducted to test the local tolerability and systemic toxicity of ND0612, a novel aqueous solution of carbidopa (CD)/levodopa (LD) intended for the treatment of Parkinson's disease by continuous subcutaneous administration using a discrete infusion pump. To evaluate tissue site reactions, we used a unique study design involving multiple infusion sites to evaluate the effect of dose per site (270/63, 360/45, and 360/84 mg LD/CD), volume of infusion per site (4.5 and 6 ml per site), formulation concentration (60/14 and 60/7.5 mg/ml LD/CD), daily rate of infusion per site (240 µl/hr for16 hr and 80 µl/hr for 8 hr, 320 µl/hr for 16 hr and 100 µl/hr for 8 hr, or 750 µl/hr for 8 hr), frequency (once every 5, 10, 15, or 20 days), and number of infusions (4, 6, or 9) to the same infusion site. No systemic adverse effects were observed. Histopathological changes at infusion sites started with localized minimal necrosis and acute inflammation that progressed to subacute and chronic inflammatory and reparative changes with evidence of progressive recovery following the final infusion. None of the infusion site effects were judged to be adverse, and clinical exposures to ND0612 are not expected to result in adverse responses.


Asunto(s)
Carbidopa/toxicidad , Agonistas de Dopamina/toxicidad , Tolerancia a Medicamentos , Reacción en el Punto de Inyección/etiología , Levodopa/toxicidad , Animales , Carbidopa/administración & dosificación , Carbidopa/sangre , Agonistas de Dopamina/administración & dosificación , Agonistas de Dopamina/sangre , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Femenino , Infusiones Subcutáneas , Reacción en el Punto de Inyección/patología , Levodopa/administración & dosificación , Levodopa/sangre , Masculino , Necrosis , Porcinos , Porcinos Enanos , Pruebas de Toxicidad Crónica
12.
Toxicol Pathol ; 45(4): 472-480, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28443458

RESUMEN

Infusion site reactions are common following subcutaneous infusion of drugs. Such reactions can lead to discontinuation of the treatment. Therefore, assessment of such reactions is essential during preclinical safety studies, and magnetic resonance imaging (MRI) can assist in evaluation. Here, in vivo and ex vivo MRI evaluations were used in addition to classical histopathology to assess the infusion site reaction to ND0701, a novel formulation of apomorphine base developed for the treatment of Parkinson's disease, in comparison to the commercial apomorphine hydrochloride (HCl) formulation. Both formulations, each at two concentrations, were continuously administered subcutaneously for 20 hr to each of 3 male and 3 female domestic pigs. Based on MRI evaluations, there was a gradual decrease in the volume of the subcutaneous lesions over 4 weeks, with smaller lesions and quicker resolution with ND0701 at concentrations 2.5- to 5-fold higher when compared to the commercial apomorphine HCl formulation. Histopathological evaluation of ND0701 revealed only minimal inflammation at the sites of infusion, whereas the commercial apomorphine HCl caused persistent inflammatory reactions and necrosis. This study provides support to the use of MRI in preclinical testing of subcutaneous drugs when evaluating local site reactions.


Asunto(s)
Antiparkinsonianos/efectos adversos , Apomorfina/efectos adversos , Reacción en el Punto de Inyección/diagnóstico por imagen , Inyecciones Subcutáneas/efectos adversos , Imagen por Resonancia Magnética , Animales , Antiparkinsonianos/administración & dosificación , Apomorfina/administración & dosificación , Biomarcadores/sangre , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Masculino , Enfermedad de Parkinson/tratamiento farmacológico , Porcinos
13.
Regul Toxicol Pharmacol ; 80: 195-209, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27177822

RESUMEN

PURPOSE: Extensive acute and subacute toxicities studies are required to evaluate the toxicological profile of the novel cardiac perfusion imaging tracer (123)I-CMICE-013 to support applications for clinical trials. METHODS: Sprague-Dawley rats and Gottingen minipigs received injections of non-radioactive 127I-CMICE-013 at two dosage levels of 1 and 5 µg/kg, and vehicle buffer as control. In the acute toxicity studies, each animal was injected on two occasions 24 h apart and then underwent a 14-day recovery period; in the subacute study, animals received daily injections for 14 days continuously. The health status and mortality of test animals were monitored daily and body weight, food consumption, physiological and biochemical parameters were measured at various time points during the study. Animals were euthanized at the end of the studies and dissected for pathologic examination of organs and tissues. RESULTS: The acute and subacute administrations of injections of the non-radioactive CMICE-013 in rats and minipigs were well tolerated. Little to no dosing-related adverse effects were observed in animal body and organ weights, hematology, coagulation, clinical chemistry, urinalysis, ophthalmoscopy, electrocardiograms, heart rates, blood pressure, macroscopic and microscopic examination of the preserved animal tissues including the brain. CONCLUSION: The lack of adverse effects from acute and subacute dosing suggest that the CMICE-013 injection solution has a reasonable safety margin within the designed concentration range to be utilized in imaging applications. The dosage level of 5 µg/kg was considered the no adverse effect level for both rats and minipigs based on our acute and subacute studies.


Asunto(s)
Cromonas/toxicidad , Compuestos Heterocíclicos de 4 o más Anillos/toxicidad , Imagen de Perfusión Miocárdica/efectos adversos , Radiofármacos/toxicidad , Pruebas de Toxicidad Aguda/métodos , Pruebas de Toxicidad Subaguda/métodos , Animales , Cromonas/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Inyecciones Intravenosas , Masculino , Imagen de Perfusión Miocárdica/métodos , Nivel sin Efectos Adversos Observados , Valor Predictivo de las Pruebas , Radiofármacos/administración & dosificación , Ratas Sprague-Dawley , Porcinos , Porcinos Enanos , Factores de Tiempo
14.
J Ethnopharmacol ; 174: 293-8, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26297844

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Limonium brasiliense (Boiss.) Kuntze, Plumbaginaceae, popularly known as baicuru, has been used in folk medicine to treat menstrual cramps and to regulate menstrual periods. However, little is known about its safety. This study evaluated the safety through in vivo tests of the acute, long-term, and liver toxicity, and the mutagenicity of the crude extract (CE) from rhizomes of L. brasiliense. MATERIALS AND METHODS: The acute toxicity was assessed in Swiss mice, and the chronic toxicity in Wistar rats. Male and female mice received the CE orally in single doses of 1.0, 2.0, 3.0, 4.0, or 5.0 g/kg. Clinical changes and mortality rate were used as parameters to assess the acute toxicity. In the long-term evaluation, male and female Wistar rats were treated orally with daily doses of the CE (50, 100, or 200 mg/kg) for 90 days. Assessments of weight, behavior and food intake, urinalysis, biochemical and hematological analyses, as well as macro- and microscopic observations of several organs were performed. The redox state of the liver was evaluated as a means of investigating the liver toxicity, and the micronucleus test to assess mutagenicity was also performed. RESULTS: Evaluation of acute toxicity indicated no apparent clinical change in the animals; the LD50 was 4.8 g/kg. Evaluation after 90 days administration showed that the CE, even in higher doses than are considered therapeutic, appeared to be safe. The micronucleus test demonstrated a low mutagenic potential for the CE. CONCLUSION: Our results showed that treatment with the CE from L. brasiliense caused low or no toxicity, as assessed using these doses and evaluation methods.


Asunto(s)
Extractos Vegetales/toxicidad , Plumbaginaceae , Rizoma/toxicidad , Pruebas de Toxicidad Aguda/métodos , Pruebas de Toxicidad Crónica/métodos , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Femenino , Masculino , Ratones , Extractos Vegetales/aislamiento & purificación , Ratas , Ratas Wistar
15.
Food Chem Toxicol ; 73: 148-56, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25139121

RESUMEN

LASSBio-596, 2-[4-(1,4-tiazinan-4-ylsulfonyl) phenylcarbamoyl] benzoic acid, is an achiral compound containing a subunit carboxylic amide, was capable of preventing induced mechanical and morphological changes in the lungs that commonly caused the onset of asthma. Previous studies to determine the acute toxicity of oral LASSBio-596 at dose of 2000mg/kg caused no deaths in any of the tested animals. To further evaluate the safety of LASSBio-596, in vitro and in vivo tests were carried out. Regarding to in vitro test were used renal, hepatic, pulmonary, cardiac, neurologic and intestinal cell lines. They were evaluated using neutral red (NR) and [3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] (MTT) assays. Micronuclei also was performed. Concerning to in vivo was performed subchronic on Wistar rats at doses of 10, 50, and 250mg/kg and zebrafish test. The in vitro tests results showed the safety of LASSBio-596. However, subchronic toxicity study results revealed changes in the blood parameters of amylase, alanine aminotransferase (ALT), aspartate aminotransferase (AST), glucose and creatine kinase (CK) which is used for cardiotoxicity evaluation, although, did not identify any histopathological alterations. However, zebrafish test demonstrated cardiac damage. It was impossible to estimate the no-observed-adverse-effect-levels and lowest observed-adverse-effect level due to the presence of cardiotoxicity in all tested doses.


Asunto(s)
Ácidos Ftálicos/toxicidad , Sulfonamidas/toxicidad , Pruebas de Toxicidad Subcrónica , Animales , Línea Celular , Técnicas In Vitro , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA