Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 198
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cutan Ocul Toxicol ; 43(2): 105-112, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38174703

RESUMEN

Purpose: Favipiravir (FAV) used against COVID-19 is an antiviral drug that causes adverse reactions, such as hyperuricaemia, liver damage, and hematopoetic toxicity. The aim of the study was to investigate the systemic and ocular side-effects of FAV in rats, for the first time.Materials and methods: A total of 18 albino male Wistar rats were used in the study. The rats were divided into 3 groups as the healthy group (HG), the group given 50 mg/kg/day favipiravir (FAV50), and the group given 200 mg/kg/d favipiravir (FAV200). These doses were given to the experimental groups for one week. At the end of the experiment histopathological examinations were performed on the conjunctiva and sclera of the eye. In addition, malondialdehyde (MDA), total glutathione (tGSH), superoxide dismutase (SOD), interleukin-1ß (IL-1ß), and tumor necrosis factor alpha (TNF-α) levels were measured in blood samples taken from rats. Results: Compared to HG, the MDA (1.37 ± 0.61 vs. 4.82 ± 1.40 µmol/mL), IL-1ß (2.52 ± 1.14 vs. 6.67 ± 1.99 pg/mL), and TNF-α levels (3.28 ± 1.42 vs. 8.53 ± 3.06 pg/mL) of the FAV200 group were higher. The levels of tGSH (7.58 ± 1.98 vs. 2.50 ± 0.98 nmol/mL) and SOD (13.63 ± 3.43 vs. 3.81 ± 1.43 U/mL) the FAV200 group were lower than the HG (p < 0.05, for all). The degree of damage to the cornea and sclera of the FAV200 group was quite high according to HG (p < 0.001). Conclusions: FAV can cause damage to rat conjunctiva and sclera by increasing oxidant stress and inflammation at high dose.


Asunto(s)
Amidas , Antivirales , Pirazinas , Ratas Wistar , Animales , Masculino , Pirazinas/toxicidad , Pirazinas/administración & dosificación , Amidas/toxicidad , Ratas , Antivirales/toxicidad , Glutatión/metabolismo , Malondialdehído/metabolismo , Superóxido Dismutasa/metabolismo , Ojo/efectos de los fármacos , Ojo/patología , Estrés Oxidativo/efectos de los fármacos , Factor de Necrosis Tumoral alfa/sangre , Interleucina-1beta/sangre , Conjuntiva/patología , Conjuntiva/efectos de los fármacos
2.
Drug Chem Toxicol ; 46(3): 546-556, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-35450509

RESUMEN

Favipiravir is a selective RNA polymerase inhibitor and a broad-spectrum antiviral drug, an important agent used in viral infections, including Ebola, Lassa, and COVID-19. This study aims to evaluate the potential toxicological effects of favipiravir administration on rats' liver and kidney tissues. Favipiravir was applied for five and ten days in the present study. During this period, it was aimed to determine possible toxic effects on the liver and kidney. For this purpose, the impact of favipiravir on liver and kidney tissues were examined using histopathologic and biochemical methods. The present study showed that favipiravir administration led to an elevation in the liver and kidney serum enzymes and oxidative and histopathologic damages. Favipiravir administration caused apoptotic cell death (Caspase-3 and Bcl-2), inflammation (NF-κB and IL-6), and a decrease in renal reabsorption (AQP2) levels. In the evaluation of the findings obtained in this study, it was determined that the favipiravir or metabolites caused liver and kidney damages.


Asunto(s)
Amidas , Antivirales , Riñón , Hígado , Pirazinas , Animales , Ratas , Antivirales/farmacología , Antivirales/toxicidad , Acuaporina 2 , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Amidas/farmacología , Amidas/toxicidad , Pirazinas/farmacología , Pirazinas/toxicidad
3.
Mol Divers ; 26(4): 2085-2101, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34561737

RESUMEN

Pyrazine-bipyrazole-based µ-oxo bridged dinuclear Au(III) complexes were synthesized and characterized by various spectrometric (1H-NMR, 13C (APT) NMR, FT-IR, Mass spectrometry) and analytical techniques (elemental analysis and conductance measurement). The evaluation of DNA binding activity by UV-Vis absorption spectra and viscosity measurement demonstrated that all the compounds intercalate in between the stacks of DNA base pair and the binding constant values were observed in the range of 5.4 × 104-2.17 × 105 M-1. The molecular docking study also supports the intercalation mode of binding. The anti-proliferation activity of complexes on A549 (Lung adenocarcinoma) cells by MTT assay demonstrated IC50 values in the range of 47.46 -298.12 µg/mL. The genotoxicity of compounds was checked by smearing observed in the DNA of S. pombe cell under the influence of complexes. The in vivo cytotoxicity of compounds against brine shrimp demonstrated the LC50 values in the range of 4.59-27.22 µg/mL. The promising results of the Au(III) complexes received significant attention and make them suitable for the new metallodrugs after the detailed mechanistic biological study.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Antineoplásicos/química , Complejos de Coordinación/química , Complejos de Coordinación/toxicidad , ADN/química , Simulación del Acoplamiento Molecular , Pirazinas/toxicidad , Espectroscopía Infrarroja por Transformada de Fourier
4.
Toxicol Appl Pharmacol ; 414: 115426, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33524445

RESUMEN

Activation of NLRP3 inflammasome is implicated in varieties of pathologies, the aim of the present study is to characterize the effect and mechanism of mitochondrial uncouplers on NLRP3 inflammasome activation by using three types of uncouplers, niclosamide, CCCP and BAM15. Niclosamide, CCCP and BAM15 inhibited LPS plus ATP-induced increases of NLRP3 protein and IL-1ß mRNA levels in RAW264.7 macrophages and THP-1 derived macrophages. Niclosamide, CCCP and BAM15 inhibited LPS plus ATP-induced increase of NFκB (P65) phosphorylation, and inhibited NFκB (P65) nuclear translocation in RAW264.7 macrophages. Niclosamide and BAM15 inhibited LPS-induced increase of IκBα phosphorylation in RAW264.7 macrophages, and the inhibitory effect was dependent on increased intracellular [Ca2+]i; however, CCCP showed no significant effect on IκBα phosphorylation in RAW264.7 macrophages stimulated with LPS. In conclusion, chemical mitochondrial uncouplers niclosamide, CCCP and BAM15 share common inhibitory effect on NLRP3 inflammasome activation through inhibiting NFκB nuclear translocation.


Asunto(s)
Inflamasomas/agonistas , Macrófagos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/agonistas , Desacopladores/toxicidad , Proteínas Quinasas Activadas por AMP/metabolismo , Transporte Activo de Núcleo Celular , Animales , Calcio/metabolismo , Carbonil Cianuro m-Clorofenil Hidrazona/toxicidad , Citocinas/genética , Citocinas/metabolismo , Diaminas/toxicidad , Humanos , Inflamasomas/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Inhibidor NF-kappaB alfa/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Niclosamida/toxicidad , Oxadiazoles/toxicidad , Fosforilación , Pirazinas/toxicidad , Células RAW 264.7 , Células THP-1
5.
Regul Toxicol Pharmacol ; 107: 104417, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31276731

RESUMEN

MB-102 was designed for measurement of real-time glomerular filtration rate (GFR). Previously reported in vitro and in vivo nonclinical studies clearly demonstrated negligible toxicity, resulting in FDA clearance for First-in Human, proof of concept clinical studies. The next tier of safety and toxicity studies are reported herein. MB-102 did not demonstrate any phototoxic potential in a BALB/c 3T3 mouse fibroblast study. Co-administration of MB-102 and iohexol resulted in pharmacokinetic parameters virtually identical to the values observed upon individual administration in beagle dogs. A single dose of MB-102 administered either intravenously (18.6 mg/mL) or perivenously (0.25 mL) was well-tolerated in NZ white rabbits, with no adverse inflammation or irritation. MB-102 did not induce micronuclei in polychromatic erythrocytes for rat bone marrow cells treated up to 450 mg/kg/day, the maximum feasible dose. Two separate optical imaging studies demonstrated that MB-102 distributes rapidly and thoroughly throughout the test subjects, followed by rapid clearance from the body without any preferential localization in any particular tissue or organ, with the exception of the bladder, which is totally consistent with a known GFR agent. In addition, two-week repeat intravenous (once-daily) toxicity and toxicokinetic studies were conducted in rats and beagles, with no MB-102- related effects. Thus, for the studies reported herein, there were no toxicological effects of concern for MB-102.


Asunto(s)
Colorantes Fluorescentes/toxicidad , Pirazinas/toxicidad , Animales , Células 3T3 BALB , Medios de Contraste/farmacocinética , Dermatitis Fototóxica , Perros , Interacciones Farmacológicas , Femenino , Colorantes Fluorescentes/farmacocinética , Tasa de Filtración Glomerular , Yohexol/farmacocinética , Masculino , Ratones , Ratones Desnudos , Pruebas de Micronúcleos , Pirazinas/farmacocinética , Conejos , Ratas Sprague-Dawley
6.
Bioorg Med Chem Lett ; 28(4): 618-625, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29395971

RESUMEN

A series of optically pure (R)- and (S)-1,3,4,12a-tetrahydropyrazino[2,1-c][1,4]benzodiazepine-6,12(2H,11H)-dione derivatives was designed and synthesized as novel anthramycin analogues in a three-step, one-pot procedure, and tested for their antiproliferative activity on nine following cell lines: MV-4-11, UMUC-3, MDA-MB-231, MCF7, LoVo, HT-29, A-549, A2780 and BALB/3T3. The key structural features responsible for exhibition of cytotoxic effect were determined: the (S)-configuration of chiral center and the presence of hydrophobic 4-biphenyl substituent in the side chain. Introduction of bromine atom into the 8 position (8g) or substitution of dilactam ring with benzyl group (8m) further improved the activity and selectivity of investigated compounds. Among others, compound 8g exhibited selective cytotoxic effect against MV-4-11 (IC50 = 8.7 µM) and HT-29 (IC50 = 17.8 µM) cell lines, while 8m showed noticeable anticancer activity against MV-4-11 (IC50 = 10.8 µM) and LoVo (IC50 = 11.0 µM) cell lines. The cell cycle arrest in G1/S checkpoint and apoptosis associated with overproduction of reactive oxygen species was also observed for 8e and 8m.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Benzodiazepinonas/farmacología , Pirazinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/toxicidad , Benzodiazepinonas/síntesis química , Benzodiazepinonas/química , Benzodiazepinonas/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Leucemia/tratamiento farmacológico , Ratones , Pirazinas/síntesis química , Pirazinas/química , Pirazinas/toxicidad , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Estereoisomerismo , Relación Estructura-Actividad
7.
Int J Mol Sci ; 19(10)2018 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-30274380

RESUMEN

Hederagenin (He) is a novel triterpene template for the development of new antitumor compounds. In this study, 26 new He⁻pyrazine derivatives were synthetized in an attempt to develop potent antitumor agents; they were screened for in vitro cytotoxicity against tumor and non-tumor cell lines. The majority of these derivatives showed much stronger cytotoxic activity than He. Remarkably, the most potent was compound 9 (half maximal inhibitory concentration (IC50) was 3.45 ± 0.59 µM), which exhibited similar antitumor activities against A549 (human non-small-cell lung cancer) as the positive drug cisplatin (DDP; IC50 was 3.85 ± 0.63 µM), while it showed lower cytotoxicity on H9c2 (murine heart myoblast; IC50 was 16.69 ± 0.12 µM) cell lines. Compound 9 could induce the early apoptosis and evoke cell-cycle arrest at the synthesis (S) phase of A549 cells. Impressively, we innovatively introduced the method of cluster analysis modeled as partial least squares discriminant analysis (PLS-DA) into the structure⁻activity relationship (SAR) evaluation, and SAR confirmed that pyrazine had a profound effect on the antitumor activity of He. The present studies highlight the importance of pyrazine derivatives of He in the discovery and development of novel antitumor agents.


Asunto(s)
Diseño de Fármacos , Ácido Oleanólico/análogos & derivados , Pirazinas/síntesis química , Pirazinas/toxicidad , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Muerte Celular , Línea Celular Tumoral , Forma de la Célula/efectos de los fármacos , Análisis por Conglomerados , Análisis Discriminante , Humanos , Concentración 50 Inhibidora , Análisis de los Mínimos Cuadrados , Ácido Oleanólico/síntesis química , Ácido Oleanólico/química , Ácido Oleanólico/toxicidad , Análisis de Componente Principal , Pirazinas/química , Coloración y Etiquetado , Relación Estructura-Actividad
8.
Respir Res ; 17(1): 57, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27184162

RESUMEN

BACKGROUND: The potential for adverse respiratory effects following exposure to electronic (e-) cigarette liquid (e-liquid) flavorings remains largely unexplored. Given the multitude of flavor permutations on the market, identification of those flavor constituents that negatively impact the respiratory tract is a daunting task. In this study we examined the impact of common e-liquid flavoring chemicals on the airway epithelium, the cellular monolayer that provides the first line of defense against inhaled particulates, pathogens, and toxicants. METHODS: We used the xCELLigence real-time cell analyzer (RTCA) as a primary high-capacity screening tool to assess cytotoxicity thresholds and physiological effects of common e-liquid flavoring chemicals on immortalized human bronchial epithelial cells (16HBE14o-). The RTCA was used secondarily to assess the capability of 16HBE14o- cells to respond to cellular signaling agonists following a 24 h exposure to select flavoring chemicals. Finally, we conducted biophysical measurements of well-differentiated primary mouse tracheal epithelial (MTE) cells with an Ussing chamber to measure the effects of e-cigarette flavoring constituents on barrier function and ion conductance. RESULTS: In our high-capacity screens five of the seven flavoring chemicals displayed changes in cellular impedance consistent with cell death at concentrations found in e-liquid. Vanillin and the chocolate flavoring 2,5-dimethylpyrazine caused alterations in cellular physiology indicative of a cellular signaling event. At subcytotoxic levels, 24 h exposure to 2,5-dimethylpyrazine compromised the ability of airway epithelial cells to respond to signaling agonists important in salt and water balance at the airway surface. Biophysical measurements of 2,5-dimethylpyrazine on primary MTE cells revealed alterations in ion conductance consistent with an efflux at the apical airway surface that was accompanied by a transient loss in transepithelial resistance. Mechanistic studies confirmed that the increases in ion conductance evoked by 2,5-dimethylpyrazine were largely attributed to a protein kinase A-dependent (PKA) activation of the cystic fibrosis transmembrane conductance regulator (CFTR) ion channel. CONCLUSIONS: Data from our high-capacity screening assays demonstrates that individual e-cigarette liquid flavoring chemicals vary in their cytotoxicity profiles and that some constituents evoke a cellular physiological response on their own independent of cell death. The activation of CFTR by 2,5-dimethylpyrazine may have detrimental consequences for airway surface liquid homeostasis in individuals that use e-cigarettes habitually.


Asunto(s)
Bronquios/efectos de los fármacos , Chocolate , Sistemas Electrónicos de Liberación de Nicotina , Células Epiteliales/efectos de los fármacos , Aromatizantes/toxicidad , Pirazinas/toxicidad , Vapeo/efectos adversos , Animales , Bronquios/metabolismo , Bronquios/patología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/efectos de los fármacos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Relación Dosis-Respuesta a Droga , Conductividad Eléctrica , Células Epiteliales/metabolismo , Células Epiteliales/patología , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones Endogámicos C57BL , Permeabilidad , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
9.
J Immunol ; 192(3): 1277-85, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24363427

RESUMEN

It was shown that the proteasome inhibitor, bortezomib, administered immediately following allogeneic bone marrow transplantation resulted in marked inhibition of acute graft-versus-host disease (aGVHD), with retention of graft-versus-tumor effects. However, continuous bortezomib administration resulted in significant acceleration of graft-versus-host disease-dependent morbidity. We carried out studies to dissect the mechanisms of aggravated aGVHD caused by delayed bortezomib administration. First, we demonstrated that IL-1ß was critically involved, and the subsequent aGVHD could be alleviated by IL-1ß blockade. Bortezomib treatment after dendritic cell (DC) activation resulted in drastically elevated IL-1ß production, whereas bortezomib treatment before DC activation inhibited IL-1ß production, suggesting that the timing of bortezomib administration significantly affected IL-1ß production by DCs. We further demonstrated that delayed administration of bortezomib accelerated aGVHD through TLR4 signaling. Because the LPS levels were much lower with reduced-intensity conditioning compared with high-dose irradiation, the accelerated graft-versus-host disease-dependent morbidity with delayed bortezomib administration could be rescued by reduced-intensity conditioning. Our studies suggested that TLR4 pathway activation and delayed bortezomib administration amplified the production of IL-1ß and other inflammatory cytokines, which resulted in accelerated aGVHD-dependent morbidity. These results indicated that decreased toxicity of continuous bortezomib administration could be achieved by reduced-intensity conditioning or by inhibiting IL-1ß.


Asunto(s)
Trasplante de Médula Ósea , Ácidos Borónicos/toxicidad , Enfermedad Injerto contra Huésped/inducido químicamente , Interleucina-1beta/fisiología , Inhibidores de Proteasas/toxicidad , Pirazinas/toxicidad , Transducción de Señal/fisiología , Receptor Toll-Like 4/fisiología , Enfermedad Aguda , Animales , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/farmacología , Bortezomib , Trasplante de Células , Ciclofosfamida/administración & dosificación , Citocinas/biosíntesis , Citocinas/genética , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Enfermedad Injerto contra Huésped/patología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/farmacología , Pirazinas/administración & dosificación , Pirazinas/farmacología , Organismos Libres de Patógenos Específicos , Bazo/citología , Acondicionamiento Pretrasplante/métodos , Irradiación Corporal Total
10.
Toxicol Appl Pharmacol ; 285(2): 98-109, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25886977

RESUMEN

Understanding how chemotherapeutic agents mediate testicular toxicity is crucial in light of compelling evidence that male infertility, one of the severe late side effects of intensive cancer treatment, occurs more often than they are expected to. Previous study demonstrated that bortezomib (BTZ), a 26S proteasome inhibitor used to treat refractory multiple myeloma (MM), exerts deleterious impacts on spermatogenesis in pubertal mice via unknown mechanisms. Here, we showed that intermittent treatment with BTZ resulted in fertility impairment in adult mice, evidenced by testicular atrophy, desquamation of immature germ cells and reduced caudal sperm storage. These deleterious effects may originate from the elevated apoptosis in distinct germ cells during the acute phase and the subsequent disruption of Sertoli-germ cell anchoring junctions (AJs) during the late recovery. Mechanistically, balance between AMP-activated protein kinase (AMPK) activation and Akt/ERK pathway appeared to be indispensable for AJ integrity during the late testicular recovery. Of particular interest, the upregulated testicular apoptosis and the following disturbance of Sertoli-germ cell interaction may both stem from the excessive oxidative stress elicited by BTZ exposure. We also provided the in vitro evidence that AMPK-dependent mechanisms counteract follicle-stimulating hormone (FSH) proliferative effects in BTZ-exposed Sertoli cells. Collectively, BTZ appeared to efficiently prevent germ cells from normal development via multiple mechanisms in adult mice. Employment of antioxidants and/or AMPK inhibitor may represent an attractive strategy of fertility preservation in male MM patients exposed to conventional BTZ therapy and warrants further investigation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antineoplásicos/toxicidad , Ácidos Borónicos/toxicidad , Células Germinativas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Pirazinas/toxicidad , Enfermedades Testiculares/inducido químicamente , Animales , Apoptosis/efectos de los fármacos , Bortezomib , Activación Enzimática/efectos de los fármacos , Fertilidad/efectos de los fármacos , Hormonas Esteroides Gonadales/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Células de Sertoli/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Enfermedades Testiculares/metabolismo , Enfermedades Testiculares/patología , Testículo/efectos de los fármacos , Testículo/patología , Regulación hacia Arriba/efectos de los fármacos
11.
Anticancer Drugs ; 26(5): 547-54, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25734832

RESUMEN

Oral cancer continues to be a major cause of morbidity and mortality worldwide. Treatment of oral cancer with combinatorial drugs is increasingly being performed as drugs with different molecular targets often exert synergistic effects, thereby enhancing treatment efficacy. Current combinatorial drug regimens often combine the tolerable dosages of individual drugs. However, the optimized ratio of a drug combination and sequence of drug administration could contribute toward the synergy, leading to increased efficacy and reduced dosages. This report aims to study the possible synergistic effects of three anticancer drugs, a proteasome inhibitor, bortezomib, a topoisomerase I inhibitor, Camptothecin, and a DNA intercalation drug, Doxorubicin, when used in combination for treating oral cancer. To rapidly optimize the three-drug regimen with minimal experimental efforts, a Feedback System Control optimization technique, a recent platform technique developed particularly for drug combination screening, was applied. The optimized regimen showed a therapeutic window (death rate difference between cancer cells and normal cells) close to 100%. This is the first report on the use of a combination of bortezomib, Camptothecin, and Doxorubicin in the treatment of oral cancer. Our results indicate that to have the most synergistic anticancer effect, the drugs in the optimized regimen should be dosage specific and ratio specific. Furthermore, the sequence of drug administration plays a vital role in ensuring that the combination is effective. The optimized regimen reported here has the potential to considerably increase the cure rate of oral cancer and reduce the toxicity of chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Camptotecina/farmacología , Doxorrubicina/farmacología , Neoplasias de la Boca/tratamiento farmacológico , Pirazinas/farmacología , Antineoplásicos/toxicidad , Ácidos Borónicos/toxicidad , Bortezomib , Camptotecina/toxicidad , Línea Celular Tumoral/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/toxicidad , Sinergismo Farmacológico , Humanos , Pirazinas/toxicidad
12.
Exp Mol Pathol ; 98(1): 18-26, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25449333

RESUMEN

Lung cancer is one of the few human diseases for which the primary etiological agent, cigarette smoke (CS), has been described; however, the precise role of individual cigarette smoke toxicant in tumor development and progression remains to be elusive. The purpose of this study was to assess in vitro the effects of previously identified cigarette smoke components, pyrazine, 2-ethylpyridine, and 3-ethylpyridine, on non-tumorigenic (MRC5) and adenocarcinomic (A549) human lung cell lines. Our data showed that the administration of three cigarette smoke components in combination perturbed the proliferation of both normal and adenocarcinomic cells. Study of malignant cells revealed that CS components were cytotoxic at high concentration (10(-6) M) and stimulatory in a dose-dependent manner at lower concentrations (10(-8) M to 10(-10) M). This adverse effect was enhanced when adenocarcinomic cells were maintained in hypoxia resembling intratumoral environment. Furthermore, exposure to pyrazine, 2-ethylpyridine, and 3-ethylpyridine induced oxidative stress in both normal and malignant cells. Finally, assessment of P-gp activity revealed that multidrug resistance was induced in CS component exposed adenocarcinomic lung cells and the induction was augmented in hypoxia. Taken together, pyrazine, 2-ethylpyridine, and 3-ethylpyridine adversely altered both normal and diseased lung cells in vitro and data collected from this study may help lung cancer patients to understand the importance of quitting smoking during lung cancer treatment.


Asunto(s)
Resistencia a Múltiples Medicamentos/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Pulmón/efectos de los fármacos , Pirazinas/toxicidad , Piridinas/toxicidad , Humo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Resistencia a Antineoplásicos , Humanos , Neoplasias Pulmonares/patología , Estrés Oxidativo/efectos de los fármacos , Nicotiana
13.
Toxicol Pathol ; 43(3): 376-83, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25112279

RESUMEN

Diacylglycerol O-acyltransferase 1 (DGAT1) plays an important role in synthesizing lipids, and inhibitors of DGAT1 have been investigated as potential treatments for diabetes and metabolic diseases. DGAT1 knockout (-/-) mice are resistant to obesity, have increased sensitivity to insulin, and exhibit sebaceous gland atrophy and alopecia. Prolonged pharmacological inhibition of DGAT1 with AZD7687 in mice results in the same skin phenotype, including sebaceous gland atrophy and alopecia, as seen in the skin of DGAT1 (-/-) mice. AZD7687-mediated effects on the skin were dose- and time-dependent and reversible. They occurred only at substantial levels of continuous DGAT1 inhibition. Prolonged treatment of dogs with AZD7687 also resulted in sebaceous gland atrophy but did not result in the more adverse skin changes of hair loss and skin lesions. Our findings highlight a significant risk of generating the same lesions that were seen in mouse skin during clinical development of DGAT1 inhibitors in humans and also reveal a species difference in the effects on the skin, indicating that the mouse may be an especially sensitive species. Therefore, although human therapeutic doses may not have the same influence on skin morphology as seen in mice, monitoring of skin changes will be essential in clinical trials with DGAT1 inhibitors.


Asunto(s)
Acetatos/toxicidad , Alopecia/patología , Diacilglicerol O-Acetiltransferasa/antagonistas & inhibidores , Pirazinas/toxicidad , Glándulas Sebáceas/patología , Piel/patología , Alopecia/inducido químicamente , Animales , Perros , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/toxicidad , Femenino , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Glándulas Sebáceas/efectos de los fármacos
14.
Regul Toxicol Pharmacol ; 72(1): 26-38, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25728979

RESUMEN

The fluorescent tracer agent 3,6-diamino-2,5-bis{N-[(1R)-1-carboxy-2-hydroxyethyl]carbamoyl}pyrazine, designated MB-102, has been developed with properties and attributes for use as a direct measure of glomerular filtration rate (GFR). In comparison to known standard exogenous GFR agents in animal models, MB-102 has demonstrated an excellent correlation. A battery of toxicity tests has been completed on this new fluorescent tracer agent, including single dose toxicity studies in rats and dogs to determine overall toxicity and toxicokinetics of the compound. Blood compatibility, mutation assay, chromosomal aberration assay, and several other assays were also completed. Toxicity assessments were based on mortality, clinical signs, body weight, food consumption and anatomical pathology. Doses of up to 200-300 times the estimated human dose were administered. No test-article related effects were noted on body weight, food consumption, ophthalmic observations and no abnormal pathology was seen in either macroscopic or microscopic evaluations of any organs or tissues. All animals survived to scheduled sacrifice. Transient discoloration of skin and urine was noted at the higher dose levels in both species as expected from a highly fluorescent compound and was not considered pathological. Thus initial toxicology studies of this new fluorescent tracer agent MB-102 have resulted in negligible demonstrable pathological test article concerns.


Asunto(s)
Colorantes/toxicidad , Colorantes Fluorescentes/toxicidad , Tasa de Filtración Glomerular/efectos de los fármacos , Pirazinas/toxicidad , Animales , Peso Corporal/efectos de los fármacos , Perros , Femenino , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Pruebas de Toxicidad/métodos
15.
J Hepatol ; 60(1): 160-6, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23978715

RESUMEN

BACKGROUND & AIMS: Oltipraz (4-methyl-5(pyrazinyl-2)-1-2-dithiole-3-thione), a promising cancer preventive agent, has an antioxidative activity and ability to enhance glutathione biosynthesis, phase II detoxification enzymes and multidrug resistance-associated protein-mediated efflux transporters. Oltipraz can protect against hepatotoxicity caused by carbon tetrachloride, acetaminophen and alpha-naphthylisothiocyanate. Whether oltipraz has hepato-protective effects on obstructive cholestasis is unknown. METHODS: We administered oltipraz to mice for 5 days prior to bile duct ligation (BDL) for 3 days. Liver histology, liver function markers, bile flow rates and hepatic expression of profibrogenic genes were evaluated. RESULTS: Mice pretreated with oltipraz prior to BDL demonstrated higher levels of serum aminotransferases and more severe liver damage than in control mice. Higher bile flow and glutathione secretion rates were observed in unoperated mice treated with oltipraz than in control mice, suggesting that liver necrosis in oltipraz-treated BDL mice may be related partially to increased bile-acid independent flow and biliary pressure. Oltipraz treatment in BDL mice enhanced α-smooth muscle actin expression, consistent with activation of hepatic stellate cells and portal fibroblasts. Matrix metalloproteinases (Mmp) 9 and 13 and tissue inhibitors of metalloproteinases (Timp) 1 and 2 levels were increased in the oltipraz-treated BDL group, suggesting that the secondary phase of liver injury induced by oltipraz might be due to excessive Mmp and Timp secretions, which induce remodeling of the extracellular matrix. CONCLUSIONS: Oltipraz treatment exacerbates the severity of liver injury following BDL and should be avoided as therapy for extrahepatic cholestatic disorders due to bile duct obstruction.


Asunto(s)
Colestasis Extrahepática/tratamiento farmacológico , Pirazinas/toxicidad , Proteínas Angiogénicas/genética , Animales , Bilis/efectos de los fármacos , Bilis/metabolismo , Ácidos y Sales Biliares/metabolismo , Conductos Biliares/cirugía , Glutatión/metabolismo , Ligadura , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Factor 2 Relacionado con NF-E2/fisiología , Tionas , Tiofenos , Factor de Crecimiento Transformador beta/fisiología
16.
Blood ; 120(8): 1589-96, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22791289

RESUMEN

The Spanish Myeloma Group conducted a trial to compare bortezomib/thalidomide/dexamethasone (VTD) versus thalidomide/dexamethasone (TD) versus vincristine, BCNU, melphalan, cyclophosphamide, prednisone/vincristine, BCNU, doxorubicin, dexamethasone/bortezomib (VBMCP/VBAD/B) in patients aged 65 years or younger with multiple myeloma. The primary endpoint was complete response (CR) rate postinduction and post-autologous stem cell transplantation (ASCT). Three hundred eighty-six patients were allocated to VTD (130), TD (127), or VBMCP/VBAD/B (129). The CR rate was significantly higher with VTD than with TD (35% vs 14%, P = .001) or with VBMCP/VBAD/B (35% vs 21%, P = .01). The median progression-free survival (PFS) was significantly longer with VTD (56.2 vs 28.2 vs 35.5 months, P = .01). In an intention-to-treat analysis, the post-ASCT CR rate was higher with VTD than with TD (46% vs 24%, P = .004) or with VBMCP/VBAD/B (46% vs 38%, P = .1). Patients with high-risk cytogenetics had a shorter PFS and overall survival in the overall series and in all treatment groups. In conclusion, VTD resulted in a higher pre- and posttransplantation CR rate and in a significantly longer PFS although it was not able to overcome the poor prognosis of high-risk cytogenetics. Our results support the use of VTD as a highly effective induction regimen prior to ASCT. The study was registered with http://www.clinicaltrials.gov (NCT00461747) and Eudra CT (no. 2005-001110-41).


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ácidos Borónicos/uso terapéutico , Dexametasona/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Mieloma Múltiple/terapia , Pirazinas/uso terapéutico , Talidomida/uso terapéutico , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/toxicidad , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/efectos adversos , Ácidos Borónicos/toxicidad , Bortezomib , Dexametasona/administración & dosificación , Dexametasona/efectos adversos , Dexametasona/toxicidad , Supervivencia sin Enfermedad , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Quimioterapia de Inducción , Masculino , Persona de Mediana Edad , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/patología , Pirazinas/administración & dosificación , Pirazinas/efectos adversos , Pirazinas/toxicidad , Células Madre/efectos de los fármacos , Células Madre/patología , Talidomida/administración & dosificación , Talidomida/efectos adversos , Talidomida/toxicidad , Trasplante Autólogo
17.
Toxicol Appl Pharmacol ; 276(2): 104-14, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24534256

RESUMEN

Glucagon Like Peptide-1 (GLP-1) drugs are currently used to treat type-2 diabetes. Safety concerns for increased risk of pancreatitis and pancreatic ductal metaplasia have accompanied these drugs. High fat diet (HFD) is a type-2 diabetes risk factor that may affect the response to GLP-1 drug treatment. The objective of the present study was to investigate the effects of diet and GLP-1 based drugs on the exocrine pancreas in mice. Experiments were designed in a mouse model of insulin resistance created by feeding a HFD or standard diet (STD) for 6weeks. The GLP-1 drugs, sitagliptin (SIT) and exenatide (EXE) were administered once daily for additional 6weeks in both mice fed HFD or STD. The results showed that body weight, blood glucose levels, and serum levels of pro-inflammatory cytokines (TNFα, IL-1ß, and KC) were significantly greater in HFD mice than in STD mice regardless of GLP-1 drug treatment. The semi-quantitative grading showed that pancreatic changes were significantly greater in EXE and SIT-treated mice compared to control and that HFD exacerbated spontaneous exocrine pancreatic changes seen in saline-treated mice on a standard diet. Exocrine pancreatic changes identified in this study included acinar cell injury (hypertrophy, autophagy, apoptosis, necrosis, and atrophy), vascular injury, interstitial edema and inflammation, fat necrosis, and duct changes. These findings support HFD as a risk factor to increased susceptibility/severity for acute pancreatitis and indicate that GLP-1 drugs cause pancreatic injury that can be exacerbated in a HFD environment.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hipoglucemiantes/toxicidad , Páncreas/efectos de los fármacos , Péptidos/toxicidad , Pirazinas/toxicidad , Receptores de Glucagón/agonistas , Triazoles/toxicidad , Ponzoñas/toxicidad , Enfermedad Aguda , Animales , Apoptosis/efectos de los fármacos , Atrofia , Exenatida , Receptor del Péptido 1 Similar al Glucagón , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis , Páncreas/patología , Pancreatitis/etiología , Fosfato de Sitagliptina
18.
Brain Behav Immun ; 38: 185-91, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24530998

RESUMEN

Bortezomib (BTZ) is a frequently used chemotherapeutic drug for the treatment of refractory multiple myeloma and hematological neoplasms. The mechanism by which the administration of BTZ leads to painful peripheral neuropathy remains unclear. In the present study, we first determined that the administration of BTZ upregulated the expression of TNF-α and phosphorylated JNK1/2 in the dorsal root ganglion (DRG) of rat. Furthermore, the TNF-α synthesis inhibitor thalidomide significantly blocked the activation of both isoforms JNK1 and JNK2 in the DRG and attenuated mechanical allodynia following BTZ treatment. Knockout of the expression of TNF-α receptor TNFR1 (TNFR1 KO mice) or TNFR2 (TNFR2 KO mice) inhibited JNK1 and JNK2 activation and decreased mechanical allodynia induced by BTZ. These results suggest that upregulated TNF-α expression may activate JNK signaling via TNFR1 or TNFR2 to mediate mechanical allodynia following BTZ treatment.


Asunto(s)
Antineoplásicos/toxicidad , Ácidos Borónicos/toxicidad , Ganglios Espinales/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/metabolismo , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Pirazinas/toxicidad , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Bortezomib , Ganglios Espinales/efectos de los fármacos , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/enzimología , Enfermedades del Sistema Nervioso Periférico/enzimología , Enfermedades del Sistema Nervioso Periférico/metabolismo , Ratas , Ratas Sprague-Dawley
19.
Anticancer Drugs ; 25(3): 282-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24300915

RESUMEN

The aim of this study was to examine the hypothesis that a combination of proteasome inhibition by bortezomib and immune therapy with interleukin-12 (IL-12) can produce enhanced antitumor efficacy relative to the effects of either of these agents alone. A mouse xenograft model of myeloma was developed. The mice were randomly divided into saline control (NS), IL-12 (0.4 µg/animal; intraperitoneal), bortezomib (0.75 mg/kg; intravenous), and bortezomib+IL-12 groups. Effects of treatments on tumor growth were assessed by before and after treatment comparisons and group comparisons. The effects of various treatments on the number of peripheral blood lymphocytes and natural killer (NK) cells were assessed by complete blood count and flow cytometry analysis. The cell-killing function of NK cells in splenocytes was evaluated using the lactate dehydrogenase release assay. IL-12 treatment alone produced a mild decrease in tumor volume compared with control (P>0.05). Bortezomib alone resulted in substantial inhibition of tumor growth at varying time points, reaching ~65 and ~60% reduction in tumor volume after 15 and 21 days of therapy, respectively. At the same time points, the combination therapy produced ~75 and ~84% decreases in tumor growth, respectively, which were significantly greater than the reduction produced by bortezomib monotherapy. Tumors resumed growth upon termination of bortezomib treatment at 2 weeks, although the tumor volume was still significantly smaller than that in the time-matched NS and IL-12 animals. This rebound of tumor growth was completely prevented with the combination therapy, and tumor volume continued to decrease throughout the time course. The percentage and total number of NK cells were significantly decreased after bortezomib monotherapy and combination therapy; however, they remained unaltered after IL-12 treatment compared with no treatment. Further, combination therapy significantly restored the bortezomib-induced functional impairment of the cell-killing capability of NK cells, relative to bortezomib alone. We conclude that the bortezomib-IL-12 combination therapy offers superior antitumor efficacy over monotherapy with either bortezomib or IL-12 in a mouse model of myeloma. Restoration of bortezomib-induced functional impairment of NK cells by IL-12 may be a mechanism for the synergetic effects of the two agents. Therefore, a combination of the two agents may represent a more rational therapeutic approach for myeloma.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Interleucina-12/farmacología , Células Asesinas Naturales/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Pirazinas/farmacología , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Apoptosis , Ácidos Borónicos/uso terapéutico , Ácidos Borónicos/toxicidad , Bortezomib , Línea Celular Tumoral , Sinergismo Farmacológico , Femenino , Xenoinjertos , Humanos , Inmunoterapia , Interleucina-12/uso terapéutico , Interleucina-12/toxicidad , Masculino , Ratones , Ratones SCID , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Trasplante de Neoplasias , Pirazinas/uso terapéutico , Pirazinas/toxicidad
20.
Biometrics ; 70(2): 389-97, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24571185

RESUMEN

While a general goal of early phase clinical studies is to identify an acceptable dose for further investigation, modern dose finding studies and designs are highly specific to individual clinical settings. In addition, as outcome-adaptive dose finding methods often involve complex algorithms, it is crucial to have diagnostic tools to evaluate the plausibility of a method's simulated performance and the adequacy of the algorithm. In this article, we propose a simple technique that provides an upper limit, or a benchmark, of accuracy for dose finding methods for a given design objective. The proposed benchmark is nonparametric optimal in the sense of O'Quigley et al. (2002, Biostatistics 3, 51-56), and is demonstrated by examples to be a practical accuracy upper bound for model-based dose finding methods. We illustrate the implementation of the technique in the context of phase I trials that consider multiple toxicities and phase I/II trials where dosing decisions are based on both toxicity and efficacy, and apply the benchmark to several clinical examples considered in the literature. By comparing the operating characteristics of a dose finding method to that of the benchmark, we can form quick initial assessments of whether the method is adequately calibrated and evaluate its sensitivity to the dose-outcome relationships.


Asunto(s)
Biometría/métodos , Ensayos Clínicos Fase I como Asunto/estadística & datos numéricos , Algoritmos , Antineoplásicos/administración & dosificación , Antineoplásicos/toxicidad , Teorema de Bayes , Benchmarking/estadística & datos numéricos , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/toxicidad , Bortezomib , Ensayos Clínicos Fase II como Asunto/estadística & datos numéricos , Simulación por Computador , Relación Dosis-Respuesta a Droga , Fibrinolíticos/administración & dosificación , Fibrinolíticos/toxicidad , Humanos , Linfoma/tratamiento farmacológico , Modelos Estadísticos , Pirazinas/administración & dosificación , Pirazinas/toxicidad , Accidente Cerebrovascular/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA