Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 288
Filtrar
1.
Sci Rep ; 11(1): 4456, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33627707

RESUMEN

The bisdioxopiperazine topoisomerase IIß inhibitor ICRF-193 has been previously identified as a more potent analog of dexrazoxane (ICRF-187), a drug used in clinical practice against anthracycline cardiotoxicity. However, the poor aqueous solubility of ICRF-193 has precluded its further in vivo development as a cardioprotective agent. To overcome this issue, water-soluble prodrugs of ICRF-193 were prepared, their abilities to release ICRF-193 were investigated using a novel UHPLC-MS/MS assay, and their cytoprotective effects against anthracycline cardiotoxicity were tested in vitro in neonatal ventricular cardiomyocytes (NVCMs). Based on the obtained results, the bis(2-aminoacetoxymethyl)-type prodrug GK-667 was selected for advanced investigations due to its straightforward synthesis, sufficient solubility, low cytotoxicity and favorable ICRF-193 release. Upon administration of GK-667 to NVCMs, the released ICRF-193 penetrated well into the cells, reached sufficient intracellular concentrations and provided effective cytoprotection against anthracycline toxicity. The pharmacokinetics of the prodrug, ICRF-193 and its rings-opened metabolite was estimated in vivo after administration of GK-667 to rabbits. The plasma concentrations of ICRF-193 reached were found to be adequate to achieve cardioprotective effects in vivo. Hence, GK-667 was demonstrated to be a pharmaceutically acceptable prodrug of ICRF-193 and a promising drug candidate for further evaluation as a potential cardioprotectant against chronic anthracycline toxicity.


Asunto(s)
Antraciclinas/efectos adversos , Cardiotónicos/farmacología , Cardiotoxicidad/tratamiento farmacológico , ADN-Topoisomerasas de Tipo II/metabolismo , Dicetopiperazinas/farmacología , Piperazina/farmacología , Inhibidores de Topoisomerasa II/farmacología , Animales , Cardiotónicos/química , Cardiotoxicidad/metabolismo , Dexrazoxano/química , Dexrazoxano/farmacología , Dicetopiperazinas/química , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Piperazina/química , Profármacos/química , Profármacos/farmacología , Conejos , Razoxano/química , Razoxano/farmacología , Inhibidores de Topoisomerasa II/química , Agua/química
2.
Open Biol ; 10(5): 190259, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32400307

RESUMEN

Topoisomerase IIα (Topo IIα), a well-conserved double-stranded DNA (dsDNA)-specific decatenase, processes dsDNA catenanes resulting from DNA replication during mitosis. Topo IIα defects lead to an accumulation of ultrafine anaphase bridges (UFBs), a type of chromosome non-disjunction. Topo IIα has been reported to resolve DNA anaphase threads, possibly accounting for the increase in UFB frequency upon Topo IIα inhibition. We hypothesized that the excess UFBs might also result, at least in part, from an impairment of the prevention of UFB formation by Topo IIα. We found that Topo IIα inhibition promotes UFB formation without affecting the global disappearance of UFBs during mitosis, but leads to an aberrant UFB resolution generating DNA damage within the next G1. Moreover, we demonstrated that Topo IIα inhibition promotes the formation of two types of UFBs depending on cell cycle phase. Topo IIα inhibition during S-phase compromises complete DNA replication, leading to the formation of UFB-containing unreplicated DNA, whereas Topo IIα inhibition during mitosis impedes DNA decatenation at metaphase-anaphase transition, leading to the formation of UFB-containing DNA catenanes. Thus, Topo IIα activity is essential to prevent UFB formation in a cell-cycle-dependent manner and to promote DNA damage-free resolution of UFBs.


Asunto(s)
ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Razoxano/farmacología , Anafase , Segregación Cromosómica , Daño del ADN , Replicación del ADN/efectos de los fármacos , Células HeLa , Humanos , No Disyunción Genética , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores
3.
Clin Exp Pharmacol Physiol ; 43(5): 569-79, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26918678

RESUMEN

Adipocytokines apelin peptide, the ligand of APJ (putative receptor related to the angiotensin receptor AT1), plays key roles in the pathogenesis and deterioration of cancer. In lung cancer, apelin elevating microvessel densities has been reported. Our previous research has characterized that apelin-13 promoted lung adenocarcinoma cell proliferation. However, the effect of apelin on metastasis in lung adenocarcinoma and the underlying mechanisms remain unclear. This study shows that apelin-13 induced human adenocarcinoma cell migration via the APJ receptor. Apelin-13 phosphorylated PAK1 and cofilin increase the migration of lung adenocarcinoma cells. Moreover, the results verify that over-expression of apelin and APJ contributed to reducing the effect of doxorubicin and razoxane on inhibiting lung adenocarcinoma cells metastasis. Hypoxia activated APJ expression and apelin release in lung adenocarcinoma cells. The results demonstrate a PAK1-cofilin phosphorylation mechanism to mediate lung adenocarcinoma cells migration promoted by apelin-13. This discovery further suggests that APJ and its downstream signalling is a potential target for anti-metastatic therapies in lung adenocarcinoma patients.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Adenocarcinoma/patología , Movimiento Celular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Neoplasias Pulmonares/patología , Quinasas p21 Activadas/metabolismo , Adenocarcinoma del Pulmón , Receptores de Apelina , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Fosforilación/efectos de los fármacos , Razoxano/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
4.
J Biol Chem ; 288(10): 7182-92, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23344961

RESUMEN

Camptothecin (CPT), a topoisomerase (Top) I-targeting drug that stabilizes Top1-DNA covalent adducts, can induce S-phase-specific cytotoxicity due to the arrest of progressing replication forks. However, CPT-induced non-S-phase cytotoxicity is less well characterized. In this study, we have identified topoisomerase IIß (Top2ß) as a specific determinant for CPT sensitivity, but not for many other cytotoxic agents, in non-S-phase cells. First, quiescent mouse embryonic fibroblasts (MEFs) lacking Top2ß were shown to be hypersensitive to CPT with prominent induction of apoptosis. Second, ICRF-187, a Top2 catalytic inhibitor known to deplete Top2ß, specifically sensitized MEFs to CPT. To explore the molecular basis for CPT hypersensitivity in Top2ß-deficient cells, we found that upon CPT exposure, the RNA polymerase II large subunit (RNAP LS) became progressively depleted, followed by recovery to nearly the original level in wild-type MEFs, whereas RNAP LS remained depleted without recovery in Top2ß-deficient cells. Concomitant with the reduction of the RNAP LS level, the p53 protein level was greatly induced. Interestingly, RNAP LS depletion has been well documented to lead to p53-dependent apoptosis. Altogether, our findings support a model in which Top2ß deficiency promotes CPT-induced apoptosis in quiescent non-S-phase cells, possibly due to RNAP LS depletion and p53 accumulation.


Asunto(s)
Apoptosis/efectos de los fármacos , Camptotecina/farmacología , ADN-Topoisomerasas de Tipo II/deficiencia , Proteínas de Unión al ADN/deficiencia , Fibroblastos/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , ADN-Topoisomerasas de Tipo II/genética , Proteínas de Unión al ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Ratones , Ratones Noqueados , Subunidades de Proteína/metabolismo , Razoxano/farmacología , Inhibidores de Topoisomerasa I/farmacología , Transcripción Genética/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
5.
Antioxid Redox Signal ; 18(8): 899-929, 2013 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-22794198

RESUMEN

SIGNIFICANCE: Anthracyclines (doxorubicin, daunorubicin, or epirubicin) rank among the most effective anticancer drugs, but their clinical usefulness is hampered by the risk of cardiotoxicity. The most feared are the chronic forms of cardiotoxicity, characterized by irreversible cardiac damage and congestive heart failure. Although the pathogenesis of anthracycline cardiotoxicity seems to be complex, the pivotal role has been traditionally attributed to the iron-mediated formation of reactive oxygen species (ROS). In clinics, the bisdioxopiperazine agent dexrazoxane (ICRF-187) reduces the risk of anthracycline cardiotoxicity without a significant effect on response to chemotherapy. The prevailing concept describes dexrazoxane as a prodrug undergoing bioactivation to an iron-chelating agent ADR-925, which may inhibit anthracycline-induced ROS formation and oxidative damage to cardiomyocytes. RECENT ADVANCES: A considerable body of evidence points to mitochondria as the key targets for anthracycline cardiotoxicity, and therefore it could be also crucial for effective cardioprotection. Numerous antioxidants and several iron chelators have been tested in vitro and in vivo with variable outcomes. None of these compounds have matched or even surpassed the effectiveness of dexrazoxane in chronic anthracycline cardiotoxicity settings, despite being stronger chelators and/or antioxidants. CRITICAL ISSUES: The interpretation of many findings is complicated by the heterogeneity of experimental models and frequent employment of acute high-dose treatments with limited translatability to clinical practice. FUTURE DIRECTIONS: Dexrazoxane may be the key to the enigma of anthracycline cardiotoxicity, and therefore it warrants further investigation, including the search for alternative/complementary modes of cardioprotective action beyond simple iron chelation.


Asunto(s)
Antraciclinas/efectos adversos , Quelantes/farmacología , Corazón/efectos de los fármacos , Metales/efectos adversos , Miocardio/metabolismo , Estrés Oxidativo , Transducción de Señal , Antraciclinas/química , Antraciclinas/farmacología , Antineoplásicos/efectos adversos , Antineoplásicos/química , Antineoplásicos/farmacología , Antioxidantes/química , Antioxidantes/farmacología , Cardiotónicos/efectos adversos , Cardiotónicos/química , Cardiotónicos/farmacología , Quelantes/efectos adversos , Quelantes/química , Humanos , Oxidación-Reducción , Razoxano/efectos adversos , Razoxano/química , Razoxano/farmacología , Especies Reactivas de Oxígeno/metabolismo
6.
Future Cardiol ; 8(4): 647-70, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22871201

RESUMEN

Advances in cancer treatment have greatly improved survival rates of children with cancer. However, these same chemotherapeutic or radiologic treatments may result in long-term health consequences. Anthracyclines, chemotherapeutic drugs commonly used to treat children with cancer, are known to be cardiotoxic, but the mechanism by which they induce cardiac damage is still not fully understood. A higher cumulative anthracycline dose and a younger age of diagnosis are only a few of the many risk factors that identify the children at increased risk of developing cardiotoxicity. While cardiotoxicity can develop at anytime, starting from treatment initiation and well into adulthood, identifying the best cardioprotective measures to minimize the long-term damage caused by anthracyclines in children is imperative. Dexrazoxane is the only known agent to date, that is associated with less cardiac dysfunction, without reducing the oncologic efficacy of the anthracycline doxorubicin in children. Given the serious long-term health consequences of cancer treatments on survivors of childhood cancers, it is essential to investigate new approaches to improving the safety of cancer treatments.


Asunto(s)
Antraciclinas/efectos adversos , Cardiotónicos/uso terapéutico , Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Corazón/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Adiponectina/farmacología , Adiponectina/uso terapéutico , Factores de Edad , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/uso terapéutico , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Carbazoles/farmacología , Carbazoles/uso terapéutico , Cardiolipinas , Carvedilol , Doxorrubicina , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Humanos , Peroxidación de Lípido , Liposomas/química , Neoplasias/epidemiología , Piperazinas/farmacología , Piperazinas/uso terapéutico , Propanolaminas/farmacología , Propanolaminas/uso terapéutico , Purinas/farmacología , Purinas/uso terapéutico , Razoxano/farmacología , Razoxano/uso terapéutico , Factores de Riesgo , Citrato de Sildenafil , Sulfonas/farmacología , Sulfonas/uso terapéutico , Sobrevivientes , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/prevención & control
7.
Can J Physiol Pharmacol ; 90(4): 473-84, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22439652

RESUMEN

Positive effects of dexrazoxane (DEX) in anthracycline cardiotoxicity have been mostly assumed to be associated with its iron-chelating properties. However, this explanation has been recently questioned. Iron plays also an important role in the catecholamine cardiotoxicity. Hence in this study, the influence of DEX on a catecholamine model of acute myocardial infarction (100 mg/kg of isoprenaline by subcutaneous injection) was assessed: (i) the effects of an intravenous dose of 20.4 mg/kg were analyzed after 24 h, (ii) the effects were monitored continuously during the first two hours after drug(s) administration to examine the mechanism(s) of cardioprotection. Additional in vitro experiments on iron chelation/reduction and influence on the Fenton chemistry were performed both with isoprenaline/DEX separately and in their combination. DEX partly decreased the mortality, reduced myocardial calcium overload, histological impairment, and peripheral haemodynamic disturbances 24 h after isoprenaline administration. Continuous 2 h experiments showed that DEX did not influence isoprenaline induced atrioventricular blocks and had little effect on the measured haemodynamic parameters. Its protective effects are probably mediated by inhibition of late myocardial impairment and ventricular fibrillation likely due to inhibition of myocardial calcium overload. Complementary in vitro experiments suggested that iron chelation properties of DEX apparently did not play the major role.


Asunto(s)
Cardiotónicos/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Razoxano/uso terapéutico , Animales , Calcio/metabolismo , Cardiotónicos/farmacología , Modelos Animales de Enfermedad , Hemodinámica/efectos de los fármacos , Hierro/metabolismo , Quelantes del Hierro/farmacología , Isoproterenol/antagonistas & inhibidores , Masculino , Infarto del Miocardio/inducido químicamente , Miocardio/metabolismo , Miocardio/patología , Ratas , Ratas Wistar , Razoxano/farmacología
8.
Am J Physiol Heart Circ Physiol ; 302(10): H2048-57, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22447943

RESUMEN

The impact of cancer therapies on adult cardiac function is becoming a concern as more children survive their initial cancer. Cardiovascular disease is now a significant problem to adult survivors of childhood cancer. Specifically, doxorubicin (DOX) may be particularly harmful in young girls. The objective of this study was to characterize DOX damage and determine the ability of dexrazoxane (DEX) to reduce DOX-mediated cardiac damage in sedentary and swim-trained female rats. Female Sprague-Dawley rats were left intact or ovariectomized (OVX) at weaning then injected with DEX (60 mg/kg) before DOX (3 mg/kg), DOX alone, or PBS. Rats were separated into sedentary and swim cohorts. Body weight was reduced in DOX:DEX- but not PBS- or DOX-treated rats. Echocardiographic parameters were similar in sedentary rats. Swim training revealed greater concentric remodeling in DOX-treated rats and reduced fractional shortening in DOX:DEX-treated rats. Calsequestrin 2 was reduced with DOX and increased with DOX:DEX postswim. Sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a was reduced and calsequestrin 2 reduced further by swim training only in intact rats. OVX rats were heavier and developed eccentric remodeling post-swim with DOX and eccentric hypertrophy with DOX:DEX. Changes in SERCA2a and calsequestrin 2 expression were not observed. Ovariectomized DOX- and DOX:DEX-treated rats stopped growing during swim training. DEX coinjection did not relieve DOX-mediated cardiotoxicity in intact or hormone-deficient rats. DOX-mediated reductions in growth, cardiac function, and expression of calcium homeostasis proteins were exacerbated by swim. DEX coadministration did not substantially relieve DOX-mediated cardiotoxicity in young female rats. Ovarian hormones reduce DOX-induced cardiotoxicity.


Asunto(s)
Antineoplásicos/farmacología , Doxorrubicina/farmacología , Corazón/efectos de los fármacos , Ovariectomía , Razoxano/farmacología , Descanso/fisiología , Natación/fisiología , Animales , Calcio/metabolismo , Calsecuestrina/metabolismo , Ecocardiografía , Femenino , Corazón/fisiología , Modelos Animales , Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
9.
J Craniomaxillofac Surg ; 40(8): e369-74, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22429609

RESUMEN

INTRODUCTION: Bisphosphonates are important and effective drugs in oncology and osteoporosis therapy. They accumulate in the bone matrix becoming released and active by bone resorption. This leads to effective inhibition of tumor cells and bone degradation. A side effect of bisphosphonates similar to other drugs like denosumab is osteonecrosis of the jaws (ONJ). This problem mostly occurs after tooth extraction. We studied the cytoprotectant dexrazoxane known from anthracycline chemotherapy for cytoprotection in nitrogen-containing bisphosphonate treated cells and in the rabbit tibia model to evaluate a possible value in ONJ management. MATERIALS & METHODS: Human osteoblasts (HOB) P2 cells and Human ginigiva fibroblasts (HGF) P2 cells were treated with zoledronic acid (50 µmol/L) and the cytoprotectant dexrazoxane (600 µmol/L). Analysis included cell viability testing with MTT assay and morphology analysis using CellTracker™ Green CMFDA. A biomaterial carrier (Bio-Oss Collagen) was implanted in the rabbit tibia of 6 female chinchilla bastard rabbits on both sides with drill hole defects (d: 3.2mm). Implants were loaded with 25 nmol zoledronic acid, with and without 300 nmol dexrazoxane and unloaded in a control group. Analysis included histological examination of undecalcified samples with toloudine blue staining after 10 days. RESULTS: In vitro experiments showed a significantly higher MTT activity in cells treated with zoledronic acid together with dexrazoxane compared to the same cells treated with the bisphosphonate alone in t-test (HOB: p=0.0003; HGF: p below 0.0001) and one-way ANOVA. Cell morphology changes were consistent with these results. In vivo results showed newly formed bone trabeculae directly growing towards the implanted hydroxylapatite particles and cortical bone interface resorption activities in the control and the experimental group only. CONCLUSION: The study suggests a possible value of this patented technology for ONJ therapy and prevention with local or systemic application.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Citoprotección/efectos de los fármacos , Difosfonatos/farmacología , Fibroblastos/efectos de los fármacos , Encía/efectos de los fármacos , Imidazoles/farmacología , Osteoblastos/efectos de los fármacos , Sustancias Protectoras/farmacología , Razoxano/farmacología , Tibia/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Resorción Ósea/patología , Técnicas de Cultivo de Célula , Línea Celular , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colágeno , Colorantes , Portadores de Fármacos , Durapatita , Femenino , Fluoresceínas , Colorantes Fluorescentes , Encía/citología , Humanos , Modelos Animales , Osteogénesis/efectos de los fármacos , Conejos , Sales de Tetrazolio , Tiazoles , Ácido Zoledrónico
10.
Biol Reprod ; 86(3): 96, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22190700

RESUMEN

Doxorubicin (DXR) is a frontline chemotherapy agent implicated in unintended ovarian failure in female cancer survivors. The fertility preservation techniques currently available for cancer patients are often time and cost prohibitive and do not necessarily preserve endocrine function. There are no drug-based ovary protection therapies clinically available. This study provides the first investigation using dexrazoxane (Dexra) to limit DXR insult in ovarian tissue. In KK-15 granulosa cells, a 3-h DXR treatment increased double-strand (ds) DNA breaks 40%-50%, as quantified by the neutral comet assay, and dose-dependent cytotoxicity. Dexra exhibited low toxicity in KK-15 cells, inducing no DNA damage and less than 20% cell loss. Cotreating KK-15 cells with Dexra prevented acute DXR-induced dsDNA damage. Similarly, Dexra attenuated the DXR-induced 40%-65% increase in dsDNA breaks in primary murine granulosa cells and cells from in vitro cultured murine ovaries. DXR can cause DNA damage either through a topoisomerase II-mediated pathway, based on DXR intercalation into DNA, or through oxidative stress. Cotreating KK-15 cells with 2 µM Dexra was sufficient to prevent DXR-induced, but not H(2)O(2)-induced, DNA damage. These data indicated the protective effects are likely due to Dexra's inhibition of topoisomerase II catalytic activity. This putative protective agent attenuated downstream cellular responses to DXR, preventing H2AFX activation in KK-15 cells and increasing viability as demonstrated by increasing the DXR lethal dose in KK-15 cells 5- to 8-fold (LD(20)) and primary murine granulosa cells 1.5- to 2-fold (LD(50)). These data demonstrate Dexra protects ovarian cells from DXR insult and suggest that it is a promising tool to limit DXR ovarian toxicity in vivo.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Doxorrubicina/efectos adversos , Infertilidad Femenina/prevención & control , Ovario/efectos de los fármacos , Razoxano/farmacología , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular , Células Cultivadas , Daño del ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/fisiología , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Femenino , Células de la Granulosa/citología , Células de la Granulosa/efectos de los fármacos , Histonas/antagonistas & inhibidores , Histonas/fisiología , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Endogámicos , Modelos Animales , Ovario/citología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología
11.
Int J Toxicol ; 30(6): 681-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21994238

RESUMEN

The cost of dexrazoxane, a drug used to provide protection from doxorubicin-induced cardiotoxicity, limits its use in low-income countries. We aimed to see whether schisandrin B, an inexpensive drug, could provide protection equivalent to that provided by dexrazoxane. New Zealand white rabbits were randomly divided into groups and treated with saline, doxorubicin, doxorubicin + dexrazoxane, or doxorubicin + schisandrin B. Doxorubicin-induced damage and the protective effects were studied by recording the echocardiographic parameters and serum levels of superoxide dismutase, malondialdehyde, cardiac troponin I, and brain natriuretic peptide and observing the histology and degree of apoptosis. Schisandrin B had dose-dependent effects in decreasing the magnitude of doxorubicin-induced indicators of cardiomyopathy to a degree that approximated the decrease produced by dexrazoxane treatment. Schisandrin B might be a useful, low-cost alternative drug for this application.


Asunto(s)
Antineoplásicos/uso terapéutico , Cardiotónicos/uso terapéutico , Cardiopatías/tratamiento farmacológico , Lignanos/uso terapéutico , Compuestos Policíclicos/uso terapéutico , Razoxano/uso terapéutico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Cardiotónicos/farmacología , Ciclooctanos/uso terapéutico , Doxorrubicina , Femenino , Cardiopatías/sangre , Cardiopatías/inducido químicamente , Cardiopatías/patología , Masculino , Malondialdehído/sangre , Péptido Natriurético Encefálico/sangre , Conejos , Razoxano/farmacología , Superóxido Dismutasa/sangre , Troponina I/sangre
12.
Future Cardiol ; 7(5): 693-704, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21929348

RESUMEN

The HER family of tyrosine kinase receptors includes several members that are clinically important targets in cancer therapies, in particular HER1 (the EGF receptor) and HER2, other members include HER3 and HER4. Trastuzumab, a humanized monoclonal antibody and lapatinib, a tyrosine kinase inhibitor, are drugs that target HER2, which is highly expressed in 20-30% of breast cancers. Trastuzumab is recommended as an adjuvant therapy for lymph node positive, HER2-positive breast cancers, or node-negative cancer with high-risk of recurrence, as well as in stage IV cancers. One serious side effect of trastuzumab is cardiomyocyte dysfunction, resulting in reduced heart contractile efficiency. The incidence of collateral effects on the heart with trastuzumab therapy increases in people with cardiovascular risk factors, heart disease and when combined with other chemotherapeutics. When cardiotoxicity was observed with trastuzumab, several studies have addressed potential cardiac damage of trastuzumab itself and lapatinib. The differences in cardiovascular effects of these two compounds are somewhat unexpected and suggest distinct mechanisms of action, which have clear implications in clinical application and prevention of cardiotoxicity in cardio-oncological approaches.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Corazón/efectos de los fármacos , Receptor ErbB-2/farmacología , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/sangre , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Lapatinib , Neoplasias Ováricas/tratamiento farmacológico , Fosforilación , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinazolinas/efectos adversos , Quinazolinas/farmacología , Razoxano/farmacología , Receptor ErbB-2/efectos adversos , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Receptor ErbB-2/uso terapéutico , Transducción de Señal/efectos de los fármacos , Trastuzumab
13.
Photochem Photobiol ; 87(6): 1419-26, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21834866

RESUMEN

Photodynamic therapy (PDT) utilizes the combined interaction of a photosensitizer, light and molecular oxygen to ablate tumor tissue. Maximizing the accumulation of the photosensitizer protoporphyrin IX (PpIX) within different cell types would be clinically useful. Dermatological PpIX-induced PDT regimes produce good clinical outcomes but this currently only applies when the lesion remains superficial. Also, as an adjuvant therapy for the treatment of primary brain tumors, fluorescence guided resection (FGR) and PDT can be used to highlight and destroy tumor cells unreachable by surgical resection. By employing iron chelators PpIX accumulation can be enhanced. Two iron-chelating agents, 1,2-diethyl-3-hydroxypyridin-4-one hydrochloride (CP94) and dexrazoxane, were individually combined with the porphyrin precursors aminolevulinic acid (ALA), methyl aminolevulinate (MAL) and hexyl aminolevulinate (HAL). Efficacies of the iron-chelating agents were compared by recording the PpIX fluorescence in human squamous epithelial carcinoma cells (A431) and human glioma cells (U-87 MG) every hour for up to 6 h. Coincubation of ALA/MAL/HAL with CP94 resulted in a greater accumulation of PpIX compared to that produced by coincubation of these congeners with dexrazoxane. Therefore the clinical employment of iron chelation, particularly with CP94 could potentially increase and/or accelerate the accumulation of ALA/MAL/HAL-induced PpIX for PDT or FGR.


Asunto(s)
Quelantes del Hierro/farmacología , Fotoquimioterapia , Protoporfirinas/metabolismo , Piridonas/farmacología , Razoxano/farmacología , Línea Celular Tumoral , Fluorescencia , Humanos , Técnicas In Vitro
14.
Mutagenesis ; 26(4): 533-43, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21430063

RESUMEN

The intention of the present study was to answer the question whether the catalytic topoisomerase-II inhibitor, dexrazoxane, can be used as a modulator of teniposide-induced DNA damage and programmed cell death (apoptosis) in the bone marrow cells in vivo. The alkaline single cell gel electrophoresis, scoring of chromosomal aberrations, micronuclei and mitotic activity were undertaken in the current study as markers of DNA damage. Apoptosis was analysed by the occurrence of a hypodiploid DNA peak and caspase-3 activity. Oxidative stress marker such as intracellular reactive oxygen species production, lipid peroxidation, reduced and oxidised glutathione were assessed in bone marrow as a possible mechanism underlying this amelioration. Dexrazoxane was neither genotoxic nor apoptogenic in mice at the tested dose. Moreover, for the first time, it has been shown that dexrazoxane affords significant protection against teniposide-induced DNA damage and apoptosis in the bone marrow cells in vivo and effectively suppresses the apoptotic signalling triggered by teniposide. Teniposide induced marked biochemical alterations characteristic of oxidative stress including accumulation of intracellular reactive oxygen species, enhanced lipid peroxidation, accumulation of oxidised glutathione and reduction in the reduced glutathione level. Prior administration of dexrazoxane ahead of teniposide challenge ameliorated these biochemical alterations. It is thus concluded that pretreatment with dexrazoxane attenuates teniposide-induced oxidative stress and subsequent DNA damage and apoptosis in bone marrow cells. Based on our data presented, strategies can be developed to decrease the teniposide-induced DNA damage in normal cells using dexrazoxane. Therefore, dexrazoxane can be a good candidate to decrease the deleterious effects of teniposide in the bone marrow cells of cancer patients treated with teniposide.


Asunto(s)
Apoptosis/efectos de los fármacos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Daño del ADN , Razoxano/farmacología , Tenipósido/toxicidad , Animales , Células de la Médula Ósea/enzimología , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Cromosomas de los Mamíferos/metabolismo , Roturas del ADN/efectos de los fármacos , Citometría de Flujo , Glutatión/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Ratones , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
15.
Br J Pharmacol ; 163(2): 299-312, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21232037

RESUMEN

BACKGROUND AND PURPOSE: Iron aggravates the cardiotoxicity of doxorubicin, a widely used anticancer anthracycline, and the iron chelator dexrazoxane is the only agent protecting against doxorubicin cardiotoxicity; however, the mechanisms underlying the role of iron in doxorubicin-mediated cardiotoxicity and the protective role of dexrazoxane remain to be established. As iron is required for the degradation of hypoxia-inducible factors (HIF), which control the expression of antiapoptotic and protective genes, we tested the hypothesis that dexrazoxane-dependent HIF activation may mediate the cardioprotective effect of dexrazoxane. EXPERIMENTAL APPROACH: Cell death, protein levels (by immunoblotting) and HIF-mediated transcription (using reporter constructs) were evaluated in the rat H9c2 cardiomyocyte cell line exposed to low doses of doxorubicin with or without dexrazoxane pretreatment. HIF levels were genetically manipulated by transfecting dominant-negative mutants or short hairpin RNA. KEY RESULTS: Treatment with dexrazoxane induced HIF-1α and HIF-2α protein levels and transactivation capacity in H9c2 cells. It also prevented the induction of cell death and apoptosis by exposure of H9c2 cells to clinically relevant concentrations of doxorubicin. Suppression of HIF activity strongly reduced the protective effect of dexrazoxane. Conversely, HIF-1α overexpression protected against doxorubicin-mediated cell death and apoptosis also in cells not exposed to the chelator. Exposure to dexrazoxane increased the expression of the HIF-regulated, antiapoptotic proteins survivin, Mcl1 and haem oxygenase. CONCLUSIONS AND IMPLICATIONS: Our results showing HIF-dependent prevention of doxorubicin toxicity in dexrazoxane-treated H9c2 cardiomyocytes suggest that HIF activation may be a mechanism contributing to the protective effect of dexrazoxane against anthracycline cardiotoxicity.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Doxorrubicina/toxicidad , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Quelantes del Hierro/farmacología , Miocitos Cardíacos/efectos de los fármacos , Razoxano/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Perfilación de la Expresión Génica , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Unión Proteica , Ratas , Activación Transcripcional
16.
Int J Cardiol ; 152(2): 196-201, 2011 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-20692056

RESUMEN

OBJECTIVES: Dexrazoxane (DZR) is a clinically approved agent for preventive treatment of doxorubicin-induced cardiotoxicity. The objective of this study was to investigate the cardioprotective effects of DZR in a rat model of myocardial infarction (MI). METHODS: Sprague-Dawley rats were randomly divided into four groups: MI (n = 16), MI + DZR (n = 16), SHAM-operated (n = 14) and DZR-only (n = 9). MI animals were subjected to left anterior descending coronary artery ligation. DZR was administered as a single dose at 125 mg/kg intraperitoneally. Four weeks after treatment, cardiac function by echocardiography, infarct size, capillary density in the infarct border zone, bone marrow-derived endothelial progenitor cells (EPCs), and cardiac expression of Bax were measured. RESULTS: Our results demonstrated that MI animals had compromised heart parameters. DZR treatment in MI animals resulted in reduction in infarct size (P = 0.013) and improved cardiac functions in terms of fractional shortening (P = 0.004) and ejection fraction (P = 0.004). The capillary density (P = 0.008) and bone marrow-derived EPCs (P < 0.05) were higher in the MI + DZR group than those in the untreated MI group. Bax expression was down-regulated in heart tissues of MI + DZR animals (P = 0.043). CONCLUSIONS: Our study demonstrated that DZR exerted a cardioprotective effect in the rat model of MI, and the mechanism might be associated with anti-apoptosis and increased neovascularization.


Asunto(s)
Apoptosis/efectos de los fármacos , Cardiotónicos/farmacología , Infarto del Miocardio/patología , Neovascularización Fisiológica/efectos de los fármacos , Razoxano/farmacología , Animales , Recuento de Células Sanguíneas , Células de la Médula Ósea/citología , Capilares/fisiología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ecocardiografía , Células Endoteliales/metabolismo , Masculino , Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley , Células Madre/metabolismo , Volumen Sistólico , Proteína X Asociada a bcl-2/metabolismo
17.
Anticancer Agents Med Chem ; 10(7): 564-70, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20950258

RESUMEN

Bisdioxopiperazine (Biz) compounds, including ICRF-154 and razoxane (ICRF-159, Raz), are anticancer agents developed in the UK specifically targeting tumor metastases. Further three bisdioxopiperazine derivatives, bimolane (Bim), probimane (Pro) and MST-16, have been synthesized at the Shanghai Institute of Materia Medica, Chinese Academy of Sciences, PR China after 1980. Since metastases, the prevailing deadliest pathologic feature of cancer in clinics, have been the main obstacle in cancer therapy, antimetastatic effects and mechanisms of Biz compounds are interesting and significant topics of all time for researchers undergoing the investigations of metastases biology, treatments and patho-physiology. This review addresses and highlights the different inhibitions against metastases in vivo and molecular mechanisms in vitro of Biz compounds especially relating to the inhibitions of tumor metastasis including pathways of inhibitions against angiogenesis, topoisomerase II, calmodulin, sialic acid, fibrinogen, cell-movement and so on. We argue hererin that the systematic exploration of antimetastatic activity and mechanisms of Biz compounds seems to be a shortcut for a final solution of cancer therapy in the future.


Asunto(s)
Antineoplásicos/farmacología , Metástasis de la Neoplasia/tratamiento farmacológico , Piperazinas/farmacología , Razoxano/análogos & derivados , Razoxano/farmacología , Inhibidores de la Angiogénesis/farmacología , Animales , Línea Celular Transformada , Fibrinógeno/fisiología , Humanos , Ratones , Terapia Molecular Dirigida , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/fisiopatología
18.
Physiol Res ; 59(5): 831-836, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20406046

RESUMEN

The matrix metalloproteinases (MMPs) play a key role during cardiac remodeling. The aim of the study was to investigate the changes in collagenous proteins and MMPs in the model of non-ischemic, anthracycline-induced chronic cardiomyopathy in rabbits using both biochemical and histological approaches. The study was carried out in three groups of Chinchilla male rabbits: 1) daunorubicin (3 mg/kg, once weekly for 10 weeks), 2) control (saline in the same schedule), 3) daunorubicin with the cardioprotectant dexrazoxane (60 mg/kg, before each daunorubicin). Morphological changes in the myocardium of daunorubicin-treated animals were characterized by focal myocardial interstitial fibrosis of different intensity. The subsequent proliferation of the fibrotic tissue was marked by an increased content of both collagen types I and III, which resulted in their typical coexpression in the majority of bundles of fibers forming either smaller or larger scars. Biochemical analysis showed a significantly increased concentration of hydroxyproline, mainly in the pepsin-insoluble fraction of collagenous proteins, in the daunorubicin-treated group (1.42+/-0.12 mg/g) as compared with the control (1.03+/-0.04 mg/g) and dexrazoxane (1.07+/-0.07 mg/g) groups. Dexrazoxane co-administration remarkably reduced the cardiotoxic effects of daunorubicin to the extent comparable with the controls in all evaluated parameters. Using zymography, it was possible to detect only a gelatinolytic band corresponding to MMP-2 (MMP-9 activity was not detectable). However, no significant changes in MMP-2 activity were determined between individual groups. Immunohistochemical analysis revealed increased MMP-2 expression in both cardiomyocytes and fibroblasts. Thus, this study has revealed specific alterations in the collagen network in chronic anthracycline cardiotoxicity in relationship to the expression and activity of major MMPs.


Asunto(s)
Cardiomiopatías , Daunorrubicina/toxicidad , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Remodelación Ventricular/fisiología , Animales , Antibióticos Antineoplásicos/toxicidad , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Cardiomiopatías/prevención & control , Fármacos Cardiovasculares/farmacología , Enfermedad Crónica , Colágeno/metabolismo , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Fibrosis , Hidroxiprolina/metabolismo , Masculino , Miocardio/enzimología , Miocardio/patología , Conejos , Razoxano/farmacología
19.
Cancer Chemother Pharmacol ; 66(2): 303-14, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19915844

RESUMEN

PURPOSE: The antineoplastic anthracycline doxorubicin can induce a dose-dependent cardiomyopathy that limits the total cumulative dose prescribed to cancer patients. In both preclinical and clinical studies, pretreatment with dexrazoxane, an intracellular iron chelator, partially protects against anthracycline-induced cardiomyopathy. To identify potential additional cardioprotective treatment strategies, we investigated early doxorubicin-induced changes in cardiac gene expression. METHODS: Spontaneously hypertensive male rats (n = 47) received weekly intravenous injections of doxorubicin (3 mg/kg) or saline 30 min after pretreatment with dexrazoxane (50 mg/kg) or saline by intraperitoneal injection. Cardiac samples were analyzed 24 h after the first (n = 20), second (n = 13), or third (n = 14) intravenous injection on days 1, 8, or 15 of the study, respectively. RESULTS: Rats receiving three doses of doxorubicin had minimal myocardial alterations that were attenuated by dexrazoxane. Cardiac expression levels of genes associated with the Nrf2-mediated stress response were increased after a single dose of doxorubicin, but not affected by cardioprotectant pretreatment. In contrast, an early repressive effect of doxorubicin on transcript levels of genes associated with mitochondrial function was attenuated by dexrazoxane pretreatment. Dexrazoxane had little effect on gene expression by itself. CONCLUSIONS: Genomic analysis provided further evidence that mitochondria are the primary target of doxorubicin-induced oxidative damage that leads to cardiomyopathy and the primary site of cardioprotective action by dexrazoxane. Additional strategies that prevent the formation of oxygen radicals by doxorubicin in mitochondria may provide increased cardioprotection.


Asunto(s)
Antibióticos Antineoplásicos , Doxorrubicina , Cardiopatías , Miocardio , Animales , Masculino , Ratas , Antibióticos Antineoplásicos/toxicidad , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Doxorrubicina/toxicidad , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Cardiopatías/inducido químicamente , Cardiopatías/metabolismo , Cardiopatías/prevención & control , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Miocardio/metabolismo , Miocardio/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas Endogámicas SHR , Razoxano/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Troponina T/sangre , Troponina T/metabolismo , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/genética
20.
Cardiovasc Toxicol ; 10(1): 1-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19915982

RESUMEN

The use of the anticancer multikinase inhibitor sorafenib is associated with cardiac ischemia or infarction and an increase in hypertension. We investigated various mechanisms that might be responsible for its cardiotoxicity in a neonatal rat myocyte model. As measured by lactate dehydrogenase release, sorafenib treatment of myocytes caused dose-dependent damage at therapeutically relevant concentrations. It had been hypothesized that inhibition of RAF1 and BRAF kinases may be responsible for sorafenib induced cardiotoxicity. However, because sorafenib treatment did not inhibit phosphorylation of ERK (extracellular signal-regulated kinase), it was concluded that sorafenib did not exert its damaging effects through RAF inhibition of the RAF/MEK/ERK kinase cascade. The clinically approved doxorubicin cardioprotective agent dexrazoxane did not protect myocytes from damage. At lower sorafenib concentrations, at least, these results are consistent with sorafenib not being able to induce significant oxidative damage. In conclusion, given the extreme lack of kinase selectivity that sorafenib exhibits, it is likely that inhibition of kinases other than RAF, or combinations of kinases, contributes to the cardiotoxic effects of sorafenib.


Asunto(s)
Bencenosulfonatos/toxicidad , Miocitos Cardíacos/patología , Inhibidores de Proteínas Quinasas/toxicidad , Piridinas/toxicidad , Adenosina Trifosfato/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Bencenosulfonatos/antagonistas & inhibidores , Western Blotting , Cardiotónicos/farmacología , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Separación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnicas In Vitro , Quelantes del Hierro/farmacología , L-Lactato Deshidrogenasa/metabolismo , Microscopía Fluorescente , Niacinamida/análogos & derivados , Oxidación-Reducción , Compuestos de Fenilurea , Fosforilación , Piridinas/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Razoxano/farmacología , Sorafenib
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...