Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Bioorg Chem ; 102: 104067, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32663671

RESUMEN

Schistosomiasis is a neglected tropical disease mainly affecting the poorest tropical and subtropical areas of the world with the impressive number of roughly 200 million infections per year. Schistosomes are blood trematode flukes of the genus Schistosoma causing symptoms in humans and animals. Organ morbidity is caused by the accumulation of parasite eggs and subsequent development of fibrosis. If left untreated, schistosomiasis can result in substantial morbidity and even mortality. Praziquantel (PZQ) is the most effective and widely used compound for the treatment of the disease, in prevention and control programs in the last 30 years. Unfortunately, it has no effect on juvenile immature schistosomes and cannot prevent reinfection or interfere with the schistosome life cycle; moreover drug-resistance represents a serious threat. The search for an alternative or complementary treatment is urgent and drug repurposing could accelerate a solution. The anti-anginal drug perhexiline maleate (PHX) has been previously shown to be effective on larval, juvenile, and adult stages of S. mansoni and to impact egg production in vitro. Since PHX is a racemic mixture of R-(+)- and S-(-)-enantiomers, we designed and realized a stereoselective synthesis of both PHX enantiomers and developed an analytical procedure for the direct quantification of the enantiomeric excess also suitable for semipreparative separation of PHX enantiomers. We next investigated the impact of each enantiomer on viability of newly transformed schistosomula (NTS) and worm pairs of S. mansoni as well as on egg production and vitellarium morphology by in vitro studies. Our results indicate that the R-(+)-PHX is mainly driving the anti-schistosomal activity but that also the S-(-)-PHX possesses a significant activity towards S. mansoni in vitro.


Asunto(s)
Perhexilina/análogos & derivados , Schistosoma mansoni/efectos de los fármacos , Animales , Larva , Estructura Molecular , Perhexilina/uso terapéutico , Estereoisomerismo , Relación Estructura-Actividad
2.
Cardiovasc Drugs Ther ; 30(4): 399-405, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27106834

RESUMEN

Approaches to the pharmacotherapy of angina pectoris have previously centred on the concept that a transient imbalance between myocardial oxygen "demand" and supply within the myocardium can best be addressed by reducing demand (for example, with ß-adrenoceptor antagonist) or by increasing availability of blood (via coronary vasomotor reactivity adjustment or coronary revascularization). However, this principle is potentially challenged by the emergence of cases of angina unsuitable for such therapies (for example because of concomitant severe systolic heart failure) and by the recognition that impaired myocardial energetics may precipitate angina in the absence of fixed or variable coronary obstruction (for example in hypertrophic cardiomyopathy). The past 20 years have seen the re-emergence of a class of anti-anginal agents which act primarily by improving efficiency of myocardial oxygen utilization, and thus can correct impaired energetics, simultaneously treating angina and heart failure symptoms. We review the principles underlying the safe use of such agents, beginning with the prototype drug perhexiline maleate, which despite complex pharmacokinetics and potential hepato- or neuro-toxicity has emerged as an attractive management option in many "complicated" cases of angina pectoris.


Asunto(s)
Angina de Pecho/tratamiento farmacológico , Fármacos Cardiovasculares/uso terapéutico , Miocardio/metabolismo , Perhexilina/análogos & derivados , Angina de Pecho/metabolismo , Animales , Fármacos Cardiovasculares/farmacología , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Humanos , Mitocondrias/metabolismo , Perhexilina/farmacología , Perhexilina/uso terapéutico
3.
Ther Drug Monit ; 37(6): 821-6, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25774703

RESUMEN

BACKGROUND: The polymorphic nature of cytochrome P450 2D6 has made therapeutic drug monitoring of the anti-anginal agent perhexiline a compulsory step in reducing adverse events associated with plasma concentrations above the therapeutic range (0.15-0.60 mg/L). The aim of this study was to develop a high-performance liquid chromatography-mass spectrometry/mass spectrometry method for the determination of plasma perhexiline concentrations and its major metabolite cis-hydroxy-perhexiline to reduce sample extraction procedures and improve sample turnaround times. METHODS: The method was validated by determining the precision and accuracy of calibrators and quality control material, comparing quality assurance program samples and patient samples measured by a previously reported liquid-liquid extraction fluorescence (FL) detection high-performance liquid chromatography method and performing matrix effects investigations. RESULTS: Replicates of calibrators at concentrations of 3.00 and 0.05 mg/L demonstrated imprecision of <10.8% and inaccuracy of <8.2% for perhexiline and <10.1% and <4.5% for cis-hydroxy-perhexiline, respectively. All samples measured by the 2 methods (n = 102) demonstrated Deming regression of perhexiline = 1.20 FL + 0.00 (Sy.x = 0.08, 1/slope = 0.67); cis-hydroxy-perhexiline = 1.48 FL - 0.20 (Sy.x = 0.40, 1/slope = 0.67). CONCLUSIONS: The assay performance was deemed acceptable and integrated into the routine therapeutic drug monitoring program of the department.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Monitoreo de Drogas/métodos , Perhexilina/análogos & derivados , Espectrometría de Masas en Tándem/métodos , Calibración , Citocromo P-450 CYP2D6/genética , Humanos , Extracción Líquido-Líquido , Perhexilina/sangre , Polimorfismo Genético , Reproducibilidad de los Resultados
4.
J Antimicrob Chemother ; 69(4): 1035-44, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24284780

RESUMEN

OBJECTIVES: Biofilms of Candida species, often formed on medical devices, are generally resistant to currently available antifungal drugs. The aim of this study was to identify compounds that increase the activity of amphotericin B and caspofungin, commonly used antifungal agents, against Candida biofilms. METHODS: A library containing off-patent drugs was screened for compounds, termed enhancers, that increase the in vitro activity of amphotericin B against Candida albicans biofilms. Biofilms were grown in 96-well plates and growth was determined by the cell titre blue assay. Synergy between identified enhancers and antifungal agents was further characterized in vitro using fractional inhibitory concentration index (FICI) values and in vivo using a worm biofilm infection model. In light of the application of these enhancers onto implants, their possible effect on the growth potential of MG63 osteoblast-like cells was assessed. RESULTS: Pre-incubation of C. albicans biofilms with subinhibitory concentrations of the enhancers drospirenone, perhexiline maleate or toremifene citrate significantly increased the activity of amphotericin B or caspofungin (FICI  < 0.5) against C. albicans and Candida glabrata biofilms. Moreover, these enhancers did not affect the growth potential of osteoblasts. Interestingly, toremifene citrate also enhanced the in vitro activity of caspofungin in a mixed biofilm consisting of C. albicans and Staphylococcus epidermidis. Furthermore, we demonstrate synergy between toremifene citrate and caspofungin in an in vivo worm C. albicans biofilm infection model. CONCLUSIONS: Our data demonstrate an in vitro and in vivo enhancement of the antibiofilm activity of caspofungin by toremifene citrate. Furthermore, our results pave the way for implant-related applications of the identified enhancers.


Asunto(s)
Anfotericina B/farmacología , Antifúngicos/farmacología , Biopelículas/efectos de los fármacos , Candida albicans/efectos de los fármacos , Reposicionamiento de Medicamentos , Sinergismo Farmacológico , Equinocandinas/farmacología , Androstenos/farmacología , Animales , Caenorhabditis elegans/microbiología , Candida albicans/fisiología , Caspofungina , Lipopéptidos , Pruebas de Sensibilidad Microbiana , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Perhexilina/análogos & derivados , Perhexilina/farmacología , Toremifeno/farmacología
5.
J Bone Miner Metab ; 31(1): 26-33, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23011467

RESUMEN

Fibrodysplasia ossificans progressiva (FOP) is a rare autosomal dominant disorder characterized by progressive heterotopic ossification. FOP is caused by a gain-of-function mutation in ACVR1 encoding the bone morphogenetic protein type II receptor, ACVR1/ALK2. The mutant receptor causes upregulation of a transcriptional factor, Id1. No therapy is available to prevent the progressive heterotopic ossification in FOP. In an effort to search for clinically applicable drugs for FOP, we screened 1,040 FDA-approved drugs for suppression of the Id1 promoter activated by the mutant ACVR1/ALK2 in C2C12 cells. We found that that two antianginal agents, fendiline hydrochloride and perhexiline maleate, suppressed the Id1 promoter in a dose-dependent manner. The drugs also suppressed the expression of native Id1 mRNA and alkaline phosphatase in a dose-dependent manner. Perhexiline but not fendiline downregulated phosphorylation of Smad 1/5/8 driven by bone morphogenetic protein (BMP)-2. We implanted crude BMPs in muscles of ddY mice and fed them fendiline or perhexiline for 30 days. Mice taking perhexiline showed a 38.0 % reduction in the volume of heterotopic ossification compared to controls, whereas mice taking fendiline showed a slight reduction of heterotopic ossification. Fendiline, perhexiline, and their possible derivatives are potentially applicable to clinical practice to prevent devastating heterotopic ossification in FOP.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Fendilina/farmacología , Células Musculares/metabolismo , Miositis Osificante/tratamiento farmacológico , Osificación Heterotópica/tratamiento farmacológico , Osteoblastos/metabolismo , Perhexilina/análogos & derivados , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Animales , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Proteína 1 Inhibidora de la Diferenciación/biosíntesis , Proteína 1 Inhibidora de la Diferenciación/genética , Ratones , Ratones Mutantes , Células Musculares/patología , Mutación , Miositis Osificante/genética , Miositis Osificante/metabolismo , Miositis Osificante/patología , Osificación Heterotópica/metabolismo , Osificación Heterotópica/patología , Osteoblastos/patología , Perhexilina/farmacología , Regiones Promotoras Genéticas/genética , Proteínas Smad/genética , Proteínas Smad/metabolismo
6.
Ther Drug Monit ; 34(2): 227-31, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22322401

RESUMEN

BACKGROUND: Concomitant treatment with amiodarone and perhsexiline has been considered to be relatively contraindicated because of the hypothetical risk of potentiated adverse effects mediated by additive inhibition of carnitine palmitoyl transferase 1. AIM: To study the prevalence of adverse effects associated with the concomitant use of perhexiline and amiodarone. METHODS: A retrospective analysis of a single hospital database of patients receiving perhexiline and amiodarone between July 2009 and April 2011. Files were reviewed for short- and long-term adverse effects requiring drug cessation. Glucose concentration, gamma glutamyl transferase activity. and perhexiline blood concentrations were recorded. RESULTS: We identified 26 patients concomitantly treated with perhexiline and amiodarone, 20 on a long-term basis. In 6 cases, amiodarone was introduced on top of preceding perhexiline. In none of the cases were drugs stopped because of adverse effects. Although blood glucose concentrations fell significantly 48 hours postadmission to hospital, this seems to reflect the resolution of "admission hyperglycemia" rather than onset of hypoglycemia; the latter was rare (5 patients), mild, and clinically silent. In 4 patients, gamma glutamyl transferase approximately doubled. CONCLUSIONS: Traditionally, concomitant treatment with amiodarone and perhexiline has been considered to be relatively contraindicated on the basis of the theoretical potential for synergistic toxicity. This cohort of 26 patients received this concomitant treatment without any excess of major adverse reactions. Our findings suggest that concomitant treatment with perhexiline and amiodarone may be safe in the setting of (1) previous tolerance of either agent, and (2) titration of plasma perhexiline concentrations to guide therapy.


Asunto(s)
Amiodarona/efectos adversos , Perhexilina/análogos & derivados , Vasodilatadores/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Amiodarona/administración & dosificación , Amiodarona/uso terapéutico , Glucemia/efectos de los fármacos , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Bases de Datos Factuales , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Perhexilina/administración & dosificación , Perhexilina/efectos adversos , Perhexilina/uso terapéutico , Prevalencia , Estudios Retrospectivos , Vasodilatadores/administración & dosificación , Vasodilatadores/uso terapéutico , gamma-Glutamiltransferasa/efectos de los fármacos , gamma-Glutamiltransferasa/metabolismo
7.
PLoS Negl Trop Dis ; 14(10): e0008767, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33044962

RESUMEN

Schistosomiasis is one of the most devastating neglected tropical parasitic diseases caused by trematodes of the genus Schistosoma. Praziquantel (PZQ) is today the only drug used in humans and animals for the treatment of schistosomiasis but unfortunately it is poorly effective on larval and juvenile stages of the parasite. Therefore, it is urgent the discovery of new drug targets and compounds. We have recently showed that the anti-anginal drug perhexiline maleate (PHX) is very active on multiple developmental stages of Schistosoma mansoni in vitro. It is well known that PHX impacts the lipid metabolism in mammals, but the final target on schistosomes still remains unknown. The aim of this study was to evaluate the ability of 1H nuclear magnetic resonance (NMR) spectroscopy in revealing metabolic perturbations due to PHX treatment of S. mansoni adult male worms. The effects of PHX were compared with the ones induced by vehicle and gambogic acid, in order to detect different metabolic profiles and specificity of the PHX action. Remarkably a list of metabolites associated to PHX-treatment was identified with enrichment in several connected metabolic pathways including also the Kennedy pathway mediating the glycerophospholipid metabolism. Our study represents the first 1H-NMR metabolomic approach to characterize the response of S. mansoni to drug treatment. The obtained "metabolic fingerprint" associated to PHX treatment could represent a strategy for displaying cellular metabolic changes for any given drug and to compare compounds targeting similar or distinct biochemical pathways.


Asunto(s)
Antihelmínticos/administración & dosificación , Monitoreo de Drogas/métodos , Espectroscopía de Protones por Resonancia Magnética/métodos , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/metabolismo , Esquistosomiasis mansoni/tratamiento farmacológico , Adulto , Animales , Femenino , Humanos , Masculino , Metaboloma/efectos de los fármacos , Ratones Endogámicos ICR , Perhexilina/administración & dosificación , Perhexilina/análogos & derivados , Praziquantel/administración & dosificación , Schistosoma mansoni/crecimiento & desarrollo , Esquistosomiasis mansoni/parasitología
8.
Cell Physiol Biochem ; 24(3-4): 167-76, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19710531

RESUMEN

Peptidoglycans (PGN) from bacterial cell walls may modify the course of an infection with bacterial pathogens. The present study explored the effect of PGN on cytosolic Ca2+ activity, cytokine production and phagocytosis of mouse dendritic cells (DCs), essential cells in the initiation and direction of antigen-specific T cell responses. Exposure of DCs to PGN was followed by a rapid increase in cytosolic Ca2+ activity ([Ca2+]i), which was due to Ca2+ release from intracellular stores and influx of extracellular Ca2+ across the cell membrane. In DCs isolated from Toll-like receptor 2 (TLR2) deficient mice the effect of PGN on [Ca2+]i was dramatically impaired. The PGN-induced increase of [Ca2+]i was dependent on voltage-gated K+ (Kv) channel activity. PGN-induced increase of [Ca2+]i was significantly blunted by margatoxin (MgTx) and perhexiline maleate (PM), inhibitors of Kv1.3 and Kv1.5, respectively. PGN further stimulated the release of tumour necrosis factor alpha (TNFalpha), interleukin-12 (IL-12) and interleukin-10 (IL-10), an effect significantly blunted by PM and the specific blocker of store-operated Ca2+ channels SKF-96365. Moreover, phagocytic capacity was dramatically increased in PGN-stimulated DCs in the presence of either Kv channel inhibitors or SKF-96365. The observations disclose Ca2+ and Kv channel-dependent cytokine production and phagocytosis in PGN-stimulated DCs.


Asunto(s)
Calcio/metabolismo , Células Dendríticas/efectos de los fármacos , Peptidoglicano/farmacología , Staphylococcus aureus/química , Animales , Bloqueadores de los Canales de Calcio/farmacología , Células Cultivadas , Citocinas/análisis , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Electrofisiología , Femenino , Fémur/citología , Colorantes Fluorescentes/metabolismo , Fura-2/metabolismo , Imidazoles/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Neurotoxinas/farmacología , Técnicas de Placa-Clamp , Perhexilina/análogos & derivados , Perhexilina/farmacología , Fagocitosis/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/metabolismo , Venenos de Escorpión/farmacología , Tibia/citología , Receptor Toll-Like 2/inmunología
9.
Toxicol In Vitro ; 22(6): 1511-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18603402

RESUMEN

Mallory-Denk bodies (MDB) are hepatocyte inclusions containing cytokeratin 8 (CK8) which can develop, along with other steatohepatitis lesions, in patients treated with amiodarone, perhexiline maleate or 4,4'-diethylaminoethoxyhexestrol. These drugs accumulate lipids, whose subsequent peroxidation liberates reactive by-products, like malondialdehyde (MDA). The formation of MDB has been previously reproduced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine or griseofulvin administration which cross-link CK8 by tissue transglutaminase, thus forming an entangled network, from which MDB progressively arise. The present study depicts the mechanisms initiating MDB formation by steatohepatitis-inducing drugs. Short incubation of hepatocytes with amiodarone (50 microM), 4,4'-diethylaminoethoxyhexestrol (50 microM) or perhexiline maleate (25 microM) increased the pool of CK8 monomers and increased cell calcium to activate Ca(++)-dependent transglutaminases which cross-linked the CK8 monomers into CK8-containing oligomers. The present study also provides the first evidence that MDA might directly participate in MDB formation, as this reactive agent cross-linked purified CK8 or albumin in vitro, disrupted the cytokeratin network of isolated hepatocytes, and bridged CK8 molecules. In conclusion, steatohepatitis-inducing drugs increase cell calcium and activate tissue transglutaminase, which cross-links CK8 to form a molecular scaffold, from which MDB might secondarily arise. Malondialdehyde also cross-links CK8, albeit through a different mechanism, and might also contribute to MDB formation.


Asunto(s)
Hepatocitos/efectos de los fármacos , Cuerpos de Inclusión/efectos de los fármacos , Queratina-8/efectos de los fármacos , Malondialdehído/metabolismo , Amiodarona/toxicidad , Animales , Calcio/metabolismo , Hígado Graso/inducido químicamente , Proteínas de Unión al GTP/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Hepatocitos/metabolismo , Hexestrol/análogos & derivados , Hexestrol/toxicidad , Cuerpos de Inclusión/metabolismo , Queratina-8/metabolismo , Masculino , Perhexilina/análogos & derivados , Perhexilina/toxicidad , Proteína Glutamina Gamma Glutamiltransferasa 2 , Proteínas , Ratas , Ratas Sprague-Dawley , Transglutaminasas/efectos de los fármacos , Transglutaminasas/metabolismo
10.
J Med Chem ; 60(7): 2780-2789, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28277663

RESUMEN

We designed and synthesized perhexiline analogues that have the same therapeutic profile as the parent cardiovascular drug but lacking its metabolic liability associated with CYP2D6 metabolism. Cycloalkyl perhexiline analogues 6a-j were found to be unsuitable for further development, as they retained a pharmacokinetic profile very similar to that shown by the parent compound. Multistep synthesis of perhexiline analogues incorporating fluorine atoms onto the cyclohexyl ring(s) provided a range of different fluoroperhexiline analogues. Of these, analogues 50 (4,4-gem-difluoro) and 62 (4,4,4',4'-tetrafluoro) were highly stable and showed greatly reduced susceptibility to CYP2D6-mediated metabolism. In vitro efficacy studies demonstrated that a number of derivatives retained acceptable potency against CPT-1. Having the best balance of properties, 50 was selected for further evaluation. Like perhexiline, it was shown to be selectively concentrated in the myocardium and, using the Langendorff model, to be effective in improving both cardiac contractility and relaxation when challenged with high fat buffer.


Asunto(s)
Fármacos Cardiovasculares/química , Fármacos Cardiovasculares/farmacocinética , Perhexilina/análogos & derivados , Perhexilina/farmacocinética , Animales , Fármacos Cardiovasculares/metabolismo , Fármacos Cardiovasculares/farmacología , Citocromo P-450 CYP2D6/metabolismo , Halogenación , Corazón/efectos de los fármacos , Corazón/fisiología , Humanos , Masculino , Ratones Endogámicos BALB C , Contracción Miocárdica/efectos de los fármacos , Miocardio/metabolismo , Perhexilina/metabolismo , Perhexilina/farmacología
11.
Artículo en Inglés | MEDLINE | ID: mdl-16837252

RESUMEN

The use of perhexiline (PHX) is limited by hepatic and neurological toxicity associated with elevated concentrations in plasma that are the result of polymorphism of the cytochrome P450 2D6 isoform (CYP2D6). PHX is cleared by hepatic oxidation that produces three 4-monohydroxy metabolites: cis-OH-PHX, trans1-OH-PHX and trans2-OH-PHX. The current study describes an HPLC-fluorescent method utilising pre-column derivatization with dansyl chloride. Following derivatization, the metabolites were resolved on a C18 column with a gradient elution using a mobile phase composed of methanol and water. The method described is suitable for the quantification of the metabolites in human plasma and urine following clinical doses and for kinetic studies using human liver microsomes. The method demonstrates sufficient sensitivity, accuracy and precision between 5.0 and 0.01, 50.0 and 0.2 and 1.0 and 0.005 mg/l in human plasma, urine and liver microsomes, respectively, with intra-assay coefficients of variation and bias <15%, except at the lowest limit of quantification (<20%). The inter-assay coefficients of variation and bias were <15%. The application of this method to plasma and urine samples of five CYP2D6 extensive metaboliser (EM) patients at steady state with respect to PHX dosing determined that the mean (+/-S.D.) renal clearances of trans1-OH-PHX and cis-OH-PHX were 1.58+/-0.35 and 0.16+/-0.06l/h, respectively. The mean (+/-S.D.) dose recovered in urine as free and glucuronidated 4-monohydroxy PHX metabolites was 20.6+/-11.6%.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Microsomas Hepáticos/química , Perhexilina/análogos & derivados , Perhexilina/metabolismo , Humanos
12.
Cancer Res ; 44(1): 144-8, 1984 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-6690032

RESUMEN

The effects of perhexiline maleate on growth and drug sensitivity were studied in the P388 murine leukemia cell line and in an anthracycline-resistant subline (P388/ADR). At noninhibitory concentrations, perhexiline maleate markedly increased the sensitivity of P388/ADR cells to doxorubicin but did not have such an effect on anthracycline-sensitive cells. The effects of perhexiline maleate on P388/ADR cells were reversible. Perhexiline maleate also increased the accumulation of another anthracycline, daunorubicin, in P388/ADR cells but did not increase its accumulation in the anthracycline-sensitive cells. Perhexiline maleate did not affect the sensitivity of either cell line to methotrexate or to 6-mercaptopurine. However, its effects on the sensitivity and on drug accumulation of vinblastine, a drug to which P388/ADR cells are cross-resistant, were similar to those observed for the anthracyclines. Although perhexiline maleate has been reported to be a calcium antagonist in other systems, our data do not suggest that this mechanism is involved in its enhancement of the sensitivity of P388/ADR cells to doxorubicin. We suggest instead that this effect might be associated with alterations of cell lipid metabolism induced by perhexiline maleate.


Asunto(s)
Doxorrubicina/toxicidad , Leucemia P388/fisiopatología , Leucemia Experimental/fisiopatología , Perhexilina/análogos & derivados , Animales , Transporte Biológico , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Daunorrubicina/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Cinética , Ratones , Ratones Endogámicos , Perhexilina/toxicidad , Vinblastina/toxicidad
13.
Cancer Res ; 50(17): 5558-66, 1990 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-2386962

RESUMEN

In the perspective of increasing the clinical potential of ricin A chain immunotoxins (RTA-ITs), perhexiline (Pex) and four structural analogues (Pex 2, Pex 3, Pex 7, and Pex 11) were evaluated for their ability to enhance RTA-IT activity in vitro. Only perhexiline significantly enhanced the cytotoxic activity of anti-CD5 RTA-ITs, T101 and T101-F(ab')2, on CEM III cell line (30- to 2000-fold), and of anti-HLA-DR RTA-IT, HNC-241, on both RAJI cell line (greater than 100-fold) and two immortalized cell lines originating from patients suffering from B-cell chronic lymphocytic leukemia, EHEB and FS2 D5 (10-fold). On 16 consecutive fresh B-cell chronic lymphocytic leukemia cell samples, significant T101-F(ab')2 RTA-IT and HNC-241 RTA-IT enhancement was observed with perhexiline which was comparable to that of NH4Cl and monensin. Perhexiline almost completely blocked RTA-IT intracellular degradation and profoundly modified its routing. These observations were linked to perhexiline-induced lipidosis via inhibition of sphingomyelinase activity. In conclusion, since the concentrations used are relevant with the pharmacokinetics of this agent, perhexiline appears to be a promising agent for in vivo enhancement of ricin A chain immunotoxins.


Asunto(s)
Inmunotoxinas/farmacología , Leucemia Linfocítica Crónica de Células B/patología , Perhexilina/farmacología , Ricina/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Cloruro de Amonio/farmacología , Anticuerpos Monoclonales , Línea Celular , Humanos , Cinética , Leucemia Linfocítica Crónica de Células B/sangre , Monensina/farmacología , Proteínas de Neoplasias/biosíntesis , Perhexilina/análogos & derivados , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/ultraestructura , Verapamilo/farmacología
14.
PLoS Negl Trop Dis ; 10(8): e0004928, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27518281

RESUMEN

BACKGROUND: Schistosomiasis, one of the world's greatest human neglected tropical diseases, is caused by parasitic trematodes of the genus Schistosoma. A unique feature of schistosome biology is that the induction of sexual maturation as well as the maintenance of the differentiation status of female reproductive organs and egg production, necessary for both disease transmission and pathogenesis, are strictly dependent on the male. The treatment and most control initiatives of schistosomiasis rely today on the long-term application of a single drug, praziquantel (PZQ), mostly by campaigns of mass drug administration. PZQ, while very active on adult parasites, has much lower activity against juvenile worms. Monotherapy also favors the selection of drug resistance and, therefore, new drugs are urgently needed. METHODS AND FINDINGS: Following the screening of a small compound library with an ATP-based luminescent assay on Schistosoma mansoni schistosomula, we here report the identification and characterization of novel antischistosomal properties of the anti-anginal drug perhexiline maleate (PHX). By phenotypic worm survival assays and confocal microscopy studies we show that PHX, in vitro, has a marked lethal effect on all S. mansoni parasite life stages (newly transformed schistosomula, juvenile and adult worms) of the definitive host. We further demonstrate that sub-lethal doses of PHX significantly impair egg production and lipid depletion within the vitellarium of adult female worms. Moreover, we highlighted tegumental damage in adult male worms and remarkable reproductive system alterations in both female and male adult parasites. The in vivo study in S. mansoni-patent mice showed a notable variability of worm burdens in the individual experiments, with an overall minimal schistosomicidal effect upon PHX treatment. The short PHX half-life in mice, together with its very high rodent plasma proteins binding could be the cause of the modest efficacy of PHX in the schistosomiasis murine model. CONCLUSIONS/SIGNIFICANCE: Overall, our data indicate that PHX could represent a promising starting point for novel schistosomicidal drug discovery programmes.


Asunto(s)
Genitales/efectos de los fármacos , Perhexilina/análogos & derivados , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/ultraestructura , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomicidas/farmacología , Animales , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Femenino , Semivida , Humanos , Estadios del Ciclo de Vida/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Perhexilina/farmacología , Praziquantel/farmacología
15.
Sci Rep ; 5: 18144, 2015 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-26674674

RESUMEN

High Risk Neuroblastoma (HR-NB) is a pediatric cancer characterized by high malignancy and remarkable cell heterogeneity within the tumour nodules. In a recent study, we demonstrated that in vitro and in vivo over-expression of the non-coding RNA NDM29 (neuroblastoma differentiation marker 29) induces NB cell differentiation, dramatically reducing their malignancy. Among gene expression changes, differentiated phenotype induced by NDM29 is characterized by decrease of the expression of ABC transporters responsible for anticancer drug resistance. Thus, the pharmacological induction of NDM29, in principle, might represent a possible novel strategy to increase cytotoxic drug responses. In this work, we identify a small molecule able to induce the expression of NDM29 in NB cells, conferring to malignant cells increased susceptibility to cisplatin cytotoxic effects. We demonstrate that the pharmacological induction of NDM29 expression in vivo enhances the antitumoral effects of chemotherapy specifically on tumour initiating/cancer stem cells sub-population, usually refractory to therapies and responsible for tumour relapse. In summary, we suggest a novel therapeutical approach possibly useful to treat very aggressive NB cases with poor prognosis. This novel pharmacological strategy aims to promote differentiation of "stem-like" cells to render them more susceptible to the killing action of cytotoxic anticancer drugs.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/genética , ARN no Traducido/genética , Transportadoras de Casetes de Unión a ATP/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cisplatino/administración & dosificación , Cisplatino/farmacología , Sinergismo Farmacológico , Humanos , Ratones Endogámicos NOD , Ratones SCID , Neuroblastoma/patología , Perhexilina/administración & dosificación , Perhexilina/análogos & derivados , Perhexilina/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Interferon Cytokine Res ; 22(3): 343-50, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12034042

RESUMEN

The novel colony-stimulating factor (CSF) inducer leustroducsin B (LSN-B), which was isolated from Streptomyces platensis, has been shown to have potent cytokine-inducing activities in clonal human bone marrow-derived stromal cell line KM-102 and in primary human bone marrow-derived stromal cells. In this study, we investigated the signal transduction pathway of LSN-B using luciferase expression plasmids linked to the 5'-flanking region of interleukin-8 (IL-8) and that of the IL-11 gene. In KM-102 cells, LSN-B induced luciferase activity both in the wild-type and in the activated protein 1 (AP-1) site point-mutated IL-8 promoter. The mutation in the nuclear factor-kappaB (NF-kappaB) site abrogated LSN-B-stimulated induction of the reporter gene. LSN-B-inducing activity was inhibited by (1) N-acetyl-L-cysteine, a well-characterized antioxidant, (2) cationic amphiphilic drugs, inhibitors of acidic sphingomyelinase (A-SMase), and (3) D609, an inhibitor of phosphatidylcholine-specific phospholipase C (PC-PLC). These observations suggest that LSN-B potentiates the A-SMase-mediated signaling pathway to stimulate NF-kappaB. In contrast, LSN-B did not induce IL-11 promoter-driven luciferase activity. The observed increase in IL-11 mRNA stability by LSN-B indicates that the inducible production of IL-11 by LSN-B is regulated at the posttranscriptional level. In addition, inhibition of LSN-B-mediated induction of IL-11 production by cationic amphiphilic drugs and D609 in KM-102 cells demonstrates that increased IL-11 mRNA stability by LSN-B might be mediated via NF-kappaB activation. From these results, we suggest that LSN-B induces cytokine production through at least two separate mechanisms, at the transcriptional level and at the posttranscriptional level via NF-kappaB activation.


Asunto(s)
Células de la Médula Ósea/metabolismo , Lactonas/farmacología , FN-kappa B/metabolismo , Compuestos Organofosforados/farmacología , Perhexilina/análogos & derivados , Esfingomielina Fosfodiesterasa/metabolismo , Células del Estroma/metabolismo , Acetilcisteína/farmacología , Antioxidantes/farmacología , Células de la Médula Ósea/citología , Hidrocarburos Aromáticos con Puentes/farmacología , Línea Celular , Células Clonales , Desipramina/farmacología , Genes Reporteros , Humanos , Interleucina-11/análisis , Interleucina-11/biosíntesis , Interleucina-11/genética , Interleucina-8/análisis , Interleucina-8/genética , Mutagénesis Sitio-Dirigida , FN-kappa B/genética , Norbornanos , Perhexilina/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Mutación Puntual , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Pironas , Transducción de Señal , Células del Estroma/citología , Tiocarbamatos , Tionas/farmacología , Activación Transcripcional/efectos de los fármacos
17.
J Med Chem ; 25(6): 709-14, 1982 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-6124638

RESUMEN

A series of 24 cyclohexylaralkylamine derivatives related to perhexiline has been synthesized and screened for cardiovascular activity. All the compounds contained an exocyclic amine which was substituted either by an alkyl, cycloalkyl, or aralkyl group. In the hope of further reducing toxicity, the synthesis of p-tolyl- and p-hydroxyphenyl derivatives 23 and 24 was undertaken. The effect of separating the cyclohexylamine moiety with respect to the aromatic nucleus has been systematically examined. The pharmacological investigations were directed to a search for compounds having an activity better than perhexiline according to the following order of criteria: (1) alpha-adrenolytic activity; (2) increase of coronary blood flow; (3) calcium antagonism. Several compounds were more potent and exhibited lower toxicity than perhexiline. Further detailed pharmacological investigations (tension time index and decreased cardiac work) have led to the selection of N,2-dicyclohexyl-2-phenethylamine (3) for clinical trials, which are now under way.


Asunto(s)
Fármacos Cardiovasculares/síntesis química , Perhexilina/síntesis química , Piperidinas/síntesis química , Antagonistas Adrenérgicos alfa/síntesis química , Animales , Bloqueadores de los Canales de Calcio/síntesis química , Fenómenos Químicos , Química , Circulación Coronaria/efectos de los fármacos , Perros , Femenino , Masculino , Perhexilina/análogos & derivados , Perhexilina/farmacología , Ratas , Porcinos
18.
J Med Chem ; 31(12): 2289-96, 1988 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2903931

RESUMEN

In the hope of reducing the toxicity of perhexiline, a series of 27 cyclohexylaralkylamines II based on the "soft drug" concept and incorporating an amide function were synthesized. In a preliminary screening, compounds were evaluated for their alpha-adrenolytic activities. Several derivatives, especially N-(cyclohexylphenylmethyl)-2-(cyclohexyl-methylamino)acetamide (3), N-(cyclohexylphenylmethyl)-2-(homoveratrylmethylamino)acetam ide (7), and N-[2-(cyclohexylamino)ethyl]-alpha-cyclohexylbenzeneacetamide (23) had the same activity range as perhexiline in vitro in rat aorta strips. The in vitro metabolism of these three molecules was then investigated and compared to that of perhexiline. The effect upon the alpha-adrenolytic activity of introducing various N-aralkylamine groups on II was examined. Structure/activity relationships are discussed.


Asunto(s)
Amidas/síntesis química , Perhexilina/análogos & derivados , Antagonistas Adrenérgicos alfa/metabolismo , Amidas/farmacología , Animales , Aorta , Disponibilidad Biológica , Fenómenos Químicos , Química , Cromatografía Líquida de Alta Presión , Técnicas In Vitro , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Perhexilina/metabolismo , Perhexilina/farmacología , Ratas , Relación Estructura-Actividad
19.
Br J Pharmacol ; 127(1): 243-51, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10369479

RESUMEN

Perhexiline has been used as an anti-anginal agent for over 25 years, and is known to cause QT prolongation and torsades de pointes. We hypothesized that the cellular basis for these effects was blockade of I(Kr). A stable transfection of HERG into a CHO-K1 cell line produced a delayed rectifier, potassium channel with similar properties to those reported for transient expression in Xenopus oocytes. Perhexiline caused voltage- and frequency-dependent block of HERG (IC50 7.8 microM). The rate of inactivation was increased and there was a 10 mV hyperpolarizing shift in the voltage-dependence of steady-state inactivation, suggestive of binding to the inactivated state. In conclusion, perhexiline potently inhibits transfected HERG channels and this is the probable mechanism for QT prolongation and torsades de pointes. Channel blockade shows greatest affinity for the inactivated state.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Proteínas de Transporte de Catión , Perhexilina/análogos & derivados , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/metabolismo , Algoritmos , Animales , Antiarrítmicos/farmacología , Células CHO , Cricetinae , Estimulación Eléctrica , Canales de Potasio Éter-A-Go-Go , Técnicas In Vitro , Potenciales de la Membrana , Oocitos , Técnicas de Placa-Clamp , Perhexilina/farmacología , Piperidinas/farmacología , Canales de Potasio/efectos de los fármacos , Piridinas/farmacología , Xenopus laevis
20.
Biochem Pharmacol ; 35(14): 2301-7, 1986 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-3729987

RESUMEN

In order to find a substitute for perhexiline maleate, an antiangina drug, for which several side effects due to poor hydroxylation have been reported, an arylalkylamine series with antianginal properties has been synthesized. The aim of the present work was to select a more rapidly hydroxylated compound than perhexiline maleate in this series. Two criteria have been retained. The binding of the molecules to liver microsomal cytochrome P-450 and their rate of hydroxylation were both studied in vitro using phenobarbital induced rat liver microsomes. Incubation with cofactors and extraction procedures have been tested on one of the molecules of the series taken as example: N-2-dicyclohexyl-2-phenethylamine. All of the molecules tested in the series substrates were type I substrates; nevertheless, no correlation was found between binding on cytochrome P-450 and oxidative metabolism of the drugs. Two of the studied molecules were more easily hydroxylated than the others and than perhexiline maleate: (N-2-dicyclohexyl-2-phenethylamine) and (II) (N-cyclohexyl-2-diphenylethylamine) with the following respective kinetics: Apparent Vmax: 0.073 and 0.32 units, apparent Km: 6.9 X 10(-5) M and 22.2 X 10(-5) M.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Ciclohexanos/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Perhexilina/análogos & derivados , Animales , Hidroxilación , Cinética , Masculino , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Perhexilina/farmacología , Fenetilaminas/metabolismo , Fenobarbital/farmacología , Ratas , Ratas Endogámicas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda