Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Negat Results Biomed ; 8: 2, 2009 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-19138416

RESUMEN

BACKGROUND: Oxaliplatin is a platinum-based chemotherapeutic drug. Neurotoxicity is the dose-limiting side effect. Previous investigations have reported that acute neurotoxicity could be mediated via voltage-gated ion channels. A possible mechanism for some of the effects is a modification of surface charges around the ion channel, either because of chelation of extracellular Ca2+, or because of binding of a charged biotransformation product of oxaliplatin to the channel. To elucidate the molecular mechanism, we investigated the effects of oxaliplatin and its chloride complex [Pt(dach)oxCl](-) on the voltage-gated Shaker K channel expressed in Xenopus oocytes. The recordings were made with the two-electrode and the cut-open oocyte voltage clamp techniques. CONCLUSION: To our surprise, we did not see any effects on the current amplitudes, on the current time courses, or on the voltage dependence of the Shaker wild-type channel. Oxaliplatin is expected to bind to cysteines. Therefore, we explored if there could be a specific effect on single (E418C) and double-cysteine (R362C/F416C) mutated Shaker channels previously shown to be sensitive to cysteine-specific reagents. Neither of these channels were affected by oxaliplatin. The clear lack of effect on the Shaker K channel suggests that oxaliplatin or its monochloro complex has no general surface-charge effect on the channels, as has been suggested before, but rather a specific effect to the channels previously shown to be affected.


Asunto(s)
Antineoplásicos/toxicidad , Compuestos Organoplatinos/toxicidad , Canales de Potasio de la Superfamilia Shaker/metabolismo , Animales , Cisteína/genética , Cisteína/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Oocitos/metabolismo , Oxaliplatino , Canales de Potasio de la Superfamilia Shaker/genética , Xenopus laevis/metabolismo
3.
Exp Biol Med (Maywood) ; 231(10): 1638-45, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17060685

RESUMEN

The anticancer drug cisplatin can cause permanent inner ear damage. We have determined the second-order degradation rate constant, k(Nu), of cisplatin and its more toxic monohydrated complex (MHC) in the presence of each of the sulfur-containing nucleophiles N-acetyl-l-cysteine, l-cysteine methyl ester, 1,3-dimethyl-2-thiourea, d-methionine, and thiosulfate, compounds that are under evaluation for local administration to prevent cisplatin-induced ototoxicity. MHC was isolated from a hydrolysis solution of cisplatin using liquid chromatography (LC). The degradations were evaluated by measuring the disappearance of MHC and cisplatin at 37 degrees C and pH 7.4 in the presence of each of the nucleophiles using LC and photometric detection. The k(Nu) of MHC and of cisplatin was 0.044 M(-1)sec(-1) and 0.012 M(-1)sec(-1) with N-acetyl-l-cysteine, 0.24 M(-1)sec(-1) and 0.067 M(-1)sec(-1) with l-cysteine methyl ester, 0.16 M(-1)sec(-1) and 0.074 M(-1)sec(-1) with 1,3-dimethyl-2-thiourea, 0.070 M(-1)sec(-1) and 0.069 M(-1)sec(-1) with d-methionine, and 3.9 M(-1)sec(-1) and 0.091 M(-1)sec(-1) with thiosulfate, respectively. Our results suggest that thiosulfate, as being the strongest nucleophile, is a promising candidate for local application in order to reduce the inner ear content of MHC and cisplatin. However, otoprotection is a multifactorial event, and it remains to be established how important nucleophilicity is for the effectiveness of the protecting agent.


Asunto(s)
Antineoplásicos/farmacocinética , Cisplatino/química , Cisplatino/farmacocinética , Oído Interno/efectos de los fármacos , Compuestos de Azufre/farmacocinética , Animales , Antineoplásicos/química , Antineoplásicos/toxicidad , Cromatografía Liquida , Cisplatino/toxicidad , Relación Dosis-Respuesta a Droga , Oído Interno/metabolismo , Concentración de Iones de Hidrógeno , Hidrólisis , Análisis de Regresión , Compuestos de Azufre/química , Compuestos de Azufre/toxicidad , Temperatura , Tiosulfatos/química , Tiosulfatos/farmacocinética , Tiosulfatos/toxicidad
4.
Eur J Pharm Sci ; 28(4): 278-83, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16621479

RESUMEN

Oxaliplatin undergoes extensive non-enzymatic chemical transformation in the body. Complexes with sulphur-containing compounds have previously been found in plasma from patients treated with oxaliplatin. We have studied the kinetics for the reactions between oxaliplatin and cysteine, methionine, and glutathione, by determination of the degradation of oxaliplatin using liquid chromatography with UV-detection. We also studied the degradation of oxaliplatin in plasma ultrafiltrate (PUF). For the degradation of oxaliplatin in the presence of glutathione, methionine, and cysteine, the second-order rate constants were 4.7M(-1)min(-1) (95% confidence interval [C.I.], 4.4-5.0M(-1)min(-1)), 5.5M(-1)min(-1) (95% C.I., 5.2-5.7M(-1)min(-1)), and 15M(-1)min(-1) (95% C.I., 14-17M(-1)min(-1)), respectively. The reaction rate was much faster than previously reported kinetics for cisplatin. The degradation rate of oxaliplatin in PUF was biphasic. The rate constant for the first phase varied from 9.5x10(-3) to 0.13min(-1) and for the second phase from (1.7 to 1.8)x10(-3)min(-1) in PUF from five healthy volunteers. The first hours of the degradation of oxaliplatin in PUF are accounted for by the degradation of oxaliplatin in a cocktail of sodium chloride and sulphur-containing compounds at physiological plasma concentrations. In conclusion, the rate of the reaction of oxaliplatin with three sulphur-containing compounds was faster for oxaliplatin than what is previously known for cisplatin. This may be important with respect to differences in the cellular effects of cisplatin and oxaliplatin treatment.


Asunto(s)
Antineoplásicos/química , Cisteína/química , Glutatión/química , Metionina/química , Compuestos Organoplatinos/química , Plasma/metabolismo , Antineoplásicos/metabolismo , Cromatografía Liquida/métodos , Estabilidad de Medicamentos , Humanos , Técnicas In Vitro , Cinética , Compuestos Organoplatinos/metabolismo , Oxaliplatino , Plasma/química , Espectrofotometría Ultravioleta , Ultrafiltración
5.
Free Radic Biol Med ; 39(5): 696-703, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16085187

RESUMEN

Mammalian thioredoxin reductase (TrxR) is important for cell proliferation, antioxidant defense, and redox signaling. Together with glutathione reductase (GR) it is the main enzyme providing reducing equivalents to many cellular processes. GR and TrxR are flavoproteins of the same enzyme family, but only the latter is a selenoprotein. With the active site containing selenocysteine, TrxR may catalyze reduction of a wide range of substrates, but can at the same time easily be targeted by electrophilic compounds due to the extraordinarily high reactivity of a selenolate moiety. Here we addressed the inhibition of the enzyme by major anticancer alkylating agents and platinum-containing compounds and we compared it to that of GR. We confirmed prior studies suggesting that the nitrosourea carmustine can inhibit both GR and TrxR. We next found, however, that nitrogen mustards (chlorambucil and melphalan) and alkyl sulfonates (busulfan) efficiently inhibited TrxR while these compounds, surprisingly, did not inhibit GR. Inhibitions were concentration and time dependent and apparently irreversible. Anticancer anthracyclines (daunorubicin and doxorubicin) were, in contrast to the alkylating agents, not inhibitors but poor substrates of TrxR. We also found that TrxR, but not GR, was efficiently inhibited by both cisplatin, its monohydrated complex, and oxaliplatin. Carboplatin, in contrast, could not inhibit any of the two enzymes. These findings lead us to conclude that representative compounds of the major classes of clinically used anticancer alkylating agents and most platinum compounds may easily target TrxR, but not GR. The TrxR inhibition should thereby be considered as a factor that may contribute to the cytotoxicity seen upon clinical use of these drugs.


Asunto(s)
Antineoplásicos/farmacología , Glutatión Reductasa/antagonistas & inhibidores , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Animales , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Alquilantes/farmacología , Antioxidantes/metabolismo , Sitios de Unión , Busulfano/farmacología , Clorambucilo/farmacología , Cisplatino/farmacología , Daunorrubicina/farmacología , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Melfalán/farmacología , Compuestos Organoplatinos/farmacología , Oxaliplatino , Ratas , Espectrofotometría/métodos , Especificidad por Sustrato , Factores de Tiempo
6.
Mol Cancer Ther ; 2(12): 1331-9, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14707274

RESUMEN

Peptichemio (PTC), a mixture of six oligopeptides all containing m-L-sarcolysin, has previously shown impressive results in clinical trials. The tripeptide P2 (L-prolyl-m-L-sarcolysyl-p-L-fluorophenylalanine ethyl ester) has been suggested as the main contributor to PTC activity. In contrast to its analogue melphalan, m-L-sarcolysin never reached clinical use. To allow a direct comparison, the corresponding melphalan containing tripeptide J3 (L-prolyl-L-melphalanyl-p-L-fluorophenylalanine ethyl ester) was synthesized and its activity was compared with that of P2; the activities of melphalan and m-L-sarcolysin were studied in parallel. Cytotoxic activity in human tumor cell lines and some fresh human tumor specimens were analyzed as well as effects on cellular metabolism, macromolecular synthesis, and preliminary evaluation of the cell death characteristics. The results show that melphalan and m-L-sarcolysin display similar activity in these systems and that the tripeptides were more active than their parent monomers. Surprisingly however, the melphalan containing tripeptide J3 demonstrated a significantly more rapid and stronger activity than the m-L-sarcolysin analogue P2. Finally, the in vivo toxicity and activity of melphalan and J3 were investigated in mice bearing human leukemia cells in s.c. fibers. The in vitro results seem translatable into the in vivo situation, demonstrating better antileukemic effect of J3 but similar side effects as melphalan.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Melfalán/análogos & derivados , Melfalán/farmacología , Oligopéptidos/farmacología , Línea Celular Tumoral , Dipéptidos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Espectroscopía de Resonancia Magnética
7.
Laryngoscope ; 125(9): E320-5, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25892279

RESUMEN

OBJECTIVE: To locate the organic cation transporter 2 (OCT2) in the cochlea of three different species and to modulate the ototoxicity of cisplatin in the guinea pig by pretreatment with phenformin, having a known affinity for OCT2. STUDY DESIGN: Immunohistochemical and in vivo study. METHODS: Sections from the auditory end organs were subjected to immunohistochemical staining in order to identify OCT2 in cochlea from untreated rats, guinea pigs, and a pig. In the in vivo study, guinea pigs were given phenformin intravenously 30 minutes before cisplatin administration. Electrophysiological hearing thresholds were determined, and hair cells loss was assessed 96 hours later. The total amount of platinum in cochlear tissue was determined using mass spectrometry. RESULTS: Organic cation transporter 2 was found in the supporting cells and in type I spiral ganglion cells in the cochlea of all species studied. Pretreatment with phenformin did not reduce the ototoxic side effect of cisplatin. Furthermore, the concentration of platinum in the cochlea was not affected by phenformin. CONCLUSIONS: The localization of OCT2 in the supporting cells and type I spiral ganglion cells suggests that this transport protein is not primarily involved in cisplatin uptake from the systemic circulation. We hypothesize that OCT2 transport intensifies cisplatin ototoxicity via transport mechanisms in alternate compartments of the cochlea. LEVEL OF EVIDENCE: N/A.


Asunto(s)
Cóclea/metabolismo , Inmunohistoquímica/métodos , Proteínas de Transporte de Catión Orgánico/metabolismo , Animales , Cóclea/citología , Femenino , Cobayas , Transportador 2 de Cátion Orgánico , Ratas , Porcinos
8.
Cancer Lett ; 180(2): 191-4, 2002 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-12175551

RESUMEN

A new way of synthesizing the dihydrated oxaliplatin complex (DOC) is presented and its cytotoxicity is compared to that of oxaliplatin and cisplatin. By hydrolyzing oxaliplatin in aqueous sodium hydroxide at 70 degrees C, DOC was formed in less than 1 h. Cytotoxicity was studied in the non-small cell lung cancer cell line A549 using the fluorescent microculture cytotoxic assay. Oxaliplatin and cisplatin had similar cytotoxicity profiles, whereas DOC was found to be considerably more toxic. The cytotoxicity of oxaliplatin might, at least in part, be mediated through the formation of DOC.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Compuestos Organoplatinos/farmacología , Oxaliplatino , Células Tumorales Cultivadas
9.
Cancer Chemother Pharmacol ; 51(1): 36-42, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12497204

RESUMEN

PURPOSE: To evaluate and compare the ototoxicity and nephrotoxicity of cisplatin and cis-diammineaquachloroplatinum(II) ion (monohydrated complex of cisplatin, MHC, formed in vivo by hydrolysis of cisplatin) after their separate administration to guinea pigs. METHODS: A dose of 4 mg/kg body weight of MHC was deemed suitable for the toxicity evaluation after dose titration. Electrophysiological hearing thresholds (auditory brainstem response, ABR), plasma creatinine and weight were measured in three groups of animals before and after receiving MHC 4 mg/kg (0.0141 mmol/kg), cisplatin 4.24 mg/kg (0.0141 mmol/kg, i.e. equimolar dose) or cisplatin 8 mg/kg (0.0267 mmol/kg) as an i.v. bolus injection. Cisplatin and MHC were analysed using liquid chromatography with post-column derivatization. RESULTS: Administration of MHC 4 mg/kg caused a moderate ABR threshold shift, a significant increase in creatinine and a significant weight loss, changes similar to those seen after administration of cisplatin 8 mg/kg. Animals given cisplatin 4.24 mg/kg had a slight increase in creatinine, but had no ABR threshold shift and gained weight during the experiment. The pharmacokinetic parameters of cisplatin and MHC were estimated after administration of cisplatin 4.24 mg/kg and MHC 4 mg/kg. The area under the blood-ultrafiltrate concentration versus time curve (AUC) for cisplatin after administration of MHC 4 mg/kg was 23% (56+/-5.0 micro g.min.ml(-1)) (means+/-SD) of that after administration of cisplatin 4.24 mg/kg (240+/-25 micro g.min.ml(-1)). The AUC for MHC after administration of cisplatin 4.24 mg/kg was 20% (30+/-4.9 micro g.min.ml(-1)) of that after administration of MHC 4 mg/kg (149+/-26 micro g.min.ml(-1)). CONCLUSIONS: MHC 4 mg/kg causes ototoxicity, nephrotoxicity and weight loss when administered to guinea pigs. The toxic effects were similar to those seen after administration of cisplatin 8 mg/kg and higher than those seen after administration of cisplatin 4.24 mg/kg.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Oído Interno/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Riñón/efectos de los fármacos , Animales , Antineoplásicos/farmacocinética , Peso Corporal/efectos de los fármacos , Cisplatino/farmacocinética , Creatinina/sangre , Femenino , Cobayas , Riñón/metabolismo , Masculino
10.
Oncol Res ; 14(3): 113-32, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14760861

RESUMEN

The strategy of using small peptides for effective targeting of tumor cells in chemotherapy has proven beneficial. Recently we showed that J1 (L-melphalanyl-p-L-fluorophenylalanine ethyl ester), an alkylating nitrogen mustard-containing dipeptide, exhibited strong cytotoxic activity in fresh human tumor samples in addition to rapid and pronounced inhibition of macromolecular syntheses and cellular respiration in the human tumor lymphoma cell line U-937 GTB. In this study, an additional series of 17 nitrogen mustard-containing dipeptides has been synthesized and analyzed for cytotoxic activity in a panel of 10 human tumor cell lines. The results were compared to the single amino acid mustard derivative melphalan and its ethyl and isopropyl esters. Also P2 (L-prolyl-m-L-sarcolysyl-p-L-fluorophenylalanine ethyl ester), a tripeptide that previously has shown impressive effects in human tumor cells, was used as reference. The tested compounds displayed various activities in the different cell lines but also showed a high correlation, indicating a similar mechanism of action. Factors like amino acid composition, amino acid sequence, modifications of the C- and N-termini, and to a minor extent the lipophilicity of the dipeptide derivatives appear to influence the in vitro activity. The results indicate that the activity of these compounds not only relies on their chemical reactivity, but also on active biological interactions such as transport across membranes and/or enzymatic liberation of reactive molecular entities.


Asunto(s)
Dipéptidos/química , Compuestos de Mostaza Nitrogenada/química , Compuestos de Mostaza Nitrogenada/toxicidad , Alquilación/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Concentración 50 Inhibidora , Estructura Molecular , Compuestos de Mostaza Nitrogenada/síntesis química , Relación Estructura-Actividad
11.
Anticancer Res ; 23(6D): 5203-8, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14981990

RESUMEN

BACKGROUND: Several studies have demonstrated the efficacy of systemic oxaliplatin (Oxa) in combination with 5-fluorouracil (5-FU) and folinic acid (FA) for the treatment of colorectal liver metastases (CRLM). However, nothing is presently known about the pharmacokinetics of Oxa administered via the hepatic artery and only very little about the feasibility and toxicity of Oxa used for hepatic artery infusion (HAI). PATIENTS AND METHODS: We designed a phase II trial using Oxa in combination with 5-FU/FA and mitomycin C (MMC) for HAI treatment of patients with isolated non-resectable CRLM. Oxa (130 mg/m2) was delivered on day (d) 1 as a 120-min infusion followed by FA (140 mg/m2) for 10 min and 5-FU (480 mg/m2) for 120 min from d1 to d5 and MMC (7 mg/m2) for 30 min on d5 every 35 days. For Oxa pharmacokinetics, peripheral venous blood was collected before, during and after arterial infusion. Oxaliplatin was determined by liquid chromatography with post-column derivatization in blood ultra filtrate. RESULTS: A total of 33 HAI cycles were administered to 5 patients with tolerable toxicity, which mainly consisted of grade I and II nausea, vomiting, leucopenia, thrombopenia and abdominal pain. During 4 cycles nausea/vomiting III degree occurred, during 3 cycles diarrhoea and abdominal pain III degree. No neurotoxicity > or = II degree and no catheter occlusion was observed. Staging showed 4 PR and 1 PD. Pharmacokinetic analysis revealed an AUC value of 85.3 micrograms x min/ml after HAI. Recalculating these values with the previously reported AUC value for systemic administration (161 micrograms x min/ml) revealed a liver extraction ratio of 0.47 for Oxa. CONCLUSION: We conclude from our results that Oxa in combination with 5-FU/FA and MMC may be a feasible protocol for HAI treatment without major toxicity, especially avoiding higher grade neurotoxicity. This is probably attributable to the low systemic bioavailability of Oxa.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Colorrectales/metabolismo , Arteria Hepática , Infusiones Intraarteriales , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Compuestos Organoplatinos/farmacocinética , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Catéteres de Permanencia , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/efectos adversos , Humanos , Leucovorina/administración & dosificación , Leucovorina/efectos adversos , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , Mitomicina/administración & dosificación , Mitomicina/efectos adversos , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/efectos adversos , Oxaliplatino
12.
Artículo en Inglés | MEDLINE | ID: mdl-14522033

RESUMEN

Oxaliplatin ([(1R,2R)-1,2-cyclohexanediamine-N,N']oxalato(2-)-O,O'-platinum) is the first platinum drug with significant activity for metastatic colon cancer. The analysis of oxaliplatin has previously almost exclusively been based on the determination of the platinum content in plasma or ultrafiltrate using flameless atomic absorption spectroscopy (FAAS) or inductively coupled plasma mass spectrometry (ICPMS). A new method for quantitative determination of the free fraction of the intact drug in blood ultrafiltrate is presented here. Blood was ultrafiltrated centripetally at 4 degrees C and the ultrafiltrate was analyzed by liquid chromatography. Oxaliplatin was separated on a Hypercarb column using a mobile phase of methanol/succinic acid buffer pH 7.0 (9/1, v/v). Post-column derivatization was performed by adding N,N-diethyldithiocarbamate in methanol and with microwave heating of a Teflon tubing. The derivative was quantified by photometric detection at 344 nm. The coefficient of variation of standard blood samples was 4.9 and 2.5% at 0.100 and 1.00 microg/ml, respectively. The limit of quantitation was 0.04 microg/ml.


Asunto(s)
Antineoplásicos/sangre , Compuestos Organoplatinos/sangre , Espectrofotometría/métodos , Humanos , Microondas , Oxaliplatino
13.
J Drug Target ; 11(6): 355-63, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-14668056

RESUMEN

Recently, we presented a series of melphalan containing di- and tripeptides with high cytotoxic activity and J1 (l-melphalanyl-p-l-fluorophenylalanine ethyl ester) was identified as one of the most interesting compounds. It was speculated that the increased activity compared to melphalan itself, demonstrated both in vitro and in vivo, resided in increased transport over the tumour cell membrane and/or hydrolytic cleavage and liberation of melphalan inside the cells. Indeed, overexpression of hydrolytic enzymes like peptidases, esterases and proteases has been described in several types of human malignancies, thus providing a target for selective chemotherapy. In this work, the details of the increased activity was further investigated and potential tumour selectivity is discussed. The intracellular delivery of melphalan is investigated in detail using peptidase resistant dipeptide derivatives, by enzyme inhibitors and probes for enzymatic activity and by studying the time dependency of drug effect as well as intracellular drug concentrations (cellular pharmacokinetics). The results show that the activity of the dipeptide mustards is highly dependent on intracellular hydrolysis, which result in rapid intracellular release of the alkylating unit (i.e. free melphalan) in cells with high enzymatic activity. The maximum intracellular melphalan concentration following J1 exposure was reached already after 15 min, thereafter declining with a half-life of approximately 1 h. This rapid intracellular loading resulted in less reduction of activity for J1 than for melphalan and six other standard drugs when human tumour cell lines were exposed to the drugs for a limited time (simulating short half-life in vivo). Peptidase inhibitors inhibited the activity and intracellular release of melphalan, and dipeptide derivatives designed to resist the action of peptidases was less active than the corresponding normal dipeptide.


Asunto(s)
Antineoplásicos Alquilantes/administración & dosificación , Antineoplásicos Alquilantes/uso terapéutico , Leucina/análogos & derivados , Melfalán/administración & dosificación , Melfalán/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Oligopéptidos/administración & dosificación , Oligopéptidos/uso terapéutico , Péptido Hidrolasas/química , Péptido Hidrolasas/metabolismo , Profármacos , Antineoplásicos Alquilantes/farmacocinética , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Dipéptidos , Fluorometría , Humanos , Hidrólisis , Indicadores y Reactivos , Leucina/farmacología , Melfalán/farmacocinética , Oligopéptidos/farmacocinética , Inhibidores de Proteasas/farmacología
14.
Hear Res ; 165(1-2): 53-61, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12031515

RESUMEN

D-Methionine has recently been advocated as a protectant against cisplatin toxicity. The use of systemic D-methionine as a protector was studied in 58 guinea pigs. Kinetics and distribution of [11CH(3)]D-methionine was analysed by positron emission tomography. Cisplatin and the monohydrated complex of cisplatin was quantified in blood ultrafiltrate using reversed-phase liquid chromatography with post-column derivatisation. Administration of 300 mg/kg of D-methionine caused a 30% decrease in the area under the concentration-time curve (AUC) of cisplatin. The toxic effect of cisplatin was studied after dose adjustment of cisplatin, i.e. with similar cisplatin AUC in the group receiving D-methionine and the saline control group. A significant ototoxic effect, measured as difference in pre- and 96 h post-treatment electrophysiological hearing threshold (auditory brainstem response), was observed at stimulus frequencies of 30 and 20 kHz. There was no difference between the groups in the extent of threshold shift. Quantitative outer hair cell counts showed a similar loss of cells in the two groups. All animals had a significant increase in plasma-creatinine but there was no difference between the groups. The results indicate that protection from cisplatin ototoxicity by systemic D-methionine can be explained by a lowered systemic exposure to the drug.


Asunto(s)
Antineoplásicos/envenenamiento , Cisplatino/envenenamiento , Citoprotección , Oído , Metionina/farmacología , Animales , Antineoplásicos/farmacocinética , Peso Corporal/efectos de los fármacos , Cisplatino/farmacocinética , Creatina/metabolismo , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Femenino , Cobayas , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Distribución Tisular , Tomografía Computarizada de Emisión
15.
J Pharm Sci ; 92(2): 436-8, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12532393

RESUMEN

Alkaline hydrolysis of the platinum anticancer drug oxaliplatin gives the oxalato monodentate complex and the dihydrated oxaliplatin complex in two consecutive steps. The acid dissociation constant for the oxalato monodentate intermediate was determined by a kinetic approach. The pK(a) value was estimated as 7.23. The monodentate intermediate is assumed to rapidly react with endogenous compounds, resulting in a continuous conversion of oxaliplatin via the monodentate form.


Asunto(s)
Antineoplásicos/química , Compuestos Organoplatinos/química , Algoritmos , Hidrólisis , Cinética , Oxaliplatino , Solubilidad , Temperatura
16.
J Pharm Sci ; 91(10): 2116-21, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12226839

RESUMEN

The alkaline degradation of the chemotherapeutic agent oxaliplatin has been studied using liquid chromatography. The oxalato ligand is lost in two consecutive steps. First, the oxalato ring is opened, forming an oxalato monodentate intermediate, as identified by electrospray ionization mass spectrometry. Subsequently, the oxalato ligand is lost and the dihydrated oxaliplatin complex is formed. The observed rate constants for the first step (k(1)) and the second step (k(2)) follow the equation k(1) or k(2) = k(0) + k(OH(-) )[OH(-)], where k(0) is the rate constant for the degradation catalyzed by water and k(OH(-) ) represents the second-order rate constant for the degradation catalyzed by the hydroxide ion. At 37 degrees C the rate constants for the first step are k(OH(-) ) = 5.5 x 10(-2) min(-1) M(-1) [95% confidence interval (CI), 2.7 x 10(-2) to 8.4 x 10(-2) min(-1) M(-1)] and k(0) = 4.3 x 10(-2) min(-1) (95% CI, 4.0 x 10(-2) to 4.7 x 10(-2) min(-1)). For the second step the rate constants are k(OH(-) ) = 1.1 x 10(-3) min(-1) M(-1) (95% CI, -1.1 x 10(-3) to 3.3 x 10(-3)) min(-1) M(-1) and k(0) = 7.5 x 10(-3) min(-1) (95% CI, 7.2 x 10(-3) to 7.8 x 10(-3) min(-1)). Thus, the ring-opening step is nearly six times faster than the step involving the loss of the oxalato ligand.


Asunto(s)
Antineoplásicos/química , Compuestos Organoplatinos/química , Compuestos Organoplatinos/síntesis química , Compuestos Organoplatinos/farmacología , Algoritmos , Cromatografía Liquida , Hidrólisis , Indicadores y Reactivos , Cinética , Oxaliplatino , Platino (Metal)/química , Hidróxido de Sodio/química , Espectrometría de Masa por Ionización de Electrospray , Espectrofotometría Ultravioleta
17.
Laryngoscope ; 114(9): 1660-7, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15475801

RESUMEN

OBJECTIVES/HYPOTHESIS: Ototoxicity is a common side effect of high-dose cisplatin treatment. Thiol-containing chemoprotectors ameliorate cisplatin ototoxicity under experimental conditions. The trial was initiated to test the efficacy of amifostine protection in high-dose cisplatin treatment (125-150 mg/m) for metastatic malignant melanoma, to correlate the ototoxic outcome with cisplatin pharmacokinetics, and to evaluate the importance of using a selective analytical method for the quantification of cisplatin. STUDY DESIGN: Prospective study of 15 patients with stage IV malignant melanoma. METHODS: Clinical follow-up of therapeutic response, pure-tone audiometry, and analysis of cisplatin and its monohydrated complex in blood ultrafiltrate by liquid chromatography with postcolumn derivatization were performed. Ultrafiltered blood platinum was analyzed by inductively coupled plasma mass spectrometry. RESULTS: Ototoxicity and gastrointestinal toxicity were the most prominent side effects. Three patients ultimately required hearing aids. All patients had audiometric changes at one or more frequencies after the second treatment course, and all but one patient reported auditory symptoms. No correlation was found between hearing loss and blood cisplatin pharmacokinetics. Platinum levels determined by inductively coupled plasma mass spectrometry were higher than total platinum levels calculated from cisplatin and monohydrated complex concentrations obtained by liquid chromatography analysis. CONCLUSION: Ototoxicity was unacceptable despite amifostine treatment. Cisplatin pharmacokinetics during the first treatment course were not predictive of hearing loss. Amifostine caused a lowering of dose-normalized area under the concentration-time curve for cisplatin and monohydrated complex. Use of the unselective inductively coupled plasma mass spectrometry analysis leads to an overestimation of active drug. Selective analysis of cisplatin is especially important when evaluating cisplatin pharmacokinetics during chemoprotector treatment.


Asunto(s)
Amifostina/uso terapéutico , Antineoplásicos/toxicidad , Cisplatino/toxicidad , Pérdida Auditiva Sensorineural/inducido químicamente , Melanoma/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Adulto , Anciano , Amifostina/efectos adversos , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Audiometría de Tonos Puros , Umbral Auditivo/efectos de los fármacos , Cisplatino/administración & dosificación , Cisplatino/farmacocinética , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Estudios de Seguimiento , Pérdida Auditiva Sensorineural/sangre , Pérdida Auditiva Sensorineural/prevención & control , Humanos , Masculino , Melanoma/sangre , Melanoma/patología , Tasa de Depuración Metabólica/efectos de los fármacos , Persona de Mediana Edad , Estadificación de Neoplasias , Fármacos Neuroprotectores/efectos adversos , Estudios Prospectivos , Neoplasias Cutáneas/sangre , Neoplasias Cutáneas/patología
18.
Acta Otolaryngol ; 134(5): 448-54, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24702224

RESUMEN

CONCLUSION: Inhibition of thioredoxin reductase (TrxR) may be a contributing factor in cisplatin-induced ototoxicity. Direct exposure of organ of Corti to cisplatin and oxaliplatin gives equal loss of hair cells. OBJECTIVES: Platinum-containing drugs are known to target the anti-oxidant selenoprotein TrxR in cancer cells. Two such anti-cancer, platinum-containing drugs, cisplatin and oxaliplatin, have different side effects. Only cisplatin induces hearing loss, i.e. has an ototoxic side effect that is not seen after treatment with oxaliplatin. The objective of this study was to evaluate if TrxR is a target in the cochlea. Loss of outer hair cells was also compared when cisplatin and oxaliplatin were administered directly to the organ of Corti. METHODS: Organ of Corti cell culture was used for direct exposure to cisplatin and oxaliplatin. Hair cells were evaluated and the level of TrxR was assessed. Immunohistochemical staining for TrxR was performed. An animal model was used to evaluate the effect on TrxR after treatment with cisplatin and oxaliplatin in vivo. RESULTS: Direct exposure of cochlear organotypic cultures to either cisplatin or oxaliplatin induced comparable levels of outer hair cell loss and inhibition of TrxR, demonstrating that both drugs are similarly ototoxic provided that the cochlea becomes directly exposed.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Células Ciliadas Auditivas Externas/efectos de los fármacos , Compuestos Organoplatinos/toxicidad , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Animales , Femenino , Cobayas , Células Ciliadas Auditivas Externas/enzimología , Técnicas de Cultivo de Órganos , Oxaliplatino , Ratas Sprague-Dawley
19.
Laryngoscope ; 123(12): 3172-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23754209

RESUMEN

OBJECTIVES/HYPOTHESIS: Cisplatin produces toxic lesions to outer hair cells (OHCs) in the cochlear base but not in the apex. The objective of this study was to compare the pharmacokinetic profile of cisplatin in scala tympani (ST) perilymph in the cochlear base and apex, respectively. STUDY DESIGN: In vivo animal study. METHODS: Forty-seven guinea pigs were given an intravenous bolus injection of an ototoxic dose of cisplatin. Ten to 240 minutes after cisplatin was given, blood, cerebrospinal fluid (CSF), and ST perilymph were aspirated within the same target time. ST perilymph was aspirated from the basal turn and from the apex of the cochlea by two different sampling techniques. Liquid chromatography with postcolumn derivatization was used for quantitative determination of the parent drug. RESULTS: Ten minutes after administration, the concentration of cisplatin in ST perilymph was 4-fold higher in the basal turn of the cochlea than in the apex. At 30 minutes, the drug concentrations did not differ. At 60 minutes, the level of cisplatin in ST perilymph and blood UF was equivalent. The perilymph-blood ratio increased thereafter with time. CONCLUSION: The pharmacokinetic findings of an early high concentration of cisplatin in the base of the cochlea and delayed elimination of cisplatin from ST perilymph compared to blood might correlate to the cisplatin-induced loss of OHCs in the base of the cochlea.


Asunto(s)
Cisplatino/farmacocinética , Cóclea/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Cromatografía Liquida , Cisplatino/administración & dosificación , Cóclea/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Estudios de Seguimiento , Cobayas , Inyecciones Intravenosas , Perilinfa/efectos de los fármacos , Perilinfa/metabolismo
20.
J Pharm Biomed Anal ; 56(1): 126-30, 2011 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-21621941

RESUMEN

The anticancer agent cisplatin (cis-diamminedichloroplatinum(II), cis-[PtCl2(NH3)2]) easily undergoes ligand-exchange reactions, resulting in mainly inactive Pt complexes. This paper presents a method for selective analysis of intact cisplatin in blood using LC and UV detection. Blood samples (hematocrit: 0.22-0.52) were spiked with cisplatin (final concentrations: 2.48 × 10⁻7 M-9.90 × 10⁻6 M) and subjected to centripetal ultrafiltration. The blood ultrafiltrate was separated (loop volume: 5 µl) with a porous graphitic carbon column and a mobile phase of HEPES-buffer (pH 9.3). Prior to UV detection (344 nm), the eluate was mixed with sodium N,N-diethyldithiocarbamate (DDTC) in a microwave field (115 °C) in order to improve the UV absorptivity. Cisplatin eluted as a Pt-DDTC complex after 11.8 min. The peak area was influenced primarily by the hematocrit, the DDTC concentration, and the temperature and residence time in the microwave cavity. The method was robust and sensitive provided preparing a fresh DDTC solution each day and, at the end of a day's run, destroying DDTC remaining in the system. It offers the main advantages of high selectivity, sensitivity, and robustness, minimal sample processing, and the possibility to use small sample volumes.


Asunto(s)
Antineoplásicos/sangre , Cisplatino/sangre , Microondas , Biotransformación , Cromatografía Liquida , Humanos , Técnicas In Vitro , Modelos Lineales , Reproducibilidad de los Resultados , Espectrofotometría Ultravioleta , Ultrafiltración
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA