Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Pharm ; 12(12): 4259-69, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26474710

RESUMEN

The impact of OATP drug uptake transporters in drug-drug interactions (DDIs) is increasingly recognized. OATP1B1 and OATP1B3 are human hepatic uptake transporters that can mediate liver uptake of a wide variety of drugs. Recently, we generated transgenic mice with liver-specific expression of human OATP1B1 or OATP1B3 in a mouse Oatp1a/1b knockout background. Here, we investigated the applicability of these mice in OATP-mediated drug-drug interaction studies using the prototypic OATP inhibitor rifampicin and a good OATP substrate, the anticancer drug methotrexate (MTX). We next assessed the possibility of OATP-mediated interactions between telmisartan and MTX, a clinically relevant drug combination. Using HEK293 cells overexpressing OATP1B1 or OATP1B3, we estimated IC50 values for both rifampicin (0.9 or 0.3 µM) and telmisartan (6.7 or 7.9 µM) in inhibiting OATP-mediated MTX uptake in vitro. Using wild-type, Oatp1a/1b-/-, and OATP1B1- or OATP1B3-humanized transgenic mice, we found that rifampicin inhibits hepatic uptake of MTX mediated by the mouse Oatp1a/1b and human OATP1B1 and OATP1B3 transporters at clinically relevant concentrations. This highlights the applicability of these mouse models for DDI studies and may be exploited in the clinic to reduce the dose and thus methotrexate-mediated toxicity. On the other hand, telmisartan inhibited only human OATP1B1-mediated hepatic uptake of MTX at concentrations higher than those used in the clinic; therefore risks for OATP-mediated clinical DDIs for this drug combination are likely to be low. Overall, we show here that OATP1B1- and OATP1B3-humanized mice can be used as in vivo tools to assess and possibly predict clinically relevant DDIs.


Asunto(s)
Interacciones Farmacológicas/fisiología , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Transportadores de Anión Orgánico/metabolismo , Animales , Antineoplásicos/metabolismo , Bencimidazoles/metabolismo , Benzoatos/metabolismo , Transporte Biológico/fisiología , Células HEK293 , Humanos , Hígado/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado , Masculino , Proteínas de Transporte de Membrana/metabolismo , Metotrexato/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos , Telmisartán
2.
Int J Cancer ; 136(1): 225-33, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24825069

RESUMEN

Organic anion transporting polypeptides (human: OATPs and mouse: Oatps) are uptake transporters with important roles in drug pharmacokinetics and toxicity. We aimed to study the in vivo impact of mouse and human OATP1A/1B transporters on docetaxel plasma clearance and liver and intestinal uptake. Docetaxel was administered to Oatp1a/1b knockout and liver-specific humanized OATP1B1, OATP1B3 and OATP1A2 transgenic mice. Experiments were conducted with a low polysorbate 80 (2.8%) formulation, as 8% polysorbate somewhat inhibited docetaxel plasma clearance after intravenous administration. After intravenous administration (10 mg/kg), Oatp1a/1b knockout mice had an approximately threefold higher plasma area under the curve (AUC). Impaired liver uptake was evident from the significantly reduced (approximately threefold) liver-to-plasma AUC ratios. Absence of mouse Oatp1a/1b transporters did not affect the intestinal absorption of orally administered docetaxel (10 mg/kg), while the systemic exposure of docetaxel was again substantially increased owing to impaired liver uptake. Most importantly, liver-specific expression of each of the human OATP1B1, OATP1B3 and OATP1A2 transporters provided a nearly complete rescue of the increased plasma levels of docetaxel in Oatp1a/1b-null mice after intravenous administration. Our data show that one or more of the mouse Oatp1a/1b transporters and each of the human OATP1A/1B transporters can mediate docetaxel uptake in vivo. This might be clinically relevant for OATP1A/1B-mediated tumor uptake of docetaxel and for docetaxel clearance in patients in whom the transport activity of OATP1A/1B transporters is reduced owing to genetic variation or pharmacological inhibition, leading to potentially altered toxicity and therapeutic efficacy of this drug.


Asunto(s)
Antineoplásicos/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/fisiología , Transportadores de Anión Orgánico/fisiología , Taxoides/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Transporte Biológico , Química Farmacéutica , Docetaxel , Prueba de Complementación Genética , Humanos , Absorción Intestinal , Transportador 1 de Anión Orgánico Específico del Hígado , Masculino , Ratones Noqueados , Polisorbatos/administración & dosificación , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos , Taxoides/administración & dosificación , Taxoides/farmacocinética
3.
Pharm Res ; 32(1): 37-46, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24962512

RESUMEN

BACKGROUND: Rucaparib is a potent, orally available, small-molecule inhibitor of poly ADP-ribose polymerase (PARP) 1 and 2. Ongoing clinical trials are assessing the efficacy of rucaparib alone or in combination with other cytotoxic drugs, mainly in breast and ovarian cancer patients with mutations in the breast cancer associated (BRCA) genes. PURPOSE: We aimed to establish whether the multidrug efflux transporters ABCG2 (BCRP) and ABCB1 (P-gp, MDR1) affect the oral availability and brain penetration of rucaparib in mice. RESULTS: In vitro, rucaparib was efficiently transported by both human ABCB1 and ABCG2, and very efficiently by mouse Abcg2. Transport could be inhibited by the small-molecule ABCB1 and ABCG2 inhibitors zosuquidar and Ko143, respectively. In vivo, oral availability (plasma AUC0-1 and AUC0-24) and brain levels of rucaparib at 1 and 24 h were increased by the absence of both Abcg2 and Abcb1a/1b after oral administration of rucaparib at 10 mg/kg. CONCLUSIONS: Our data show to our knowledge for the first time that oral availability and brain accumulation of a PARP inhibitor are markedly and additively restricted by Abcg2 and Abcb1a/1b. This may have clinical relevance for improvement of rucaparib therapy in PARP inhibitor-resistant tumors with ABCB1 and/or ABCG2 expression and in patients with brain (micro)metastases positioned behind a functional blood-brain barrier.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Encéfalo/metabolismo , Indoles/farmacocinética , Proteínas de Neoplasias/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Administración Oral , Animales , Disponibilidad Biológica , Transporte Biológico , Técnicas de Cultivo de Célula , Perros , Femenino , Humanos , Indoles/administración & dosificación , Indoles/sangre , Células de Riñón Canino Madin Darby , Ratones Noqueados , Proteínas de Neoplasias/genética , Especificidad por Sustrato , Distribución Tisular
4.
Mol Pharmacol ; 85(3): 520-30, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24334255

RESUMEN

The multidrug transporters breast cancer resistance protein (BCRP), multidrug-resistance protein 1 (MDR1), and multidrug-resistance-associated protein (MRP) 2 and 3 eliminate toxic compounds from tissues and the body and affect the pharmacokinetics of many drugs and other potentially toxic compounds. The food-derived carcinogen PhIP (2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine) is transported by BCRP, MDR1, and MRP2. To investigate the overlapping functions of Bcrp1, Mdr1a/b, and Mrp2 in vivo, we generated Bcrp1;Mdr1a/b;Mrp2(-/-) mice, which are viable and fertile. These mice, together with Bcrp1;Mrp2;Mrp3(-/-) mice, were used to study the effects of the multidrug transporters on the pharmacokinetics of PhIP and its metabolites. Thirty minutes after oral or intravenous administration of PhIP (1 mg/kg), the PhIP levels in the small intestine were reduced 4- to 6-fold in Bcrp1;Mdr1a/b;Mrp2(-/) (-) and Bcrp1;Mrp2;Mrp3(-/-) mice compared with wild-type mice. Fecal excretion of PhIP was reduced 8- to 20-fold in knockouts. Biliary PhIP excretion was reduced 41-fold in Bcrp1;Mdr1a/b;Mrp2(-/-) mice. Biliary and small intestine levels of PhIP metabolites were reduced in Bcrp1;Mrp2-deficient mice. Furthermore, in both knockout strains, kidney levels and urinary excretion of genotoxic PhIP-metabolites were significantly increased, suggesting that reduced biliary excretion of PhIP and PhIP metabolites leads to increased urinary excretion of these metabolites and increased systemic exposure. Bcrp1 and Mdr1a limited PhIP brain accumulation. In Bcrp1;Mrp2;Mrp3(-/-), but not Bcrp1;Mdr1a/b;Mrp(-/-) mice, the carcinogenic metabolites N2-OH-PhIP (2-hydroxyamino-1-methyl-6-phenylimidazo[4,5-b]pyridine) and PhIP-5-sulfate (a genotoxicity marker) accumulated in liver tissue, indicating that Mrp3 is involved in the sinusoidal secretion of these compounds. We conclude that Bcrp1, Mdr1a/b, Mrp2, and Mrp3 significantly affect tissue disposition and biliary and fecal elimination of PhIP and its carcinogenic metabolites and may affect PhIP-induced carcinogenesis as a result.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/genética , Carcinógenos/metabolismo , Imidazoles/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Carcinógenos/farmacocinética , Imidazoles/farmacocinética , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
5.
Mol Cancer Ther ; 13(2): 492-503, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24194565

RESUMEN

Organic anion-transporting polypeptides (OATP) mediate the hepatic uptake of many drugs, thus codetermining their clearance. Impaired hepatic clearance due to low-activity polymorphisms in human OATP1B1 may increase systemic exposure to SN-38, the active and toxic metabolite of the anticancer prodrug irinotecan. We investigated the pharmacokinetics and toxicity of irinotecan and SN-38 in Oatp1a/1b-null mice: Plasma exposure of irinotecan and SN-38 was increased 2 to 3-fold after irinotecan dosing (10 mg/kg, i.v.) compared with wild-type mice. Also, liver-to-plasma ratios were significantly reduced, suggesting impaired hepatic uptake of both compounds. After 6 daily doses of irinotecan, Oatp1a/1b-null mice suffered from increased toxicity. However, Oatp1a/1b-null mice had increased levels of carboxylesterase (Ces) enzymes, which caused higher conversion of irinotecan to SN-38 in plasma, potentially complicating pharmacokinetic analyses. Ces inhibitors blocked this increased conversion. Interestingly, liver-specific humanized OATP1B1 and OATP1B3 transgenic mice had normalized hepatic expression of Ces1 genes. While irinotecan liver-to-plasma ratios in these humanized mice were similar to those in Oatp1a/1b-null mice, SN-38 liver-to-plasma ratios returned to wild-type levels, suggesting that human OATP1B proteins mediate SN-38, but not irinotecan uptake in vivo. Upon direct administration of SN-38 (1 mg/kg, i.v.), Oatp1a/1b-null mice had increased SN-38 plasma levels, lower liver concentrations, and decreased cumulative biliary excretion of SN-38. Mouse Oatp1a/1b transporters have a role in the plasma clearance of irinotecan and SN-38, whereas human OATP1B transporters may only affect SN-38 disposition. Oatp1a/1b-null mice have increased expression and activity of Ces1 enzymes, whereas humanized mice provide a rescue of this phenotype.


Asunto(s)
Camptotecina/análogos & derivados , Hidrolasas de Éster Carboxílico/genética , Transportadores de Anión Orgánico Sodio-Independiente/genética , Proteínas de Transporte de Catión Orgánico/genética , Animales , Apoptosis/efectos de los fármacos , Área Bajo la Curva , Camptotecina/sangre , Camptotecina/farmacocinética , Femenino , Expresión Génica , Humanos , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Intestino Delgado/patología , Irinotecán , Hígado/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado , Ratones Noqueados , Ratones Transgénicos , Profármacos/efectos adversos , Profármacos/farmacocinética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Timo/efectos de los fármacos , Timo/metabolismo , Timo/patología , Regulación hacia Arriba
6.
Mol Pharmacol ; 83(5): 919-29, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23429889

RESUMEN

Organic anion-transporting polypeptides (OATPs) mediate the liver uptake and hence plasma clearance of a broad range of drugs. For rosuvastatin, a cholesterol-lowering drug and OATP1A/1B substrate, the liver represents both its main therapeutic target and its primary clearance organ. Here we studied the impact of Oatp1a/1b uptake transporters on the pharmacokinetics of rosuvastatin using wild-type and Oatp1a/1b-null mice. After oral administration (15 mg/kg), intestinal absorption of rosuvastatin was not impaired in Oatp1a/1b-null mice, but systemic exposure (area under the curve) was 8-fold higher in these mice compared with wild-type. Although liver exposure was comparable between the two mouse strains (despite the increased blood exposure), the liver-to-blood ratios were markedly decreased (>10-fold) in the absence of Oatp1a/1b transporters. After intravenous administration (5 mg/kg), systemic exposure was 3-fold higher in Oatp1a/1b-null mice than in the wild-type mice. Liver, small intestinal, and kidney exposure were slightly, but not significantly, increased in Oatp1a/1b-null mice. The biliary excretion of rosuvastatin was very fast, with 60% of the dose eliminated within 15 minutes after intravenous administration, and also not significantly altered in Oatp1a/1b-null mice. Rosuvastatin renal clearance, although still minor, was increased ∼15-fold in Oatp1a/1b-null males, suggesting a role of Oatp1a1 in the renal reabsorption of rosuvastatin. Absence of Oatp1a/1b uptake transporters increases the systemic exposure of rosuvastatin by reducing its hepatic extraction ratio. However, liver concentrations are not significantly affected, most likely due to the compensatory activity of high-capacity, low-affinity alternative uptake transporters at higher systemic rosuvastatin levels and the absence of efficient alternative rosuvastatin clearance mechanisms.


Asunto(s)
Fluorobencenos/farmacocinética , Hígado/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Transportadores de Anión Orgánico/metabolismo , Pirimidinas/farmacocinética , Sulfonamidas/farmacocinética , Animales , Transporte Biológico , Femenino , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Absorción Intestinal , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Rosuvastatina Cálcica
7.
Clin Cancer Res ; 19(4): 821-32, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23243220

RESUMEN

PURPOSE: Organic anion-transporting polypeptide (OATP) drug uptake transporters are thought to play an important role in drug pharmacokinetics and toxicokinetics. We aimed to determine the influence of the individual human OATP1B1, OATP1B3, and OATP1A2 transporters on the in vivo disposition of the anticancer drugs methotrexate and paclitaxel by using liver-specific humanized OATP1A/1B transgenic mice. EXPERIMENTAL DESIGN: Wild-type, Slco1a/1b(-/-) (Oatp1a/1b knockout), Slco1a/1b(-/-);1B1(tg), Slco1a/1b(-/-);1B3(tg), and newly generated Slco1a/1b(-/-);1A2(tg) (humanized OATP1B1, OATP1B3, and OATP1A2 transgenic) mice were characterized biochemically and physiologically, and subsequently intravenously dosed with methotrexate or paclitaxel (2 or 10 mg/kg each) for pharmacokinetic analyses. RESULTS: Humanized OATP1B1, OATP1B3, and OATP1A2 transgenic mice all showed partial or complete rescue of increased plasma bilirubin levels, but also of the increased plasma levels and decreased liver and small intestinal accumulation of methotrexate observed in Slco1a/1b(-/-) mice. Furthermore, hepatic expression of OATP1B3 and OATP1A2, but not OATP1B1, resulted in increased liver uptake of paclitaxel (2 mg/kg). At 10 mg/kg, a modest effect of only OATP1A2 on paclitaxel liver uptake was observed. CONCLUSION: Human OATP1A/1B transporters play an important role in plasma and tissue distribution of the structurally diverse chemotherapeutics methotrexate (organic anion) and paclitaxel (hydrophobic, bulky). Variation in OATP1A/1B activity due to genetic variation and pharmacologic inhibition, or differences in tumor-specific expression levels might therefore affect plasma, tissue, and tumor levels of these drugs in patients, and hence their therapeutic efficacy. Humanized transgenic OATP1A/1B mice will provide excellent tools to further study these aspects in vivo for many (anticancer) drugs.


Asunto(s)
Metotrexato/farmacocinética , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Transportadores de Anión Orgánico/metabolismo , Paclitaxel/farmacocinética , Animales , Bilirrubina/sangre , Humanos , Hígado/efectos de los fármacos , Transportador 1 de Anión Orgánico Específico del Hígado , Metotrexato/administración & dosificación , Ratones , Ratones Transgénicos , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico Sodio-Independiente/genética , Paclitaxel/administración & dosificación , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos , Distribución Tisular
8.
Mol Pharm ; 9(9): 2497-504, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22812517

RESUMEN

Organic anion-transporting polypeptides (OATPs) mediate the hepatic uptake of many drugs. Hepatic uptake is crucial for the therapeutic effect of pravastatin, a cholesterol-lowering drug and OATP1A/1B substrate. We aimed to gain empirical insight into the relationship between OATPs and pravastatin pharmacokinetics and toxicity. We therefore compared the distribution and toxicity of pravastatin in wild-type and Oatp1a/1b-null mice. Intestinal absorption of pravastatin was not affected by Oatp1a/1b absence, but systemic plasma exposure (AUC) increased up to 30-fold after oral bolus administration. This increased plasma exposure resulted from reduced hepatic uptake, as evident from 10 to 100-fold lower liver-to-plasma concentration ratios. However, the reductions in liver exposure were far smaller (<2-fold) than the increases in plasma exposure. Reduced pravastatin liver uptake in Oatp1a/1b-null mice was more obvious shortly after intravenous administration, with 8-fold lower biliary pravastatin excretion. Although mice chronically exposed to pravastatin for 60 days evinced little muscular toxicity, Oatp1a/1b-null mice displayed 10-fold higher plasma concentrations and 8-fold lower liver concentrations than wild-type mice. Thus, Oatp1a/1b transporters importantly control the hepatic uptake of pravastatin. Activity-reducing human OATP1B polymorphisms may therefore both reduce pravastatin therapeutic efficacy in the liver and increase systemic toxicity risks, thus compromising its therapeutic index in a two-edged way.


Asunto(s)
Hígado/metabolismo , Transportadores de Anión Orgánico/metabolismo , Pravastatina/farmacocinética , Administración Intravenosa , Administración Oral , Animales , Absorción Intestinal , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Transportadores de Anión Orgánico/deficiencia , Transportadores de Anión Orgánico/genética
9.
J Clin Invest ; 122(2): 519-28, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22232210

RESUMEN

Bilirubin, a breakdown product of heme, is normally glucuronidated and excreted by the liver into bile. Failure of this system can lead to a buildup of conjugated bilirubin in the blood, resulting in jaundice. The mechanistic basis of bilirubin excretion and hyperbilirubinemia syndromes is largely understood, but that of Rotor syndrome, an autosomal recessive disorder characterized by conjugated hyperbilirubinemia, coproporphyrinuria, and near-absent hepatic uptake of anionic diagnostics, has remained enigmatic. Here, we analyzed 8 Rotor-syndrome families and found that Rotor syndrome was linked to mutations predicted to cause complete and simultaneous deficiencies of the organic anion transporting polypeptides OATP1B1 and OATP1B3. These important detoxification-limiting proteins mediate uptake and clearance of countless drugs and drug conjugates across the sinusoidal hepatocyte membrane. OATP1B1 polymorphisms have previously been linked to drug hypersensitivities. Using mice deficient in Oatp1a/1b and in the multispecific sinusoidal export pump Abcc3, we found that Abcc3 secretes bilirubin conjugates into the blood, while Oatp1a/1b transporters mediate their hepatic reuptake. Transgenic expression of human OATP1B1 or OATP1B3 restored the function of this detoxification-enhancing liver-blood shuttle in Oatp1a/1b-deficient mice. Within liver lobules, this shuttle may allow flexible transfer of bilirubin conjugates (and probably also drug conjugates) formed in upstream hepatocytes to downstream hepatocytes, thereby preventing local saturation of further detoxification processes and hepatocyte toxic injury. Thus, disruption of hepatic reuptake of bilirubin glucuronide due to coexisting OATP1B1 and OATP1B3 deficiencies explains Rotor-type hyperbilirubinemia. Moreover, OATP1B1 and OATP1B3 null mutations may confer substantial drug toxicity risks.


Asunto(s)
Bilirrubina/análogos & derivados , Hiperbilirrubinemia Hereditaria/fisiopatología , Hígado/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/deficiencia , Transportadores de Anión Orgánico/deficiencia , Animales , Bilirrubina/metabolismo , Análisis Mutacional de ADN , Femenino , Humanos , Hiperbilirrubinemia Hereditaria/sangre , Hiperbilirrubinemia Hereditaria/genética , Transportador 1 de Anión Orgánico Específico del Hígado , Masculino , Ratones , Ratones Noqueados , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico Sodio-Independiente/genética , Linaje , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos
10.
Drug Metab Dispos ; 39(8): 1338-44, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21566011

RESUMEN

The ATP-binding cassette (ABC) transporters ABCC2 [multidrug resistance-associated protein (MRP) 2], ABCC3 (MRP3), and ABCG2 (breast cancer resistance protein) are involved in the efflux of potentially toxic compounds from the body. We have shown before that ABCC2, ABCC3, and ABCG2 together influence the pharmacokinetics of the anticancer and antirheumatic drug methotrexate (MTX) and its toxic metabolite 7-hydroxymethotrexate (7OH-MTX) after intravenous MTX administration. We now have used Abcc2;Abcc3;Abcg2(-/-) and corresponding single and double knockout mice to investigate the relative impact of these transporters on MTX and 7OH-MTX pharmacokinetics after oral MTX administration (50 mg/kg). The plasma areas under the curve (AUC(plasma)) in Abcg2(-/-) and Abcc2;Abcg2(-/-) mice were 1.7- and 3.0-fold higher than those in wild-type mice, respectively, suggesting additive effects of Abcc2 and Abcg2 on oral MTX pharmacokinetics. However, the AUC(plasma) in Abcc2;Abcc3;Abcg2(-/-) mice was not different from that in wild-type mice, indicating that Abcc3 protein is necessary for increased MTX plasma concentrations in the absence of Abcc2 and/or Abcg2. Furthermore, 2 h after administration, MTX liver levels were increased in Abcg2-deficient strains and MTX kidney levels were 2.2-fold increased in Abcc2;Abcg2(-/-) mice compared with those in wild-type mice. The absence of Abcc2 and/or Abcg2 also led to significantly increased liver and kidney levels of 7OH-MTX. Our results suggest that inhibition of ABCG2 and/or ABCC2, genetic polymorphisms or mutations reducing expression or activity of these proteins may increase the oral availability of MTX. Such conditions may also present risk factors for increased MTX-related toxicity in patients treated with oral MTX.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Antimetabolitos Antineoplásicos/farmacocinética , Metotrexato/análogos & derivados , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Administración Oral , Animales , Antimetabolitos Antineoplásicos/sangre , Antimetabolitos Antineoplásicos/metabolismo , Área Bajo la Curva , Disponibilidad Biológica , Femenino , Inyecciones Intravenosas , Metotrexato/sangre , Metotrexato/metabolismo , Metotrexato/farmacocinética , Ratones , Ratones Noqueados , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Especificidad de Órganos , Distribución Tisular
11.
Clin Cancer Res ; 17(2): 294-301, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21097690

RESUMEN

PURPOSE: Organic anion-transporting polypeptides (OATP) mediate the cellular uptake of a broad range of drugs. The hydrophobic anticancer drug, paclitaxel (PTX), was recently identified as a substrate for OATP1B3 in vitro. We investigated the role of Oatp1a/1b transporters in the pharmacokinetics of PTX in vivo, as well as their impact at different dose levels of PTX and methotrexate (MTX). EXPERIMENTAL DESIGN: Recently generated Slco1a/1b(-/-) (lacking all Oatp1a/1b transporters) and wild-type mice were intravenously dosed with 2, 10, or 50 mg/kg of PTX, or with 10, 50, or 500 mg/kg of MTX, and plasma and tissue drug concentrations were measured. RESULTS: In spite of its hydrophobicity, PTX systemic exposure (at 10 mg/kg) was increased by greater than 2-fold in Slco1a/1b(-/-) mice compared with wild-type, whereas PTX liver uptake was reduced by about 2-fold. Oatp1a/1b transporters displayed a high impact on PTX and MTX pharmacokinetics over a broad dose range. For MTX, even at 500 mg/kg, saturation of Oatp1a/1b was not observed, with a 3.4-fold increase in plasma and 30-fold decrease in liver levels in Slco1a/1b(-/-) mice compared with wild-type. Although beginning saturation of Oatp1a/1b was observed at the highest dose of PTX, plasma levels in Slco1a/1b(-/-) mice were still 1.7-fold increased and liver levels 1.5-fold decreased compared with wild-type. CONCLUSION: Oatp1a/1b transporters play a pronounced role in determining plasma levels and tissue distribution of MTX and PTX, thus affecting even highly hydrophobic drugs. Variation in OATP1A/1B transporter activity, due to genetic variation, inhibition, and/or tumor expression might affect toxicity and therapeutic efficacy of these anticancer drugs.


Asunto(s)
Transportadores de Anión Orgánico Sodio-Independiente/farmacología , Transportadores de Anión Orgánico/farmacología , Transportadores de Anión Orgánico/fisiología , Proteínas de Transporte de Catión Orgánico/farmacología , Animales , Antineoplásicos/farmacocinética , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Interacciones Hidrofóbicas e Hidrofílicas , Hígado/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado , Metotrexato/farmacocinética , Ratones , Ratones Noqueados , Paclitaxel/farmacocinética
12.
Mol Cancer Ther ; 8(12): 3350-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19996279

RESUMEN

The multidrug transporters ABCC2, ABCC3, and ABCG2 can eliminate potentially toxic compounds from the body and have overlapping substrate specificities. To investigate the overlapping functions of Abcc2, Abcc3, and Abcg2 in vivo, we generated and characterized Abcc3;Abcg2-/- and Abcc2;Abcc3;Abcg2-/- mice. We subsequently analyzed the relative impact of these transport proteins on the pharmacokinetics of the anticancer drug methotrexate (MTX) and its main, toxic, metabolite 7-hydroxymethotrexate (7OH-MTX) after i.v. administration of MTX (50 mg/kg). Whereas in single and double knockout mice, the plasma and liver concentrations of MTX and 7OH-MTX decreased rapidly after MTX administration, in the Abcc2;Abcc3;Abcg2-/- mice, they remained very high. One hour after administration, 67% of the MTX dose was still present in livers of Abcc2;Abcc3;Abcg2-/- mice as MTX or 7OH-MTX versus 7% in wild-type, showing dramatic liver accumulation of these toxic compounds when Abcc2, Abcc3, and Abcg2 were all absent. Furthermore, the urinary and fecal excretion of the nephrotoxic metabolite 7OH-MTX were increased 27- and 7-fold, respectively, in Abcc2;Abcc3;Abcg2-/- mice. Thus, Abcc2, Abcc3, and Abcg2 together mediate the rapid elimination of MTX and 7OH-MTX after i.v. administration and can to a large extent compensate for each other's absence. This may explain why it is still comparatively safe to use a toxic drug such as MTX in the clinic, as the risk of highly increased toxicity due to dysfunctioning of ABCC2, ABCC3, or ABCG2 alone is limited. Nevertheless, cotreatment with possible inhibitors of ABCC2, ABCC3, and ABCG2 should be done with utmost caution when treating patients with methotrexate.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Metotrexato/análogos & derivados , Metotrexato/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Alanina Transaminasa/sangre , Análisis de Varianza , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/sangre , Antimetabolitos Antineoplásicos/farmacocinética , Área Bajo la Curva , Western Blotting , Cromatografía Líquida de Alta Presión , Creatinina/sangre , Heces/química , Femenino , Intestino Delgado/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Masculino , Tasa de Depuración Metabólica , Metotrexato/sangre , Metotrexato/farmacocinética , Metotrexato/orina , Ratones , Ratones Noqueados , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular
13.
Clin Cancer Res ; 15(9): 3084-93, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19383815

RESUMEN

PURPOSE: ABCC2 (MRP2) and ABCG2 (BCRP) transport various endogenous and exogenous compounds, including many anticancer drugs, into bile, feces, and urine. We investigated the possibly overlapping roles of Abcg2 and Abcc2 in the elimination of the anticancer drug methotrexate (MTX) and its toxic metabolite 7-hydroxymethotrexate (7OH-MTX). EXPERIMENTAL DESIGN: We generated and characterized Abcc2;Abcg2(-/-) mice, and used these to determine the overlapping roles of Abcc2 and Abcg2 in the elimination of MTX and 7OH-MTX after i.v. administration of 50 mg/kg MTX. RESULTS: Compared with wild-type, the plasma areas under the curve (AUC) for MTX were 1.6-fold and 2.0-fold higher in Abcg2(-/-) and Abcc2(-/-) mice, respectively, and 3.3-fold increased in Abcc2;Abcg2(-/-) mice. The biliary excretion of MTX was 23-fold reduced in Abcc2;Abcg2(-/-) mice, and the MTX levels in the small intestine were dramatically decreased. Plasma levels of 7OH-MTX were not significantly altered in Abcg2(-/-) mice, but the areas under the curve were 6.2-fold and even 12.4-fold increased in Abcc2(-/-) and Abcc2;Abcg2(-/-) mice, respectively. This indicates that Abcc2 compensates for Abcg2 deficiency but that Abcg2 can only partly compensate for Abcc2 absence. Furthermore, 21-fold decreased biliary 7OH-MTX excretion in Abcc2;Abcg2(-/-) mice and substantial 7OH-MTX accumulation in the liver and kidney were seen. We additionally found that in the absence of Abcc2, Abcg2 mediated substantial urinary excretion of MTX and 7OH-MTX. CONCLUSIONS: Abcc2 and Abcg2 together are major determinants of MTX and 7OH-MTX pharmacokinetics. Variations in ABCC2 and/or ABCG2 activity due to polymorphisms or coadministered inhibitors may therefore substantially affect the therapeutic efficacy and toxicity in patients treated with MTX.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Sistema Biliar/metabolismo , Hígado/metabolismo , Metotrexato/análogos & derivados , Metotrexato/farmacocinética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Área Bajo la Curva , Western Blotting , Cromatografía Líquida de Alta Presión , Heces/química , Femenino , Antagonistas del Ácido Fólico/farmacocinética , Antagonistas del Ácido Fólico/toxicidad , Antagonistas del Ácido Fólico/orina , Masculino , Metotrexato/toxicidad , Metotrexato/orina , Ratones , Ratones Noqueados , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular
14.
Clin Cancer Res ; 14(24): 8152-60, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19088030

RESUMEN

PURPOSE: ATP-binding cassette sub-family C member 2 [ABCC2; multidrug resistance-associated protein 2 (MRP2)] and ABCC3 (MRP3) mediate the elimination of toxic compounds, such as drugs and carcinogens, and have a large overlap in substrate specificity. We investigated the roles of Abcc2 and Abcc3 in the elimination of the anticancer drug methotrexate (MTX) and its toxic metabolite 7-hydroxymethotrexate (7OH-MTX) in vivo. EXPERIMENTAL DESIGN: Abcc2;Abcc3(-/-) mice were generated, characterized, and used to investigate possibly overlapping or complementary roles of Abcc2 and Abcc3 in the elimination of MTX and 7OH-MTX after i.v. administration of 50 mg/kg MTX. RESULTS: Abcc2;Abcc3(-/-) mice were viable and fertile. In Abcc2(-/-) mice, the plasma area under the curve (AUCi.v.) for MTX was 2.0-fold increased compared with wild type, leading to 1.6-fold increased urinary excretion, which was not seen in Abcc2;Abcc3(-/-) mice. Biliary excretion of MTX was 3.7-fold reduced in Abcc2(-/-) but unchanged in Abcc2;Abcc3(-/-) mice. The plasma AUCi.v.s of 7OH-MTX were 6.0-fold and 4.3-fold increased in Abcc2(-/-) and Abcc2;Abcc3(-/-) mice, respectively, leading to increased urinary excretion. The biliary excretion of 7OH-MTX was 5.8-fold reduced in Abcc2(-/-) but unchanged in Abcc2;Abcc3(-/-) mice. 7OH-MTX accumulated substantially in the liver of Abcc2(-/-) and especially Abcc2;Abcc3(-/-) mice. CONCLUSIONS: Abcc2 is important for (biliary) excretion of MTX and its toxic metabolite 7OH-MTX. When Abcc2 is absent, Abcc3 transports MTX and 7OH-MTX back from the liver into the circulation, leading to increased plasma levels and urinary excretion. Variation in ABCC2 and/or ABCC3 activity may therefore have profound effects on the elimination and severity of toxicity of MTX and 7OH-MTX after MTX treatment of patients.


Asunto(s)
Metotrexato/análogos & derivados , Metotrexato/farmacocinética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Animales , Bilis/metabolismo , Femenino , Hígado/metabolismo , Masculino , Metotrexato/toxicidad , Ratones , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos
15.
Am J Pathol ; 167(1): 47-58, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15972951

RESUMEN

Wegener's granulomatosis, microscopic polyangiitis, Churg-Strauss syndrome, and idiopathic pauci-immune necrotizing crescentic glomerulonephritis are associated with myeloperoxidase (MPO)-specific anti-neutrophil cytoplasmic autoantibodies (ANCAs). Clinical and experimental evidence indicates that ANCA and proinflammatory stimuli of infectious origin act synergistically to cause vasculitis. We tested this hypothesis in a recently developed mouse model of anti-MPO IgG-induced glomerulonephritis by using bacterial lipopolysaccharide (LPS) as the proinflammatory stimulus. Systemic administration of LPS dose dependently increased renal injury induced by anti-MPO IgG as demonstrated by increased glomerular crescent formation and glomerular necrosis. In the early phase, LPS enhanced anti-MPO IgG-induced glomerular neutrophil accumulation. Furthermore, a transient induction of circulating tumor necrosis factor (TNF)-alpha levels, followed by a marked increase in circulating MPO levels, was observed on administration of LPS. In vitro, anti-MPO IgG induced a respiratory burst in murine neutrophils only after priming with TNF-alpha. Finally, anti-TNF-alpha treatment attenuated, but did not prevent, the LPS-mediated aggravation of anti-MPO IgG-induced glomerulonephritis. In conclusion, our study demonstrates that ANCA and proinflammatory stimuli act synergistically to induce vasculitic disease and suggests potential benefits of inhibiting TNF-alpha bioactivity in treating human ANCA-associated necrotizing crescentic glomerulonephritis.


Asunto(s)
Anticuerpos Anticitoplasma de Neutrófilos/inmunología , Glomerulonefritis/inmunología , Lipopolisacáridos/inmunología , Peroxidasa/inmunología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Glomerulonefritis/patología , Técnicas In Vitro , Ratones , Neutrófilos/inmunología , Peroxidasa/sangre , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/inmunología , Vasculitis/inmunología , Vasculitis/patología
16.
Int J Cancer ; 105(1): 20-5, 2003 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-12672025

RESUMEN

The current research aimed to define hypothesis-based anti-angiogenic properties of the vascular targeting agent combretastatin A-4 phosphate (combreAp). The in vitro wound assay indicated that combreAp potently inhibited migration of endothelial cells (EC). A significant inhibition of migration could already be measured after 2 hr of treatment. In a three-dimensional (3D) tube formation assay, combreAp inhibited sprout formation at concentrations that did not inhibit the proliferation of EC. At sub-ng concentrations the half-maximal response was reached. Interestingly, although combreAp is considered a vascular targeting agent, the human tumor cell lines tested were found to be 20-30 times more sensitive for combreAp than the human umbilical vein endothelial cells (HUVEC). A similar response difference between rat EC and R1 rat rhabdomyosarcoma tumor cells was observed. The growth inhibition in EC was only in part mediated by induction of apoptosis. The growth delay results obtained with the in vivo rodent tumor models involving repeat dosing of combreAp can partly be explained by anti-angiogenic activity of the compound. The results obtained with the various in vitro and in vivo assays substantiate an anti-angiogenic profile of combreAp, largely at the level of EC migration. This mechanism may operate to a different extent in different tumor types.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neovascularización Patológica , Estilbenos/farmacología , Animales , Apoptosis , División Celular , Movimiento Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Endotelio Vascular/metabolismo , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos C57BL , Ratas , Factores de Tiempo , Células Tumorales Cultivadas , Venas Umbilicales/citología , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...