Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Control Release ; 349: 109-117, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35798092

RESUMEN

P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) are two efflux transporters which are expressed in the apical (i.e. airway lumen-facing) membranes of lung epithelial cells. To assess the influence of P-gp and BCRP on the pulmonary disposition of inhaled drugs, we performed positron emission tomography (PET) imaging in rats after intratracheal aerosolization of two model P-gp/BCRP substrate radiotracers (i.e. [11C]erlotinib and [11C]tariquidar). We studied rat groups in which both transporters were active (i.e. wild-type rats), either of the two transporters was inactive (Abcb1a/b(-/-) and Abcg2(-/-) rats) or both transporters were inactive (Abcg2(-/-) rats in which pulmonary P-gp activity was inhibited by treatment with unlabeled tariquidar). PET-measured lung distribution data were compared with brain-to-plasma radioactivity concentration ratios measured in a gamma counter at the end of the PET scan. For [11C]erlotinib, lung exposure (AUClungs) was moderately but not significantly increased in Abcb1a/b(-/-) rats (1.6-fold) and Abcg2(-/-) rats (1.5-fold), and markedly (3.6-fold, p < 0.0001) increased in tariquidar-treated Abcg2(-/-) rats, compared to wild-type rats. Similarly, the brain uptake of [11C]erlotinib was substantially (4.5-fold, p < 0.0001) increased when both P-gp and BCRP activities were impaired. For [11C]tariquidar, differences in AUClungs between groups pointed into a similar direction as for [11C]erlotinib, but were less pronounced and lacked statistical significance. Our study demonstrates functional P-gp and BCRP activity in vivo in the lungs and further suggests functional redundancy between P-gp and BCRP in limiting the pulmonary uptake of a model P-gp/BCRP substrate, analogous to the blood-brain barrier. Our results suggest that pulmonary efflux transporters are important for the efficacy and safety of inhaled drugs and that their modulation may be exploited in order to improve the pharmacokinetic and pharmacodynamic performance of pulmonary delivered drugs.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Proteínas de Neoplasias , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Clorhidrato de Erlotinib , Pulmón/diagnóstico por imagen , Pulmón/metabolismo , Proteínas de Neoplasias/metabolismo , Tomografía de Emisión de Positrones/métodos , Ratas
2.
J Control Release ; 342: 44-52, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34971693

RESUMEN

Several drugs approved for inhalation for the treatment of pulmonary diseases are substrates of the adenosine triphosphate-binding cassette (ABC) transporter P-glycoprotein (P-gp). P-gp is expressed in the apical membrane of pulmonary epithelial cells and could play a role in modulating the pulmonary absorption and distribution of inhaled drugs, thereby potentially contributing to variability in therapeutic response and/or systemic side effects. We developed a new in vivo experimental approach to assess the functional impact of P-gp on the pulmonary delivery of inhaled drugs in rats. By using positron emission tomography (PET) imaging, we measured the intrapulmonary pharmacokinetics of the model P-gp substrates (R)-[11C]verapamil ([11C]VPM) and [11C]-N-desmethyl-loperamide ([11C]dLOP) administered by intratracheal aerosolization in three rat groups: wild-type, Abcb1a/b(-/-) and wild-type treated with the P-gp inhibitor tariquidar. Lung exposure (AUClung_right) to [11C]VPM was 64% and 50% lower (p < 0.05) in tariquidar-treated and in Abcb1a/b(-/-) rats, respectively, compared to untreated wild-type rats. For [11C]dLOP, AUClung_right was 59% and 34% lower (p < 0.05) in tariquidar-treated and in Abcb1a/b(-/-) rats, respectively. Our results show that P-gp can affect the pulmonary disposition of inhaled P-gp substrates, whereby a decrease in P-gp activity may lead to lower lung exposure and potentially to a decrease in therapeutic efficacy. Our study highlights the potential of PET imaging with intratracheally aerosolized radiotracers to assess the impact of membrane transporters on pulmonary drug delivery, in rodents and potentially also in humans.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Barrera Hematoencefálica , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Pulmón/diagnóstico por imagen , Pulmón/metabolismo , Tomografía de Emisión de Positrones/métodos , Ratas
3.
Alzheimers Res Ther ; 13(1): 175, 2021 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-34656177

RESUMEN

BACKGROUND: To better understand the etiology and pathomechanisms of Alzheimer's disease, several transgenic animal models that overexpress human tau or human amyloid-beta (Aß) have been developed. In the present study, we generated a novel transgenic rat model by cross-breeding amyloid precursor protein (APP) rats with tau rats. We characterized this model by performing positron emission tomography scans combined with immunofluorescent labeling and cerebrospinal fluid analyses. METHODS: APP/Tau rats were generated by cross-breeding male McGill-R-Thy1-APP transgenic rats with female hTau-40/P301L transgenic rats. APP/Tau double transgenic rats and non-transgenic (ntg) littermates aged 7, 13, and 21 months were subjected to dynamic [11C] PiB scan and dynamic [18F]THK-5317 scans. For regional brain analysis, a template was generated from anatomical MR images of selected animals, which was co-registered with the PET images. Regional analysis was performed by application of the simplified reference tissue model ([11C]PiB data), whereas [18F]THK-5317 data were analyzed using a 2-tissue compartment model and Logan graphical analysis. In addition, immunofluorescent labeling (tau, amyloid) and cerebrospinal fluid analyses were performed. RESULTS: [11C]PiB binding potential (BPND) and [18F]THK-5317 volume of distribution (VT) showed an increase with age in several brain regions in the APP/Tau group but not in the ntg control group. Immunohistochemical analysis of brain slices of PET-scanned animals revealed a positive correlation between Aß labeling and [11C]PiB regional BPND. Tau staining yielded a trend towards higher levels in the cortex and hippocampus of APP/Tau rats compared with ntg littermates, but without reaching statistical significance. No correlation was found between tau immunofluorescence labeling results and the respective [18F]THK-5317 VT values. CONCLUSIONS: We thoroughly characterized a novel APP/Tau rat model using combined PET imaging and immunofluorescence analysis. We observed an age-related increase in [11C]PiB and [18F]THK-5317 binding in several brain regions in the APP/Tau group but not in the ntg group. Although we were able to reveal a positive correlation between amyloid labeling and [11C]PiB regional brain uptake, we observed relatively low human tau and amyloid fibril expression levels and a somewhat unstable brain pathology which questions the utility of this animal model for further studies.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides , Precursor de Proteína beta-Amiloide/genética , Animales , Femenino , Masculino , Tomografía de Emisión de Positrones , Ratas , Ratas Transgénicas
4.
Pharmaceutics ; 13(8)2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34452247

RESUMEN

P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) are co-localized at the blood-brain barrier, where they display functional redundancy to restrict the brain distribution of dual P-gp/BCRP substrate drugs. We used positron emission tomography (PET) with the metabolically stable P-gp/BCRP substrates [11C]tariquidar, [11C]erlotinib, and [11C]elacridar to assess whether a similar functional redundancy as at the BBB exists in the liver, where both transporters mediate the biliary excretion of drugs. Wild-type, Abcb1a/b(-/-), Abcg2(-/-), and Abcb1a/b(-/-)Abcg2(-/-) mice underwent dynamic whole-body PET scans after i.v. injection of either [11C]tariquidar, [11C]erlotinib, or [11C]elacridar. Brain uptake of all three radiotracers was markedly higher in Abcb1a/b(-/-)Abcg2(-/-) mice than in wild-type mice, while only moderately changed in Abcb1a/b(-/-) and Abcg2(-/-) mice. The transfer of radioactivity from liver to excreted bile was significantly lower in Abcb1a/b(-/-)Abcg2(-/-) mice and almost unchanged in Abcb1a/b(-/-) and Abcg2(-/-) mice (with the exception of [11C]erlotinib, for which biliary excretion was also significantly reduced in Abcg2(-/-) mice). Our data provide evidence for redundancy between P-gp and BCRP in controlling both the brain distribution and biliary excretion of dual P-gp/BCRP substrates and highlight the utility of PET as an upcoming tool to assess the effect of transporters on drug disposition at a whole-body level.

5.
Eur J Pharm Sci ; 163: 105854, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-33865975

RESUMEN

Ciprofloxacin is a commonly prescribed fluoroquinolone antibiotic which is cleared by active tubular secretion and intestinal excretion. Ciprofloxacin is a known substrate of the ATP-binding cassette (ABC) transporters breast cancer resistance protein (BCRP) and multidrug resistance-associated protein 4 (MRP4). In this work, we used positron emission tomography (PET) imaging to investigate the influence of BCRP, MRP4, MRP2 and P-glycoprotein (P-gp) on the excretion of [18F]ciprofloxacin in mice. Dynamic 90-min PET scans were performed after intravenous injection of [18F]ciprofloxacin in wild-type mice without and with pre-treatment with the broad-spectrum MRP inhibitor MK571. Moreover, [18F]ciprofloxacin PET scans were performed in Abcc4(-/-), Abcc2(-/-), Abcc4(-/-)Abcg2(-/-) and Abcb1a/b(-/-)Abcg2(-/-) mice. In addition to non-compartmental pharmacokinetic (PK) analysis, a novel three-compartment PK model was developed for a detailed assessment of the renal disposition of [18F]ciprofloxacin. In MK571 pre-treated mice, a significant increase in the blood exposure to [18F]ciprofloxacin was observed along with a significant reduction in the renal and intestinal clearances. PK modelling revealed a significant reduction in renal radioactivity uptake (CL1) and in the rate constants for transfer of radioactivity from the corticomedullary renal region into blood (k2) and urine (k3), respectively, after MK571 administration. No changes in the renal clearance or in the estimated kidney PK model parameters were observed in any of the studied knockout models, while a significant reduction in the intestinal clearance was observed in Abcc2(-/-) and Abcc4(-/-)Abcg2(-/-) mice. Our data failed to reveal a role of any of the studied ABC transporters in the tubular secretion of ciprofloxacin. This may indicate that ciprofloxacin is handled in the kidneys by more than one transporter family, most likely with a great degree of mutual functional redundancy. Our study highlights the potential of PET imaging for an assessment of transporter-mediated renal excretion of radiolabelled drugs.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Ciprofloxacina , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportadoras de Casetes de Unión a ATP/genética , Animales , Ratones , Ratones Noqueados , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas de Neoplasias/metabolismo , Tomografía de Emisión de Positrones
6.
Pharm Res ; 38(1): 127-140, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33559045

RESUMEN

PURPOSE: To investigate the role of cation transporters (OCTs, MATEs) in the renal and hepatic disposition of the radiolabeled antiemetic drug [11C]metoclopramide in mice with PET. METHODS: PET was performed in wild-type mice after administration of an intravenous microdose (<1 µg) of [11C]metoclopramide without and with co-administration of either unlabeled metoclopramide (5 or 10 mg/kg) or the prototypical cation transporter inhibitors cimetidine (150 mg/kg) or sulpiride (25 mg/kg). [11C]Metoclopramide PET was also performed in wild-type and Slc22a1/2(-/-) mice. Radiolabeled metabolites were measured at 15 min after radiotracer injection and PET data were corrected for radiolabeled metabolites. RESULTS: [11C]Metoclopramide was highly metabolized and [11C]metoclopramide-derived radioactivity was excreted into the urine. The different investigated treatments decreased (~2.5-fold) the uptake of [11C]metoclopramide from plasma into the kidney and liver, inhibited metabolism and decreased (up to 3.8-fold) urinary excretion, which resulted in increased plasma concentrations of [11C]metoclopramide. Kidney and liver uptake were moderately (~1.3-fold) reduced in Slc22a1/2(-/-) mice. CONCLUSIONS: Our results suggest a contribution of OCT1/2 to the kidney and liver uptake and of MATEs to the urinary excretion of [11C]metoclopramide in mice. Cation transporters may contribute, next to variability in the activity of metabolizing enzymes, to variability in metoclopramide pharmacokinetics and side effects.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Eliminación Hepatobiliar , Metoclopramida/farmacocinética , Transportador 2 de Cátion Orgánico/metabolismo , Eliminación Renal , Animales , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Femenino , Células HEK293 , Humanos , Masculino , Metoclopramida/administración & dosificación , Ratones , Ratones Noqueados , Modelos Animales , Transportador 2 de Cátion Orgánico/genética
7.
J Cereb Blood Flow Metab ; 41(7): 1634-1646, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33081568

RESUMEN

P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) restrict at the blood-brain barrier (BBB) the brain distribution of the majority of currently known molecularly targeted anticancer drugs. To improve brain delivery of dual ABCB1/ABCG2 substrates, both ABCB1 and ABCG2 need to be inhibited simultaneously at the BBB. We examined the feasibility of simultaneous ABCB1/ABCG2 inhibition with i.v. co-infusion of erlotinib and tariquidar by studying brain distribution of the model ABCB1/ABCG2 substrate [11C]erlotinib in mice and rhesus macaques with PET. Tolerability of the erlotinib/tariquidar combination was assessed in human embryonic stem cell-derived cerebral organoids. In mice and macaques, baseline brain distribution of [11C]erlotinib was low (brain distribution volume, VT,brain < 0.3 mL/cm3). Co-infusion of erlotinib and tariquidar increased VT,brain in mice by 3.0-fold and in macaques by 3.4- to 5.0-fold, while infusion of erlotinib alone or tariquidar alone led to less pronounced VT,brain increases in both species. Treatment of cerebral organoids with erlotinib/tariquidar led to an induction of Caspase-3-dependent apoptosis. Co-infusion of erlotinib/tariquidar may potentially allow for complete ABCB1/ABCG2 inhibition at the BBB, while simultaneously achieving brain-targeted EGFR inhibition. Our protocol may be applicable to enhance brain delivery of molecularly targeted anticancer drugs for a more effective treatment of brain tumors.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Radioisótopos de Carbono/metabolismo , Clorhidrato de Erlotinib/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Quinolinas/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Permeabilidad de la Membrana Celular , Sistemas de Liberación de Medicamentos , Quimioterapia Combinada , Clorhidrato de Erlotinib/administración & dosificación , Femenino , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Macaca mulatta , Masculino , Ratones
8.
Int J Mol Sci ; 21(21)2020 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-33153231

RESUMEN

BACKGROUND: ABCB1 (P-glycoprotein) and ABCG2 (breast cancer resistance protein) are co-localized at the blood-brain barrier (BBB), where they restrict the brain distribution of many different drugs. Moreover, ABCB1 and possibly ABCG2 play a role in Alzheimer's disease (AD) by mediating the brain clearance of beta-amyloid (Aß) across the BBB. This study aimed to compare the abundance and activity of ABCG2 in a commonly used ß-amyloidosis mouse model (APP/PS1-21) with age-matched wild-type mice. METHODS: The abundance of ABCG2 was assessed by semi-quantitative immunohistochemical analysis of brain slices of APP/PS1-21 and wild-type mice aged 6 months. Moreover, the brain distribution of two dual ABCB1/ABCG2 substrate radiotracers ([11C]tariquidar and [11C]erlotinib) was assessed in APP/PS1-21 and wild-type mice with positron emission tomography (PET). [11C]Tariquidar PET scans were performed without and with partial inhibition of ABCG2 with Ko143, while [11C]erlotinib PET scans were only performed under baseline conditions. RESULTS: Immunohistochemical analysis revealed a significant reduction (by 29-37%) in the number of ABCG2-stained microvessels in the brains of APP/PS1-21 mice. Partial ABCG2 inhibition significantly increased the brain distribution of [11C]tariquidar in APP/PS1-21 and wild-type mice, but the brain distribution of [11C]tariquidar did not differ under both conditions between the two mouse strains. Similar results were obtained with [11C]erlotinib. CONCLUSIONS: Despite a reduction in the abundance of cerebral ABCG2 and ABCB1 in APP/PS1-21 mice, the brain distribution of two dual ABCB1/ABCG2 substrates was unaltered. Our results suggest that the brain distribution of clinically used ABCB1/ABCG2 substrate drugs may not differ between AD patients and healthy people.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Amiloidosis/metabolismo , Amiloidosis/patología , Encéfalo/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Amiloidosis/diagnóstico por imagen , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/diagnóstico por imagen , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tomografía de Emisión de Positrones , Quinolinas/farmacocinética , Distribución Tisular
9.
Zootaxa ; 4731(4): zootaxa.4731.4.1, 2020 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-32230280

RESUMEN

Schindleria (Giltay (1934), Schindler's fishes (or infantfishes), is a genus of small (< 22 mm) paedomorphic species of the family Gobiidae which mature extremely fast. These fishes occur from the eastern Pacific (Cocos Islands off Costa Rica, seamounts Nazca and Sala y Gómez) to the southwestern Indian Ocean (southeast Africa). Nevertheless, there is a large gap in the distributional area between the Philippines (western Pacific) and India/Sri Lanka (Central Indian Ocean) which spans nearly 5000 km. We present the first comprehensive documentation of published records of Schindleria together with samples collected during the Dana-Expedition, between 1928 and 1930 at 44 stations from Polynesia to southeast Africa, with 8 records from the western Pacific to the Central Indian Ocean. We present three first records, 18 new records and the southernmost record for the Indian Ocean. Although Schindler's fishes were generally documented from or close to islands and reefs, we present 23 offshore records (at least 30 km distant to a shore or reef) and 27 deep-water records (at least 65 m deep). Records between 320 and 360 km offshore are the most extreme offshore records of Schindleria ever documented. The records from about 500- and 1000-m depths are the deepest ever documented for Schindler's fishes.


Asunto(s)
Peces , Animales , Expediciones , Océano Pacífico , Agua
10.
J Nucl Med ; 61(11): 1650-1657, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32284394

RESUMEN

Multidrug resistance-associated protein 1 (adenosine triphosphate-binding cassette subfamily C member 1 [ABCC1]) is abundantly expressed at the lung epithelial barrier, where it may influence the pulmonary disposition of inhaled drugs and contribute to variability in therapeutic response. The aim of this study was to assess the impact of ABCC1 on the pulmonary disposition of 6-bromo-7-11C-methylpurine (11C-BMP), a prodrug radiotracer that is intracellularly conjugated with glutathione to form the ABCC1 substrate S-(6-(7-11C-methylpurinyl))glutathione (11C-MPG). Methods: Groups of Abcc1(-/-) rats, wild-type rats pretreated with the ABCC1 inhibitor MK571, and wild-type control rats underwent dynamic PET scans after administration of 11C-BMP intravenously or by intratracheal aerosolization. In vitro transport experiments were performed with unlabeled BMP on the human distal lung epithelial cell line NCI-H441. Results: The pulmonary kinetics of radioactivity significantly differed between wild-type and Abcc1(-/-) rats, but differences were more pronounced after intratracheal than after intravenous administration. After intravenous administration, lung exposure (area under the lung time-activity curve from 0 to 80 min after radiotracer administration [AUClung]) was 77% higher and the elimination slope of radioactivity washout from the lungs (kE,lung) was 70% lower in Abcc1(-/-) rats, whereas after intratracheal administration, AUClung was 352% higher and kE,lung was 86% lower in Abcc1(-/-) rats. Pretreatment with MK571 decreased kE,lung by 20% after intratracheal radiotracer administration. Intracellular accumulation of MPG in NCI-H441 cells was significantly higher and extracellular efflux was lower in the presence than in the absence of MK571. Conclusion: PET with pulmonary administered 11C-BMP can measure ABCC1 activity at the lung epithelial barrier and may be applicable in humans to assess the effects of disease, genetic polymorphisms, or concomitant drug intake on pulmonary ABCC1 activity.


Asunto(s)
Radioisótopos de Carbono/farmacocinética , Pulmón/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Purinas/farmacocinética , Animales , Células Cultivadas , Femenino , Humanos , Tomografía de Emisión de Positrones , Propionatos/farmacología , Quinolinas/farmacología , Ratas , Ratas Sprague-Dawley
11.
Nucl Med Biol ; 84-85: 28-32, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31981857

RESUMEN

INTRODUCTION: Tau deposition is one of the hallmarks of Alzheimer's disease (AD) and can be visualized and quantified using [18F]THK-5317 together with kinetic modeling. To determine the feasibility of this approach, we measured blood/plasma pharmacokinetics and radiotracer metabolism in female and male rats. METHODS: Female and male rats (n = 11-12) were cannulated via the femoral artery for continuous blood sampling. Blood sampling was performed at regular intervals after intravenous injection of [18F]THK-5317. After collection of the last blood sample, animals were sacrificed, and organs were excised. Blood from minute 5, 20 and 60 was centrifuged to obtain plasma. Radiolabeled metabolites in plasma, brain, liver and urine were analyzed by radio-thin-layer chromatography (radio-TLC). RESULTS: Plasma pharmacokinetics and metabolism were significantly different between female and male rats. [18F]THK-5317 plasma clearance was faster in female (0.66 ± 0.08 mL/h/kg BW) than in male (0.52 ± 0.11 mL/h/kg BW) rats (p = .005). The percentage of unmetabolized parent was significantly different between both sexes at 20 min and 60 min p.i. In the liver, a 1.6-fold higher radioactivity concentration was found in male versus female animals and in addition also the percentage of unmetabolized parent was different. CONCLUSION: Our results show pronounced sex differences in blood/plasma pharmacokinetics and metabolism of [18F]THK-5317 in rats. Female animals showed a faster plasma clearance compared to males. These results underline the importance of investigating both sexes and also support the notion that individual input functions or sex-specific population-based input functions are needed for kinetic modeling analyses. ADVANCES IN KNOWLEDGE: First preclinical study in rats showing pronounced sex differences in blood/plasma pharmacokinetics and metabolism of [18F]THK-5317. IMPLICATIONS FOR PATIENT CARE: Sex-specific differences might also be present in humans and thus clinical trials should have adequate sample size to account for effects in men and women separately.


Asunto(s)
Compuestos de Anilina/farmacocinética , Quinolinas/farmacocinética , Caracteres Sexuales , Enfermedad de Alzheimer/metabolismo , Compuestos de Anilina/sangre , Compuestos de Anilina/metabolismo , Animales , Femenino , Masculino , Quinolinas/sangre , Quinolinas/metabolismo , Trazadores Radiactivos , Ratas , Distribución Tisular , Proteínas tau/metabolismo
12.
J Cereb Blood Flow Metab ; 40(1): 150-162, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30354871

RESUMEN

P-glycoprotein (P-gp, ABCB1) is an efflux transporter at the blood-brain barrier (BBB), which mediates clearance of beta-amyloid (Aß) from brain into blood. We used (R)-[11C]verapamil PET in combination with partial P-gp inhibition with tariquidar to measure cerebral P-gp function in a beta-amyloidosis mouse model (APPtg) and in control mice at three different ages (50, 200 and 380 days). Following tariquidar pre-treatment (4 mg/kg), whole brain-to-plasma radioactivity concentration ratios (Kp,brain) were significantly higher in APPtg than in wild-type mice aged 50 days, pointing to decreased cerebral P-gp function. Moreover, we found an age-dependent decrease in cerebral P-gp function in both wild-type and APPtg mice of up to -50%. Alterations in P-gp function were more pronounced in Aß-rich brain regions (hippocampus, cortex) than in a control region with negligible Aß load (cerebellum). PET results were confirmed by immunohistochemical staining of P-gp in brain microvessels. Our results confirm previous findings of reduced P-gp function in Alzheimer's disease mouse models and show that our PET protocol possesses adequate sensitivity to measure these functional changes in vivo. Our PET protocol may find use in clinical studies to test the efficacy of drugs to induce P-gp function at the human BBB to enhance Aß clearance.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Amiloidosis/metabolismo , Química Encefálica , Tomografía de Emisión de Positrones/métodos , Factores de Edad , Enfermedad de Alzheimer/metabolismo , Animales , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Modelos Animales de Enfermedad , Ratones , Quinolinas/farmacología
13.
J Cereb Blood Flow Metab ; 40(5): 954-965, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31195936

RESUMEN

Previous data suggest a possible link between multidrug resistance-associated protein 1 (ABCC1) and brain clearance of beta-amyloid (Aß). We used PET with 6-bromo-7-[11C]methylpurine ([11C]BMP) to measure cerebral ABCC1 transport activity in a beta-amyloidosis mouse model (APP/PS1-21) and in wild-type mice aged 50 and 170 days, without and with pretreatment with the ABCC1 inhibitor MK571. One hundred seventy days-old-animals additionally underwent [11C]PiB PET scans to measure Aß load. While baseline [11C]BMP PET scans detected no differences in the elimination slope of radioactivity washout from the brain (kelim) between APP/PS1-21 and wild-type mice of both age groups, PET scans after MK571 pretreatment revealed significantly higher kelim values in APP/PS1-21 mice than in wild-type mice aged 170 days, suggesting increased ABCC1 activity. The observed increase in kelim occurred across all investigated brain regions and was independent of the presence of Aß plaques measured with [11C]PiB. Western blot analysis revealed a trend towards increased whole brain ABCC1 levels in 170 days-old-APP/PS1-21 mice versus wild-type mice and a significant positive correlation between ABCC1 levels and kelim. Our data point to an upregulation of ABCC1 in APP/PS1-21 mice, which may be related to an induction of ABCC1 in astrocytes as a protective mechanism against oxidative stress.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Neuroimagen/métodos , Tomografía de Emisión de Positrones/métodos , Precursor de Proteína beta-Amiloide/genética , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Presenilina-1/genética , Radiofármacos
14.
J Nucl Med ; 61(7): 1050-1057, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31806767

RESUMEN

P-glycoprotein (ABC subfamily B member 1, ABCB1) plays an important role at the blood-brain barrier (BBB) in promoting clearance of neurotoxic ß-amyloid (Aß) peptides from the brain into the blood. ABCB1 expression and activity were found to be decreased in the brains of Alzheimer disease patients. Treatment with drugs that induce cerebral ABCB1 activity may be a promising approach to delay the build-up of Aß deposits in the brain by enhancing clearance of Aß peptides from the brain. The aim of this study was to investigate whether PET with the weak ABCB1 substrate radiotracer 11C-metoclopramide can measure ABCB1 induction at the BBB in a ß-amyloidosis mouse model (APP/PS1-21 mice) and in wild-type mice. Methods: Groups of wild-type and APP/PS1-21 mice aged 50 or 170 d underwent 11C-metoclopramide baseline PET scans or scans after intraperitoneal treatment with the rodent pregnane X receptor activator 5-pregnen-3ß-ol-20-one-16α-carbonitrile (PCN, 25 mg/kg) or its vehicle over 7 d. At the end of the PET scans, brains were harvested for immunohistochemical analysis of ABCB1 and Aß levels. In separate groups of mice, radiolabeled metabolites of 11C-metoclopramide were determined in plasma and brain at 15 min after radiotracer injection. As an outcome parameter of cerebral ABCB1 activity, the elimination slope of radioactivity washout from the brain (kE,brain) was calculated. Results: PCN treatment resulted in an increased clearance of radioactivity from the brain as reflected by significant increases in kE,brain (from +26% to +54% relative to baseline). Immunohistochemical analysis confirmed ABCB1 induction in the brains of PCN-treated APP/PS1-21 mice with a concomitant decrease in Aß levels. There was a significant positive correlation between kE,brain and ABCB1 levels in the brain. In wild-type mice, a significant age-related decrease in kE,brain was found. Metabolite analysis showed that most radioactivity in the brain comprised unmetabolized 11C-metoclopramide in all animal groups. Conclusion:11C-metoclopramide can measure ABCB1 induction in the mouse brain without the need to consider an arterial input function and may find potential application in Alzheimer disease patients to noninvasively evaluate strategies to enhance the clearance properties of the BBB.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Aciltransferasas , Amiloidosis/diagnóstico por imagen , Amiloidosis/metabolismo , Barrera Hematoencefálica/metabolismo , Radioisótopos de Carbono , Metoclopramida , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Modelos Animales de Enfermedad , Femenino , Ratones
15.
Mol Pharm ; 16(3): 1282-1293, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30694684

RESUMEN

P-Glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) are two efflux transporters at the blood-brain barrier (BBB), which effectively restrict brain distribution of diverse drugs, such as tyrosine kinase inhibitors. There is a crucial need for pharmacological ABCB1 and ABCG2 inhibition protocols for a more effective treatment of brain diseases. In the present study, seven marketed drugs (osimertinib, erlotinib, nilotinib, imatinib, lapatinib, pazopanib, and cyclosporine A) and one nonmarketed drug (tariquidar), with known in vitro ABCB1/ABCG2 inhibitory properties, were screened for their inhibitory potency at the BBB in vivo. Positron emission tomography (PET) using the model ABCB1/ABCG2 substrate [11C]erlotinib was performed in mice. Tested inhibitors were administered as i.v. bolus injections at 30 min before the start of the PET scan, followed by a continuous i.v. infusion for the duration of the PET scan. Five of the tested drugs increased total distribution volume of [11C]erlotinib in the brain ( VT,brain) compared to vehicle-treated animals (tariquidar, + 69%; erlotinib, + 19% and +23% for the 21.5 mg/kg and the 43 mg/kg dose, respectively; imatinib, + 22%; lapatinib, + 25%; and cyclosporine A, + 49%). For all drugs, increases in [11C]erlotinib brain distribution were lower than in Abcb1a/b(-/-)Abcg2(-/-) mice (+149%), which suggested that only partial ABCB1/ABCG2 inhibition was reached at the mouse BBB. The plasma concentrations of the tested drugs at the time of the PET scan were higher than clinically achievable plasma concentrations. Some of the tested drugs led to significant increases in blood radioactivity concentrations measured at the end of the PET scan (erlotinib, + 103% and +113% for the 21.5 mg/kg and the 43 mg/kg dose, respectively; imatinib, + 125%; and cyclosporine A, + 101%), which was most likely caused by decreased hepatobiliary excretion of radioactivity. Taken together, our data suggest that some marketed tyrosine kinase inhibitors may be repurposed to inhibit ABCB1 and ABCG2 at the BBB. From a clinical perspective, moderate increases in brain delivery despite the administration of high i.v. doses as well as peripheral drug-drug interactions due to transporter inhibition in clearance organs question the translatability of this concept.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Clorhidrato de Erlotinib/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Radiofármacos/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Animales , Permeabilidad Capilar/fisiología , Ciclosporina/administración & dosificación , Ciclosporina/sangre , Ciclosporina/metabolismo , Ciclosporina/farmacología , Interacciones Farmacológicas , Clorhidrato de Erlotinib/administración & dosificación , Clorhidrato de Erlotinib/sangre , Clorhidrato de Erlotinib/farmacología , Femenino , Ratones , Modelos Animales , Tomografía de Emisión de Positrones/métodos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/administración & dosificación , Quinolinas/sangre , Quinolinas/metabolismo , Quinolinas/farmacología , Radiofármacos/administración & dosificación , Radiofármacos/sangre , Radiofármacos/farmacología , Solubilidad , Distribución Tisular
16.
Mol Imaging Biol ; 21(2): 306-316, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29942989

RESUMEN

PURPOSE: Multidrug resistance-associated proteins (MRPs) mediate the hepatobiliary and renal excretion of many drugs and drug conjugates. The positron emission tomography (PET) tracer 6-bromo-7-[11C]methylpurine is rapidly converted in tissues by glutathione-S-transferases into its glutathione conjugate, and has been used to measure the activity of Abcc1 in the brain and the lungs of mice. Aim of this work was to investigate if the activity of MRPs in excretory organs can be measured with 6-bromo-7-[11C]methylpurine. PROCEDURES: We performed PET scans with 6-bromo-7-[11C]methylpurine in groups of wild-type, Abcc4(-/-) and Abcc1(-/-) mice, with and without pre-treatment with the prototypical MRP inhibitor MK571. RESULTS: 6-Bromo-7-[11C]methylpurine-derived radioactivity predominantly underwent renal excretion. In blood, MK571 treatment led to a significant increase in the AUC and a decrease in the elimination rate constant of radioactivity (kelimination,blood). In the kidneys, there were significant decreases in the rate constant for radioactivity uptake from the blood (kuptake,kidney), kelimination,kidney, and the rate constant for tubular secretion of radioactivity (kurine). Experiments in Abcc4(-/-) mice indicated that Abcc4 contributed to renal excretion of 6-bromo-7-[11C]methylpurine-derived radioactivity. CONCLUSIONS: Our data suggest that 6-bromo-7-[11C]methylpurine may be useful to assess the activity of MRPs in the kidneys as well as in other organs (brain, lungs), although further work is needed to identify the MRP subtypes involved in the disposition of 6-bromo-7-[11C]methylpurine-derived radioactivity.


Asunto(s)
Radioisótopos de Carbono/química , Sondas Moleculares/química , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Tomografía de Emisión de Positrones , Purinas/química , Animales , Glutatión/metabolismo , Ratones Endogámicos C57BL , Propionatos/farmacocinética , Quinolinas/farmacocinética , Factores de Tiempo , Distribución Tisular
17.
Zootaxa ; 4429(2): 357-371, 2018 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-30313274

RESUMEN

Two new gobiid species, Thorogobius alvheimi sp. nov. and Thorogobius laureatus sp. nov. (Teleostei: Gobiiformes: Gobiidae), are described from the outer continental shelf and upper slope of the Eastern Central Atlantic off Angola, Ghana and São Tomé and Príncipe. The specimens were trawled from depths between 57 and 208 m. The new species are most similar to Thorogobius angolensis. Thorogobius alvheimi sp. nov. is distinguished from its congeners in the combination of following characters. Fins: first dorsal fin with the second and third spines distinctly elongated; pectoral fin ray count 20-21; pelvic disc complete and short with well-developed anterior membrane (frenum) and pointed lateral lobes. Scales: nape and predorsal area naked; no scales on opercle; scales in longitudinal series 27-31. Pattern of free neuromasts: supratemporal rows tr and trp developed, extending transversally between pores H and K; longitudinal row g short, not passing row m posteriorly and distinctly distant from row h; infraorbital row 6 long, ventrally extending to lower margin of preopercle, its ventral section 6i originating anterior to its dorsal section 6s; posterior lateral row h reaching anteriorly above posterior third of opercle. Coloration: body uniformly pale fawn and brown; margin of scale pockets dark brown pigmented, yielding a reticulated pattern; pale spots on nape and predorsal area; caudal fin uniformly dusky greyish. Thorogobius laureatus sp. nov. is distinguished from its congeners in the combination of following characters. Fins: first dorsal fin the second and third spines distinctly elongated; pectoral fin ray count 19-22; pelvic disc complete, short, with well-developed anterior membrane (frenum) with pointed lateral lobes. Scales: scales on sides of predorsal area and midline mostly naked; no scales on opercle; scales in longitudinal series 24-27. Pattern of free neuromasts (sensory papillae): supratemporal rows tr and trp developed, extending transversally between pores H and K; longitudinal row g long, passing row m posteriorly and relatively close to row h; infraorbital row 6 long, ventrally extending to lower margin of preopercle, its ventral (6i) and dorsal (6s) originating opposite to each other; posterior lateral rows: row h long, reaching anteriorly above middle of opercle. Coloration: body pale fawn and brownish; margin of scale pockets dark brown pigmented, yielding a reticulated pattern; five brown blotches on flanks in lateral midline from below first dorsal fin to caudal peduncle; caudal fin with six or seven distinct dark vertical bands.


Asunto(s)
Perciformes , Angola , Animales , Peces , Ghana , Agua
18.
Eur J Pharm Sci ; 115: 212-222, 2018 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-29360507

RESUMEN

Ko143 is a reference inhibitor of the adenosine triphosphate-binding cassette (ABC) transporter breast cancer resistance protein (humans: ABCG2, rodents: Abcg2) for in vitro and in vivo use. Previous in vitro data indicate that Ko143 binds specifically to ABCG2/Abcg2, suggesting a potential utility of Ko143 as a positron emission tomography (PET) tracer to assess the density (abundance) of ABCG2 in different tissues. In this work we radiolabeled Ko143 with carbon-11 (11C) and performed small-animal PET experiments with [11C]Ko143 in wild-type, Abcg2(-/-), Abcb1a/b(-/-) and Abcb1a/b(-/-)Abcg2(-/-) mice to assess the influence of Abcg2 and Abcb1a/b on tissue distribution and excretion of [11C]Ko143. [11C]Ko143 was extensively metabolized in vivo and unidentified radiolabeled metabolites were found in all investigated tissues. We detected no significant differences between wild-type and Abcg2(-/-) mice in the distribution of [11C]Ko143-derived radioactivity to Abcg2-expressing organs (brain, liver and kidney). [11C]Ko143 and possibly its radiolabeled metabolites were transported by Abcb1a and not by Abcg2 at the mouse blood-brain barrier. [11C]Ko143-derived radioactivity underwent both hepatobiliary and urinary excretion, with Abcg2 playing a possible role in mediating the transport of radiolabeled metabolites of [11C]Ko143 from the kidney into urine. Experiments in which a pharmacologic dose of unlabeled Ko143 (10 mg/kg) was co-administered with [11C]Ko143 revealed pronounced effects of the vehicle used for Ko143 formulation (containing polyethylene glycol 300 and polysorbate 80) on radioactivity distribution to the brain and the liver, as well as on hepatobiliary and urinary excretion of radioactivity. Our results highlight the challenges associated with the development of PET tracers for ABC transporters and emphasize that inhibitory effects of pharmaceutical excipients on membrane transporters need to be considered when performing in vivo drug-drug interaction studies. Finally, our study illustrates the power of small-animal PET to assess the interaction of drug molecules with membrane transporters on a whole body level.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Neoplasias de la Mama/metabolismo , Dicetopiperazinas/administración & dosificación , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Distribución Tisular/efectos de los fármacos , Animales , Transporte Biológico/efectos de los fármacos , Encéfalo/metabolismo , Radioisótopos de Carbono/administración & dosificación , Interacciones Farmacológicas/fisiología , Femenino , Riñón/metabolismo , Hígado/metabolismo , Ratones , Tomografía de Emisión de Positrones/métodos
19.
Zootaxa ; 4531(1): 95-108, 2018 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-30651452

RESUMEN

Schindleria macrodentata sp. nov., a new species of the paedomorphic gobiid fish genus Schindleria, is described from the Malay Archipelago. The two specimens were collected in 1929 during the "Dana Expedition" in the Molucca Sea between Sulawesi and Halmahera islands (00°29'N, 125°54'E) (Indonesia) and in the Sulu Sea close to Panay Island (11°43'N, 121°43'E) (Philippines). The new species is characterized by a slender body (body depth at pectoral fin base 3.6-3.7 % of SL and at anal fin origin 3.9-4.8 % of SL), a long second dorsal fin (first dorsal fin absent) originating distinctly anterior to the origin of the anal fin (predorsal length 61.8-65.2 % of SL and preanal length 71.7-75.8 % of SL), a short tail (from anus to tip of longest caudal fin ray) (22.1 % of SL), 19-20 dorsal fin rays and 10 anal fin rays, first anal fin ray below 9th dorsal fin ray, few large, widely spaced teeth in the upper and the lower jaws (7 on the premaxilla and 6 on the dentary), an elongated pectoral radial plate (length 3.7-4.5 % of SL) and, in the caudal skeleton, a procurrent ray with an additional spiny process at its base. No distinct urogenital papilla and no pigmentation on body are developed. In this study we present three morphological traits until now not considered as diagnostic characters for Schindleria, the shape of the pectoral radial plate, the shape of the last procurrent caudal spine and the shape of the arch formed by the lower jaw. We also discuss the caudal-fin skeleton of Schindleria, a character developing beyond the larval stages. The record of S. macrodentata is the first of the genus Schindleria for Indonesia and, at more than 100 km distant to the next shore, it is the first offshore record of a Schindler's fish.


Asunto(s)
Peces , Perciformes , Animales , Indonesia , Islas , Filipinas
20.
Nucl Med Biol ; 52: 7-15, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28575795

RESUMEN

INTRODUCTION: [11C]Erlotinib PET has shown promise to distinguish non-small cell lung cancer (NSCLC) tumors harboring the activating epidermal growth factor receptor (EGFR) mutation delE746-A750 from tumors with wild-type EGFR. To assess the suitability of [11C]erlotinib PET to detect the emergence of acquired erlotinib resistance in initially erlotinib-responsive tumors, we performed in vitro binding and PET experiments in mice bearing tumor xenografts using a range of different cancer cells, which were erlotinib-sensitive or exhibited clinically relevant resistance mechanisms to erlotinib. METHODS: The following cell lines were used for in vitro binding and PET experiments: the epidermoid carcinoma cell line A-431 (erlotinib-sensitive, wild-type EGFR) and the three NSCLC cell lines HCC827 (erlotinib-sensitive, delE746-A750), HCC827EPR (erlotinib-resistant, delE746-A750 and T790M) and HCC827ERLO (erlotinib-resistant, delE746-A750 and MET amplification). BALB/c nude mice with subcutaneous tumor xenografts underwent two consecutive [11C]erlotinib PET scans, a baseline scan and a second scan in which unlabeled erlotinib (10mg/kg) was co-injected. Logan graphical analysis was used to estimate total distribution volume (VT) of [11C]erlotinib in tumors. RESULTS: In vitro experiments revealed significantly higher uptake of [11C]erlotinib (5.2-fold) in the three NSCLC cell lines as compared to A-431 cells. In all four cell lines co-incubation with unlabeled erlotinib (1µM) led to significant reductions in [11C]erlotinib uptake (-19% to -66%). In both PET scans and for all four studied cell lines there were no significant differences in tumoral [11C]erlotinib VT values. For all three NSCLC cell lines, but not for the A-431 cell line, tumoral VT was significantly reduced following co-injection of unlabeled erlotinib (-20% to -35%). CONCLUSIONS: We found no significant differences in the in vitro and in vivo binding of [11C]erlotinib between erlotinib-sensitive and erlotinib-resistant NSCLC cells. Our findings suggest that [11C]erlotinib PET will not be suitable to distinguish erlotinib-sensitive NSCLC tumors from tumors with acquired resistance to erlotinib.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Transformación Celular Neoplásica , Resistencia a Antineoplásicos , Clorhidrato de Erlotinib , Neoplasias Pulmonares/diagnóstico por imagen , Tomografía de Emisión de Positrones , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA