Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Antimicrob Chemother ; 78(11): 2723-2730, 2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37757461

RESUMO

BACKGROUND: The glycopeptide teicoplanin is considered first-line treatment for severe infections caused by Gram-positive bacteria. Individualized treatment of teicoplanin is gaining interest. As only protein-unbound drug is pharmacologically active, a sensitive assay measuring unbound and total teicoplanin is indispensable for pharmacological research and dose optimization. OBJECTIVES: To develop and validate a UPLC-MS/MS method to quantify unbound and total teicoplanin in human serum. METHODS: The developed assay was validated according to the ICH guideline M10 on Bioanalytical Method Validation and study sample analysis. Unbound teicoplanin was obtained by ultrafiltration. The assay was cross-validated with a quantitative microsphere (QMS) immunoassay in a side-by-side comparison using 40 patient samples. RESULTS: With the developed and validated method, all main teicoplanin components (A2-1, A2-2/A2-3, A2-4/A2-5 and A3-1) can be quantified. Total run time was 5.5 min. Concentration range was 2.5-150 mg/L for total and 0.1-25 mg/L for unbound teicoplanin. Precision (coefficient of variation) and accuracy (bias) of total teicoplanin were 5.97% and 107%, respectively, and 7.17% and 108%, respectively, for unbound teicoplanin.Bland-Altman analysis showed total concentrations measured with the UPLC-MS/MS method were equivalent to the results of the QMS immunoassay. A total of 188 samples from 30 patients admitted to the ICU and haematology department were measured; total concentrations ranged between 2.92 and 98.5 mg/L, and unbound concentrations ranged between 0.37 and 30.7 mg/L. CONCLUSIONS: The developed method provided rapid, precise and accurate measurement of unbound and total teicoplanin. The developed method is now routinely applied in pharmacological research and clinical practice.


Assuntos
Espectrometria de Massas em Tandem , Teicoplanina , Humanos , Cromatografia Líquida , Glicopeptídeos
5.
Lung Cancer ; 146: 30-35, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32505078

RESUMO

INTRODUCTION: Pemetrexed is a pharmacotherapeutic cornerstone in the treatment of non-small cell lung cancer. As it is primarily eliminated by renal excretion, adequate renal function is essential to prevent toxic exposure. There is growing evidence for the nephrotoxic potential of pemetrexed, which even becomes a greater issue now combined immuno-chemotherapy prolongs survival. Therefore, the aim of this study was to describe the incidence of nephrotoxicity and related treatment consequences during pemetrexed-based treatment. METHODS: A retrospective cohort study was conducted in the Jeroen Bosch Hospital, Den Bosch, the Netherlands. All patients that received at least 1 cycle of pemetrexed based therapy were included in the dataset. The primary outcome was defined as a ≥25 % reduction in eGFR. Additionally, the treatment consequences of decreased renal function were assessed. Logistic regression was used to identify risk factors for nephrotoxicity during treatment with pemetrexed. RESULTS: Of the 359 patients included in this analysis, 21 % patients had a clinically relevant decline in renal function after treatment and 8.1 % of patients discontinued treatment due to nephrotoxicity. Cumulative dose (≥10 cycles of pemetrexed based therapy) was identified as a risk factor for the primary outcome measure (adjusted OR 5.66 (CI 1.73-18.54)). CONCLUSION: This study shows that patients on pemetrexed-based treatment are at risk of developing renal impairment. Risk significantly increases with prolonged treatment. Renal impairment is expected to become an even greater issue now that pemetrexed-based immuno-chemotherapy results in longer survival and thus longer treatment duration.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Países Baixos/epidemiologia , Pemetrexede/efeitos adversos , Estudos Retrospectivos
6.
Biochem Pharmacol ; 178: 114050, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32446887

RESUMO

Efavirenz (EFV) is used for antiretroviral treatment of HIV infection, and successfully inhibits viral replication and mother-to-child transmission of HIV during pregnancy and childbirth. Unfortunately, the drug induces neuropsychiatric symptoms such as anxiety and depressed mood and potentially affects cognitive performance. EFV acts on, among others, the serotonin transporter and serotonin receptors that are expressed in the developing brain. Yet, how perinatal EFV exposure affects brain cytoarchitecture remains unclear. Here, we exposed pregnant and lactating rats to EFV, and examined in the medial prefrontal cortex (mPFC) of their adult offspring the effects of the maternal EFV exposure on cortical architecture. We observed a significant decrease in the number of cells, mainly mature neurons, in the infra/prelimbic and cingulate cortices of adult offspring. Next, we found an altered cortical cytoarchitecture characterized by a significant reduction in deep- and superficial-layer cells. This was accompanied by a sharp increase in programmed cell death, as we identified a significantly higher number of cleaved Caspase-3-positive cells. Finally, the serotonergic and dopaminergic innervation of the mPFC subdomains was increased. Thus, the perinatal exposure to EFV provoked in the mPFC of adult offspring cell death, significant changes in cytoarchitecture, and disturbances in serotonergic and dopaminergic innervation. Our results are important in the light of EFV treatment of HIV-positive pregnant women, and its effect on brain development and cognitive behavior.


Assuntos
Alcinos/toxicidade , Benzoxazinas/toxicidade , Ciclopropanos/toxicidade , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/patologia , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/patologia , Inibidores da Transcriptase Reversa/toxicidade , Animais , Animais Recém-Nascidos , Fármacos Anti-HIV/toxicidade , Feminino , Masculino , Córtex Pré-Frontal/crescimento & desenvolvimento , Gravidez , Ratos , Ratos Wistar
7.
Lung Cancer ; 130: 156-158, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30885337

RESUMO

OBJECTIVES: Pemetrexed is indicated for non-small cell lung cancer and mesothelioma. Dosing is based on body surface are (BSA), while renal function is the only determinant for exposure and thus toxicity. BSA-based dosing introduces large variability in exposure and may lead to (hemato)toxicity in patients with impaired renal function. Therefore, pemetrexed is contraindicated in renal impairment. The presented cases provide proof-of-concept for pharmacokinetically-guided dosing of pemetrexed in a haemodialysis patient and a patient with mild renal impairment. METHODS: The pharmacokinetic target was an area under the concentration-time curve (AUC) of 123-205 mg·h/L. Using a previously developed population pharmacokinetic model, individual pharmacokinetics were estimated. RESULTS: Both patients had an exposure above target after the initial dose, but a proportional dose reduction resulted in a therapeutic exposure in both patients (185 and 166 mg·h/L, respectively), that was well-tolerated. Interestingly, a threefold increase in systemic clearance of pemetrexed was observed during hemodialysis (from 1.00 L/h to 3.01 L/h), which approximates the population clearance of pemetrexed. CONCLUSION: Altogether, we showed that pharmacokinetically-guided dosing of pemetrexed may be a feasible strategy for patients with lung cancer and renal impairment.


Assuntos
Antineoplásicos/farmacocinética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Nefropatias/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Pemetrexede/farmacocinética , Idoso , Antineoplásicos/uso terapêutico , Área Sob a Curva , Superfície Corporal , Carcinoma Pulmonar de Células não Pequenas/complicações , Cálculos da Dosagem de Medicamento , Estudos de Viabilidade , Feminino , Humanos , Nefropatias/complicações , Neoplasias Pulmonares/complicações , Masculino , Taxa de Depuração Metabólica , Pemetrexede/uso terapêutico , Diálise Renal
8.
J Antimicrob Chemother ; 73(11): 3095-3101, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30137340

RESUMO

Background: Extended dosing intervals for micafungin could overcome the need for hospitalization for antifungal prophylaxis. Objectives: This multicentre, open-label, randomized trial compared the pharmacokinetics of 300 mg of micafungin given twice weekly with 100 mg once daily as antifungal prophylaxis in adult haematology patients at risk of developing invasive fungal disease. Secondary objectives were assessment of adequate exposure with an alternative dosing regimen of micafungin (700 mg once weekly) through Monte Carlo simulations and assessment of safety in this patient population. Patients and methods: Twenty adult patients were randomized to receive either 300 mg of micafungin twice weekly or 100 mg once daily for 8 days. Blood samples were drawn daily and pharmacokinetic curves were determined on days 4/5 and 8. Monte Carlo simulations were performed for both investigated regimens as well as a frequently proposed alternative regimen (700 mg once weekly). Results: The predicted median AUC0-168h (IQR) for a typical patient on the investigated regimens of 100 mg once daily and 300 mg twice weekly and the hypothetical regimen of 700 mg once weekly were 690 (583-829), 596 (485-717) and 704 (585-833) mg·h/L, respectively. Conclusions: We observed comparable exposure with 300 mg of micafungin twice weekly and 100 mg of micafungin once daily. We provide the pharmacokinetic proof for an extended dosing regimen, which now needs to be tested in a clinical trial with hard endpoints.


Assuntos
Antifúngicos/administração & dosagem , Antifúngicos/farmacocinética , Doenças Hematológicas/microbiologia , Infecções Fúngicas Invasivas/prevenção & controle , Micafungina/administração & dosagem , Micafungina/farmacocinética , Adulto , Idoso , Área Sob a Curva , Esquema de Medicação , Feminino , Doenças Hematológicas/complicações , Hematologia , Humanos , Masculino , Pessoa de Meia-Idade , Método de Monte Carlo , Estudos Prospectivos
9.
Br J Clin Pharmacol ; 84(7): 1575-1586, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29574974

RESUMO

AIMS: Everolimus is a drug from the class of mammalian target of rapamycin inhibitors used for both immunosuppressant and oncological indications. We postulate that there is room for improvement of dosing, as the optimal immunosuppressive dose in calcineurin-free regimens is unknown and since the once daily dosing regimen for oncological indications is often associated with treatment-limiting toxicity. METHODS: We developed a mechanistic population pharmacokinetic model for everolimus in cancer and transplant patients and explored alternative dosing regimens. RESULTS: We found that formulation did not influence bioavailability and that use of >20 mg prednisolone daily increased everolimus clearance. In transplant patients, the approved dose of 0.75-1 mg twice daily (BID) results in subtherapeutic trough levels (<6 µg l-1 ) and that a higher starting dose of 2.25-3 mg BID is required. CONCLUSION: For oncological indications, our results encourage the investigation of dosing everolimus 3.75 mg BID in terms of superiority in safety and noninferiority in efficacy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Everolimo/administração & dosagem , Rejeição de Enxerto/prevenção & controle , Imunossupressores/administração & dosagem , Neoplasias/tratamento farmacológico , Administração Oral , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Disponibilidade Biológica , Relação Dose-Resposta a Droga , Esquema de Medicação , Quimioterapia Combinada/efeitos adversos , Quimioterapia Combinada/métodos , Everolimo/efeitos adversos , Everolimo/farmacocinética , Feminino , Rejeição de Enxerto/imunologia , Humanos , Imunossupressores/efeitos adversos , Imunossupressores/farmacocinética , Transplante de Rim/efeitos adversos , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Modelos Biológicos , Neoplasias/imunologia , Prednisolona/administração & dosagem , Prednisolona/farmacocinética , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/imunologia , Tacrolimo/administração & dosagem , Tacrolimo/efeitos adversos , Tacrolimo/farmacocinética , Transplante Homólogo/efeitos adversos , Resultado do Tratamento
10.
BMJ Open ; 7(9): e018148, 2017 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-28963315

RESUMO

INTRODUCTION: Oral corticosteroids are the first-line treatment for idiopathic childhood nephrotic syndrome. Most children experience several relapses, needing repeated courses of corticosteroid therapy. This exposes them to side effects and long-term complications. For most patients, long-term prognosis is for complete resolution of the disease over time and maintenance of normal kidney function. Therefore, it is vital to focus on minimising adverse events of the disease and its therapy. Unfortunately, no randomised controlled trials are available to determine the optimal corticosteroid treatment of an infrequent relapse of nephrotic syndrome. Recent studies show that treatment schedules for the first episode can safely be shortened to 2 months. The hypothesis of the REducing STEroids in Relapsing Nephrotic syndrome (RESTERN) study is that a 4-week reduction of alternate-day steroids after inducing remission is effective and safe, reduces steroid exposure by 35% on average and is therefore preferable. METHODS AND ANALYSIS: The RESTERN study is a nationwide, double-blind, randomised, placebo-controlled, non-inferiority intervention study. Children aged 1-18 years with a relapse of steroid-sensitive nephrotic syndrome are eligible for this study. Study subjects (n=144) will be randomly assigned to either current standard therapy in the Netherlands or a reduced prednisolone schedule. The primary outcome of the RESTERN study is the time to first relapse after the final prednisolone dose. The secondary outcomes are the number or relapses, progression to frequent relapsing or steroid dependent nephrotic syndrome and the cumulative dosage of prednisolone during the study period. ETHICS AND DISSEMINATION: This non-inferiority trial will be performed in accordance with the Declaration of Helsinki and has been approved by the medical ethical committee of Arnhem-Nijmegen and the Dutch Competent Authority (Central Committee on Research Involving Human Subjects, CCMO). After completion of this study, results will be published in national and international peer-reviewed scientific journals. Papers will be published according to CCMO guidelines. The final report will be made available to trial participants. TRIAL REGISTRATION NUMBER: NTR5670, EudraCT no 2016-002430-76.


Assuntos
Síndrome Nefrótica/tratamento farmacológico , Síndrome Nefrótica/fisiopatologia , Prednisolona/administração & dosagem , Prevenção Secundária/métodos , Esteroides/administração & dosagem , Adolescente , Criança , Pré-Escolar , Método Duplo-Cego , Monitoramento de Medicamentos , Feminino , Humanos , Lactente , Masculino , Países Baixos , Recidiva , Projetos de Pesquisa , Resultado do Tratamento
11.
Br J Cancer ; 116(10): 1312-1317, 2017 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-28399110

RESUMO

BACKGROUND: Cabazitaxel is approved for treatment of castration-resistant metastatic prostate cancer. The current dosing strategy of cabazitaxel is based on body surface area (BSA). Body surface area is known as a poor predictor for total systemic exposure to drugs, since it does not take into account variability in activity of metabolising enzymes, necessary for clearance of drugs. As exposure to cabazitaxel is related to treatment response, it is essential to develop a better individualised dosing strategy. METHODS: Ten patients with metastatic castration-resistant prostate cancer, who received cabazitaxel dosed on BSA as a part of routine palliative care, were enrolled in this study. Midazolam was administered as phenotyping probe for cytochrome P450 isoenzyme 3A (CYP3A). The relationship between midazolam and cabazitaxel clearance was investigated using non-linear mixed effects modelling. RESULTS: The clearance of Midazolam highly correlated with cabazitaxel clearance (R=0.74). Midazolam clearance significantly (P<0.004) explained the majority (∼60%) of the inter-individual variability in cabazitaxel clearance in the studied population. CONCLUSIONS: Metabolic phenotyping of CYP3A using midazolam is a promising strategy to individualise cabazitaxel dosing. Before clinical application, a randomised study is warranted.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Carcinoma/tratamento farmacológico , Carcinoma/metabolismo , Citocromo P-450 CYP3A/metabolismo , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/metabolismo , Taxoides/administração & dosagem , Taxoides/farmacocinética , Idoso , Ansiolíticos/farmacocinética , Superfície Corporal , Humanos , Masculino , Midazolam/farmacocinética , Fenótipo
12.
Nucl Med Biol ; 42(5): 465-469, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25662844

RESUMO

INTRODUCTION: (188)Rhenium-HEDP is an effective bone-targeting therapeutic radiopharmaceutical, for treatment of osteoblastic bone metastases. It is known that the presence of carrier (non-radioactive rhenium as ammonium perrhenate) in the reaction mixture during labeling is a prerequisite for adequate bone affinity, but little is known about the optimal carrier concentration. METHODS: We investigated the influence of carrier concentration in the formulation on the radiochemical purity, in-vitro hydroxyapatite affinity and the in-vivo bone accumulation of (188)Rhenium-HEDP in mice. RESULTS: The carrier concentration influenced hydroxyapatite binding in-vitro as well as bone accumulation in-vivo. Variation in hydroxyapatite binding with various carrier concentrations seemed to be mainly driven by variation in radiochemical purity. The in-vivo bone accumulation appeared to be more complex: satisfactory radiochemical purity and hydroxyapatite affinity did not necessarily predict acceptable bio-distribution of (188)Rhenium-HEDP. CONCLUSIONS: For development of new bisphosphonate-based radiopharmaceuticals for clinical use, human administration should not be performed without previous animal bio-distribution experiments. Furthermore, our clinical formulation of (188)Rhenium-HEDP, containing 10 µmol carrier, showed excellent bone accumulation that was comparable to other bisphosphonate-based radiopharmaceuticals, with no apparent uptake in other organs. ADVANCES IN KNOWLEDGE: Radiochemical purity and in-vitro hydroxyapatite binding are not necessarily predictive of bone accumulation of (188)Rhenium-HEDP in-vivo. IMPLICATIONS FOR PATIENT CARE: The formulation for (188)Rhenium-HEDP as developed by us for clinical use exhibits excellent bone uptake and variation in carrier concentration during preparation of this radiopharmaceutical should be avoided.


Assuntos
Durapatita/química , Ácido Etidrônico/química , Radioquímica/métodos , Radioisótopos/química , Compostos Radiofarmacêuticos/química , Rênio/química , Animais , Osso e Ossos/metabolismo , Durapatita/farmacocinética , Durapatita/uso terapêutico , Ácido Etidrônico/farmacocinética , Ácido Etidrônico/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Compostos Radiofarmacêuticos/farmacocinética , Compostos Radiofarmacêuticos/uso terapêutico , Distribuição Tecidual
13.
Lung Cancer ; 75(3): 391-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22101147

RESUMO

INTRODUCTION: Erlotinib is an agent in the class of oral epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors. Although this class of agents is considered to be relatively safe, the most serious, but rare, adverse reaction is drug-associated interstitial lung disease (ILD). This potentially fatal adverse reaction has been often described with gefitinib, but has been less well described for erlotinib. We here describe a case report of fatal interstitial lung disease in a Caucasian man associated with erlotinib and high erlotinib and metabolite plasma levels and discuss it in the context of all documented cases of erlotinib associated ILD. METHODS: Our case was described and for the literature review a Pubmed and Google Scholar search was conducted for cases of erlotinib associated ILD. The retrieved publications were screened for relevant literature. RESULTS: Besides our case, a total of 19 cases of erlotinib-associated ILD were found. Eleven out 19 cases had a fatal outcome and in only one case erlotinib plasma concentrations were measured and found to be high. CONCLUSION: Erlotinib-associated ILD is a rare, serious and often fatal adverse reaction. Most likely, the cause for erlotinib-associated ILD is multifactorial and high drug levels may be present in patients without serious adverse reactions. However, considering the pharmacology of EGFR inhibitors, high drug and metabolite levels may play a role and future studies are warranted to identify risk factors and to investigate the role of elevated levels of erlotinib and its metabolites in the development of pulmonary toxicity.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Quinazolinas/efeitos adversos , Idoso , Carcinoma Pulmonar de Células não Pequenas/patologia , Cloridrato de Erlotinib , Evolução Fatal , Gefitinibe , Humanos , Doenças Pulmonares Intersticiais/induzido quimicamente , Neoplasias Pulmonares/patologia , Masculino , Infarto do Miocárdio/diagnóstico , Neoplasias Pleurais/tratamento farmacológico , Neoplasias Pleurais/patologia , Inibidores de Proteínas Quinases/efeitos adversos , Quinazolinas/metabolismo
14.
Br J Pharmacol ; 160(5): 1224-33, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20590614

RESUMO

BACKGROUND AND PURPOSE: Lopinavir is extensively metabolized by cytochrome P450 3A (CYP3A) and is considered to be a substrate for the drug transporters ABCB1 (P-glycoprotein) and ABCC2 (MRP2). Here, we have assessed the individual and combined effects of CYP3A, ABCB1 and ABCC2 on the pharmacokinetics of lopinavir and the relative importance of intestinal and hepatic metabolism. We also evaluated whether ritonavir increases lopinavir oral bioavailability by inhibition of CYP3A, ABCB1 and/or ABCC2. EXPERIMENTAL APPROACH: Lopinavir transport was measured in Madin-Darby canine kidney cells expressing ABCB1 or ABCC2. Oral lopinavir kinetics (+/- ritonavir) was studied in mice with genetic deletions of Cyp3a, Abcb1a/b and/or Abcc2, or in transgenic mice expressing human CYP3A4 exclusively in the liver and/or intestine. KEY RESULTS: Lopinavir was transported by ABCB1 but not by ABCC2 in vitro. Lopinavir area under the plasma concentration - time curve (AUC)(oral) was increased in Abcb1a/b(-/-) mice (approximately ninefold vs. wild-type) but not in Abcc2(-/-) mice. Increased lopinavir AUC(oral) (>2000-fold) was observed in cytochrome P450 3A knockout (Cyp3a(-/-)) mice compared with wild-type mice. No difference in AUC(oral) between Cyp3a(-/-) and Cyp3a/Abcb1a/b/Abcc2(-/-) mice was observed. CYP3A4 activity in intestine or liver, separately, reduced lopinavir AUC(oral) (>100-fold), compared with Cyp3a(-/-) mice. Ritonavir markedly increased lopinavir AUC(oral) in all CYP3A-containing mouse strains. CONCLUSIONS AND IMPLICATIONS: CYP3A was the major determinant of lopinavir pharmacokinetics, far more than Abcb1a/b. Both intestinal and hepatic CYP3A activity contributed importantly to low oral bioavailability of lopinavir. Ritonavir increased lopinavir bioavailability primarily by inhibiting CYP3A. Effects of Abcb1a/b were only detectable in the presence of CYP3A, suggesting saturation of Abcb1a/b in the absence of CYP3A activity.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Citocromo P-450 CYP3A/efeitos dos fármacos , Inibidores da Protease de HIV/farmacocinética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Pirimidinonas/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Disponibilidade Biológica , Citocromo P-450 CYP3A/genética , Cães , Interações Medicamentosas , Humanos , Mucosa Intestinal/metabolismo , Intestinos/enzimologia , Fígado/enzimologia , Fígado/metabolismo , Lopinavir , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Ritonavir/farmacologia
15.
Drug Metab Dispos ; 37(9): 1826-40, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19546238

RESUMO

Atazanavir is a commonly prescribed protease inhibitor for treatment of HIV-1 infection. Thus far, only limited data are available on the in vivo metabolism of the drug. Three systemic circulating metabolites have been reported, but their chemical structures have not been released publicly. Atazanavir metabolites may contribute to its effectiveness but also to its toxicity and interactions. Thus, there is a need for extensive metabolic profiling of atazanavir. Our goals were to screen and identify previously unknown atazanavir metabolites and to develop a sensitive metabolite profiling method in plasma. Five atazanavir metabolites were detected and identified in patient samples using liquid chromatography coupled to linear ion trap mass spectrometry: one N-dealkylation product (M1), two metabolites resulting from carbamate hydrolysis (M2 and M3), a hydroxylated product (M4), and a keto-metabolite (M5). For sensitive semiquantitative analysis of the metabolites in plasma, the method was transferred to liquid chromatography coupled to triple quadrupole mass spectrometry. In 12 patient samples, all the metabolites could be detected, and possible other potential atazanavir keto-metabolites were found. Atazanavir metabolite levels were positively correlated with atazanavir levels, but interindividual variability was high. The developed atazanavir metabolic screening method can now be used for further clinical pharmacological research with this antiretroviral agent.


Assuntos
Inibidores da Protease de HIV/metabolismo , Oligopeptídeos/farmacocinética , Piridinas/farmacocinética , Sulfato de Atazanavir , Biotransformação , Cromatografia Líquida de Alta Pressão , Remoção de Radical Alquila , Inibidores da Protease de HIV/análise , Humanos , Hidrólise , Hidroxilação , Indicadores e Reagentes , Oligopeptídeos/análise , Piridinas/análise , Manejo de Espécimes , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
16.
J Pharm Biomed Anal ; 49(2): 451-8, 2009 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-19128911

RESUMO

For the quantification of the HIV-integrase inhibitor raltegravir in human plasma, dried blood spots and peripheral blood mononuclear cell (PBMC) lysate, an assay was developed and validated, using liquid chromatography coupled with tandem mass spectrometry. The assay also allowed detection, but no quantification due to absence of reference substance, of the main metabolite, raltegravir-glucuronide. Raltegravir was extracted from plasma by means of protein precipitation with a mixture of methanol and acetonitrile using only 50microL plasma. Extraction from dried blood spots was performed with a simple one-step extraction with a mixture of methanol, acetonitrile and 0.2M zincsulphate in water (1:1:2, v/v/v) and extraction from cell lysate was performed in 50% methanol in water. Chromatographic separation was performed on a reversed phase C18 column (150mmx2.0mm, particle size 5microm) with a quick stepwise gradient using an acetate buffer (pH 5) and methanol, at a flow rate of 0.25mL/min. The analytical run time was 10min. The triple quadrupole mass spectrometer was operated in the positive ion-mode and multiple reaction monitoring was used for drug quantification. The method was validated over a range of 50-10,000ng/mL in plasma and dried blood spots and a range of 1-500ng/mL in PBMC lysate. Dibenzepine was used as the internal standard. The method was proven to be specific, accurate, precise and robust. Accuracies ranged from 104% to 105% in plasma, from 93% to 105% in dried blood spots and from 82% to 113% in PBMC lysate. Precision over the complete concentration range was less than 6%, 11% and 13% in plasma, dried blood spots and PBMC lysate, respectively. The method is now applied for therapeutic drug monitoring and pharmacological research in HIV-infected patients treated with raltegravir.


Assuntos
Antivirais/sangue , Inibidores de Integrase de HIV/sangue , Leucócitos Mononucleares/química , Pirrolidinonas/sangue , Espectrometria de Massas em Tandem/métodos , Acetonitrilas/química , Antivirais/química , Bioensaio , Calibragem , Cromatografia Líquida de Alta Pressão/instrumentação , Cromatografia Líquida de Alta Pressão/métodos , Dessecação , Monitoramento de Medicamentos , Estabilidade de Medicamentos , Armazenamento de Medicamentos , Congelamento , Infecções por HIV/sangue , Inibidores de Integrase de HIV/química , Humanos , Metanol/química , Estrutura Molecular , Tamanho da Partícula , Pirrolidinonas/química , Raltegravir Potássico , Padrões de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Soluções/química , Água/química , Sulfato de Zinco/química
17.
J Chromatogr B Analyt Technol Biomed Life Sci ; 877(5-6): 575-80, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19168402

RESUMO

For pharmacokinetic monitoring, measurement of antiretroviral agents in plasma is the gold standard. However, human immunodeficiency virus protease inhibitors (PIs) or non-nucleoside reverse transcriptase inhibitors (NNRTIs) exert their action within the infected cell. Cell-associated concentrations may therefore more adequately reflect therapy outcome. Therefore, for the quantification of nine PIs (amprenavir, atazanavir, darunavir, indinavir, lopinavir, nelfinavir, ritonavir, saquinavir and tipranavir), 1 active PI metabolite (nelfinavir M8) and 2 NNRTIs (efavirenz and nevirapine) in lysate of peripheral blood mononuclear cells (PBMCs) an assay was developed and validated, using liquid chromatography coupled with tandem mass spectrometry. Analytes were extracted from a PBMC pellet by means of a one-step extraction with 50% methanol containing the internal standards D6-indinavir, D5-saquinavir, 13C6-efavirenz and dibenzepine. Chromatographic separation was performed on a reversed phase C18 column (150mmx2.0mm, particle size 5microm) with a quick stepwise gradient using an acetate buffer (pH 5) and methanol, at a flow rate of 0.25mL/min. The analytical run time was 10min. The triple quadrupole mass spectrometer was operated in the positive ion-mode and multiple reaction monitoring was used for drug quantification. The method was validated over a range of 1-500ng/mL in PBMC lysate for all analytes. The method was proven to be specific, accurate, precise and robust. The mean precision and accuracy was less than +/-12% at all concentration levels. Using the developed assay and a previously developed assay for these analytes in plasma, the relationship between plasma and intracellular pharmacokinetics and their relationship with therapy outcome can now be determined.


Assuntos
Antivirais/sangue , Extratos Celulares/química , Inibidores da Protease de HIV/sangue , Leucócitos Mononucleares/química , Nucleosídeos/sangue , Inibidores da Transcriptase Reversa/sangue , Espectrometria de Massas em Tandem/métodos , Contagem de Células Sanguíneas , Cromatografia Líquida , Estabilidade de Medicamentos , Humanos , Reprodutibilidade dos Testes
18.
J Pharm Biomed Anal ; 49(2): 393-400, 2009 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-19059744

RESUMO

For the quantification of the novel non-nucleoside reverse transcriptase inhibitor etravirine in human plasma, dried blood spots and peripheral blood mononuclear cell (PBMC) lysate, an assay was developed and validated, using liquid chromatography coupled with tandem mass spectrometry. Etravirine was extracted from plasma by means of protein precipitation with a mixture of methanol and acetonitrile using only 50 microL plasma. Extraction from dried blood spots was performed with a one-step extraction with a mixture of methanol, acetonitrile and 0.2M zinc sulphate in water (1:1:2, v/v/v) and extraction from cell lysate was performed in 50% methanol in water. Chromatographic separation was performed on a reversed phase C18 column (150mmx2.0mm, particle size 5microm) with a quick stepwise gradient using an acetate buffer (pH 5) and methanol, at a flow rate of 0.25mL/min. (13)C(6)-efavirenz was used as an internal standard. The analytical run time was only 10min. The triple quadrupole mass spectrometer was operated in the positive ion-mode and multiple reaction monitoring was used for drug quantification. The method was validated over a range of 25-5000ng/mL in plasma, 50-10,000ng/mL in dried blood spots and a range of 5-2500ng/mL in PBMC lysate. Accuracies ranged from 89% to 106% in plasma, from 94% to 109% in dried blood spots and from 91% to 105% in PBMC lysate. Precisions at the all concentration levels ranged from 1.9% to 14% in plasma, 4.7% to 20% in dried blood spots and from 3.1% to 11% in PBMC lysate. The bioanalytical assay was successfully incorporated with previously developed assays for the determination of all currently approved PIs and NNRTIs in plasma and dried blood spots and it is now applied for therapeutic drug monitoring and pharmacological research in HIV-infected patients treated with etravirine.


Assuntos
Dessecação/métodos , Inibidores da Protease de HIV/sangue , Leucócitos Mononucleares/química , Piridazinas/sangue , Espectrometria de Massas em Tandem/métodos , Acetonitrilas/química , Pressão Atmosférica , Bioensaio , Calibragem , Extratos Celulares/química , Cromatografia Líquida/instrumentação , Cromatografia Líquida/métodos , Monitoramento de Medicamentos/métodos , Estabilidade de Medicamentos , Armazenamento de Medicamentos , Congelamento , Inibidores da Protease de HIV/química , Inibidores da Protease de HIV/farmacocinética , Humanos , Concentração de Íons de Hidrogênio , Metanol/química , Estrutura Molecular , Nitrilas , Tamanho da Partícula , Piridazinas/química , Piridazinas/farmacocinética , Pirimidinas , Padrões de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Temperatura , Água/química , Sulfato de Zinco/química
19.
Bioanalysis ; 1(7): 1329-38, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21083054

RESUMO

Therapeutic drug monitoring is a way to pharmacokinetically guide drug therapy to assure a certain exposure to a drug when this exposure is related to treatment effectiveness or toxicity. Routinely, drug concentrations are measured in plasma obtained by venipuncture. However, venous sampling is difficult in some populations, such as neonates and patients suffering from phlebitis, and there may be logistical challenges, for example when nonhospital-based sampling is warranted (e.g., resource-limited settings). A proper bioanalytical method is crucial for measurements of drug level matrices suitable for patient-friendly drug monitoring. Special attention must be paid to bioanalytical methods in these patient-friendly matrices, since specific matrix-associated issues may have important implications. In this review, we will discuss these issues and give an overview of published bioanalytical methods with a focus on patient-friendly drug monitoring of antiretroviral drugs, where dried blood spots, hair and saliva have been the most important matrices for patient-friendly therapeutic drug monitoring. Furthermore, we will point out considerations for proper assay development and validation.


Assuntos
Antivirais/análise , Bioensaio/métodos , Sangue/metabolismo , Monitoramento de Medicamentos/métodos , Cabelo/metabolismo , Saliva/metabolismo , Antivirais/metabolismo , Cabelo/química , Humanos , Saliva/química
20.
Artigo em Inglês | MEDLINE | ID: mdl-18456582

RESUMO

A bioanalytical method for the determination of most commonly prescribed protease inhibitors (atazanavir, darunavir, lopinavir and ritonavir) and non-nucleoside reverse transcriptase inhibitors (efavirenz and nevirapine) was developed and validated according to FDA guidelines. In brief, dried blood spots were punched out of a collection paper with a 0.25 in. diameter punch. The analytes were extracted from the punched-out disc using a mixture of acetonitrile, methanol and 0.2M zinc sulphate in water (1:1:2, v/v/v) containing the internal standards dibenzepine, 13C6-efavirenz and D5-saquinavir. 20 microL of the extract was injected onto the reversed-phase C18 column (150 mm x 2.0 mm) for separation from endogenous compounds and the analytes were quantified using a triple quadrupole mass spectrometer. The analytical run time was only 10 min. Validated concentration ranges covered the ranges encountered in routine clinical practice. The assay was linear over the concentration ranges tested (0.1-20 mg/L for atazanavir, lopinavir, nevirapine and efavirenz and 0.05-10 mg/L for darunavir and ritonavir). Accuracies and inter- and intra-run precisions at all levels ranged from 96.2 to 113.9% and 3.1 to 13.3%, respectively. Analytes in dried blood spots were stable for at least 7 days at 30 degrees C. The method enabled patient-friendly sample collection, easy and cheap sample shipment and non-hospital based sampling for therapeutic drug monitoring and pharmacokinetic studies.


Assuntos
Cromatografia Líquida/métodos , Inibidores da Protease de HIV/sangue , Espectrometria de Massas/métodos , Inibidores da Transcriptase Reversa/sangue , Inibidores da Protease de HIV/farmacocinética , Humanos , Padrões de Referência , Reprodutibilidade dos Testes , Inibidores da Transcriptase Reversa/farmacocinética , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...