Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Clin Pharmacol ; 89(2): 898-902, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36354135

RESUMEN

The inhibition of cytochrome P450 (CYP) enzymes is the most frequent cause of drug-drug interactions. Many safe, inexpensive and widely available therapeutic drugs can inhibit CYP enzymes (e.g., azoles). Also, the specific potency of inhibition and the targeted CYP enzyme have been well described (e.g., itraconazole strongly inhibits CYP enzyme 3A4 and, in turn, CYP3A4 metabolizes venetoclax and ibrutinib). CYP enzyme inhibitors increase the plasma concentration of other drugs via shared metabolic pathways. We herein present the effects of inhibiting CYP enzymes with itraconazole-venetoclax for the treatment of refractory acute myeloid leukaemia, as well as itraconazole-ibrutinib to treat steroid-refractory acute graft vs. host disease in the same patient. Both of the patient's conditions responded completely. This appears to be a feasible strategy that decreases treatment costs by 75%. Previous Food and Drug Administration recommendations and clinical data support these subsequent dose reductions. Eleven months after the transplant, the patient remains in complete response and with no minimal residual disease. Another patient had been effectively treated before with CYP enzyme inhibition prior to venetoclax-itraconazole administration for orbital myeloid sarcoma. Thus, this case study furthers information on the CYP enzyme inhibition strategy when associated with another costly drug, ibrutinib. The CYP enzyme inhibition strategy could be applied to many more anticancer drugs (e.g., ruxolitinib and ponatinib) and facilitate the availability of expensive oncological treatments in low- and middle-income countries. Also, this strategy could be further generalized by using different CYP enzyme inhibitors with varied pharmacokinetic and pharmacodynamic properties (i.e., grapefruit, azoles and clarithromycin).


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450 , Interacciones Farmacológicas , Humanos , Citocromo P-450 CYP3A/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Sistema Enzimático del Citocromo P-450/metabolismo , Itraconazol/farmacología
2.
Pharmacol Res ; 163: 105332, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33271294

RESUMEN

Aldosterone, the main mineralocorticoid hormone, plays a fundamental role in maintaining blood pressure (BP)and volume under hypovolemic conditions. However, in numerous diseases, where it is produced in excess, it plays a detrimental role and contributes to cardiovascular events and ultimately to death in a multitude of patients. The seminal observation that the fungicide-derivative fadrozole blunted steroidogenesis has led to develop several agents to inhibit aldosterone synthase (AS, CYP11B2), the mitochondrial NADH-dependent enzyme that is necessary for aldosterone biosynthesis. Aldosterone synthase inhibitors (ASI) have, thereafter, been conceived and investigated in phase I and phase II studies. We herein reviewed the ASIs available so far considering their chemical structure, the related aldosterone synthase binding and pharmacodynamic properties. We also examined the promising results obtained with ASIs that have already been tested in phase II human studies.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Citocromo P-450 CYP11B2/antagonistas & inhibidores , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Animales , Enfermedades Cardiovasculares/metabolismo , Simulación por Computador , Citocromo P-450 CYP11B2/química , Citocromo P-450 CYP11B2/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/clasificación , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Humanos , Unión Proteica
3.
Cancer Metastasis Rev ; 37(2-3): 409-423, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30066055

RESUMEN

While cytochrome P450 (CYP)-mediated biosynthesis of arachidonic acid (AA) epoxides promotes tumor growth by driving angiogenesis, cancer cell intrinsic functions of CYPs are less understood. CYP-derived AA epoxides, called epoxyeicosatrienoic acids (EETs), also promote the growth of tumor epithelia. In cancer cells, CYP AA epoxygenase enzymes are associated with STAT3 and mTOR signaling, but also localize in mitochondria, where they promote the electron transport chain (ETC). Recently, the diabetes drug metformin was found to inhibit CYP AA epoxygenase activity, allowing the design of more potent biguanides to target tumor growth. Biguanide inhibition of EET synthesis suppresses STAT3 and mTOR pathways, as well as the ETC. Convergence of biguanide activity and eicosanoid biology in cancer has shown a new pathway to attack cancer metabolism and provides hope for improved treatments that target this vulnerability. Inhibition of EET-mediated cancer metabolism and angiogenesis therefore provides a dual approach for targeted cancer therapeutics.


Asunto(s)
Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Autofagia/genética , Ensayos Clínicos como Asunto , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Descubrimiento de Drogas , Interacciones Farmacológicas , Reposicionamiento de Medicamentos , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Microambiente Tumoral
4.
Drug Metab Dispos ; 46(1): 26-32, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29038231

RESUMEN

The 2016 World Health Organization treatment recommendations for drug-resistant tuberculosis (DR-TB) positioned clofazimine as a core second-line drug. Being identified as a cytochrome P450 (P450) inhibitor in vitro, a P450-mediated drug interaction may be likely when clofazimine is coadministered with substrates of these enzymes. The P450-mediated drug interaction potential of clofazimine was evaluated using both static [estimation of the R1 and area under the plasma concentration-time curve ratio (AUCR) values] and dynamic [physiologically based pharmacokinetics (PBPK)] modeling approaches. For static and dynamic predictions, midazolam, repaglinide, and desipramine were used as probe substrates for CYP3A4/5, CYP2C8, and CYP2D6, respectively. The AUCR static model estimations for clofazimine with the substrates midazolam, repaglinide, and desipramine were 5.59, 1.34, and 1.69, respectively. The fold increases in the area under the curve (AUC) predicted for midazolam, repaglinide, and desipramine with clofazimine (based on PBPK modeling) were 2.69, 1.60, and 1.47, respectively. Clofazimine was predicted to be a moderate-to-strong CYP3A4/5 inhibitor and weak CYP2C8 and CYP2D6 inhibitor based on the calculated AUCR by static and PBPK modeling. Additionally, for selected antiretroviral, antitubercular, antihypertensive, antidiabetic, antileprotics, and antihyperlipidemic CYP3A4/5 substrate drugs, approximately 2- to 6-fold increases in the AUC were predicted with static modeling when coadministered with 100 mg of clofazimine. Therefore, the possibility of an increase in the AUC of CYP3A4/5 substrates when coadministered with clofazimine cannot be ignored.


Asunto(s)
Antituberculosos/farmacocinética , Clofazimina/farmacocinética , Sistema Enzimático del Citocromo P-450/metabolismo , Modelos Biológicos , Tuberculosis Resistente a Múltiples Medicamentos/tratamiento farmacológico , Antituberculosos/uso terapéutico , Área Bajo la Curva , Carbamatos/farmacocinética , Clofazimina/uso terapéutico , Inhibidores Enzimáticos del Citocromo P-450/farmacocinética , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Desipramina/farmacocinética , Interacciones Farmacológicas , Humanos , Midazolam/farmacocinética , Piperidinas/farmacocinética
5.
Epilepsia ; 59(7): e103-e108, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29897632

RESUMEN

We prospectively examined the effect of antiepileptic (AED) cotherapy on steady state plasma concentrations of perampanel (PMP) in epileptic patients. We classified AEDs as strong enzyme inducers (carbamazepine, phenobarbital, phenytoin, oxcarbazepine), not strong enzyme inducers/not inhibitors (levetiracetam, lamotrigine, topiramate, rufinamide, lacosamide, zonisamide, clobazam), and enzyme inhibitors (valproic acid [VPA]). The main outcome was the comparison of PMP plasma concentration to weight-adjusted dose ratio (C/D; [µg/mL]/mg kg-1  d-1 ) among comedication subgroups. From 79 patients (42 females, 37 males) aged (mean ± standard deviation) 33 ± 13 years (range = 12-66 years), 114 plasma samples were collected. Twenty-eight patients (44 samples) were cotreated with enzyme inducers (group A), 21 (27 samples) with not strong enzyme inducers/not inhibitors (group B), 21 (31 samples) with not strong enzyme inducers/not inhibitors + VPA (group C), and 9 (12 samples) with enzyme inducers + VPA (group D). PMP C/D was reduced (-56%, P < .001) in group A (1.79 ± 0.80) versus group B (4.05 ± 2.16) and increased (P < .001) in group C (6.72 ± 4.04) compared with groups A (+275%), B (+66%), and D (2.76 ± 2.00, +143%). Our study documents the unpublished higher PMP C/D in patients cotreated with VPA. These findings have both theoretical relevance, suggesting better characterization of PMP metabolic pathways with ad hoc studies, and clinical usefulness in managing patients on AED polytherapy.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia/tratamiento farmacológico , Piridonas/farmacocinética , Piridonas/uso terapéutico , Ácido Valproico/uso terapéutico , Adolescente , Adulto , Anciano , Anticonvulsivantes/efectos adversos , Anticonvulsivantes/clasificación , Niño , Inductores de las Enzimas del Citocromo P-450/efectos adversos , Inductores de las Enzimas del Citocromo P-450/uso terapéutico , Inhibidores Enzimáticos del Citocromo P-450/efectos adversos , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Interacciones Farmacológicas , Quimioterapia Combinada , Epilepsia/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nitrilos , Estudios Prospectivos , Ácido Valproico/efectos adversos , Adulto Joven
6.
Int J Mol Sci ; 19(12)2018 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-30558157

RESUMEN

Cancer still remains one of the leading causes of death worldwide. In spite of significant advances in treatment options and the advent of novel targeted therapies, there still remains an unmet need for the identification of novel pharmacological agents for cancer therapy. This has led to several studies evaluating the possible application of natural agents found in vegetables, fruits, or plant-derived products that may be useful for cancer treatment. Bergamottin is a furanocoumarin derived from grapefruits and is also a well-known cytochrome P450 inhibitor. Recent studies have demonstrated potent anti-oxidative, anti-inflammatory, and anti-cancer properties of grapefruit furanocoumarin both in vitro and in vivo. The present review focuses on the potential anti-neoplastic effects of bergamottin in different tumor models and briefly describes the molecular targets affected by this agent.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Furocumarinas/uso terapéutico , Neoplasias/terapia , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Citrus paradisi/química , Inhibidores Enzimáticos del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Furocumarinas/química , Furocumarinas/farmacología , Humanos , Estructura Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/prevención & control , Estrés Oxidativo/efectos de los fármacos
7.
Xenobiotica ; 47(2): 127-132, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27052193

RESUMEN

1. Diclofenac sodium (DIC) is a widely used anti-inflammatory drug and its administration in humans receiving long-term therapy with herbal drugs containing piperine (PIP) may occur, which leads to drug-phytochemical interactions. The purpose of the present study was to investigate the influence of PIP treatment on the pharmacokinetics of DIC in healthy volunteers. 2. The open-label, two period, sequential study was conducted in 12 healthy volunteers. PIP 20 mg was administered once daily for 10 days during treatment phase. A single dose of DIC 100 mg was administered during control and after treatment phases under fasting conditions. The blood samples were collected after DIC dosing at predetermined time intervals and analyzed by HPLC. 3. Treatment with PIP significantly enhanced maximum plasma concentration (Cmax) (2.24-3.68 µg/mL, p < 0.05), area under the curve (AUC) (7.09-11.81 µg h/mL, p < 0.05), half-life (T1/2) (1.23-1.65 h, p < 0.05) and significantly decreased elimination rate constant (Kel) (0.62-0.41 h-1, p < 0.05), apparent oral clearance (CL/F) (7.57-4.52 L/h, p < 0.05) of DIC as compared to that of control phase. 4. The results suggest that the altered pharmacokinetics of DIC might be attributed to PIP mediated inhibition of CYP2C9 enzyme, which indicates the clinically significant interaction present between DIC and PIP. Therefore, the combination therapy of DIC along with PIP may represent a novel approach to reduce dosage and result in reduced incidence of gastrointestinal side effects seen with DIC alone at higher doses.


Asunto(s)
Alcaloides/uso terapéutico , Benzodioxoles/uso terapéutico , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Diclofenaco/farmacocinética , Piperidinas/uso terapéutico , Alcamidas Poliinsaturadas/uso terapéutico , Adulto , Alcaloides/efectos adversos , Área Bajo la Curva , Benzodioxoles/efectos adversos , Inhibidores Enzimáticos del Citocromo P-450/efectos adversos , Interacciones Farmacológicas , Femenino , Semivida , Voluntarios Sanos , Humanos , Masculino , Piperidinas/efectos adversos , Alcamidas Poliinsaturadas/efectos adversos
8.
Toxicol Mech Methods ; 27(1): 1-17, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27919191

RESUMEN

Polycyclic aromatic hydrocarbons (PAHs) comprise the major class of cancer-causing chemicals and are ranked ninth among the chemical compounds threatening to humans. Moreover, interest in PAHs has been mainly due to their genotoxic, teratogenic, mutagenic and carcinogenic property. Polymorphism in cytochrome P450 (CYP450) and aryl hydrocarbon receptor (AhR) has the capacity to convert procarcinogens into carcinogens, which is an imperative factor contributing to individual susceptibility to cancer development. The carcinogenicity potential of PAHs is related to their ability to bind to DNA, thereby enhances DNA cross-linking, causing a series of disruptive effects which can result in tumor initiation. They induce cellular toxicity by regulating the generation of reactive oxygen species (ROS), which arbitrate apoptosis. Additionally, cellular toxicity-mediated apoptotic and autophagic cell death and immune suppression by industrial pollutants PAH, provide fertile ground for the proliferation of mutated cells, which results in cancer growth and progression. PAHs play a foremost role in angiogenesis necessary for tumor metastasization by promoting the upregulation of metalloproteinase-9 (MMP-9), vascular endothelial growth factor (VEGF) and hypoxia inducible factor (HIF) in human cancer cells. This review sheds light on the molecular mechanisms of PAHs induced cancer development as well as autophagic and apoptotic cell death. Besides that authors have unraveled how phytotherapeutics is an alternate potential therapeutics acting as a savior from the toxic effects of PAHs for safer and cost effective perspectives.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Carcinógenos/toxicidad , Neoplasias/inducido químicamente , Fitoterapia/métodos , Hidrocarburos Policíclicos Aromáticos/toxicidad , Carcinogénesis , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Sistema Enzimático del Citocromo P-450/biosíntesis , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Preparaciones de Plantas/uso terapéutico , Receptores de Hidrocarburo de Aril/metabolismo
9.
Arch Toxicol ; 90(1): 119-36, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26525395

RESUMEN

The incidence, prevalence, and hospitalization rates associated with cardiovascular diseases (CVDs) are projected to increase substantially in the world. Understanding of the biological and pathophysiological mechanisms of survival can help the researchers to develop new management modalities. Numerous experimental studies have demonstrated that mid-chain HETEs are strongly involved in the pathogenesis of the CVDs. Mid-chain HETEs are biologically active eicosanoids that result from the metabolism of arachidonic acid (AA) by both lipoxygenase and CYP1B1 (lipoxygenase-like reaction). Therefore, identifying the localizations and expressions of the lipoxygenase and CYP1B1 and their associated AA metabolites in the cardiovascular system is of major importance in understanding their pathological roles. Generally, the expression of these enzymes is shown to be induced during several CVDs, including hypertension and cardiac hypertrophy. The induction of these enzymes is associated with the generation of mid-chain HETEs and subsequently causation of cardiovascular events. Of interest, inhibiting the formation of mid-chain HETEs has been reported to confer a protection against different cardiac hypertrophy and hypertension models such as angiotensin II, Goldblatt, spontaneously hypertensive rat and deoxycorticosterone acetate (DOCA)-salt-induced models. Although the exact mechanisms of mid-chain HETEs-mediated cardiovascular dysfunction are not fully understood, the present review proposes several mechanisms which include activating G-protein-coupled receptor, protein kinase C, mitogen-activated protein kinases, and nuclear factor kappa B. This review provides a clear understanding of the role of mid-chain HETEs in the pathogenesis of cardiovascular diseases and their importance as novel targets in the treatment for hypertension and cardiac hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Sistema Cardiovascular/metabolismo , Ácidos Hidroxieicosatetraenoicos/metabolismo , Hipertensión/metabolismo , Transducción de Señal , Animales , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/epidemiología , Cardiomegalia/fisiopatología , Fármacos Cardiovasculares/uso terapéutico , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/fisiopatología , Citocromo P-450 CYP1B1/antagonistas & inhibidores , Citocromo P-450 CYP1B1/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Diseño de Fármacos , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/epidemiología , Hipertensión/fisiopatología , Lipooxigenasa/metabolismo , Inhibidores de la Lipooxigenasa/uso terapéutico , Terapia Molecular Dirigida , Transducción de Señal/efectos de los fármacos
10.
J Antimicrob Chemother ; 70(10): 2693-708, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26203180

RESUMEN

The NNRTI efavirenz has long been one of the most frequently employed antiretroviral drugs in the multidrug regimens used to treat HIV infection, in accordance with its well-demonstrated antiretroviral efficacy and favourable pharmacokinetics. However, growing concern about its adverse effects has sometimes led to efavirenz being replaced by other drugs in the initial treatment selection or to switching of therapy to efavirenz-free regimens in experienced patients. Neurological and neuropsychiatric reactions are the manifestations most frequently experienced by efavirenz-treated patients and range from transitory effects, such as nightmares, dizziness, insomnia, nervousness and lack of concentration, to more severe symptoms including depression, suicidal ideation or even psychosis. In addition, efavirenz has recently been associated with mild/moderate neurocognitive impairment, which is of specific relevance given that half of the patients receiving ART eventually suffer some form of HIV-associated neurocognitive disorder. The mechanisms responsible for efavirenz-induced neurotoxicity are unclear, although growing evidence points to disturbances in brain mitochondrial function and bioenergetics. This review offers a comprehensive overview of the current evidence on the interaction that efavirenz displays with the CNS, including the penetration and concentration of the drug in the brain. We discuss the prevalence, types and specificities of its side effects and recently uncovered cellular mechanisms that may be involved in their development.


Asunto(s)
Fármacos Anti-VIH/efectos adversos , Benzoxazinas/efectos adversos , Sistema Nervioso Central/efectos de los fármacos , Inhibidores Enzimáticos del Citocromo P-450/efectos adversos , Inhibidores de la Transcriptasa Inversa/efectos adversos , Alquinos , Animales , Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/uso terapéutico , Terapia Antirretroviral Altamente Activa/efectos adversos , Benzoxazinas/farmacología , Benzoxazinas/uso terapéutico , Enfermedades del Sistema Nervioso Central/etiología , Ciclopropanos , Citocromo P-450 CYP2B6/genética , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Modelos Animales de Enfermedad , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Humanos , Farmacogenética , Polimorfismo de Nucleótido Simple , Inhibidores de la Transcriptasa Inversa/farmacología , Inhibidores de la Transcriptasa Inversa/uso terapéutico
11.
Pharm Res ; 32(6): 1864-83, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25446771

RESUMEN

PURPOSE: To develop a population physiologically-based pharmacokinetic (PBPK) model for simvastatin (SV) and its active metabolite, simvastatin acid (SVA), that allows extrapolation and prediction of their concentration profiles in liver (efficacy) and muscle (toxicity). METHODS: SV/SVA plasma concentrations (34 healthy volunteers) were simultaneously analysed with NONMEM 7.2. The implemented mechanistic model has a complex compartmental structure allowing inter-conversion between SV and SVA in different tissues. Prior information for model parameters was extracted from different sources to construct appropriate prior distributions that support parameter estimation. The model was employed to provide predictions regarding the effects of a range of clinically important conditions on the SV and SVA disposition. RESULTS: The developed model offered a very good description of the available plasma SV/SVA data. It was also able to describe previously observed effects of an OATP1B1 polymorphism (c.521 T > C) and a range of drug-drug interactions (CYP inhibition) on SV/SVA plasma concentrations. The predicted SV/SVA liver and muscle tissue concentrations were in agreement with the clinically observed efficacy and toxicity outcomes of the investigated conditions. CONCLUSIONS: A mechanistically sound SV/SVA population model with clinical applications (e.g., assessment of drug-drug interaction and myopathy risk) was developed, illustrating the advantages of an integrated population PBPK approach.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Modelos Biológicos , Simvastatina/análogos & derivados , Activación Metabólica , Disponibilidad Biológica , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Interacciones Farmacológicas , Voluntarios Sanos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/sangre , Inhibidores de Hidroximetilglutaril-CoA Reductasas/química , Hígado/efectos de los fármacos , Hígado/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/metabolismo , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Farmacogenética , Polimorfismo Genético , Reproducibilidad de los Resultados , Simvastatina/administración & dosificación , Simvastatina/efectos adversos , Simvastatina/sangre , Simvastatina/química , Simvastatina/farmacocinética , Distribución Tisular
12.
Prostate ; 74(15): 1544-50, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25175831

RESUMEN

BACKGROUND: Questions about optimal sequencing of systemic therapy in metastatic castration-resistant prostate cancer (mCRPC) and whether cross-resistance occurs between different drugs remain largely unanswered. Previous studies have produced conflicting data on the activity of docetaxel in patients who did not attain a prostate-specific antigen (PSA) response to abiraterone acetate (abiraterone). We investigated whether the biochemical response to abiraterone is associated with efficacy of subsequent docetaxel therapy. METHODS: mCRPC patients treated with docetaxel after abiraterone were retrospectively identified at three Canadian institutions. Patients who had also received docetaxel prior to abiraterone were termed "docetaxel-experienced," while those not treated with docetaxel prior to abiraterone were termed "docetaxel-naïve." Treatment outcomes on docetaxel were stratified by prior response to abiraterone and compared using χ(2) -square test for confirmed PSA response rate (≥ 50% decline from baseline maintained for ≥ 3 weeks) and the log-rank method for progression-free survival (PFS) and overall survival (OS). RESULTS: Eighty-six patients were treated with abiraterone, of whom 49 were docetaxel-experienced and 37 were docetaxel-naïve. Prior PSA response to abiraterone was no decline, <50% decline and ≥ 50% decline in 37%, 26%, and 37% of patients respectively. The overall PSA response rate to docetaxel was 34.9%, median PFS was 4.0 months and median OS was 11.66 months. Notably, no differences were seen in confirmed PSA response rates (38% vs. 36% vs. 31%, P = 0.86), median PFS (4.04 months vs. 3.94 months vs. 4.24 months, P = 0.43) and median OS (11.86 months vs. 15.38 months vs. 11.00 months, P = 0.56) on docetaxel for patients with no PSA decline, <50% decline and ≥ 50% decline on abiraterone respectively. Importantly, PSA response rates to docetaxel were comparable in the docetaxel-experienced and docetaxel-naïve cohorts and were not linked to prior response to abiraterone in either group. CONCLUSION: Activity of docetaxel was not associated with the biochemical response to prior abiraterone therapy. These data suggest that prior response to abiraterone should not influence decisions on subsequent use of docetaxel in mCRPC.


Asunto(s)
Androstenoles/uso terapéutico , Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Taxoides/uso terapéutico , Anciano , Anciano de 80 o más Años , Androstenos , Canadá , Supervivencia sin Enfermedad , Docetaxel , Humanos , Masculino , Persona de Mediana Edad , Antígeno Prostático Específico/sangre , Estudios Retrospectivos , Análisis de Supervivencia , Resultado del Tratamiento
14.
Curr Drug Metab ; 23(2): 137-149, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35184709

RESUMEN

BACKGROUND AND OBJECTIVE: Concurrent usage of proton pump inhibitors and their effect on survival and medication termination has been found in individuals receiving protein kinase inhibitor chemotherapy. To investigate the drug-drug interaction mechanism between CDK inhibitors and proton pump inhibitors, the in-silico docking approach was designed by applying computer simulation modules to predict the binding and inhibitory potential. METHODS: The interaction potential of proton pump inhibitors and CDK inhibitors was predicted utilising molecular docking techniques that employed Schrödinger algorithms to capture the dynamics of the CYP450 enzyme-inhibitor interaction between proton pump inhibitors and CDK inhibitors. Additionally, the human liver microsomes assay was used to determine the in vitro half-maximal inhibitory concentration (IC50) of proton pump inhibitors and the inactivation of CDK inhibitors via CYP3A4. RESULTS: Proton pump inhibitors alter the conformation of the CYP3A4 and CYP2C19 enzymes and interact with the heme prosthetic group, as determined by docking studies. It may result in the suppression of CDK inhibitors' metabolism via competitive inhibition at the binding site of an enzyme. Omeprazole and rabeprazole both significantly block midazolam's 1'-hydroxylation by CYP3A4 in vitro, with IC50 values of 9.86µM and 9.71µM, respectively. When omeprazole and rabeprazole are co-incubated in human liver microsomes at a 30µM concentration equivalent to the Cmax of omeprazole and rabeprazole, rabeprazole significantly prolongs the metabolic clearance of palbociclib, whereas omeprazole affects the ribociclib CYP3A4-mediated metabolism. CONCLUSION: Using dynamic models, we determined that proton pump inhibitors such as rabeprazole and omeprazole indeed have the potential to cause clinically significant drug-drug interactions with CDK inhibitors in the treatment of estrogen receptor (ER) positive and HER2-positive breast cancer. As a result, it is suggested to use caution when prescribing proton pump inhibitors to these individuals.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas , Neoplasias de la Mama , Inhibidores Enzimáticos del Citocromo P-450 , Inhibidores de la Bomba de Protones , Hidrocarburo de Aril Hidroxilasas/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Simulación por Computador , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP3A , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Femenino , Humanos , Simulación del Acoplamiento Molecular , Omeprazol , Inhibidores de la Bomba de Protones/uso terapéutico , Rabeprazol , Sustancias Reductoras
16.
Clin Pharmacol Ther ; 109(1): 82-86, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32452536

RESUMEN

Agents that modify cytochrome P-450 (CYP) enzyme activity are characterized as strong, moderate, or weak inhibitors or inducers based on the magnitude of their impact on substrate exposure in clinical studies. Criteria for these classifications are simple and semiquantitative. However, assignment of a given agent to a CYP inhibitor or inducer category is often complicated by limitations of the published data, inconsistent study findings, and other factors. CYP inhibitor and inducer categories are commonly used as a basis for differentiating drug interaction management recommendations. For example, product labeling for a CYP substrate may recommend avoidance in combination with strong inhibitors and dose reduction in combination with moderate inhibitors. When such recommendations exist, ambiguity or variability in placement of inhibitors or inducers into categories can introduce potentially harmful variations in clinical drug interaction management. Failure to adequately reflect the drug interaction potential of an agent by under-categorizing it (e.g., calling it weak when data point to moderate effects), for example, may lead clinicians to respond inadequately to real risks, or to ignore potential interactions altogether. Over-categorization may lead to actions such as over-adjustment of substrate doses or unnecessary avoidance of optimal treatments. This review describes the current criteria for assignment of CYP inhibitor and inducer categories, summarizes common circumstances leading to ambiguous or variable CYP inhibitor and inducer categorizations, and proposes an approach to data interpretation and application of current criteria under uncertainty. When applied to > 1,000 CYP reviews, the approach described has identified a clear categorization in almost all cases.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/farmacología , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Sistema Enzimático del Citocromo P-450/metabolismo , Animales , Interacciones Farmacológicas/fisiología , Humanos
17.
AAPS J ; 23(4): 91, 2021 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-34181150

RESUMEN

Medicinal cannabis use has increased exponentially with widespread legalization around the world. Cannabis-based products are being used for numerous health conditions, often in conjunction with prescribed medications. The risk of clinically significant drug-drug interactions (DDIs) increases in this setting of polypharmacy, prompting concern among health care providers. Serious adverse events can result from DDIs, specifically those affecting CYP-mediated drug metabolism. Both cannabidiol (CBD) and Δ9-tetrahydrocannabinol (Δ9-THC), major constituents of cannabis, potently inhibit CYPs. Cannabis-based products contain an array of cannabinoids, many of which have limited data available regarding potential DDIs. This study assessed the inhibitory potential of 12 cannabinoids against CYP-mediated drug metabolism to predict the likelihood of clinically significant DDIs between cannabis-based therapies and conventional medications. Supersomes™ were used to screen the inhibitory potential of cannabinoids in vitro. Twelve cannabinoids were evaluated at the predominant drug-metabolizing isoforms: CYP3A4, CYP2D6, CYP2C9, CYP1A2, CYP2B6, and CYP2C19. The cannabinoids exhibited varied effects and potencies across the CYP isoforms. CYP2C9-mediated metabolism was inhibited by nearly all the cannabinoids with estimated Ki values of 0.2-3.2 µM. Most of the cannabinoids inhibited CYP2C19, whereas CYP2D6, CYP3A4, and CYP2B6 were either not affected or only partially inhibited by the cannabinoids. Effects of the cannabinoids on CYP2D6, CYP1A2, CYP2B6, and CYP3A4 metabolism were limited so in vivo DDIs mediated by these isoforms would not be predicted. CYP2C9-mediated metabolism was inhibited by cannabinoids at clinically relevant concentrations. In vivo DDI studies may be justified for CYP2C9 substrates with a narrow therapeutic index.


Asunto(s)
Cannabinoides/farmacología , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Marihuana Medicinal/química , Cannabinoides/uso terapéutico , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Interacciones Farmacológicas , Pruebas de Enzimas , Humanos , Polifarmacia
18.
Neurochem Int ; 149: 105138, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34284077

RESUMEN

Ischemic stroke (IS) is a rapidly increasing global burden and is associated with severe neurological decline and mortality. There is urgent requirement of the efforts, aimed to identify therapeutic strategies that are effective in clinic to promote significant recovery from IS. Studies have shown that mitochondria mediated neuroprotection can be a competent target against ischemic damage. Therefore, we examined whether mitochondrial impairment is regulated by Piperine (PIP), an alkaloid of Piper Longum, which has neuroprotective activity against ischemic brain injury. In this study, transient middle cerebral artery occlusion (tMCAO) surgery was performed on male Wistar rats for 90 min followed by 22.5 h of reperfusion for mimicking the IS condition. This study consisted of three groups: sham, tMCAO and tMCAO + PIP (10 mg/kg b.wt., p.o/day for 15 days), and studied for behavioral tests, infarct volume, brain pathological changes, mitochondrial dysfunction, inflammation alongwith cell survival status. PIP pre-treatment showed reduction in neurological alterations and infarct volume. In addition, PIP pre-treatment suppressed the mitochondrial dysfunction and might have anti-apoptotic potential by preventing Cytochrome c (Cyt c) release from mitochondria to cytoplasm and caspase 3 activation. It also regulates pro-apoptotic, Bax and anti-apoptotic, Bcl-2 proteins accompanied by glial fibrillary acidic protein (GFAP) positive cells in cortex region. Quantitative Reverse transcription-polymerase chain reaction (qRT-PCR) results also showed that PIP reduced the expression of pro-inflammatory protein, interleukin-1 ß (IL-1ß) and enhanced cell survival by restoring the activity of brain derived neurotrophic factor (BDNF) and its transcription protein, cAMP response element binding protein (CREB). Taken together, PIP reduced the mitochondrial dysfunction, neurological impairment, and enhanced neuronal survival. In conclusion, our findings reinforce PIP as an effective neuroprotective agent and provide important evidence about its role as a potential target to serve as a promising therapy for treatment of IS.


Asunto(s)
Alcaloides/uso terapéutico , Benzodioxoles/uso terapéutico , Isquemia Encefálica/prevención & control , Accidente Cerebrovascular Isquémico/prevención & control , Mitocondrias/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Piperidinas/uso terapéutico , Alcamidas Poliinsaturadas/uso terapéutico , Alcaloides/farmacología , Animales , Benzodioxoles/farmacología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Masculino , Mitocondrias/metabolismo , Neuroprotección/fisiología , Fármacos Neuroprotectores/farmacología , Piperidinas/farmacología , Alcamidas Poliinsaturadas/farmacología , Ratas , Ratas Wistar
19.
Drug Discov Today ; 25(9): 1651-1667, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32763499

RESUMEN

'Multidrug resistance' (MDR) is a difficult challenge for cancer treatment. The combined role of cytochrome P450 enzymes (CYPs) and active efflux transporters (AETs) in cancer cells appears relevant in inducing MDR. Chemotherapeutic drugs can be substrates of both CYPs and AETs and CYP inducers or inhibitors can produce the same effects on AETs. In addition, a small subpopulation of cancer stem-like cells (CSCs) appears to survive conventional chemotherapy, leading to recurrent disease. Natural products appear efficacious against CSCs; their combinational treatments with standard chemotherapy are promising for cancer eradication, in particular when supported by nanotechnologies.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Nanomedicina , Fitoquímicos/uso terapéutico
20.
Curr Drug Metab ; 21(14): 1127-1135, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33292107

RESUMEN

BACKGROUND: In clinical practice, chloroquine and hydroxychloroquine are often co-administered with other drugs in the treatment of malaria, chronic inflammatory diseases, and COVID-19. Therefore, their metabolic properties and the effects on the activity of cytochrome P450 (P450, CYP) enzymes and drug transporters should be considered when developing the most efficient treatments for patients. METHODS: Scientific literature on the interactions of chloroquine and hydroxychloroquine with human P450 enzymes and drug transporters, was searched using PUBMED.Gov (https://pubmed.ncbi.nlm.nih.gov/) and the ADME database (https://life-science.kyushu.fujitsu.com/admedb/). RESULTS: Chloroquine and hydroxychloroquine are metabolized by P450 1A2, 2C8, 2C19, 2D6, and 3A4/5 in vitro and by P450s 2C8 and 3A4/5 in vivo by N-deethylation. Chloroquine effectively inhibited P450 2D6 in vitro; however, in vivo inhibition was not apparent except in individuals with limited P450 2D6 activity. Chloroquine is both an inhibitor and inducer of the transporter MRP1 and is also a substrate of the Mate and MRP1 transport systems. Hydroxychloroquine also inhibited P450 2D6 and the transporter OATP1A2. CONCLUSIONS: Chloroquine caused a statistically significant decrease in P450 2D6 activity in vitro and in vivo, also inhibiting its own metabolism by the enzyme. The inhibition indicates a potential for clinical drug-drug interactions when taken with other drugs that are predominant substrates of the P450 2D6. When chloroquine and hydroxychloroquine are used clinically with other drugs, substrates of P450 2D6 enzyme, attention should be given to substrate-specific metabolism by P450 2D6 alleles present in individuals taking the drugs.


Asunto(s)
Cloroquina/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Hidroxicloroquina/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Animales , COVID-19/metabolismo , Cloroquina/uso terapéutico , Inhibidores Enzimáticos del Citocromo P-450/uso terapéutico , Interacciones Farmacológicas/fisiología , Humanos , Hidroxicloroquina/uso terapéutico , Preparaciones Farmacéuticas/metabolismo , Tratamiento Farmacológico de COVID-19
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA