- Home
- A-Z Publications
- Current Drug Metabolism
- Previous Issues
- Volume 11, Issue 8, 2010
Current Drug Metabolism - Volume 11, Issue 8, 2010
Volume 11, Issue 8, 2010
-
-
Disposition of Drugs of Abuse and Their Metabolites in Wastewater as a Method of the Estimation of Drug Consumption
Authors: Emilia Daghir and Michal J. MarkuszewskiThe occurrence of drugs and trace amounts of their metabolites in the aqueous environment has become a global problem. Nowadays, general information about drug use patterns results from indirect methods such as anonymous surveys and police crime statistics. Unfortunately, these sources of information determine drug use consumption indirectly and give only rough estimations about drug use trends. Therefore, in order to assess the real extent of this phenomenon, new objective tools are needed to monitor drug abuse on a large social and international scale. Several analytical methods have been developed to diagnose illicit drug consumption. GC-MS and HPLC-MS are the techniques of choice for the quantitative analysis of illicit drugs and their metabolites in clinical and forensic toxicology. These separation methods have been widely used for the determination of the occurrence of stimulatory drugs in different biological matrixes such as blood, urine, sweat, saliva and hair. Recently, a new direct and objective approach of monitoring drug use patterns has been proposed to estimate illicit drug consumption involving the measurement of urinary breakdown products in waste- and surface water. The approach proposed seems to be suitable for monitoring consumption in real time so that it is possible to identify trends in drug use patterns. The measurement of illicit drug residue in wastewater and surface water might become a standardized tool for the comprehensive assessment of drug abuse in populations.
-
-
-
Human Carbonyl Reductases
Authors: Petra Malatkova, Edmund Maser and Vladimir WsolEnzymatic carbonyl reduction means the formation of a hydroxy function out of a ketone or aldehyde moiety and applies for the metabolism of physiological (endogenous) or xenobiotic (exogenous) molecules. As for endogenous substrates, carbonyl reduction is often part of a reversible oxidoreductase process and involves the activation or inactivation of important signal molecules like steroids, prostaglandins, retinoids and biogenic amines. These reactions are carried out by NAD(P)(H)-dependent dehydrogenases belonging to two protein superfamilies, the aldo-keto reductases (AKR) and the short-chain dehydrogenases/reductases (SDR). With regard to exogenous substrates, carbonyl reduction of xenobiotics is generally a “one-way” detoxification reaction, since the resulting alcohol is easier to conjugate and to eliminate. Interestingly, the participating enzymes do also belong to the AKR and SDR superfamilies. Moreover, some enzymes from the two protein superfamilies exhibit pluripotency in that they are able to catalyze the oxidoreduction of endobiotics but do also function in the reductive metabolism of carbonyl group bearing xenobiotics. A special case are carbonyl reductases per se which belong to the SDR superfamily and whose substrates or physiological roles are not quite clear. Usually, carbonyl reductases have a broad and diverse substrate spectrum for xenobiotics, however, for some of them a specific physiological function has been speculated. In the human genome, three SDR genes have been identified to code for the carbonyl reductases CBR1 (SDR21C1), CBR3 (SDR21C2) and CBR4 (SDR45C1). The present review summarizes the current knowledge on these enzymes with special emphasis on their role as a defence system against toxicants, as well as their possible physiological function and medical application. In detail, we have screened the recent literature on these three enzymes with regard to endogenous and exogenous substrates, their three-dimensional structure, tissues specific expression, polymorphisms, transcriptional regulation, occurrence in pathological states, and their possible association with cancer. Combined, this review contributes to understanding the complex nature and biological role(s) of the human carbonyl reductases CBR1, CBR3 and CBR4.
-
-
-
Psychedelic 5-Methoxy-N,N-Dimethyltryptamine: Metabolism, Pharmacokinetics, Drug Interactions, and Pharmacological Actions
Authors: Hong-Wu Shen, Xi-Ling Jiang, Jerrold C. Winter and Ai-Ming Yu5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT) belongs to a group of naturally-occurring psychoactive indolealkylamine drugs. It acts as a nonselective serotonin (5-HT) agonist and causes many physiological and behavioral changes. 5-MeO-DMT is O-demethylated by polymorphic cytochrome P450 2D6 (CYP2D6) to an active metabolite, bufotenine, while it is mainly inactivated through the deamination pathway mediated by monoamine oxidase A (MAO-A). 5-MeO-DMT is often used with MAO-A inhibitors such as harmaline. Concurrent use of harmaline reduces 5-MeO-DMT deamination metabolism and leads to a prolonged and increased exposure to the parent drug 5-MeO-DMT, as well as the active metabolite bufotenine. Harmaline, 5-MeO-DMT and bufotenine act agonistically on serotonergic systems and may result in hyperserotonergic effects or serotonin toxicity. Interestingly, CYP2D6 also has important contribution to harmaline metabolism, and CYP2D6 genetic polymorphism may cause considerable variability in the metabolism, pharmacokinetics and dynamics of harmaline and its interaction with 5-MeO-DMT. Therefore, this review summarizes recent findings on biotransformation, pharmacokinetics, and pharmacological actions of 5-MeO-DMT. In addition, the pharmacokinetic and pharmacodynamic drug-drug interactions between harmaline and 5-MeO-DMT, potential involvement of CYP2D6 pharmacogenetics, and risks of 5- MeO-DMT intoxication are discussed.
-
-
-
Impact of Genetic Polymorphisms and Drug-Drug Interactions on Clopidogrel and Prasugrel Response Variability
Authors: V. Ancrenaz, Y. Daali, P. Fontana, M. Besson, C. Samer, P. Dayer and J. DesmeulesThienopyridine antiaggregating platelet agents (clopidogrel and prasugrel) act as irreversible P2Y12 receptor inhibitors. They are used with aspirin to prevent thrombotic complications after an acute coronary syndrome or percutaneous coronary intervention. A large interindividual variability in response to clopidogrel and to a lesser extent to prasugrel is observed and may be related to their metabolism. Clopidogrel and prasugrel are indeed prodrugs converted into their respective active metabolites by several cytochromes P450 (CYPs). Besides clopidogrel inactivation (85%) by esterases to the carboxylic acid, clopidogrel is metabolized by CYPs to 2-oxoclopidogrel (15%) and further metabolized to an unstable but potent platelet-aggregating inhibitor. Prasugrel is more potent than clopidogrel with a better bioavailability and lower pharmacodynamic variability. Prasugrel is completely converted by esterases to an intermediate oxo-metabolite (R-95913) further bioactivated by CYPs. Numerous clinical studies have shown the influence of CYP2C19 polymorphism on clopidogrel antiplatelet activity. Moreover, unwanted drug-drug pharmacokinetic interactions influencing CYP2C19 activity and clopidogrel bioactivation such as with proton pump inhibitors remain a matter of intense controversy. Several studies have also demonstrated that CYP3A4/5 and CYP1A2 are important in clopidogrel bioactivation and should also be considered as potential targets for unwanted drug-drug interactions. Prasugrel bioactivation is mainly related to CYP3A4 and 2B6 activity and therefore the question of the effect of drug-drug interaction on its activity is open. The purpose of this review is to critically examine the current literature evaluating the influence of genetic and environmental factors such as unwanted drug-drug interaction affecting clopidogrel and prasugrel antiplatelet activity.
-
-
-
Methodologies for Investigating Drug Metabolism at the Early Drug Discovery Stage: Prediction of Hepatic Drug Clearance and P450 Contribution
Authors: Chie Emoto, Norie Murayama, Amin Rostami-Hodjegan and Hiroshi YamazakiThe attrition rate in drug development is being reduced by continuous advances in science and technology introduced by various academic institutions and pharmaceutical companies. This has been certainly noticeable in reducing the frequency with which unfavorable absorption, distribution, metabolism, and elimination (ADME) characteristics of any candidate drug causes failure in clinical development. Nonetheless, it is important that the objectives in reducing attrition during later stages of development are matched by information generated in the earliest stage of discovery. In this review, we summarize the methodologies employed during the early stages of drug discovery and discuss new findings in the areas of (1) drug metabolism enzymes, (2) the contribution of cytochrome P450 enzymes (P450, CYP) to hepatic metabolism, (3) prediction of hepatic intrinsic clearance, (4) reaction phenotyping, and (5) the metabolic differences between highly homologous enzymes such as CYP3A4 and CYP3A5. The total contribution of P450 and UDPglucuronosyltransferases to drug metabolism is reported to be more than 80%; therefore, glucuronidation is increasingly recognized as an important clearance pathway in addition to that of P450 enzymes. When estimating the contribution of P450, interpreting the results of inhibition studies using a single P450 inhibitor can lead to false conclusions. For instance, 1-aminobenzotriazole and SKF-525A have a varying range of IC50 values for inhibition of drug oxidation reactions by different CYP450 enzymes. There are disparities between methodologies at early stage drug discovery and late stage development. For example, although the drug depletion approach for the prediction of hepatic intrinsic clearance may not be desirable at late stages of development, it is suitable at the early drug discovery stage since kinetic characterization and measurement of specific drug metabolites are not required. Data from protein binding assays in plasma and/or liver microsomes is an integral part to predicting hepatic clearance; therefore, the prediction methods for protein binding have been addressed in terms of automation and in silico prediction. The approach to reaction phenotyping using recombinant P450 microsome data are reviewed as this approach enables combining the drug depletion method with appropriate scaling factors to predict clearance values. CYP3A enzymes have broad substrate specificities and are responsible for the oxidative metabolism of more than 50% of clinically used drugs. Although CYP3A4 is the most abundant CYP3A isoform in adult human liver, CYP3A5 may contribute more to CYP3A-mediated drug oxidation by human liver microsomes than CYP3A4 does, especially in Japanese subjects, who typically have a relatively high frequency of genetic CYP3A5 expression. Lack of efficacy and presence of serious side effects in some sub-group of patients remain the biggest sources of drug failure at late stage of drug development. Advances in appreciation of inter-individual variabilities in ADME, by creation of virtual individuals and use of appropriate information from early discovery may lead to a better anticipation of variable clinical and toxicological outcome following administration of any new drug candidate. Thus may also help with dosing strategies which minimize the potential side effects and maximize the clinical benefits. Accordingly, front-loading of efforts for characterizing the candidate drugs at early stages of discovery is recommended.
-
-
-
The Role of Metabonomics at the Interface Between Drug Metabolism and Safety Assessment
More LessSafety assessment of candidate drugs is a key stage in drug development and one which represents a significant attritional hurdle. There is also a focused effort in drug metabolism studies to assess bioactivation potential based on the notion this could lead to the risk of macromolecule adduct formation and subsequent toxicological consequences. However, characterization of the molecular mechanisms of drug toxicity still remains an enormous challenge. Recent advancements in ‘omics sciences, and in particular metabonomics, has enabled some elucidation or insights into toxicological sequelae. Metabonomics is a global metabolic profiling framework which utilizes high resolution analytics (typically NMR and mass spectrometry) together with chemometric statistical tools (such as principal component analysis and partial least squares) to derive an integrated picture of both endogenous and xenobiotic metabolism. This review details some of the current progress in the application of metabonomics in drug safety and metabolism.
-
-
-
Impact of Sex Hormone Metabolism on the Vascular Effects of Menopausal Hormone Therapy in Cardiovascular Disease
Authors: Durr-e-Nayab Masood, Emir C. Roach, Katie G. Beauregard and Raouf A. KhalilEpidemiological studies have shown that cardiovascular disease (CVD) is less common in pre-menopausal women (Pre-MW) compared to men of the same age or post-menopausal women (Post-MW), suggesting cardiovascular benefits of estrogen. Estrogen receptors (ERs) have been identified in the vasculature, and experimental studies have demonstrated vasodilator effects of estrogen/ER on the endothelium, vascular smooth muscle (VSM) and extracellular matrix. Several natural and synthetic estrogenic preparations have been developed for relief of menopausal vasomotor symptoms. However, whether menopausal hormone therapy (MHT) is beneficial in postmenopausal CVD remains controversial. Despite reports of vascular benefits of MHT from observational and experimental studies, randomized clinical trials (RCTs), such as the Heart and Estrogen/progestin Replacement Study (HERS) and the Women's Health Initiative (WHI), have suggested that, contrary to expectations, MHT may increase the risk of CVD. These discrepancies could be due to agerelated changes in sex hormone synthesis and metabolism, which would influence the effective dose of MHT and the sex hormone environment in Post-MW. Age-related changes in the vascular ER subtype, structure, expression, distribution, and post-ER signaling pathways in the endothelium and VSM, along with factors related to the design of RCTs, preexisting CVD condition, and structural changes in the blood vessels architecture have also been suggested as possible causes of MHT failure in CVD. Careful examination of these factors should help in identifying the causes of the changes in the vascular effects of estrogen with age. The sex hormone metabolic pathways, the active versus inactive estrogen metabolites, and their effects on vascular function, the mitochondria, the inflammatory process and angiogenesis should be further examined. Also, the genomic and non-genomic effects of estrogenic compounds should be viewed as integrated rather than discrete responses. The complex interactions between these factors highlight the importance of careful design of MHT RCTs, and the need of a more customized approach for each individual patient in order to enhance the vascular benefits of MHT in postmenopausal CVD.
-
Volumes & issues
-
Volume 25 (2024)
-
Volume 24 (2023)
-
Volume 23 (2022)
-
Volume 22 (2021)
-
Volume 21 (2020)
-
Volume 20 (2019)
-
Volume 19 (2018)
-
Volume 18 (2017)
-
Volume 17 (2016)
-
Volume 16 (2015)
-
Volume 15 (2014)
-
Volume 14 (2013)
-
Volume 13 (2012)
-
Volume 12 (2011)
-
Volume 11 (2010)
-
Volume 10 (2009)
-
Volume 9 (2008)
-
Volume 8 (2007)
-
Volume 7 (2006)
-
Volume 6 (2005)
-
Volume 5 (2004)
-
Volume 4 (2003)
-
Volume 3 (2002)
-
Volume 2 (2001)
-
Volume 1 (2000)