Current Drug Metabolism - Volume 10, Issue 3, 2009
Volume 10, Issue 3, 2009
-
-
Biological Actions and Metabolism of Currently Used Pharmacological Agents for the Treatment of Congestive Heart Failure
Authors: Harjot K. Saini-Chohan and Grant M. HatchCongestive heart failure (CHF), a complex clinical syndrome with impaired cardiac pump function, occurs as a consequence of mechanical deformities (pressure and volume overload), myocardial abnormalities (neurohormonal disorders, myocarditis, cardiomyopathies, inflammation and loss of cardiomyocytes) and rhythmic defects (conduction disturbances, fibrillation and tachycardia). Several studies have demonstrated that chronic activation of sympathetic and renin-angiotensin systems, alteration in myocardial substrate utilization, increase in intracellular Ca2+ concentration, development of oxidative stress, release of pro-inflammatory cytokines and increased production of endothelin are responsible for the maladaptive cardiac and subcellular remodeling depending upon the type and stage of heart failure. A variety of pharmacological agents have been used to prevent the development and progression of CHF under different experimental and clinical settings. Although these drugs belong to specific classes, depending on their mechanism of action, individual drug biotransformation into different metabolites makes them distinct chemical moieties. Thorough understanding of biological effects of these pharmacological agents and metabolism is necessary to establish the basis for their preeminent use in clinical settings. The purpose of this review is to present a mechanistic understanding for the biological activities of different drugs used to treat CHF and to provide an insight of different metabolites formed after biotransformation of these chemical entities. Since development of CHF is a multifactorial and heterogeneous process, induction of combination regimens and improvement in patient compliance are the major challenges for future drug development.
-
-
-
Foreign Compounds and Intermediary Metabolism: Sulfoxidation Bridges the Divide
Authors: S. C. Mitchell and G. B. SteventonIt is widely appreciated that as a xenobiotic travels through an organism and interacts with the biochemical machinery of a living system, it most probably will undergo a number of metabolic alterations usually leading to a cluster of differing chemical species. Indeed, the modern ‘metabonomic’ approach, where earlier studied drug metabolism profiles have been reassessed, has indicated that there are normally many more previously unrecognised minor metabolites, and when all such biotransformation products are considered, then their total number is legion. It is now being recognised also that the same metabolic alteration of a substrate, especially a xenobiotic substrate, may be catalysed by more than one enzyme and that the previously sacrosanct notion of an enzyme's ‘substrate specificity’ may well be inverted to read a substrate's ‘enzyme preference’. The following brief article attempts to highlight another aspect where our general acceptance of the ‘status quo’ needs to be reconsidered. The conventionally acknowledged division between the collection of enzymes that undertake intermediary metabolism and the group of enzymes responsible for xenobiotic metabolism may be becoming blurred. It may well be a prudent time to reassess the current dichotomous view. Overcoming inertia, with a realignment of ideas or alteration of perception, may permit new concepts to emerge leading to a more profound understanding and hopefully eventual benefits for mankind.
-
-
-
Cutaneous Metabolism in Transdermal Drug Delivery
Authors: Qian Zhang, Jeffrey E. Grice, Guangji Wang and Michael S. RobertsThe skin is the major interface between the body and the environment. The cutaneous metabolic activity has been identified and widely studied in recent years. It is clear that active enzymes in viable skin tissues have a capacity for bio-transforming topically applied compounds, with a consequence of an altered pharmacological effect. Although the extent of cutaneous metabolism is modest compared to major metabolism in liver, it is important to consider the effect of inherent metabolic function on both local and systematic transdermal delivery. In this review, recent literatures concerning in vitro & in vivo models and techniques used in the study of skin metabolic processes were summarized. The potential influence from skin transporters, diseased conditions, and the chemicals used in skin absorption studies on cutaneous metabolic function, was then discussed. We also reviewed the prodrug design strategy and its applications in transdermal drug delivery.
-
-
-
Polymorphic Drug Metabolism in Anaesthesia
Authors: Juan G. Restrepo, Elena Garcia-Martin, Carmen Martinez and Jose A.G. AgundezA substantial part of the interindividual variability in response to drugs and xenobiotics is related to genetically-determined impairment in drug metabolism. Several drug-metabolising enzymes are polymorphic in humans and often polymorphisms are strongly related to altered drug biodisposition and to the risk of developing adverse effects. Drugs used in general anaesthesia undergo polymorphic metabolism. Among these, halothane is metabolized by cytochrome P450 (CYP) 2E1 and, to a lesser extent, by CYP3A4 and CYP2A6. CYP2E1 also plays a key role in the metabolism of isoflurane, sevoflurane, enflurane and desflurane. CYP2B6, CYP3A4 and CYP2C9 play a relevant role in the metabolism of ketamine. The enzymes involved in the metabolism of thiopental and etomidate remains to be elucidated. Propofol is metabolized mainly by glucuronidation by uridine diphosphate- glucuronosyltransferases (UGTs) and by hydroxylation by CYP2B6 and CYP2C enzymes. The enzymes SULT1A1 and NQO1 participate in later steps in propofol metabolism. All the above-mentioned anaesthetic-metabolising enzymes are polymorphic in man. The present review analyzes the importance of enzymes in the metabolism of anaesthetics and common polymorphisms related to the biotransformation of general anaesthetics and it raises hypotheses on genetic and non-genetics factors related to altered response to anaesthetics that require further investigation. Based on functional relevance and allele frequencies, we identify the most promising targets for the clinical use of pharmacogenomic techniques in anaesthesia to prevent altered pharmacokinetics or adverse drug effects.
-
-
-
Cellular Transport and Lipid Interactions of Miltefosine
Authors: Gillian Barratt, Michele Saint-Pierre-Chazalet and Philippe M. LoiseauMiltefosine (hexadecylphosphocholine, HePC) is an alkyl phospholipid which was first developed as an anticancer agent for local treatment of skin metastases. It was later found to have remarkable activity against Leishmania parasites by the oral route and is marketed as Impavido® for this indication. The mechanism of action of HePC involves interaction with lipids and in particular membrane lipids - phospholipids and sterols. Studies of interactions between HePC and these lipids carried out in model systems suggest an affinity of HePC for cholesterol-rich lipid rafts. The uptake of HePC by cancer cells begins by insertion into the plasma membrane which may be followed by internalization. Within the plasma membrane, HePC interferes with the functioning of a number of enzymes involved in phospholipid metabolism, including protein kinase C and the phospholipases A2, C and D, and can also induce apoptosis. Effects on lipid metabolism have also been observed in Leishmania parasites. In these organisms, a proposed mechanism of HePC uptake can be proposed: HePC inserts into the outer leaflet of the plasma membrane as monomers when its concentration is below the critical micellar concentration (CMC) and as both monomers and oligomers when it is above the CMC. Thereafter, a two-subunit aminophospholipid translocase, LdMT-LdRos3, internalizes the drug. Some evidence obtained in the Caco-2 intestinal cell model suggests that a similar process may occur during the oral absorption of HePC. Finally, the use of phospholipid vesicles (liposomes) as carrier systems for HePC, reducing its toxic side-effects, is reviewed.
-
-
-
Potential of Flavonoids as Anti-inflammatory Agents: Modulation of Pro- Inflammatory Gene Expression and Signal Transduction Pathways
Authors: M. J. Tunon, M. V. Garcia-Mediavilla, S. Sanchez-Campos and J. Gonzalez-GallegoFlavonoids are a large class of naturally occurring compounds widely present in fruits, vegetables, and beverages derived from plants. Reports have suggested that these compounds might be useful for the prevention of a number of diseases, partly due to their antiinflammatory properties. It has been demonstrated that flavonoids are able to inhibit expression of isoforms of inducible nitric oxide synthase, ciclooxygenase and lipooxygenase, which are responsible for the production of a great amount of nitric oxide, prostanoids and leukotrienes, as well as other mediators of the inflammatory process such as cytokines, chemokines or adhesion molecules. Modulation of the cascade of molecular events leading to the over-expression of those mediators include inhibition of transcription factors such as nuclear factor kappa B, activator protein 1, signal transducers and activators of transcription, CCAAT/enhancer binding protein and others. Effects on the binding capacity of transcription factors may be regulated through the inhibition of protein kinases involved in signal transduction, such as mitogen activated protein kinases. Although the numerous studies published with in vitro approaches allow identifying molecular mechanisms of flavonoid effects, the limited bioavailability of these molecules makes necessary validation in humans. Whatever the case, the data available make clear the potential utility of dietary flavonoids or new flavonoid-based agents for the possible treatment of inflammatory diseases. The present review summarizes recent research data focusing on the modulation of the expression of different inflammatory mediators by flavonoids and the effects on cell signaling pathways responsible for their anti-inflammatory activity.
-
-
-
Interaction of Macrocyclic Lactones with the Multidrug Transporters: The Bases of the Pharmacokinetics of Lipid-Like Drugs
Authors: A. Lespine, J. Dupuy, M. Alvinerie, C. Comera, T. Nagy, P. Krajcsi and S. OrlowskiLike most drugs, macrocyclic lactone endectocides (MLs) exert their antiparasitic effects within the defined target tissues where parasites are located, and whose drug concentrations correlate with those in the plasma compartment. The process of drug distribution to the active site constitutes the link in the pharmacokinetic/pharmacodynamic relationship. In the past few years it has become evident that transporter proteins play a major role in regulating the distribution, elimination and metabolism of the antiparasitic macrocyclic lactones. The efflux transporter P-glycoprotein (P-gp) has received the most attention with regards to its strong interaction with ivermectin and other MLs. P-gp has been reported to be involved in restricting the absorption of these drugs, in enhancing their intestinal elimination, in the protection against their neurotoxicity and in the ML resistance mechanisms in parasites. This review focuses on the interaction of MLs with P-glycoprotein and with other multidrug resistance transporters. Given the structural and physicochemical diversity of these drugs, they constitute models of interest to study the major molecular determinants for the interaction with transporters. We will discuss the consequences of such interactions on ML pharmacokinetics and the possibility of benefiting from of drug/drug interaction to reverse multidrug resistance in several therapeutic fights such as against parasites and tumors.
-
-
-
Pharmacokinetics and Pharmacodynamics of Endoperoxide Antimalarials
Authors: Anirudh Gautam, Tausif Ahmed, Vijay Batra and Jyoti PaliwalThere are several clinically useful endoperoxides, mainly artemisinin derivatives available in market for the treatment of malaria. These are highly potent drugs, with fastest parasite reduction ratio, broadest parasite stage specificity and effectiveness against all species of plasmodium in human. Endoperoxides are crystalline compounds having poor aqueous solubility. Several theories have been proposed for their mechanism of action, but the understanding is still incomplete. The major limitation of this class of compounds is the short half-life, requiring frequent administration, leading to noncompliance and recrudescence. Therefore, WHO recommends their use in combination with long acting antimalarial drugs (Artemisinin based combination therapy, ACT) to manage drug resistance, recrudescence, and non compliance. Endoperoxide compounds bind selectively to malaria-infected red blood cells and moderately to human plasma proteins. Artemisinin derivatives are converted primarily to the bioactive metabolite dihydroartemisinin after parenteral, oral or rectal administration. The rate of conversion is lowest for artelinic acid and highest for the water-soluble artesunate. Such conversion occurs largely in the liver by CYP enzymes. Oral bioavailability in animals ranges between 19 to 35%. Based on their liphophilicity, they tend to cross the blood-brain barrier, causing neurotoxicity in animal models. Efforts have been made to understand and develop pharmacokineticpharmacodynamic (PK-PD) correlation and identify PK-PD indices of endoperoxides. In the absence of the above, the selection of doses in ACTs has been empirical. There are several reports on clinical pharmacokinetic interactions of endoperoxides and their long acting partner drugs but as on date no clinically significant interaction has been reported. This review is an update on physicochemical, pharmacokinetic and pharmacodynamic properties of the endoperoxide antimalarials.
-
-
-
Methodological Uncertainty in Quantitative Prediction of Human Hepatic Clearance from In Vitro Experimental Systems
Authors: D. Hallifax and J. B. HoustonMechanistic prediction of unbound drug clearance from human hepatic microsomes and hepatocytes correlates with in vivo clearance but is both systematically low (10 - 20 % of in vivo clearance) and highly variable, based on detailed assessments of published studies. Metabolic capacity (Vmax) of commercially available human hepatic microsomes and cryopreserved hepatocytes is log-normally distributed within wide (30 - 150-fold) ranges; Km is also log-normally distributed and effectively independent of Vmax, implying considerable variability in intrinsic clearance. Despite wide overlap, average capacity is 2 - 20-fold (dependent on P450 enzyme) greater in microsomes than hepatocytes, when both are normalised (scaled to whole liver). The in vitro ranges contrast with relatively narrow ranges of clearance among clinical studies. The high in vitro variation probably reflects unresolved phenotypical variability among liver donors and practicalities in processing of human liver into in vitro systems. A significant contribution from the latter is supported by evidence of low reproducibility (several fold) of activity in cryopreserved hepatocytes and microsomes prepared from the same cells, between separate occasions of thawing of cells from the same liver. The large uncertainty which exists in human hepatic in vitro systems appears to dominate the overall uncertainty of in vitro-in vivo extrapolation, including uncertainties within scaling, modelling and drug dependent effects. As such, any notion of quantitative prediction of clearance appears severely challenged.
-
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)
Most Read This Month
