Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cytokine ; 88: 136-143, 2016 12.
Article in English | MEDLINE | ID: mdl-27619517

ABSTRACT

Thymic epithelial cells (TECs) play a key role in the regulation of central immune tolerance by expressing autoantigens and eliminating self-reactive T cells. In a previous paper we reported that adrenomedullin (ADM) and its co-receptor protein RAMP2 are located intracellularly in newborn human thymic epithelial cells (TECs). This work has two main aims: (1) to examine the cellular localization of ADM and its receptor in TECs of adult Wistar rats to validate this animal model for the study of the ADM system and its function(s) in thymus; (2) to investigate the potential modulating effect of ADM on the NF-kB pathway, which is involved through the production of cytokines such as IL-6, in the maturation of T-lymphocytes and immunological tolerance. Our results show that, similarly to human newborn TECs, ADM is localized to the cytoplasm of adult rat TECs, and RAMP2 is expressed in the nucleus but not in the plasma membrane. Pretreatment of TECs for 4h with ADM significantly reduced lipopolysaccharide (LPS)-induced release of IL-6 (P<0.001) and expression of the p65 subunit of NF-kB, while doubled the expression of IkBα (P<0.001), the physiological inhibitor of NF-kB nuclear translocation. These effects were not mediated by activation of the cAMP pathway, a signalling cascade that is rapidly activated by ADM in cells that express plasma membrane RAMP2, but were the consequence of a reduction in the transcription of p65 (P<0.001) and an increase in the transcription of IkBα (P<0.05). On the basis of these findings we propose that in rat TECs ADM reduces IL-6 secretion by modulating NF-kB genes transcription through an interaction with a receptor localized to the nucleus. This may partly explain the protective effects of ADM in autoimmune diseases and points to the ADM system of TECs as a novel potential target for immunomodulating drugs.


Subject(s)
Adrenomedullin/metabolism , Cyclic AMP/metabolism , Epithelial Cells/metabolism , Interleukin-6/metabolism , NF-kappa B/metabolism , Second Messenger Systems , Thymus Gland/metabolism , Animals , Lipopolysaccharides/toxicity , Male , Rats , Rats, Wistar , Receptor Activity-Modifying Protein 2/metabolism , Th1 Cells/metabolism , Th17 Cells/metabolism
2.
World J Gastroenterol ; 22(3): 1260-78, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26811663

ABSTRACT

Inhibition and induction of drug-metabolizing enzymes are the most frequent and dangerous drug-drug interactions. They are an important cause of serious adverse events that have often resulted in early termination of drug development or withdrawal of drugs from the market. Management of such interactions by dose adjustment in clinical practice is extremely difficult because of the wide interindividual variability in their magnitude. This review examines the genetic, physiological, and environmental factors responsible for this variability, focusing on an important but so far neglected cause of variability, liver functional status. Clinical studies have shown that liver disease causes a reduction in the magnitude of interactions due to enzyme inhibition, which is proportional to the degree of liver function impairment. The effect of liver dysfunction varies quantitatively according to the nature, reversible or irreversible, of the inhibitory interaction. The magnitude of reversible inhibition is more drastically reduced and virtually vanishes in patients with advanced hepatocellular insufficiency. Two mechanisms, in order of importance, are responsible for this reduction: decreased hepatic uptake of the inhibitory drug and reduced enzyme expression. The extent of irreversible inhibitory interactions is only partially reduced, as it is only influenced by the decreased expression of the inhibited enzyme. Thus, for appropriate clinical management of inhibitory drug interactions, both the liver functional status and the mechanism of inhibition must be taken into consideration. Although the inducibility of drug-metabolizing enzymes in liver disease has long been studied, very conflicting results have been obtained, mainly because of methodological differences. Taken together, the results of early animal and human studies indicated that enzyme induction is substantially preserved in compensated liver cirrhosis, whereas no definitive conclusion as to whether it is significantly reduced in the decompensated state of cirrhosis was provided. Since ethical constraints virtually preclude the possibility of performing methodologically rigorous investigations in patients with severe liver dysfunction, studies have recently been performed in animals rigorously stratified according to the severity of liver insufficiency. The results of these studies confirmed that enzyme induction is virtually unaffected in compensated cirrhosis and indicated that the susceptibility of enzyme induction to severe liver dysfunction depends on the type of nuclear receptor involved and also varies among enzyme isoforms under the transcriptional control of the same nuclear receptor. These findings make it clear that no general conclusion can be reached from the study of any particular enzyme and partly explain the conflicting results obtained by previous studies. Since no general guidelines can be provided for the management of drug interactions resulting from enzyme induction, both the effects and the plasma concentration of the induced drug should be strictly monitored. The findings discussed in this review have important methodological implications as they indicate that, contrary to current guidelines, the magnitude of metabolic drug-drug interactions in patients with liver disease cannot be inferred from studies in healthy subjects.


Subject(s)
Drug Interactions , Drug-Related Side Effects and Adverse Reactions/etiology , Enzyme Inhibitors/adverse effects , Liver Diseases/metabolism , Liver/enzymology , Membrane Transport Proteins/metabolism , Biological Transport , Biotransformation/genetics , Cytochrome P-450 Enzyme Inducers/adverse effects , Cytochrome P-450 Enzyme Inhibitors/adverse effects , Drug Dosage Calculations , Drug-Related Side Effects and Adverse Reactions/enzymology , Drug-Related Side Effects and Adverse Reactions/genetics , Enzyme Induction , Enzyme Inhibitors/administration & dosage , Humans , Liver Diseases/complications , Liver Diseases/diagnosis , Liver Function Tests , Pharmacokinetics , Polymorphism, Genetic , Protein Binding , Risk Assessment , Risk Factors
3.
Drug Metab Dispos ; 42(10): 1617-26, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25030308

ABSTRACT

Conflicting results have been obtained by clinical studies investigating the effect of liver cirrhosis on enzyme induction. Because ethical concerns do not give consent for methodologically rigorous studies in humans, we addressed this question by examining the effect of the prototypical inducer dexamethasone (DEX) on the pregnane X receptor (PXR)-mediated induction of CYP3A1 and 3A2 in a validated animal model of liver cirrhosis obtained by exposure of rats to carbon tetrachloride. For this purpose, we assessed mRNA levels, protein expressions, and enzymatic activities of both CYP3A enzymes, as well as mRNA and protein expressions of PXR in rat populations rigorously stratified according to the severity of liver insufficiency. Constitutive mRNA and protein expressions of CYP3A1 and CYP3A2 and their basal enzyme activities were not affected by liver dysfunction. DEX treatment markedly increased steady-state mRNA level, protein content, and enzymatic activity of CYP3A1 in healthy and cirrhotic rats, irrespective of the degree of liver dysfunction. On the contrary, the inducing effect of DEX on gene and protein expressions and enzyme activity of CYP3A2 was preserved in moderate liver insufficiency, whereas it was greatly curtailed when liver insufficiency became severe. mRNA and protein expressions of PXR were neither reduced by liver dysfunction nor increased by DEX treatment. These results indicate that even the inducibility of cytochrome P450 isoforms under the transcriptional control of the same nuclear receptor may be differentially affected by cirrhosis and may partly explain why conflicting results were obtained by human studies.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Liver Cirrhosis, Experimental/metabolism , Receptors, Steroid/biosynthesis , Animals , Carbon Tetrachloride , Dexamethasone/pharmacology , Enzyme Induction , Gene Expression/drug effects , Liver Cirrhosis, Experimental/chemically induced , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Pregnane X Receptor , Rats , Receptors, Steroid/drug effects
4.
PLoS One ; 9(5): e97592, 2014.
Article in English | MEDLINE | ID: mdl-24831942

ABSTRACT

Adrenomedullin (AM) is a multifunctional peptide endowed with various biological actions mediated by the interaction with the calcitonin receptor-like receptor (CLR), which couples to the receptor activity-modifying proteins 2 or 3 (RAMP2 or RAMP3) to form the functional plasma membrane receptors AM1 and AM2, respectively. In this study, we investigated for the first time the expression and localization of AM, CLR, RAMP2 and RAMP3 in human thymic tissue from newborns and in primary cultures of thymic epithelial cells (TECs) and thymocytes. Immunohistochemical analysis of thymic tissue showed that both AM and RAMP2 are abundantly expressed in the epithelial cells of medulla and cortex, blood vessels and mastocytes. In contrast, RAMP3 could not be detected. In cultured TECs, double immunofluorescence coupled to confocal microscopy revealed that AM is present in the cytoplasmic compartment, whereas RAMP2 could be detected in the cytoplasm and nucleus, but not in the cell membrane. At variance with RAMP2, CLR was not only present in the nucleus and cytoplasm of TECs, but could also be detected in the cell membrane. The nuclear and cytoplasmic localizations of RAMP2 and CLR and the absence of RAMP2 in the cell membrane were confirmed by western-blot analysis performed on cell fractions. AM, RAMP2 and CLR could also be detected in thymocytes by means of double immunofluorescence coupled to confocal microscopy, although these proteins were not present in the whole thymocyte population. In these cells, AM and RAMP2 were detected in the cytoplasm, whereas CLR could be observed in the cytoplasm and the plasma membrane. In conclusion, our results show that the AM system is widely expressed in human thymus from newborns and suggest that both AM1 receptor components CLR and RAMP2 are not associated with the plasma membrane of TECs and thymocytes but are located intracellularly, notably in the nucleus.


Subject(s)
Adrenomedullin/metabolism , Gene Expression Regulation , Thymus Gland/growth & development , Calcitonin Receptor-Like Protein/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cytoplasm/metabolism , Epithelial Cells/cytology , Humans , Immunohistochemistry , Infant, Newborn , Mast Cells/cytology , Microscopy, Fluorescence , Protein Binding , Receptor Activity-Modifying Protein 2/metabolism , Receptor Activity-Modifying Protein 3/metabolism , Thymocytes/cytology , Thymus Gland/metabolism
5.
PLoS One ; 8(4): e61983, 2013.
Article in English | MEDLINE | ID: mdl-23626760

ABSTRACT

Although the induction of cytochrome P450 (CYP) has long been investigated in patients with cirrhosis, the question whether liver dysfunction impairs the response to CYP inducers still remains unresolved. Moreover, the mechanism underlying the possible effect of cirrhosis on induction has not been investigated. Since ethical constraints do not permit methodologically rigorous studies in humans, this question was addressed by investigating the effect of the prototypical inducer benzo[a]pyrene (BP) on CYP1A1 and CYP1A2 in cirrhotic rats stratified according to the severity of liver dysfunction. We simultaneously assessed mRNA level, protein expression and enzymatic activity of the CYP1A enzymes, as well as mRNA and protein expressions of the aryl hydrocarbon receptor (AhR), which mediates the BP effect. Basal mRNA and protein expressions of CYP1A1 were virtually absent in both healthy and cirrhotic rats. On the contrary, CYP1A2 mRNA, protein and enzyme activity were constitutively present in healthy rats and decreased significantly as liver function worsened. BP treatment markedly increased the concentrations of mRNA and immunodetectable protein, and the enzymatic activities of both CYP1A enzymes to similar levels in healthy and non-ascitic cirrhotic rats. Induced mRNA levels, protein expressions and enzymatic activities of both CYPs were much lower in ascitic rats and were proportionally reduced. Both constitutive and induced protein expressions of AhR were significantly lower in ascitic than in healthy rats. These results indicate that the inducibility of CYP1A enzymes is well preserved in compensated cirrhosis, whereas it is markedly reduced when liver dysfunction becomes severe. Induction appears to be impaired at the transcriptional level, due to the reduced expression of AhR, which controls the transcription of CYP1A genes.


Subject(s)
Ascites/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochromes/metabolism , Enzyme Induction/genetics , Liver Cirrhosis, Experimental/genetics , Liver/enzymology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Ascites/chemically induced , Ascites/enzymology , Ascites/pathology , Benzo(a)pyrene , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2 , Cytochromes/genetics , Gene Expression Regulation , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/enzymology , Liver Cirrhosis, Experimental/pathology , Liver Function Tests , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Aryl Hydrocarbon/genetics , Severity of Illness Index , Transcription, Genetic
6.
Drug Metab Pharmacokinet ; 27(6): 640-52, 2012.
Article in English | MEDLINE | ID: mdl-22785257

ABSTRACT

The objective of this study was to compare RT-PCR, Western blot and determination of enzyme activity in the assessment of the induction of cytochromes P450 (CYPs) 1A1 and 1A2 by benzo[a]pyrene (BaP) in Sprague-Dawley and Wistar rats. Inhibition studies and kinetic analyses confirmed literature data indicating that methoxyresorufin is a specific CYP1A2 substrate in both uninduced and BaP-treated rats, whereas ethoxyresorufin is a specific CYP1A1 substrate only in BaP-treated rats. BaP treatment increased mRNA and protein expressions of both CYP1A enzymes to a greater extent in Wistar than Sprague-Dawley rats. It consistently caused a higher increase in mRNA and protein expression of the aryl hydrocarbon receptor in the former rats. By contrast, CYP1A2 enzyme activity was much more markedly increased in Sprague-Dawley than Wistar rats and CYP1A1 activity was induced to similar levels. A BaP-induced increase in the turnover number of CYP1A enzymes in Sprague-Dawley rats, relative to Wistar rats, may provide a plausible explanation for the differential effect of BaP on gene expression and enzyme activity. These results have methodological implications, since they show that RT-PCR and Western blot may not provide a quantitative measure of induction of CYP1A activity, which is the actual measure of the change in CYP1A-mediated metabolism.


Subject(s)
Benzo(a)pyrene/pharmacology , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochromes/genetics , Cytochromes/metabolism , Animals , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A2 , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytochromes/biosynthesis , Gene Expression/drug effects , Kinetics , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Oxazines/pharmacology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
7.
Drug Metab Pharmacokinet ; 26(6): 559-68, 2011.
Article in English | MEDLINE | ID: mdl-21791871

ABSTRACT

The aim of this study was to assess the effects of diosmetin and hesperetin, two flavonoids present in various medicinal products, on CYP2C8 activity of human liver microsomes using paclitaxel oxidation to 6α-hydroxy-paclitaxel as a probe reaction. Diosmetin and hesperetin inhibited 6α-hydroxy-paclitaxel production in a concentration-dependent manner, diosmetin being about 16-fold more potent than hesperetin (mean IC(50) values 4.25 ± 0.02 and 68.5 ± 3.3 µM for diosmetin and hesperetin, respectively). Due to the low inhibitory potency of hesperetin, we characterized the mechanism of diosmetin-induced inhibition only. This flavonoid proved to be a reversible, dead-end, full inhibitor of CYP2C8, its mean inhibition constant (K(i)) being 3.13 ± 0.11 µM. Kinetic analysis showed that diosmetin caused mixed-type inhibition, since it significantly decreased the V(max) (maximum velocity) and increased the K(m) value (substrate concentration yielding 50% of V(max)) of the reaction. The results of kinetic analyses were consistent with those of molecular docking simulation, which showed that the putative binding site of diosmetin coincided with the CYP2C8 substrate binding site. The demonstration that diosmetin inhibits CYP2C8 at concentrations similar to those observed after in vivo administration (in the low micromolar range) is of potential clinical relevance, since it may cause pharmacokinetic interactions with co-administered drugs metabolized by this CYP.


Subject(s)
Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/metabolism , Flavonoids/pharmacology , Flavonoids/pharmacokinetics , Binding Sites , Cytochrome P-450 CYP2C8 , Hesperidin/pharmacokinetics , Hesperidin/pharmacology , Humans , Inactivation, Metabolic , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Dynamics Simulation , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology
8.
Drug Metab Pharmacokinet ; 25(5): 466-76, 2010.
Article in English | MEDLINE | ID: mdl-20877134

ABSTRACT

The aim of this study was to examine in vitro, by means of kinetic analysis and molecular docking simulations, the effects of the flavone diosmetin and its flavanone analog hesperetin on CYP (cytochrome P450) 2C9-mediated drug metabolism. To this purpose, the conversion of diclofenac to 4'-hydroxydiclofenac by human liver microsomes was used as a model assay for assessing the CYP2C9 inhibitory activity of these two flavonoids. Kinetic analyses showed that diosmetin and hesperetin were reversible, dead-end inhibitors of 4'-hydroxydiclofenac formation; their mean K(i) (inhibitor dissociation constant) values were 1.71 ± 0.58 and 21.50 ± 3.62 µM, respectively. Diosmetin behaved as a competitive inhibitor, since it increased markedly the K(m) (substrate concentration yielding 50% of V(max)) of the reaction without affecting the V(max) (maximum velocity of reaction). Hesperetin modified markedly K(m) and to a lesser extent also modified V(max), thus acting as a mixed competitive-noncompetitive inhibitor. The results of molecular docking simulations were consistent with those of kinetic analysis, since they showed that the putative binding sites of both diosmetin and hesperetin coincided with the CYP2C9 substrate binding site. The demonstration that diosmetin and hesperetin inhibit CYP2C9-mediated diclofenac metabolism at low micromolar concentrations is of potential clinical relevance because CYP2C9 is responsible for the biotransformation of various therapeutically important drugs that have narrow therapeutic indexes.


Subject(s)
Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Flavonoids/pharmacology , Hesperidin/pharmacology , Pharmaceutical Preparations/metabolism , Aryl Hydrocarbon Hydroxylases/chemistry , Aryl Hydrocarbon Hydroxylases/drug effects , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Binding, Competitive , Biocatalysis/drug effects , Cytochrome P-450 CYP2C9 , Diclofenac/analogs & derivatives , Diclofenac/metabolism , Female , Flavonoids/chemistry , Flavonoids/metabolism , Flurbiprofen/chemistry , Hesperidin/chemistry , Hesperidin/metabolism , Humans , Hydroxylation/drug effects , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Models, Molecular , NADP/metabolism , Recombinant Proteins/metabolism , Thermodynamics
9.
Expert Opin Drug Metab Toxicol ; 6(10): 1215-30, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20604736

ABSTRACT

IMPORTANCE OF THE FIELD: Inhibition of CYP-mediated metabolism is the most frequent and dangerous drug interaction, which causes serious problems in drug development, drug approval and clinical practice. The wide interindividual variability in the magnitude of such interactions constitutes the main hurdle to their management by dose adjustment. AREA COVERED IN THIS REVIEW: This review focuses on a recently identified source of variability in the extent of inhibitory drug interactions, namely liver functional status. It examines the differential effect of liver dysfunction on reversible and mechanism-based CYP inhibition, as well as on inhibition accompanied by plasma protein-binding displacement. WHAT THE READER WILL GAIN: Liver disease progression is accompanied by a proportional, drastic reduction in the magnitude of reversible inhibitory drug interactions due to decreased hepatic uptake of the inhibitor and reduced CYP expression. The degree of mechanism-based inhibition is reduced to a much lesser extent as it is only influenced by the decreased hepatic content of the inhibited CYP enzyme. Metabolic inhibition accompanied by plasma protein-binding displacement results in a disproportionate rise in free drug concentration, the extent of which increases as liver function worsens. TAKE HOME MESSAGE: For an appropriate management of inhibitory drug interactions, the patient's liver functional status and the mechanism of inhibition must be taken into consideration.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Liver Diseases/physiopathology , Pharmaceutical Preparations/metabolism , Animals , Blood Proteins/metabolism , Cytochrome P-450 Enzyme System/genetics , Disease Progression , Drug Interactions , Gene Expression Regulation, Enzymologic , Humans , Protein Binding
10.
Br J Clin Pharmacol ; 69(3): 279-86, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20233199

ABSTRACT

AIMS: To investigate the effects of age and chronic heart failure (CHF) on the oral disposition kinetics of fluvoxamine. METHODS: A single fluvoxamine dose (50 mg) was administered orally to 10 healthy young adults, 10 healthy elderly subjects and 10 elderly patients with CHF. Fluvoxamine concentration in plasma was measured for up to 96 h. RESULTS: With the exception of apparent distribution volume, ageing modified all main pharmacokinetic parameters of fluvoxamine. Thus, peak concentration was about doubled {31 +/- 19 vs. 15 +/- 9 ng ml(-1); difference [95% confidence interval (CI)] 16 (3, 29), P < 0.05}, and area under the concentration-time curve was almost three times higher [885 +/- 560 vs. 304 +/- 84 ng h ml(-1); difference (95% CI) 581 (205, 957), P < 0.05]; half-life was prolonged by 63% [21.1 +/- 6.2 vs. 12.9 +/- 6.4 h; difference (95% CI) 8.2 (2.3, 14.1), P < 0.01], and oral clearance was halved (1.12 +/- 0.77 vs. 2.25 +/- 0.66 l h(-1) kg(-1); difference (95% CI) -1.13 (-1.80, -0.46), P < 0.001]. A significant inverse correlation was consistently observed between age and oral clearance (r=-0.67; P < 0.001). The coexistence of CHF had no significant effect on any pharmacokinetic parameters in elderly subjects. CONCLUSIONS: Ageing results in considerable impairment of fluvoxamine disposition, whereas CHF causes no significant modifications. Therefore, adjustment of initial dose and subsequent dose titrations may be required in elderly subjects, whereas no further dose reduction is necessary in elderly patients with CHF.


Subject(s)
Depression/drug therapy , Fluvoxamine/pharmacokinetics , Heart Failure/metabolism , Administration, Oral , Adult , Age Factors , Aged , Aged, 80 and over , Aging , Chronic Disease , Fluvoxamine/administration & dosage , Fluvoxamine/blood , Humans , Male , Metabolic Clearance Rate
11.
Biochem Pharmacol ; 76(6): 784-95, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18671948

ABSTRACT

We recently demonstrated that nemorubicin (MMDX), an investigational antitumor drug, is converted to an active metabolite, PNU-159682, by human liver cytochrome P450 (CYP) 3A4. The objectives of this study were: (1) to investigate MMDX metabolism by liver microsomes from laboratory animals (mice, rats, and dogs of both sexes) to ascertain whether PNU-159682 is also produced in these species, and to identify the CYP form(s) responsible for its formation; (2) to compare the animal metabolism of MMDX with that by human liver microsomes (HLMs), in order to determine which animal species is closest to human beings; (3) to explore whether differences in PNU-159682 formation are responsible for previously reported species- and sex-related differences in MMDX host toxicity. The animal metabolism of MMDX proved to be qualitatively similar to that observed with HLMs since, in all tested species, MMDX was mainly converted to PNU-159682 by a single CYP3A form. However, there were marked quantitative inter- and intra-species differences in kinetic parameters. The mouse and the male rat exhibited V(max) and intrinsic metabolic clearance (CL(int)) values closest to those of human beings, suggesting that these species are the most suitable animal models to investigate MMDX biotransformation. A close inverse correlation was found between MMDX CL(int) and previously reported values of MMDX LD(50) for animals of the species, sex and strain tested here, indicating that differences in the in vivo toxicity of MMDX are most probably due to sex- and species-related differences in the extent of PNU-159682 formation.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Doxorubicin/analogs & derivatives , Microsomes, Liver/metabolism , Animals , Antineoplastic Agents/chemistry , Biotransformation , Dogs , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Female , Humans , Male , Mice , Microsomes, Liver/chemistry , Rats , Rats, Sprague-Dawley , Species Specificity
12.
Curr Clin Pharmacol ; 3(1): 56-69, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18690879

ABSTRACT

This article reviews the influence of liver functional status on pharmacokinetic interactions due to inhibition and induction of drug-metabolizing enzymes. Recent human studies have shown that the magnitude of inhibitory interactions caused by the reversible CYP1A2 inhibitor fluvoxamine decreases as liver function worsens, and virtually vanishes in patients with more advanced hepatocellular insufficiency. This effect of liver dysfunction is independent of the pharmacokinetic characteristics of the CYP1A2 substrate, since it has been observed with both high- and low-clearance drugs. It is most probably due to reduced uptake of the inhibitory drug by the cirrhotic liver. In order to ascertain whether this is a general phenomenon, the following questions remain to be addressed: 1) whether the inhibition of any CYP isoform is reduced in liver disease; 2) whether the effect of liver dysfunction depends on the chemical nature of the inhibitory drug, since both reduced in vivo inhibition and in vitro uptake by the cirrhotic liver have so far been shown only for basic drugs; 3) lastly, if similar effects can also be observed with irreversible and quasi-irreversible inhibitors, as their accumulation kinetics in the hepatocyte may differ from those of a reversible inhibitor. Although many in vivo and in vitro studies have examined the inducibility of drug-metabolizing enzymes in liver disease, available data are incomplete and conflicting, since both well-preserved and severely curtailed responses to inducing agents have been reported. The reasons for these variable responses are most probably methodological, i.e., differences in the type and degree of liver dysfunction of the animals and patients examined, and in the type and dosage of the inducing agent used. Nonetheless, the results of those few studies which used pathologically homogeneous animal or patient groups suggest that, like basal enzyme expression, drug-inducible expression is also substantially preserved in mild to moderate liver disease, whereas it is lost in severe hepatic dysfunction. For a definitive conclusion, further studies are necessary which examine etiologically homogeneous patient groups and stratify patients rigorously according to their functional hepatic reserve. Such studies should also examine inducers with different physicochemical properties and acting by different mechanisms, since the expression of both hepatic transporters and nuclear receptors may be differentially affected by liver function impairment.


Subject(s)
Enzyme Induction , Enzyme Inhibitors/pharmacology , Liver Diseases/enzymology , Animals , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/biosynthesis , Drug Interactions , Fluvoxamine/pharmacology , Humans , Lidocaine/metabolism , Liver/metabolism , Theophylline/metabolism
13.
Biochem Pharmacol ; 75(6): 1426-37, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18191104

ABSTRACT

We evaluated the effects of increasing concentrations of the flavonoids salvigenin, diosmetin and luteolin on the in vitro metabolism of midazolam (MDZ), a probe substrate for cytochrome P450 (CYP) 3A enzymes, which is converted into 1'-hydroxy-midazolam (1'-OH-MDZ) and 4-hydroxy-midazolam (4-OH-MDZ) by human liver microsomes. Salvigenin had only a modest effect on MDZ metabolism, whereas diosmetin and luteolin inhibited in a concentration-dependent manner the formation of both 1'-OH-MDZ and 4-OH-MDZ, with apparent K(i) values in the 30-50mumol range. Both diosmetin and luteolin decreased 1'-OH-MDZ formation by human recombinant CYP3A4, but not CYP3A5, whereas they decreased 4-OH-MDZ formation by both recombinant enzymes. To assess whether any relationship exists between the physico-chemical characteristics of flavones and their effects on MDZ metabolism, we tested the effects of three other flavones (flavone, tangeretin, chrysin) on MDZ metabolism by human liver microsomes. Whereas flavones possessing more than two hydroxyl groups (luteolin, diosmetin) inhibited MDZ biotransformation, flavones lacking hydroxyl groups in their A and B rings (flavone, tangeretin) stimulated MDZ metabolism. We also found close relationships between the maximum stimulatory or inhibitory effects of flavones on 1'-OH-MDZ and 4-OH-MDZ formation rates and their log of octanol/water partition coefficients (logP) or their total number of hydroxyl groups. The results of the study may be of clinical relevance since they suggest that luteolin and diosmetin may cause pharmacokinetic interactions with co-administered drugs metabolized via CYP3A.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Flavonoids/pharmacology , Luteolin/pharmacology , Midazolam/antagonists & inhibitors , Cytochrome P-450 CYP3A/genetics , Drug Interactions , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Midazolam/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
Clin Pharmacol Ther ; 80(6): 597-606, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17178261

ABSTRACT

BACKGROUND AND OBJECTIVES: The effect of chronic renal failure (CRF) on the pharmacokinetics of lidocaine, a drug cleared almost exclusively by hepatic metabolism, has thus far only been evaluated in patients undergoing regular hemodialysis. This study had 2 objectives: (1) to investigate the effect of CRF on the pharmacokinetics of lidocaine in both patients undergoing hemodialysis and patients not undergoing hemodialysis and (2) to test the effects of plasma from the patients examined and of lidocaine metabolites possibly accumulated in vivo on lidocaine biotransformation in vitro. METHODS: In a clinical investigation we studied the kinetics of lidocaine and its metabolites, monoethylglycinexylidide (MEGX) and glycinexylidide (GX), after intravenous injection of 1 mg/kg lidocaine in 15 healthy volunteers (creatinine clearance [CL(cr)] >80 mL/min x 1.73 m(-2)), 10 subjects with moderate renal insufficiency (CL(cr) between 30 and 60 mL/min x 1.73 m(-2)), 10 subjects with severe renal insufficiency (CL(cr) <30 mL/min x 1.73 m(-2)), and 10 functionally anephric patients undergoing long-term hemodialysis. In experiments in vitro we determined the effects of plasma and GX on the formation rate of the primary lidocaine metabolite, MEGX, by use of human liver microsomes. RESULTS: In patients not undergoing hemodialysis, lidocaine kinetic parameters were altered in proportion to the degree of renal function impairment, but only in patients with severe renal insufficiency were differences statistically significant: clearance was about half that of control subjects (mean +/- SD, 6.01 +/- 2.54 mL/min x kg versus 11.87 +/- 2.97 mL/min x kg; P < .001), and half-life was approximately doubled (4.55 +/- 1.71 hours versus 2.24 +/- 0.55 hours, P < .001). No such alterations were observed in patients undergoing regular hemodialysis, whose values were similar to those of the control group. The steady-state volume of distribution and MEGX levels were independent of renal function, whereas GX levels were more than double those of control subjects (P < .05) in all CRF groups. No inhibitory effect of plasma was observed, for any of the subjects examined, on lidocaine biotransformation in vitro. GX was found to be a competitive inhibitor, but its apparent inhibition constant value (52 +/- 6 micromol/L) was 2 orders of magnitude higher than its concentrations in vivo. CONCLUSIONS: Our in vivo findings have both clinical and methodologic implications: (1) Lidocaine dose adjustment may be required in patients with severe renal insufficiency who are not receiving hemodialysis. (2) Results of studies evaluating the effect of CRF on metabolic drug disposition are not of general validity, unless both patients undergoing hemodialysis and patients not undergoing hemodialysis have been examined. Our in vitro observations exclude that impairment of lidocaine disposition is the result of direct inhibition of metabolizing enzymes by accumulated metabolites or uremic toxins. Alternative mechanisms, suggested by the results of recent in vitro studies, are discussed.


Subject(s)
Kidney Failure, Chronic/metabolism , Lidocaine/pharmacokinetics , Renal Dialysis , Area Under Curve , Biotransformation , Half-Life , Humans , Lidocaine/analogs & derivatives , Lidocaine/blood , Lidocaine/metabolism , Male , Metabolic Clearance Rate , Middle Aged
15.
Pharmacol Res ; 54(3): 181-5, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16750634

ABSTRACT

A combination of tacrolimus (TAC) and sirolimus (SIR) has recently proved to be a very effective immunosuppressive regimen in organ transplantation. In pediatric transplant recipients, co-administration of these two drugs has been shown to result in a significant decrease of exposure to TAC, whereas conflicting data have been obtained regarding this pharmacokinetic interaction in adults. The aim of this study was to investigate the effect of SIR on TAC pharmacokinetics in adult transplant recipients. Sixteen adult patients (mean age 38+/-8 years), who had been on standard TAC plus low-dose SIR immunosuppressive treatment for 6 months after renal transplantation, were enrolled for a TAC pharmacokinetic study before and 15 days after discontinuing SIR. Eight patients had received SIR 0.5 mg day(-1) and eight patients 2 mg day(-1). TAC doses remained the same in all patients after SIR withdrawal. After discontinuing SIR, statistically significant, dose-dependent increases were observed in area under the curve (AUC), peak (C(max)) and trough (C(min)) TAC concentrations (+15-20% and +27-32%, after discontinuing the 0.5 and the 2 mg day(-1) doses, respectively). Proportional decreases were consistently observed in apparent oral clearance (-13% and -23%). Very good correlations were found between TAC AUC and C(min), both before and after SIR withdrawal (R(2)=0.94, P<0.0001 and R(2)=0.97, P<0.0001, respectively). Our findings clearly demonstrate that the SIR-induced reduction in TAC exposure also takes place in adults and is, therefore, a general, age-independent phenomenon. Hence, TAC levels need to be carefully monitored in transplant recipients of any age, in order to avoid possible TAC overexposure upon SIR discontinuation.


Subject(s)
Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/pharmacokinetics , Kidney Transplantation , Sirolimus/pharmacology , Tacrolimus/pharmacokinetics , Adult , Drug Interactions , Female , Humans , Kidney/metabolism , Male , Multicenter Studies as Topic , Randomized Controlled Trials as Topic , Sirolimus/blood , Tacrolimus/blood
16.
Clin Pharmacol Ther ; 79(5): 489-99, 2006 May.
Article in English | MEDLINE | ID: mdl-16678550

ABSTRACT

BACKGROUND AND OBJECTIVES: In vivo inhibition of cytochrome P450 (CYP) 1A2 by fluvoxamine causes a reduction in the clearance of the high-extraction drug lidocaine, which decreases in proportion to the degree of liver dysfunction. The objectives of this study were (1) to evaluate the effect of liver cirrhosis on the inhibition by fluvoxamine of the metabolic disposition of theophylline, a CYP1A2 substrate with a low-extraction ratio, to assess whether decreased sensitivity to CYP1A2 inhibition in liver disease is a general characteristic of CYP1A2 substrates, regardless of their pharmacokinetic properties, and (2) to investigate the mechanism(s) underlying the effect of liver dysfunction on CYP1A2 inhibition. METHODS: The study was carried out in 10 healthy volunteers and 20 patients with cirrhosis, 10 with mild liver dysfunction (Child class A) and 10 with severe liver dysfunction (Child class C), according to a randomized, double-blind, 2-phase, crossover design. In one phase all participants received placebo for 7 days; in the other phase they received one 50-mg fluvoxamine dose for 2 days and two 50-mg fluvoxamine doses, 12 hours apart, in the next 5 days. On day 6, 4 mg/kg of theophylline was administered orally 1 hour after the morning fluvoxamine dose. Concentrations of theophylline and its metabolites, 3-methylxanthine, 1-methyluric acid, and 1,3-dimethyluric acid, were then measured in plasma and urine up to 48 hours. RESULTS: Fluvoxamine-induced inhibition of theophylline clearance decreased from 62% in healthy subjects to 52% and 12% in patients with mild cirrhosis and those with severe cirrhosis, respectively. CYP1A2-mediated formations of 3-methylxanthine and 1-methyluric acid were almost totally inhibited in control subjects, whereas they were only reduced by one third in patients with Child class C cirrhosis. Inhibition of 1,3-dimethyluric acid formation, which is catalyzed by CYP1A2 and CYP2E1, progressively decreased from 58% in healthy subjects to 43% and 7% in patients with mild cirrhosis and those with severe cirrhosis, respectively. CONCLUSIONS: The effect of liver dysfunction on the inhibition of CYP1A2-mediated drug elimination is a general phenomenon, independent of the pharmacokinetic characteristics of the CYP1A2 substrate. Therefore, for any drug metabolized by CYP1A2, the clinical consequences of enzyme inhibition are expected to become less and less important as liver function worsens. Two mechanisms, as follows in order of importance, are responsible for the effect of liver dysfunction: (1) decreased sensitivity to fluvoxamine of CYP1A2-mediated biotransformations in the cirrhotic liver, probably resulting from reduced uptake of the inhibitory drug, and (2) reduced hepatic expression of CYP1A2, which makes its contribution to overall drug elimination less important.


Subject(s)
Antidepressive Agents/pharmacology , Bronchodilator Agents/pharmacokinetics , Cytochrome P-450 CYP1A2 Inhibitors , Fluvoxamine/pharmacology , Liver Cirrhosis/metabolism , Theophylline/pharmacokinetics , Adult , Cross-Over Studies , Double-Blind Method , Humans , Middle Aged , Severity of Illness Index
17.
Clin Pharmacol Ther ; 75(1): 80-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14749694

ABSTRACT

OBJECTIVES: This study was designed (1) to evaluate the effect of a cytochrome P450 (CYP) 1A2 inhibitor, fluvoxamine, on the pharmacokinetics of intravenous lidocaine and its 2 pharmacologically active metabolites, monoethylglycinexylidide (MEGX) and glycinexylidide (GX), to confirm recent in vitro results indicating that CYP1A2 is the main isoform responsible for lidocaine biotransformation and (2) to assess whether liver function has any influence on the fluvoxamine-lidocaine interaction. METHODS: The study was carried out in 10 healthy volunteers and 20 patients with cirrhosis, 10 with mild (Child grade A) and 10 with severe (Child grade C) liver dysfunction, according to a randomized, double-blind, 2-phase, crossover design. In one phase all participants received placebo for 6 days; in the other phase they received 50 mg fluvoxamine for 2 days and 100 mg fluvoxamine for the next 4 days. On day 6, a 1-mg/kg lidocaine dose was administered intravenously 2 hours after the last dose of fluvoxamine or placebo. Plasma concentrations of lidocaine, MEGX, GX, and fluvoxamine were measured up to 12 hours after lidocaine injection. RESULTS: The effects of fluvoxamine coadministration were dependent on liver function. Lidocaine clearance was decreased on average by 60% (from 12.1 mL/min.kg to 4.85 mL/min.kg, P <.001) in healthy subjects and by 44% (from 9.83 mL/min.kg to 5.06 mL/min.kg, P <.001) in patients with mild liver dysfunction, with proportional increases in terminal half-lives, whereas virtually no effect was produced in patients with severe liver dysfunction (4.21 mL/min.kg versus 3.65 mL/min.kg, P >.05). Analogous effects were observed on MEGX and GX formation kinetics, which were drastically impaired in healthy subjects and patients with mild liver cirrhosis but virtually unaffected in patients with severe cirrhosis. CONCLUSION: CYP1A2 is the enzyme principally responsible for the metabolic disposition of lidocaine in subjects with normal liver function. The extent of fluvoxamine-lidocaine interaction decreases as liver function worsens, most likely because of the concomitant decrease in the hepatic level of CYP1A2. These observations indicate that results obtained in healthy subjects cannot be extended a priori to patients with liver dysfunction, but the clinical consequences of inhibition of drug metabolism must also be assessed in such patients.


Subject(s)
Anti-Arrhythmia Agents/pharmacokinetics , Cytochrome P-450 CYP1A2 Inhibitors , Fluvoxamine/pharmacology , Lidocaine/analogs & derivatives , Lidocaine/pharmacokinetics , Selective Serotonin Reuptake Inhibitors/pharmacokinetics , Anti-Arrhythmia Agents/administration & dosage , Anti-Arrhythmia Agents/blood , Area Under Curve , Cross-Over Studies , Double-Blind Method , Fluvoxamine/blood , Humans , Infusions, Intravenous , Lidocaine/administration & dosage , Lidocaine/blood , Liver/drug effects , Liver/enzymology , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Liver Function Tests , Male , Middle Aged , Selective Serotonin Reuptake Inhibitors/blood , Severity of Illness Index
18.
Surg Oncol Clin N Am ; 12(3): 781-94, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14567031

ABSTRACT

Intraperitoneal chemotherapy, mainly when performed during HIIC after cytoreductive surgery, is considered potentially curative for the treatment of solid tumors with spread to the peritoneal surface. When selecting antiblastic agents to be administered intraperitoneally, it is important to bear in mind that a low lipophility and a high molecular weight are the ideal drug characteristics. Drugs with these features allow a favorable ratio to be achieved between peritoneal and plasma concentrations, due to the reduced tendency to diffuse through the plasma-peritoneal barrier, even after extensive removal of the peritoneum. Moreover, a low rate of diffusion through the tumor capillaries implies a low rate of drug clearance, with a higher intratumoral drug accumulation. Among the drugs used so far for intraperitoneal chemotherapy, the combination of CDDP and DXR appears to be one of the most effective available regimens with acceptable local-regional toxicity. CDDP has also been extensively employed as a single agent for ovarian and gastrointestinal cancers, under both normal and hyperthermic conditions, while intraperitoneally administered DXR appears to be of greater potential efficacy when associated with CDDP and hyperthermia (41.5 degrees C) following cytoreductive surgery. In our clinical experience with this drug combination, DXR showed a much more advantageous plasma/peritoneal AUC ratio than CDDP (162 +/- 113 and 20 +/- 6, respectively). On the other hand, it has been demonstrated that very high intraperitoneal concentrations of CDDP can be achieved without incurring significant systemic toxicity by using intravenous injection of sodium thiosulphate during HIIC. Penetration of the tumor mass by CDDP is greater than DXR. This phenomenon is enhanced by hyperthermia and by hypotonic solutions of sodium chloride used as the perfusate. Following experimental and clinical results of TNF alpha-based isolated limb perfusion for locally advanced soft tissue sarcoma or melanoma, greater efforts are being made to exploit the potential effect of this cytokine used in association with hyperthermia and other drugs (i.e., CDDP and DXR) suitable for intraperitoneal infusion/perfusion. However, it is not yet clear whether the observed effect of TNF alpha on the peritoneal-plasma barrier, which seems to favor the passage of both drugs into the systemic circulation, is overcome by the positive effect of this agent on drug penetration into tumor. Further pharmacologic studies should be undertaken to clarify whether or not these interactions will be of benefit to the patient. Likewise, liposomes, which in animal models seem to favor tumor uptake of encapsulated DXR, should now be tested in the clinical setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carcinoma/drug therapy , Infusions, Parenteral , Peritoneal Neoplasms/drug therapy , Biological Availability , Carcinoma/mortality , Carcinoma/pathology , Carcinoma/surgery , Cisplatin/administration & dosage , Combined Modality Therapy , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Female , Humans , Intraoperative Period , Male , Maximum Tolerated Dose , Neoplasm Staging , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/surgery , Peritoneum/drug effects , Peritoneum/surgery , Prognosis , Randomized Controlled Trials as Topic , Risk Assessment , Survival Rate , Treatment Outcome
19.
J Laparoendosc Adv Surg Tech A ; 13(3): 181-4, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12855100

ABSTRACT

Laparoscopy is a relatively safe invasive procedure, but complications can occur, mainly related to Veress needle and trocar insertion. The rate of these complications is generally reported to be low, but the true incidence may be higher because of underreporting. We retrospectively studied the records of 2650 consecutive diagnostic laparoscopies performed by the same operator with the aim of assessing the true incidence and nature of these complications. Major complications occurred in 0.41% of cases and included bladder injury, bowel perforation, hemoperitoneum, and abdominal wall hematoma. Minor complications, including omental and subcutaneous emphysema, occurred in 1.58% of cases. Some of these resolved spontaneously, whereas others required surgical or medical treatment. We believe that all laparoscopic complications should be reported to a registry so that their potential risk can be quantified. Simply reporting complications as major or minor on the basis of the follow-up does not allow laparoscopists to understand their true incidence completely.


Subject(s)
Anesthesia, Local , Laparoscopy/adverse effects , Needles/adverse effects , Surgical Instruments/adverse effects , Adolescent , Adult , Aged , Aged, 80 and over , Female , Hematoma/epidemiology , Humans , Incidence , Intestinal Perforation/epidemiology , Male , Middle Aged , Postoperative Complications/epidemiology , Retrospective Studies
20.
Life Sci ; 72(24): 2741-50, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12679191

ABSTRACT

Resveratrol (3,4',5-trihydroxy-trans-stilbene) is a natural phytoalexin found in grapes and wine. It has antioxidant and antiproliferative activities, and has been shown to induce NAD(P)H:quinone oxidoreductase, also known as DT-diaphorase, in cultured mouse hepatoma cells. DT-diaphorase is a detoxifying enzyme for quinone-containing substances, due to its ability to prevent their one-electron reduction and the consequent generation of reactive oxygen species (ROS). The aim of the present study was to investigate whether oral administration of trans-resveratrol to guinea pigs (60 mg/l in tap water for 16 days, ad libitum) increases cardiac DT-diaphorase and, consequently, reduces the response of isolated atria to 2-methyl-1,4-naphthoquinone (menadione), the positive inotropic effect of which is related to the amount of ROS generated by its cardiac metabolism. In the cardiac tissue of resveratrol-treated animals, DT-diaphorase activity was significantly higher than that measured in control animals, the V(max) of the enzyme reaction being 75.47 +/- 3.87 and 50.73 +/- 0.63 nmoles/mg protein/min, respectively (p < 0.05). Resveratrol administration also significantly increased the activity of cardiac catalase (32.20 +/- 2.39 vs. 25.14 +/- 3.85 units/mg protein in treated and control animals, respectively; p < 0.001). As a consequence, menadione metabolism by the cardiac homogenate obtained from resveratrol-treated animals generated a smaller amount of ROS and, in electrically driven left atria, menadione produced a significantly lower increase in the force of contraction than in atria isolated from control animals. These results indicate that oral administration of resveratrol exerts cardioprotection against ROS-mediated menadione toxicity.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/pharmacology , Catalase/metabolism , Heart/drug effects , Myocardium/enzymology , NAD(P)H Dehydrogenase (Quinone)/metabolism , Stilbenes/pharmacology , Vitamin K 3/antagonists & inhibitors , Vitamin K 3/toxicity , Animals , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Guinea Pigs , Heart Atria/drug effects , Heart Atria/pathology , In Vitro Techniques , Male , Myocardial Contraction/drug effects , Reactive Oxygen Species/metabolism , Resveratrol , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology , Subcellular Fractions/metabolism , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...