epha.health
Feedback
Neuer Fall
Anmelden
busy
3
Wirkstoffe
|
11
0
Befunde
|
Vorgehen

Pharmakologische Bewertung zu Pravastatin, Gemfibrozil und Amiodaron

Plan
Drucken
Version 6.0.32 (Beta Preview)

Zusammenfassung Zusammenfassung info 82%

Pharmakokinetik -1%
Pravastatin
Gemfibrozil
Amiodaron
Scores -7%
Verlängerung der QT-Zeit
Anticholinerge Effekte
Serotonerge Effekte
Unerwünschte Arzneimittelwirkungen -10%
Übelkeit
Erbrechen
Dyspepsie

Varianten ✨

Für die rechenintensive Bewertung der Varianten bitte das kostenpflichtige Standard Abonnement wählen.

Medikation Anwendungsgebiete

Eklärungen für Patienten zu den Wirkstoffen

undefined Pharmakokinetik info -1%

∑ ExpositionaPraGemAmi
Pravastatin 1.42 [1.42,1.86] 1 1.4 1.02
Gemfibrozil 1 1 1
Amiodaron n.a. 1 n.a.
Relevante Genotypen: 1OATP1B1
Symbol (a): x-fache Veränderung der AUC
Legende (n.a.): Angaben nicht verfügbar

Die genannten Expositionsveränderungen beziehen sich jeweils auf Veränderungen der Plasmakonzentrations-Zeit-Kurve [ AUC ]. Die Exposition von Pravastatin erhöht sich auf 142%, wenn eine Kombination mit Gemfibrozil (140%) und Amiodaron (102%) erfolgt. Für Gemfibrozil erwarten wir keine Veränderung der Exposition, wenn eine Kombination mit Pravastatin (100%) und Amiodaron (100%) erfolgt. Eine Veränderung der Exposition von Amiodaron haben wir nicht erkannt, wenn eine Kombination mit Pravastatin (100%) erfolgt. Den Einfluss von Gemfibrozil können wir aktuell nicht abschätzen.

Bewertung: Für die Berechnung der individuellen Expositionsveränderungen durch die Wechselwirkungen werden als Ausgangsbasis die pharmakokinetischen Parameter der durchschnittlichen Population verwendet.
Pravastatin hat eine tiefe orale Bioverfügbarkeit [ F ] von 18%, weshalb die maximalen Plasmaspiegel [ Cmax ] sich bei einer Interaktion tendentiell stark verändern. Die terminale Halbwertszeit [ t12 ] ist mit 0.79 Stunden eher kurz und konstante Plasmaspiegel [ Css ] werden schnell erreicht. Die Proteinbindung [ Pb ] ist mit 48.5% eher schwach und das Verteilungsvolumen [ Vd ] liegt mit 34 Liter im mittleren Bereich. weshalb bei einer mittleren hepatische Extraktionsrate von 0.65 sowohl der Leberblutfluss [ Q ] als auch eine Veränderung der Proteinbindung [ Pb ] relevant sind. Ungefähr 47.0% einer verabreichten Dosis werden unverändert über die Niere ausgeschieden und dieser Anteil wird selten durch Interaktionen verändert. Die Metabolisierung erfolgt nicht über die gängigen Cytochrome und der aktive Transport erfolgt zum Teil über BCRP, MRP2, MRP4, OATP1A2, OATP1B1, OATP2B1 und PGP.
Gemfibrozil hat eine hohe orale Bioverfügbarkeit [ F ] von 98%, weshalb die maximalen Plasmaspiegel [ Cmax ] sich bei einer Interaktion tendentiell wenig verändern. Die terminale Halbwertszeit [ t12 ] ist mit 1.5 Stunden eher kurz und konstante Plasmaspiegel [ Css ] werden schnell erreicht. Die Proteinbindung [ Pb ] ist mit 97% stark und das Verteilungsvolumen [ Vd ] liegt mit 36 Liter im mittleren Bereich, Die Metabolisierung erfolgt nicht über die gängigen Cytochrome und der aktive Transport erfolgt zum Teil über MRP2, MRP4 und UGT2B7. Unter anderem ist Gemfibrozil ein Hemmer von OATP1B1, OATP2B1 und CYP2C8.
Amiodaron hat eine mittlere orale Bioverfügbarkeit [ F ] von 55%, weshalb die maximalen Plasmaspiegel [ Cmax ] sich bei einer Interaktion tendentiell verändern. Die terminale Halbwertszeit [ t12 ] ist mit 1884 Stunden eher lang und konstante Plasmaspiegel [ Css ] werden erst nach mehr als 7536 Stunden erreicht. Die Proteinbindung [ Pb ] ist mit 96% stark. Die Metabolisierung findet unter anderem über CYP2C8 und CYP3A4 statt und der aktive Transport erfolgt insbesondere über PGP. Unter anderem ist Amiodaron ein Hemmer von BCRP und PGP.

Transmitter Serotonerge Effekte info -0%

Scores ∑ Punkte PraGemAmi
Serotonerge Effekte a 0 Ø Ø Ø
Symbol (a): Risiko ab 5 Punkten erhöht.

Bewertung: Gemäss unseren Erkenntnissen erhöhen weder Pravastatin, Gemfibrozil noch Amiodaron die serotonerge Aktivität.

Transmitter Anticholinerge Effekte info -0%

Scores ∑ Punkte PraGemAmi
Kiesel b 0ØØØ
Symbol (b): Risiko ab 3 Punkten erhöht.

Bewertung: Gemäss unseren Erkenntnissen erhöhen weder Pravastatin, Gemfibrozil noch Amiodaron die anticholinerge Aktivität.

Elektrokardiogramm Verlängerung der QT-Zeit info -9%

Scores ∑ Punkte PraGemAmi
RISK-PATH c 3ØØ+++
Symbol (c): Risiko ab 10 Punkten erhöht. Fragen zu Risikofaktoren sollten beantwortet werden.

Empfehlung: Um das individuelle Risiko für Arrhythmien abschätzen zu können, empfehlen wir Ihnen folgende vollständig zu beantworten.

Bewertung: Amiodaron kann potentiell ventrikuläre Arrhythmien vom Typ Torsades de pointes auslösen. Für Pravastatin und Gemfibrozil ist uns kein QT-Zeit verlängerndes Potential bekannt.

Weitere Nebenwirkungen Allgemeine Nebenwirkungen info -10%

Nebenwirkungen ∑ Häufigkeit PraGemAmi
Übelkeit21.5 %n.a.n.a.21.5
Erbrechen21.5 %n.a.n.a.21.5
Dyspepsie19.6 %n.a.19.6n.a.
Muskuloskelettale Schmerzen14.4 %14.4n.a.n.a.
Infektion der oberen Atemwege12.7 %12.7n.a.n.a.
Hypothyreose10.0 %n.a.n.a.10.0
Bauchschmerzen9.8 %n.a.9.8n.a.
Schwindel7.4 %n.a.+6.5
Lichtempfindlichkeit6.5 %n.a.n.a.6.5
Verstopfung6.5 %n.a.n.a.6.5
Tablellarischer Auszug der häufigsten Nebenwirkungen
Zeichen (+): Nebenwirkung beschrieben, aber Häufigkeit nicht bekannt
Zeichen (↑/↓): Häufigkeit aufgrund der Exposition eher höher / tiefer

Gastrointestinal
Verlust von Appetit (6.5%): Amiodaron
Pankreatitis: Gemfibrozil, Pravastatin
Durchfall: Pravastatin

Neurologisch
Ataxia (6.5%): Amiodaron
Koordinationsstörungen (6.5%): Amiodaron
Parästhesie (6.5%): Amiodaron
Periphere Neuropathie: Amiodaron
Kopfschmerzen: Gemfibrozil, Pravastatin
Pseudotumor cerebri: Amiodaron

Opthalmologisch
Verschwommenes Sehen (6.5%): Amiodaron
Optikusneuritis: Amiodaron
Sehverlust: Amiodaron

Endokrin
Hyperthyreose (2%): Amiodaron

Respiratorisch
Akutes Lungenversagen (2%): Amiodaron
Lungenfibrose: Amiodaron
Husten: Pravastatin
Nasopharyngitis: Pravastatin

Kardiologisch
Hypotonie: Amiodaron
Bradykardie: Amiodaron
Herzinsuffizienz: Amiodaron
Ventrikuläre Arrhythmie: Amiodaron

Dermatologisch
Hautausschlag: Gemfibrozil, Pravastatin
Stevens Johnson-Syndrom: Amiodaron
Toxische epidermale Nekrolyse: Amiodaron

Hepatisch
Erhöhte alkalische Phosphatase: Gemfibrozil
Erhöhte Transaminasen: Gemfibrozil
Hepatotoxizität: Amiodaron
Cholestatische Hepatitis: Gemfibrozil
Hyperbilirubinämie: Gemfibrozil

Muskuloskeletal
Erhöhte Kreatinkinase: Gemfibrozil
Myopathie: Gemfibrozil, Pravastatin
Rhabdomyolyse: Gemfibrozil, Pravastatin
Sehnenruptur: Pravastatin

Hämatologisch
Thrombozytopenie: Amiodaron, Gemfibrozil
Anämie: Gemfibrozil
Leukopenie: Gemfibrozil

Immunologisch
Überempfindlichkeitsreaktion: Amiodaron
Angioödem: Gemfibrozil

Renal
Nierenversagen: Amiodaron

Vaskulär
Vaskulitis: Amiodaron

Einschränkungen Einschränkungen

Basierend auf Ihren und wissenschaftlichen Informationen bewerten wir das individuelle Risiko für unerwünschte Nebenwirkungen. Die orange ausgefüllten Balken signalisieren das grundsätzliche Potential der Medikamente diese Nebenwirkung hervorzurufen. Diese Empfehlungen sollen Fachpersonen beraten und ersetzen nicht die Konsultation durch einen Arzt. In der eingeschränkten Testversion (alpha) ist das Risiko aller Substanzen noch nicht abschliessend bewertet.

Literatur Referenzen

1. Singhvi SM et al. Disposition of pravastatin sodium, a tissue-selective HMG-CoA reductase inhibitor, in healthy subjects. British journal of clinical pharmacology. 1990
Authors: Singhvi SM Pan HY Morrison RA Willard DA
Abstract: Pravastatin sodium, a competitive inhibitor of HMG-CoA reductase, is a new orally effective hypocholesterolaemic agent. In a two-way crossover study, eight healthy male subjects each received an intravenous and an oral dose of [14C]-pravastatin sodium. The oral absorption of [14C] activity from pravastatin sodium was about 34% and the oral bioavailability was about 18%, suggesting first-pass metabolism of pravastatin. After the intravenous dose, the recovery of radioactivity averaged 60% and 34% in urine and faeces, respectively. Corresponding values were 20% (urine) and 71% (faeces) for the oral dose. The estimated average plasma elimination half-life of pravastatin was 0.8 and 1.8 h for the intravenous and oral routes, respectively. The average values for total and renal clearances were 13.5 and 6.3 ml min-1 kg-1, respectively, and the steady-state volume of distribution averaged 0.51 kg-1. These results suggest that both kidney and liver are important sites of elimination for pravastatin.
Pubmed Id: 2106337
2. Middlekauff HR et al. Amiodarone and torsades de pointes in patients with advanced heart failure. The American journal of cardiology. 1995
Authors: Middlekauff HR Stevenson WG Saxon LA Stevenson LW
Abstract: Amiodarone is considered to be safe in patients with prior QT prolongation and torsades de pointes taking class I antiarrhythmic agents who require continued antiarrhythmic drug therapy. However, the safety of amiodarone in advanced heart failure patients with a history of drug-induced torsades de pointes, who may be more susceptible to proarrhythmia, is unknown. Therefore, the objective of this study was to assess amiodarone safety and efficacy in heart failure patients with prior antiarrhythmic drug-induced torsades de pointes. We determined the history of torsades de pointes in 205 patients with heart failure treated with amiodarone, and compared the risk of sudden death in patients with and without such a history. To evaluate the possibility that all patients with a history of torsades de pointes would be at high risk for sudden death regardless of amiodarone treatment, we compared this risk in patients with a history of torsades de pointes who were and were not subsequently treated with amiodarone. Of 205 patients with advanced heart failure, 8 (4%) treated with amiodarone had prior drug-induced torsades de pointes. Despite similar severity of heart failure, the 1-year actuarial sudden death risk was markedly increased in amiodarone patients with than without prior torsades de pointes (55% vs 15%, p = 0.0001). Similarly, the incidence of 1-year sudden death was markedly increased in patients with prior torsades de pointes taking amiodarone compared with such patients who were not subsequently treated with amiodarone (55% vs 0%, p = 0.09).(ABSTRACT TRUNCATED AT 250 WORDS)
Pubmed Id: 7653452
3. Hsiang B et al. A novel human hepatic organic anion transporting polypeptide (OATP2). Identification of a liver-specific human organic anion transporting polypeptide and identification of rat and human hydroxymethylglutaryl-CoA reductase inhibitor transporters. The Journal of biological chemistry. 1999
Authors: Hsiang B Zhu Y Wang Z Wu Y Sasseville V Yang WP Kirchgessner TG
Abstract: A novel human organic transporter, OATP2, has been identified that transports taurocholic acid, the adrenal androgen dehydroepiandrosterone sulfate, and thyroid hormone, as well as the hydroxymethylglutaryl-CoA reductase inhibitor, pravastatin. OATP2 is expressed exclusively in liver in contrast to all other known transporter subtypes that are found in both hepatic and nonhepatic tissues. OATP2 is considerably diverged from other family members, sharing only 42% sequence identity with the four other subtypes. Furthermore, unlike other subtypes, OATP2 did not transport digoxin or aldosterone. The rat isoform oatp1 was also shown to transport pravastatin, whereas other members of the OATP family, i.e. rat oatp2, human OATP, and the prostaglandin transporter, did not. Cis-inhibition studies indicate that both OATP2 and roatp1 also transport other statins including lovastatin, simvastatin, and atorvastatin. In summary, OATP2 is a novel organic anion transport protein that has overlapping but not identical substrate specificities with each of the other subtypes and, with its liver-specific expression, represents a functionally distinct OATP isoform. Furthermore, the identification of oatp1 and OATP2 as pravastatin transporters suggests that they are responsible for the hepatic uptake of this liver-specific hydroxymethylglutaryl-CoA reductase inhibitor in rat and man.
Pubmed Id: 10601278
4. Hatanaka T Clinical pharmacokinetics of pravastatin: mechanisms of pharmacokinetic events. Clinical pharmacokinetics. 2000
Authors: Hatanaka T
Abstract: Pravastatin, one of the 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (statins) widely used in the management of hypercholesterolaemia, has unique pharmacokinetic characteristics among the members of this class. Many in vivo and in vitro human and animal studies suggest that active transport mechanisms are involved in the pharmacokinetics of pravastatin. The oral bioavailability of pravastatin is low because of incomplete absorption and a first-pass effect. The drug is rapidly absorbed from the upper part of the small intestine, probably via proton-coupled carrier-mediated transport, and then taken up by the liver by a sodium-independent bile acid transporter. About half of the pravastatin that reaches the liver via the portal vein is extracted by the liver, and this hepatic extraction is mainly attributed to biliary excretion which is performed by a primary active transport mechanism. The major metabolites are produced by chemical degradation in the stomach rather than by cytochrome P450-dependent metabolism in the liver. The intact drug and its metabolites are cleared through both hepatic and renal routes, and tubular secretion is a predominant mechanism in renal excretion. The dual routes of pravastatin elimination reduce the need for dosage adjustment if the function of either the liver or kidney is impaired, and also reduce the possibility of drug interactions compared with other statins. which are largely eliminated by metabolism. The lower protein binding than other statins weakens the tendency for displacement of highly protein-bound drugs. Although all statins show a hepatoselective disposition, the mechanism for pravastatin is different from that of the others. There is high uptake of pravastatin by the liver via an active transport mechanism, but not by other tissues because of its hydrophilicity, whereas the disposition characteristics of other statins result from high hepatic extraction because of high lipophilicity. These pharmacokinetic properties of pravastatin may be the result of the drug being given in the pharmacologically active open hydroxy acid form and the fact that its hydrophilicity is markedly higher than that of other statins. The nature of the pravastatin transporters, particularly in humans, remains unknown at present. Further mechanistic studies are required to establish the pharmacokinetic-pharmacodynamic relationships of pravastatin and to provide the optimal therapeutic efficacy for various types of patients with hypercholesterolaemia.
Pubmed Id: 11192473
5. Nakai D et al. Human liver-specific organic anion transporter, LST-1, mediates uptake of pravastatin by human hepatocytes. The Journal of pharmacology and experimental therapeutics. 2001
Authors: Nakai D Nakagomi R Furuta Y Tokui T Abe T Ikeda T Nishimura K
Abstract: Involvement of LST-1 (a human liver-specific transporter, also called OATP2) as the major transporter in the uptake of pravastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, by human liver was demonstrated. The hepatic uptake of pravastatin evaluated using human hepatocytes was Na(+)-independent and reached saturation with a Michaelis constant (K(m)) of 11.5 +/- 2.2 microM. The uptake of pravastatin was temperature-dependent and was inhibited by estradiol-17beta-D-glucuronide, taurocholic acid, bromosulfophthalein, and simvastatin acid, but not by p-aminohippurate. Estradiol-17beta-D-glucuronide competitively inhibited pravastatin uptake with an inhibition constant comparable to the K(m) value for estradiol-17beta-D-glucuronide transport, indicating that a common transporter mediates the transport of pravastatin and estradiol-17beta-D-glucuronide in human hepatocytes. The results obtained with human hepatocytes agreed with those obtained with LST-1 expressing Xenopus oocytes. Oocytes microinjected with human liver polyadenylated mRNA showed Na(+)-independent uptake of pravastatin and estradiol-17beta-D-glucuronide. A simultaneous injection of LST-1 antisense oligonucleotides completely abolished this uptake. Expression of LST-1 was immunohistochemically demonstrated in the human hepatocytes, but not in Hep G2 cells, which showed very low uptake of pravastatin. Therefore, LST-1 was regarded as a key molecule for pravastatin in liver-specific inhibition of cholesterol synthesis, making pravastatin accessible to the target enzyme, which would otherwise not be inhibited by this hydrophilic drug.
Pubmed Id: 11356905
6. Sasaki M et al. Transcellular transport of organic anions across a double-transfected Madin-Darby canine kidney II cell monolayer expressing both human organic anion-transporting polypeptide (OATP2/SLC21A6) and Multidrug resistance-associated protein 2 (MRP2/ABCC2). The Journal of biological chemistry. 2002
Authors: Sasaki M Suzuki H Ito K Abe T Sugiyama Y
Abstract: Human organic anion transporting polypeptide 2 (OATP2/SLC21A6) and multidrug resistance-associated protein 2 (MRP2/ABCC2) play important roles in the vectorial transport of organic anions across hepatocytes. In the present study, we have established a double-transfected Madin-Darby canine kidney (MDCK II) cell monolayer, which expresses both OATP2 and MRP2 on basal and apical membranes, respectively. The basal-to-apical transport of 17 beta estradiol 17 beta-d-glucuronide (E(2)17 beta G), pravastatin, and leukotriene C(4) (LTC(4)), which are substrates of OATP2 and MRP2, was significantly higher than that in the opposite direction in the double-transfected cells. Such vectorial transport was also observed for taurolithocholate sulfate, which is transported by rat oatp1 and Mrp2. The K(m) values of E(2)17 beta G and pravastatin for the basal-to-apical flux were 27.9 and 24.3 microm, respectively, which were comparable with those reported for OATP2. Moreover, the MRP2-mediated export of E(2)17 beta G across the apical membrane was not saturated. In contrast, basal-to-apical transport of estrone-3-sulfate and dehydroepiandrosterone sulfate, which are significantly transported by OATP2, but not by MRP2, was not stimulated by MRP2 expression. The double-transfected MDCK II monolayer expressing both OATP2 and MRP2 may be used to analyze the hepatic vectorial transport of organic anions and to screen the transport profiles of new drug candidates.
Pubmed Id: 11748225
7. Nozawa T et al. Functional characterization of pH-sensitive organic anion transporting polypeptide OATP-B in human. The Journal of pharmacology and experimental therapeutics. 2004
Authors: Nozawa T Imai K Nezu J Tsuji A Tamai I
Abstract: The pH-sensitive activity of human organic anion transporting polypeptide OATP-B, which is expressed at the apical membrane of human small intestinal epithelial cells, was functionally characterized. When initial uptake of estrone-3-sulfate, a typical substrate of OATP, was studied kinetically, we observed an increase in V(max) with decrease of pH from 7.4 to 5.0, whereas the change in K(m) was negligible. OATP-B-mediated uptake of estrone-3-sulfate was independent of sodium, chloride, bicarbonate, or glutathione, whereas the proton ionophore carbonylcyanide p-trifluoromethoxyphenylhydrazone exhibited a pH-dependent inhibitory effect, suggesting that a proton gradient is a driving force for OATP-B. When OATP-B was expressed in human embryonic kidney 293 cells, uptake activities for anionic compounds showed various kinds of pH sensitivity. Dehydroepiandrosterone-sulfate, estrone-3-sulfate, and fexofenadine were transported by OATP-B at both neutral and acidic pH, whereas estradiol-17beta-glucuronide, acetic acid, and lactic acid were not transported at all. Transport of taurocholic acid and pravastatin by OATP-B was observed only at acidic pH, demonstrating a pH-sensitive substrate specificity of OATP-B. Because the physiological pH close to the surface of intestinal epithelial cells is acidic, the roles of OATP-B in the small intestine might be different from those in other tissues, such as liver basolateral membrane. Although the driving force for OATP-B has not been fully established, the clarification of factors, such as pH, that affect the OATP-B-activity is essential for an understanding of the physiological and pharmacological relevance of the transporter in the small intestine.
Pubmed Id: 14610227
8. Holtzman CW et al. Role of P-glycoprotein in statin drug interactions. Pharmacotherapy. 2006
Authors: Holtzman CW Wiggins BS Spinler SA
Abstract: Understanding the mechanisms of drug interactions with 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) has become increasingly important because of the potential for serious adverse effects, most notably myopathy. Most of the evidence supports the role of cytochrome P450 (CYP) isoenzymes in many of these drug interactions. However, P-glycoprotein (P-gp), an efflux protein located in the gastrointestinal tract, placenta, kidneys, brain, and liver, may also play a role. Results of several studies with in vitro models have shown that lovastatin, simvastatin, and atorvastatin are inhibitors for P-gp and may be substrates for this transporter as well. Pravastatin and fluvastatin consistently demonstrate no significant inhibition of P-gp. Drug interaction studies involving statins and digoxin support a role for P-gp. Many additional drugs such as diltiazem, verapamil, itraconazole, ketoconazole, and cyclosporine, as well as dietary supplements such as St. John's wort and grapefruit juice, interact with statins and are modulators of both CYP3A4 and P-gp. However, the role of P-gp in these specific drug interactions remains unclear.
Pubmed Id: 17064205
9. Mano Y et al. The UDP-glucuronosyltransferase 2B7 isozyme is responsible for gemfibrozil glucuronidation in the human liver. Drug metabolism and disposition: the biological fate of chemicals. 2007
Authors: Mano Y Usui T Kamimura H
Abstract: Gemfibrozil, a fibrate hypolipidemic agent, is eliminated in humans by glucuronidation. A gemfibrozil glucuronide has been reported to show time-dependent inhibition of cytochrome P450 2C8. Comprehensive assessment of the drug interaction between gemfibrozil and cytochrome P450 2C8 substrates requires a clear understanding of gemfibrozil glucuronidation. However, the primary UDP-glucuronosyltransferase (UGT) isozymes responsible for gemfibrozil glucuronidation remain to be determined. Here, we identified the main UGT isozymes involved in gemfibrozil glucuronidation. Evaluation of 12 recombinant human UGT isozymes shows gemfibrozil glucuronidation activity in UGT1A1, UGT1A3, UGT1A9, UGT2B4, UGT2B7, and UGT2B17, with UGT2B7 showing the highest activity. The kinetics of gemfibrozil glucuronidation in pooled human liver microsomes (HLMs) follows Michaelis-Menten kinetics with high and low affinity components. The high affinity K(m) value was 2.5 microM, which is similar to the K(m) value of gemfibrozil glucuronidation in recombinant UGT2B7 (2.2 microM). In 16 HLMs, a significant correlation was observed between gemfibrozil glucuronidation and both morphine 3-OH glucuronidation (r = 0.966, p < 0.0001) and flurbiprofen glucuronidation (r = 0.937, p < 0.0001), two reactions mainly catalyzed by UGT2B7, whereas no significant correlation was observed between gemfibrozil glucuronidation and either estradiol 3beta-glucuronidation and propofol glucuronidation, two reactions catalyzed by UGT1A1 and UGT1A9, respectively. Flurbiprofen and mefenamic acid inhibited gemfibrozil glucuronidation in HLMs with similar IC(50) values to those reported in recombinant UGT2B7. These results suggest that UGT2B7 is the main isozyme responsible for gemfibrozil glucuronidation in humans.
Pubmed Id: 17670842
10. Neuvonen PJ et al. Pharmacokinetic comparison of the potential over-the-counter statins simvastatin, lovastatin, fluvastatin and pravastatin. Clinical pharmacokinetics. 2008
Authors: Neuvonen PJ Backman JT Niemi M
Abstract: HMG-CoA reductase inhibitors (statins) dose-dependently lower both the level of low-density lipoprotein cholesterol and risk of cardiovascular disease. In 2004, the UK approved a low-dose over-the-counter (OTC) simvastatin, but the US has rejected applications for non-prescription preparations of statins. The pharmacokinetics and interaction potentials of the possible OTC candidate statins simvastatin, lovastatin, fluvastatin and pravastatin are clearly different. Simvastatin and lovastatin are mainly metabolized by cytochrome P450 (CYP) 3A, fluvastatin is metabolized by CYP2C9, and pravastatin is excreted largely unchanged. Several cell membrane transporters can influence the disposition of statins, e.g. the organic anion transporting polypeptide (OATP) 1B1 enhances their hepatic uptake. The c.521T>C (p.Val174Ala) genetic polymorphism of SLCO1B1 (encoding OATP1B1) considerably increases the plasma concentrations of simvastatin acid and moderately increases those of pravastatin but seems to have no significant effect on fluvastatin. Strong inhibitors of CYP3A (itraconazole, ritonavir) greatly (up to 20-fold) increase plasma concentrations of simvastatin, lovastatin and their active acid forms, thus enhancing the risk of myotoxicity. Weak or moderately potent CYP3A inhibitors such as verapamil, diltiazem and grapefruit juice can be used cautiously with low doses of simvastatin or lovastatin, but their concomitant use needs medical supervision. Potent inducers of CYP3A can greatly decrease plasma concentrations of simvastatin and simvastatin acid, and probably those of lovastatin and lovastatin acid. Although fluvastatin is metabolized by CYP2C9, its concentrations are changed less than 2-fold by inhibitors or inducers of CYP2C9. Pravastatin plasma concentrations are not significantly affected by any CYP inhibition and only slightly affected by inducers. Ciclosporin inhibits CYP3A, P-glycoprotein and OATP1B1. Gemfibrozil and its glucuronide inhibit CYP2C8 and OATP1B1. Ciclosporin and gemfibrozil increase plasma concentrations of statins and the risk of their myotoxicity, but fluvastatin seems to carry a smaller risk than other statins. Inhibitors of OATP1B1 may decrease the benefit-risk ratio of simvastatin, lovastatin and pravastatin by interfering with their (active acid forms) entry into hepatocytes. Understanding the differences in the pharmacokinetics and interaction potential of various statins helps in their selection for possible non-prescription status. On the pharmacokinetic basis, fluvastatin and pravastatin can be better choices than simvastatin or lovastatin for an OTC statin.
Pubmed Id: 18563955
11. Busse KH et al. Gemfibrozil concentrations are significantly decreased in the presence of lopinavir-ritonavir. Journal of acquired immune deficiency syndromes (1999). 2009
Authors: Busse KH Hadigan C Chairez C Alfaro RM Formentini E Kovacs JA Penzak SR
Abstract: OBJECTIVE: The objective of this study was to determine the influence of a 2-week course of lopinavir-ritonavir on the pharmacokinetics of the triglyceride-lowering agent, gemfibrozil. METHODS: The study was conducted as an open label, single-sequence pharmacokinetic study in healthy human volunteers. Gemfibrozil pharmacokinetic parameter values were compared using a Student t test after a single 600-mg dose was administered to healthy volunteers before and after 2 weeks of lopinavir-ritonavir (400/100 mg) twice daily. RESULTS: Fifteen healthy volunteers (eight males) completed the study. All study drugs were generally well tolerated and no subjects withdrew participation. The geometric mean ratio (90% confidence interval) for gemfibrozil area under the plasma concentration-time curve after 14 days of lopinavir-ritonavir compared with baseline was 0.59 (0.52, 0.67) (P < 0.001). All 15 study subjects experienced a reduction in gemfibrozil area under the plasma concentration-time curve after lopinavir-ritonavir (range, -6% to -74%). The geometric mean ratios for gemfibrozil apparent oral clearance and maximum concentration were 1.69 (1.41, 1.97) and 0.67 (0.49, 0.86) after 14 days of lopinavir-ritonavir versus baseline, respectively (P < 0.0001 and 0.01, respectively). Gemfibrozil elimination half-life did not change after lopinavir-ritonavir administration (P = 0.60). CONCLUSION: Lopinavir-ritonavir significantly reduced the systemic exposure of gemfibrozil by reducing gemfibrozil absorption. Clinicians treating HIV-infected patients with hypertriglyceridemia should be aware of this drug interaction.
Pubmed Id: 19648824
12. Roth M et al. OATPs, OATs and OCTs: the organic anion and cation transporters of the SLCO and SLC22A gene superfamilies. British journal of pharmacology. 2012
Authors: Roth M Obaidat A Hagenbuch B
Abstract: The human organic anion and cation transporters are classified within two SLC superfamilies. Superfamily SLCO (formerly SLC21A) consists of organic anion transporting polypeptides (OATPs), while the organic anion transporters (OATs) and the organic cation transporters (OCTs) are classified in the SLC22A superfamily. Individual members of each superfamily are expressed in essentially every epithelium throughout the body, where they play a significant role in drug absorption, distribution and elimination. Substrates of OATPs are mainly large hydrophobic organic anions, while OATs transport smaller and more hydrophilic organic anions and OCTs transport organic cations. In addition to endogenous substrates, such as steroids, hormones and neurotransmitters, numerous drugs and other xenobiotics are transported by these proteins, including statins, antivirals, antibiotics and anticancer drugs. Expression of OATPs, OATs and OCTs can be regulated at the protein or transcriptional level and appears to vary within each family by both protein and tissue type. All three superfamilies consist of 12 transmembrane domain proteins that have intracellular termini. Although no crystal structures have yet been determined, combinations of homology modelling and mutation experiments have been used to explore the mechanism of substrate recognition and transport. Several polymorphisms identified in members of these superfamilies have been shown to affect pharmacokinetics of their drug substrates, confirming the importance of these drug transporters for efficient pharmacological therapy. This review, unlike other reviews that focus on a single transporter family, briefly summarizes the current knowledge of all the functionally characterized human organic anion and cation drug uptake transporters of the SLCO and the SLC22A superfamilies.
Pubmed Id: 22013971
13. Kudo T et al. Analysis of the repaglinide concentration increase produced by gemfibrozil and itraconazole based on the inhibition of the hepatic uptake transporter and metabolic enzymes. Drug metabolism and disposition: the biological fate of chemicals. 2013
Authors: Kudo T Hisaka A Sugiyama Y Ito K
Abstract: The plasma concentration of repaglinide is reported to increase greatly when given after repeated oral administration of itraconazole and gemfibrozil. The present study analyzed this interaction based on a physiologically based pharmacokinetic (PBPK) model incorporating inhibition of the hepatic uptake transporter and metabolic enzymes involved in repaglinide disposition. Firstly, the plasma concentration profiles of inhibitors (itraconazole, gemfibrozil, and gemfibrozil glucuronide) were reproduced by a PBPK model to obtain their pharmacokinetic parameters. The plasma concentration profiles of repaglinide were then analyzed by a PBPK model, together with those of the inhibitors, assuming a competitive inhibition of CYP3A4 by itraconazole, mechanism-based inhibition of CYP2C8 by gemfibrozil glucuronide, and inhibition of organic anion transporting polypeptide (OATP) 1B1 by gemfibrozil and its glucuronide. The plasma concentration profiles of repaglinide were well reproduced by the PBPK model based on the above assumptions, and the optimized values for the inhibition constants (0.0676 nM for itraconazole against CYP3A4; 14.2 μM for gemfibrozil against OATP1B1; and 5.48 μM for gemfibrozil glucuronide against OATP1B1) and the fraction of repaglinide metabolized by CYP2C8 (0.801) were consistent with the reported values. The validity of the obtained parameters was further confirmed by sensitivity analyses and by reproducing the repaglinide concentration increase produced by concomitant gemfibrozil administration at various timings/doses. The present findings suggested that the reported concentration increase of repaglinide, suggestive of synergistic effects of the coadministered inhibitors, can be quantitatively explained by the simultaneous inhibition of the multiple clearance pathways of repaglinide.
Pubmed Id: 23139378
14. Varma MV et al. Mechanistic modeling to predict the transporter- and enzyme-mediated drug-drug interactions of repaglinide. Pharmaceutical research. 2013
Authors: Varma MV Lai Y Kimoto E Goosen TC El-Kattan AF Kumar V
Abstract: PURPOSE: Quantitative prediction of complex drug-drug interactions (DDIs) is challenging. Repaglinide is mainly metabolized by cytochrome-P-450 (CYP)2C8 and CYP3A4, and is also a substrate of organic anion transporting polypeptide (OATP)1B1. The purpose is to develop a physiologically based pharmacokinetic (PBPK) model to predict the pharmacokinetics and DDIs of repaglinide. METHODS: In vitro hepatic transport of repaglinide, gemfibrozil and gemfibrozil 1-O-β-glucuronide was characterized using sandwich-culture human hepatocytes. A PBPK model, implemented in Simcyp (Sheffield, UK), was developed utilizing in vitro transport and metabolic clearance data. RESULTS: In vitro studies suggested significant active hepatic uptake of repaglinide. Mechanistic model adequately described repaglinide pharmacokinetics, and successfully predicted DDIs with several OATP1B1 and CYP3A4 inhibitors (<10% error). Furthermore, repaglinide-gemfibrozil interaction at therapeutic dose was closely predicted using in vitro fraction metabolism for CYP2C8 (0.71), when primarily considering reversible inhibition of OATP1B1 and mechanism-based inactivation of CYP2C8 by gemfibrozil and gemfibrozil 1-O-β-glucuronide. CONCLUSIONS: This study demonstrated that hepatic uptake is rate-determining in the systemic clearance of repaglinide. The model quantitatively predicted several repaglinide DDIs, including the complex interactions with gemfibrozil. Both OATP1B1 and CYP2C8 inhibition contribute significantly to repaglinide-gemfibrozil interaction, and need to be considered for quantitative rationalization of DDIs with either drug.
Pubmed Id: 23307347
15. Varma MV et al. Quantitative Rationalization of Gemfibrozil Drug Interactions: Consideration of Transporters-Enzyme Interplay and the Role of Circulating Metabolite Gemfibrozil 1-O-β-Glucuronide. Drug metabolism and disposition: the biological fate of chemicals. 2015
Authors: Varma MV Lin J Bi YA Kimoto E Rodrigues AD
Abstract: Gemfibrozil has been suggested as a sensitive cytochrome P450 2C8 (CYP2C8) inhibitor for clinical investigation by the U.S. Food and Drug Administration and the European Medicines Agency. However, gemfibrozil drug-drug interactions (DDIs) are complex; its major circulating metabolite, gemfibrozil 1-O-β-glucuronide (Gem-Glu), exhibits time-dependent inhibition of CYP2C8, and both parent and metabolite also behave as moderate inhibitors of organic anion transporting polypeptide 1B1 (OATP1B1) in vitro. Additionally, parent and metabolite also inhibit renal transport mediated by OAT3. Here, in vitro inhibition data for gemfibrozil and Gem-Glu were used to assess their impact on the pharmacokinetics of several victim drugs (including rosiglitazone, pioglitazone, cerivastatin, and repaglinide) by employing both static mechanistic and dynamic physiologically based pharmacokinetic (PBPK) models. Of the 48 cases evaluated using the static models, about 75% and 98% of the DDIs were predicted within 1.5- and 2-fold of the observed values, respectively, when incorporating the interaction potential of both gemfibrozil and its 1-O-β-glucuronide. Moreover, the PBPK model was able to recover the plasma profiles of rosiglitazone, pioglitazone, cerivastatin, and repaglinide under control and gemfibrozil treatment conditions. Analyses suggest that Gem-Glu is the major contributor to the DDIs, and its exposure needed to bring about complete inactivation of CYP2C8 is only a fraction of that achieved in the clinic after a therapeutic gemfibrozil dose. Overall, the complex interactions of gemfibrozil can be quantitatively rationalized, and the learnings from this analysis can be applied in support of future predictions of gemfibrozil DDIs.
Pubmed Id: 25941268
16. Ivanyuk A et al. Renal Drug Transporters and Drug Interactions. Clinical pharmacokinetics. 2017
Authors: Ivanyuk A Livio F Biollaz J Buclin T
Abstract: Transporters in proximal renal tubules contribute to the disposition of numerous drugs. Furthermore, the molecular mechanisms of tubular secretion have been progressively elucidated during the past decades. Organic anions tend to be secreted by the transport proteins OAT1, OAT3 and OATP4C1 on the basolateral side of tubular cells, and multidrug resistance protein (MRP) 2, MRP4, OATP1A2 and breast cancer resistance protein (BCRP) on the apical side. Organic cations are secreted by organic cation transporter (OCT) 2 on the basolateral side, and multidrug and toxic compound extrusion (MATE) proteins MATE1, MATE2/2-K, P-glycoprotein, organic cation and carnitine transporter (OCTN) 1 and OCTN2 on the apical side. Significant drug-drug interactions (DDIs) may affect any of these transporters, altering the clearance and, consequently, the efficacy and/or toxicity of substrate drugs. Interactions at the level of basolateral transporters typically decrease the clearance of the victim drug, causing higher systemic exposure. Interactions at the apical level can also lower drug clearance, but may be associated with higher renal toxicity, due to intracellular accumulation. Whereas the importance of glomerular filtration in drug disposition is largely appreciated among clinicians, DDIs involving renal transporters are less well recognized. This review summarizes current knowledge on the roles, quantitative importance and clinical relevance of these transporters in drug therapy. It proposes an approach based on substrate-inhibitor associations for predicting potential tubular-based DDIs and preventing their adverse consequences. We provide a comprehensive list of known drug interactions with renally-expressed transporters. While many of these interactions have limited clinical consequences, some involving high-risk drugs (e.g. methotrexate) definitely deserve the attention of prescribers.
Pubmed Id: 28210973
17. Etchegoyen CV et al. Drug-induced QT Interval Prolongation in the Intensive Care Unit. Current clinical pharmacology. 2017
Authors: Etchegoyen CV Keller GA Mrad S Cheng S Di Girolamo G
Abstract: BACKGROUND: The most common acquired cause of Long QT syndrome (LQTS) is drug induced QT interval prolongation. It is an electrophysiological entity, which is characterized by an extended duration of the ventricular repolarization. Reflected as a prolonged QT interval in a surface ECG, this syndrome increases the risk for polymorphic ventricular tachycardia (Torsade de Pointes) and sudden death. METHOD: Bibliographic databases as MEDLINE and EMBASE, reports and drug alerts from several regulatory agencies (FDA, EMEA, ANMAT) and drug safety guides (ICH S7B, ICH E14) were consulted to prepare this article. The keywords used were: polymorphic ventricular tachycardia, adverse drug events, prolonged QT, arrhythmias, intensive care unit and Torsade de Pointes. Such research involved materials produced up to December 2017. RESULTS: Because of their mechanism of action, antiarrhythmic drugs such as amiodarone, sotalol, quinidine, procainamide, verapamil and diltiazem are associated to the prolongation of the QTc interval. For this reason, they require constant monitoring when administered. Other noncardiovascular drugs that are widely used in the Intensive Care Unit (ICU), such as ondansetron, macrolide and fluoroquinolone antibiotics, typical and atypical antipsychotics agents such as haloperidol, thioridazine, and sertindole are also frequently associated with the prolongation of the QTc interval. As a consequence, critical patients should be closely followed and evaluated. CONCLUSION: ICU patients are particularly prone to experience a QTc interval prolongation mainly for two reasons. In the first place, they are exposed to certain drugs that can prolong the repolarization phase, either by their mechanism of action or through the interaction with other drugs. In the second place, the risk factors for TdP are prevalent clinical conditions among critically ill patients. As a consequence, the attending physician is expected to perform preventive monitoring and ECG checks to control the QTc interval.
Pubmed Id: 29473523
18. Yau S et al. Short-term use of oral amiodarone causing torsades de pointes. Clinical case reports. 2018
Authors: Yau S Chan P Sapkin J Hsieh E
Abstract: Amiodarone is one of the most commonly used antiarrhythmic drugs. Despite its well-known side effects, amiodarone is considered to be a relatively safe drug, especially in short-term usage to prevent life-threatening ventricular arrhythmias. Our case demonstrates an instance where short-term usage can yield drug side effect.
Pubmed Id: 30147903

epha.ch AG

Stadelhoferstrasse 40 8001 Zürich Schweiz
Github LinkedIn Facebook Twitter
kontakt@epha.ch

Allgemeines

Impressum Datenschutz Nutzungsbedingungen Über uns API

Sprachen

Deutsch English Français Italiano Español
Copyright © 2020 epha.ch - Alle Rechte vorbehalten