Skip to main content
Erschienen in: Clinical Pharmacokinetics 10/2021

18.05.2021 | Original Research Article

Effect of Liver Cirrhosis on the Pharmacokinetics, Metabolism, and Tolerability of Daridorexant, A Novel Dual Orexin Receptor Antagonist

verfasst von: Benjamin Berger, Jasper Dingemanse, Giancarlo Sabattini, Stéphane Delahaye, Urs Duthaler, Clemens Muehlan, Stephan Krähenbühl

Erschienen in: Clinical Pharmacokinetics | Ausgabe 10/2021

Einloggen, um Zugang zu erhalten

Abstract

Background and Objective

Daridorexant is a dual orexin receptor antagonist in clinical development for insomnia. As daridorexant is cleared mainly via cytochrome P450 (CYP) 3A4, the effect of hepatic impairment on the pharmacokinetics (PK), metabolism, and tolerability of daridorexant was evaluated. Sleep disorders are common in patients with liver cirrhosis and, therefore, sleep-promoting drugs with a better tolerability than currently available would be preferable, a premise that dual orexin receptor antagonists may fulfill.

Methods

This was a single-dose, open-label, phase I study. Subjects with mild (Child–Pugh A, N = 8) or moderate (Child–Pugh B, N = 8) liver cirrhosis and matched healthy control subjects (N = 8) received 25 mg of daridorexant orally. Blood samples were collected for 72 h post-dose for PK assessments of daridorexant and three major metabolites.

Results

Compared with healthy subjects, patients showed a decrease in total daridorexant area under the plasma concentration–time curve from zero to infinity (AUC0-inf) and maximum plasma concentration with a geometric mean ratio (GMR, 90% confidence interval [CI]) of 0.51 (0.28–0.92) and 0.50 (0.35–0.72) in Child–Pugh A and 0.74 (0.39–1.41) and 0.42 (0.29–0.60) in Child–Pugh B patients, respectively. Furthermore, the median time to reach maximum plasma concentration was slightly delayed (1.0 h [90% CI 0.0–2.0] in Child–Pugh A patients and 0.5 h [90% CI 0.0–1.5] in Child–Pugh B patients), while for Child–Pugh B patients, a doubling in half-life was observed (GMR [90% CI]: 2.09 [1.32–3.30]). Considering the high plasma protein binding (> 99%) and a 1.9-fold to 2.3-fold increase in the unbound fraction in patients, the PK of unbound daridorexant was also assessed. Compared with healthy subjects, Child–Pugh B patients had a higher AUC0-inf (GMR [90% CI] 1.60 [0.93–2.73]), a lower apparent plasma clearance (GMR [90% CI] 0.63 [0.37–1.07]), and the same doubling in the half-life observed for total daridorexant, whereas maximum plasma concentration and apparent volume of distribution were not different. Unbound daridorexant PK in Child–Pugh A patients did not differ from healthy subjects. In addition, the metabolic ratios (parent to metabolite), i.e., a marker of CYP 3A4 activity, of the two most abundant daridorexant metabolites were higher in patients with liver cirrhosis compared with healthy subjects. All treatment-emergent adverse events were transient and of mild or moderate intensity and no other treatment-related effects were apparent.

Conclusions

No safety issue of concern was detected following administration of 25 mg of daridorexant in the study population. Moderate liver cirrhosis causes impaired hepatic clearance of unbound daridorexant, which prolongs the half-life. A 25-mg dose of daridorexant should, therefore, not be exceeded in Child–Pugh B patients. A dose adjustment is not required in Child–Pugh A patients, while avoidance of daridorexant in patients with Child–Pugh C cirrhosis is recommended.

Clinical Trial Registration

ClinicalTrials.gov ID: NCT03713242.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka H, et al. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell. 1998;92(5):573–85.PubMedCrossRef Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka H, et al. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell. 1998;92(5):573–85.PubMedCrossRef
2.
Zurück zum Zitat Sakurai T. The neural circuit of orexin (hypocretin): maintaining sleep and wakefulness. Nat Rev Neurosci. 2007;8(3):171–81.PubMedCrossRef Sakurai T. The neural circuit of orexin (hypocretin): maintaining sleep and wakefulness. Nat Rev Neurosci. 2007;8(3):171–81.PubMedCrossRef
3.
Zurück zum Zitat Treiber A, de Kanter R, Roch C, Gatfield J, Boss C, von Raumer M, et al. The use of physiology-based pharmacokinetic and pharmacodynamic modeling in the discovery of the dual orexin receptor antagonist ACT-541468. J Pharmacol Exp Ther. 2017;362(3):489–503.PubMedCrossRef Treiber A, de Kanter R, Roch C, Gatfield J, Boss C, von Raumer M, et al. The use of physiology-based pharmacokinetic and pharmacodynamic modeling in the discovery of the dual orexin receptor antagonist ACT-541468. J Pharmacol Exp Ther. 2017;362(3):489–503.PubMedCrossRef
4.
Zurück zum Zitat Wang C, Wang Q, Ji B, Pan Y, Xu C, Cheng B, et al. The orexin/receptor system: molecular mechanism and therapeutic potential for neurological diseases. Front Mol Neurosci. 2018;11:220.PubMedPubMedCentralCrossRef Wang C, Wang Q, Ji B, Pan Y, Xu C, Cheng B, et al. The orexin/receptor system: molecular mechanism and therapeutic potential for neurological diseases. Front Mol Neurosci. 2018;11:220.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Muehlan C, Vaillant C, Zenklusen I, Kraehenbuehl S, Dingemanse J. Clinical pharmacology, efficacy, and safety of orexin receptor antagonists for the treatment of insomnia disorders. Expert Opin Drug Metab Toxicol. 2020;16(11):1063–78.PubMedCrossRef Muehlan C, Vaillant C, Zenklusen I, Kraehenbuehl S, Dingemanse J. Clinical pharmacology, efficacy, and safety of orexin receptor antagonists for the treatment of insomnia disorders. Expert Opin Drug Metab Toxicol. 2020;16(11):1063–78.PubMedCrossRef
6.
Zurück zum Zitat Mieda M, Sakurai T. Orexin (hypocretin) receptor agonists and antagonists for treatment of sleep disorders: rationale for development and current status. CNS Drugs. 2013;27(2):83–90.PubMedCrossRef Mieda M, Sakurai T. Orexin (hypocretin) receptor agonists and antagonists for treatment of sleep disorders: rationale for development and current status. CNS Drugs. 2013;27(2):83–90.PubMedCrossRef
7.
Zurück zum Zitat Brisbare-Roch C, Dingemanse J, Koberstein R, Hoever P, Aissaoui H, Flores S, et al. Promotion of sleep by targeting the orexin system in rats, dogs and humans. Nat Med. 2007;13(2):150–5.PubMedCrossRef Brisbare-Roch C, Dingemanse J, Koberstein R, Hoever P, Aissaoui H, Flores S, et al. Promotion of sleep by targeting the orexin system in rats, dogs and humans. Nat Med. 2007;13(2):150–5.PubMedCrossRef
8.
Zurück zum Zitat Hoever P, Dorffner G, Beneš H, Penzel T, Danker-Hopfe H, Barbanoj MJ, et al. Orexin receptor antagonism, a new sleep-enabling paradigm: a proof-of-concept clinical trial. Clin Pharmacol Ther. 2012;91(6):975–85.PubMedPubMedCentralCrossRef Hoever P, Dorffner G, Beneš H, Penzel T, Danker-Hopfe H, Barbanoj MJ, et al. Orexin receptor antagonism, a new sleep-enabling paradigm: a proof-of-concept clinical trial. Clin Pharmacol Ther. 2012;91(6):975–85.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Dauvilliers Y, Zammit G, Fietze I, Mayleben D, Seboek Kinter D, Pain S, et al. Daridorexant, a new dual orexin receptor antagonist to treat insomnia disorder. Ann Neurol. 2020;87(3):347–56.PubMedCrossRef Dauvilliers Y, Zammit G, Fietze I, Mayleben D, Seboek Kinter D, Pain S, et al. Daridorexant, a new dual orexin receptor antagonist to treat insomnia disorder. Ann Neurol. 2020;87(3):347–56.PubMedCrossRef
10.
Zurück zum Zitat Muehlan C, Brooks S, Zuiker R, van Gerven J, Dingemanse J. Multiple-dose clinical pharmacology of ACT-541468, a novel dual orexin receptor antagonist, following repeated-dose morning and evening administration. Eur Neuropsychopharmacol. 2019;29(7):847–57.PubMedCrossRef Muehlan C, Brooks S, Zuiker R, van Gerven J, Dingemanse J. Multiple-dose clinical pharmacology of ACT-541468, a novel dual orexin receptor antagonist, following repeated-dose morning and evening administration. Eur Neuropsychopharmacol. 2019;29(7):847–57.PubMedCrossRef
11.
Zurück zum Zitat Zammit G, Dauvilliers Y, Pain S, Sebök Kinter D, Mansour Y, Kunz D. Daridorexant, a new dual orexin receptor antagonist, in elderly subjects with insomnia disorder. Neurology. 2020;94(21):e2222–32.PubMedCrossRef Zammit G, Dauvilliers Y, Pain S, Sebök Kinter D, Mansour Y, Kunz D. Daridorexant, a new dual orexin receptor antagonist, in elderly subjects with insomnia disorder. Neurology. 2020;94(21):e2222–32.PubMedCrossRef
12.
Zurück zum Zitat Boof ML, Alatrach A, Ufer M, Dingemanse J. Interaction potential of the dual orexin receptor antagonist ACT-541468 with CYP3A4 and food: results from two interaction studies. Eur J Clin Pharmacol. 2019;75(2):195–205.PubMedCrossRef Boof ML, Alatrach A, Ufer M, Dingemanse J. Interaction potential of the dual orexin receptor antagonist ACT-541468 with CYP3A4 and food: results from two interaction studies. Eur J Clin Pharmacol. 2019;75(2):195–205.PubMedCrossRef
13.
Zurück zum Zitat Zenklusen I, Muehlan C, Ulc I, Liska J, Dingemanse J. The dual orexin receptor antagonist daridorexant does not affect the pharmacokinetics of the BCRP substrate rosuvastatin. Clin Exp Pharmacol Physiol. 2020;47(11):1843–9.PubMedCrossRef Zenklusen I, Muehlan C, Ulc I, Liska J, Dingemanse J. The dual orexin receptor antagonist daridorexant does not affect the pharmacokinetics of the BCRP substrate rosuvastatin. Clin Exp Pharmacol Physiol. 2020;47(11):1843–9.PubMedCrossRef
14.
Zurück zum Zitat Muehlan C, Zuiker R, Peeters P, Rowles R, Dingemanse J. Pharmacokinetics and pharmacodynamics of the dual orexin receptor antagonist daridorexant in Japanese and Caucasian subjects. J Clin Psychopharmacol. 2020;40(2):157–66.PubMedCrossRef Muehlan C, Zuiker R, Peeters P, Rowles R, Dingemanse J. Pharmacokinetics and pharmacodynamics of the dual orexin receptor antagonist daridorexant in Japanese and Caucasian subjects. J Clin Psychopharmacol. 2020;40(2):157–66.PubMedCrossRef
15.
Zurück zum Zitat Muehlan C, Heuberger J, Juif PE, Croft M, van Gerven J, Dingemanse J. Accelerated development of the dual orexin receptor antagonist ACT-541468: integration of a microtracer in a first-in-human study. Clin Pharmacol Ther. 2018;104(5):1022–9.PubMedCrossRef Muehlan C, Heuberger J, Juif PE, Croft M, van Gerven J, Dingemanse J. Accelerated development of the dual orexin receptor antagonist ACT-541468: integration of a microtracer in a first-in-human study. Clin Pharmacol Ther. 2018;104(5):1022–9.PubMedCrossRef
16.
Zurück zum Zitat Muehlan C, Boehler M, Brooks S, Zuiker R, van Gerven J, Dingemanse J. Clinical pharmacology of the dual orexin receptor antagonist ACT-541468 in elderly subjects: exploration of pharmacokinetics, pharmacodynamics and tolerability following single-dose morning and repeated-dose evening administration. J Psychopharmacol. 2020;34(3):326–35.PubMedCrossRef Muehlan C, Boehler M, Brooks S, Zuiker R, van Gerven J, Dingemanse J. Clinical pharmacology of the dual orexin receptor antagonist ACT-541468 in elderly subjects: exploration of pharmacokinetics, pharmacodynamics and tolerability following single-dose morning and repeated-dose evening administration. J Psychopharmacol. 2020;34(3):326–35.PubMedCrossRef
17.
Zurück zum Zitat Muehlan C, Fischer H, Zimmer D, Aissaoui H, Grimont J, Boss C, et al. Metabolism of the dual orexin receptor antagonist ACT-541468, based on microtracer/accelerator mass spectrometry. Curr Drug Metab. 2019;20(4):254–65.PubMedCrossRef Muehlan C, Fischer H, Zimmer D, Aissaoui H, Grimont J, Boss C, et al. Metabolism of the dual orexin receptor antagonist ACT-541468, based on microtracer/accelerator mass spectrometry. Curr Drug Metab. 2019;20(4):254–65.PubMedCrossRef
18.
Zurück zum Zitat Tapper EB, Henderson JB, Parikh ND, Ioannou GN, Lok AS. Incidence of and risk factors for hepatic encephalopathy in a population-based cohort of Americans with cirrhosis. Hepatol Commun. 2019;3(11):1510–9.PubMedPubMedCentralCrossRef Tapper EB, Henderson JB, Parikh ND, Ioannou GN, Lok AS. Incidence of and risk factors for hepatic encephalopathy in a population-based cohort of Americans with cirrhosis. Hepatol Commun. 2019;3(11):1510–9.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Córdoba J, Cabrera J, Lataif L, Penev P, Zee P, Blei AT. High prevalence of sleep disturbance in cirrhosis. Hepatology. 1998;27(2):339–45.PubMedCrossRef Córdoba J, Cabrera J, Lataif L, Penev P, Zee P, Blei AT. High prevalence of sleep disturbance in cirrhosis. Hepatology. 1998;27(2):339–45.PubMedCrossRef
20.
Zurück zum Zitat Martin Mateos R, Garcia de la Filia Molina I, Albillos A. Pre-surgical risk assessment in patients with cirrhosis. Acta Gastroenterol Belg. 2020;83(3):449–53.PubMed Martin Mateos R, Garcia de la Filia Molina I, Albillos A. Pre-surgical risk assessment in patients with cirrhosis. Acta Gastroenterol Belg. 2020;83(3):449–53.PubMed
21.
Zurück zum Zitat Pugh RN, Murray-Lyon IM, Dawson JL, Pietroni MC, Williams R. Transection of the oesophagus for bleeding oesophageal varices. Br J Surg. 1973;60(8):646–9.PubMedCrossRef Pugh RN, Murray-Lyon IM, Dawson JL, Pietroni MC, Williams R. Transection of the oesophagus for bleeding oesophageal varices. Br J Surg. 1973;60(8):646–9.PubMedCrossRef
22.
Zurück zum Zitat Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16(1):31–41.PubMedCrossRef Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16(1):31–41.PubMedCrossRef
23.
Zurück zum Zitat Donzelli M, Derungs A, Serratore MG, Noppen C, Nezic L, Krahenbuhl S, et al. The basel cocktail for simultaneous phenotyping of human cytochrome P450 isoforms in plasma, saliva and dried blood spots. Clin Pharmacokinet. 2014;53(3):271–82.PubMedCrossRef Donzelli M, Derungs A, Serratore MG, Noppen C, Nezic L, Krahenbuhl S, et al. The basel cocktail for simultaneous phenotyping of human cytochrome P450 isoforms in plasma, saliva and dried blood spots. Clin Pharmacokinet. 2014;53(3):271–82.PubMedCrossRef
24.
Zurück zum Zitat Åkerstedt T, Gillberg M. Subjective and objective sleepiness in the active individual. Int J Neurosci. 1990;52(1–2):29–37.PubMedCrossRef Åkerstedt T, Gillberg M. Subjective and objective sleepiness in the active individual. Int J Neurosci. 1990;52(1–2):29–37.PubMedCrossRef
25.
Zurück zum Zitat Boss C, Gatfield J, Brotschi C, Heidmann B, Sifferlen T, von Raumer M, et al. The quest for the best dual orexin receptor antagonist (daridorexant) for the treatment of insomnia disorders. Chem Med Chem. 2020;15(23):2286–305.PubMedCrossRef Boss C, Gatfield J, Brotschi C, Heidmann B, Sifferlen T, von Raumer M, et al. The quest for the best dual orexin receptor antagonist (daridorexant) for the treatment of insomnia disorders. Chem Med Chem. 2020;15(23):2286–305.PubMedCrossRef
26.
Zurück zum Zitat Delco F, Tchambaz L, Schlienger R, Drewe J, Krahenbuhl S. Dose adjustment in patients with liver disease. Drug Saf. 2005;28(6):529–45.PubMedCrossRef Delco F, Tchambaz L, Schlienger R, Drewe J, Krahenbuhl S. Dose adjustment in patients with liver disease. Drug Saf. 2005;28(6):529–45.PubMedCrossRef
27.
Zurück zum Zitat Quigley EM. Gastrointestinal dysfunction in liver disease and portal hypertension: gut-liver interactions revisited. Dig Dis Sci. 1996;41(3):557–61.PubMedCrossRef Quigley EM. Gastrointestinal dysfunction in liver disease and portal hypertension: gut-liver interactions revisited. Dig Dis Sci. 1996;41(3):557–61.PubMedCrossRef
28.
Zurück zum Zitat Sadik R, Abrahamsson H, Björnsson E, Gunnarsdottir A, Stotzer PO. Etiology of portal hypertension may influence gastrointestinal transit. Scand J Gastroenterol. 2003;38(10):1039–44.PubMedCrossRef Sadik R, Abrahamsson H, Björnsson E, Gunnarsdottir A, Stotzer PO. Etiology of portal hypertension may influence gastrointestinal transit. Scand J Gastroenterol. 2003;38(10):1039–44.PubMedCrossRef
29.
Zurück zum Zitat Thalheimer U, De Iorio F, Capra F, del Mar LM, Zuliani V, Ghidini V, et al. Altered intestinal function precedes the appearance of bacterial DNA in serum and ascites in patients with cirrhosis: a pilot study. Eur J Gastroenterol Hepatol. 2010;22(10):1228–34.PubMedCrossRef Thalheimer U, De Iorio F, Capra F, del Mar LM, Zuliani V, Ghidini V, et al. Altered intestinal function precedes the appearance of bacterial DNA in serum and ascites in patients with cirrhosis: a pilot study. Eur J Gastroenterol Hepatol. 2010;22(10):1228–34.PubMedCrossRef
30.
Zurück zum Zitat Isobe H, Sakai H, Satoh M, Sakamoto S, Nawata H. Delayed gastric emptying in patients with liver cirrhosis. Dig Dis Sci. 1994;39(5):983–7.PubMedCrossRef Isobe H, Sakai H, Satoh M, Sakamoto S, Nawata H. Delayed gastric emptying in patients with liver cirrhosis. Dig Dis Sci. 1994;39(5):983–7.PubMedCrossRef
31.
Zurück zum Zitat Pimpo MT, Frieri G, Saltarelli P, Ciccocioppo R, Aggio A, Marchetti G, et al. Effects of cisapride on abnormally prolonged endogastric alkalinity time and delayed gastric emptying in cirrhotic patients. Hepatogastroenterology. 1996;43(12):1678–84.PubMed Pimpo MT, Frieri G, Saltarelli P, Ciccocioppo R, Aggio A, Marchetti G, et al. Effects of cisapride on abnormally prolonged endogastric alkalinity time and delayed gastric emptying in cirrhotic patients. Hepatogastroenterology. 1996;43(12):1678–84.PubMed
32.
Zurück zum Zitat Usami A, Mizukami Y, Onji M. Abnormal gastric motility in liver cirrhosis: roles of secretin. Dig Dis Sci. 1998;43(11):2392–7.PubMedCrossRef Usami A, Mizukami Y, Onji M. Abnormal gastric motility in liver cirrhosis: roles of secretin. Dig Dis Sci. 1998;43(11):2392–7.PubMedCrossRef
33.
Zurück zum Zitat Taegtmeyer AB, Haschke M, Tchambaz L, Buylaert M, Tschopl M, Beuers U, et al. A study of the relationship between serum bile acids and propranolol pharmacokinetics and pharmacodynamics in patients with liver cirrhosis and in healthy controls. PLoS ONE. 2014;9(6):e97885.PubMedPubMedCentralCrossRef Taegtmeyer AB, Haschke M, Tchambaz L, Buylaert M, Tschopl M, Beuers U, et al. A study of the relationship between serum bile acids and propranolol pharmacokinetics and pharmacodynamics in patients with liver cirrhosis and in healthy controls. PLoS ONE. 2014;9(6):e97885.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Albarmawi A, Czock D, Gauss A, Ehehalt R, Lorenzo Bermejo J, Burhenne J, et al. CYP3A activity in severe liver cirrhosis correlates with Child-Pugh and model for end-stage liver disease (MELD) scores. Br J Clin Pharmacol. 2014;77(1):160–9.PubMedCrossRef Albarmawi A, Czock D, Gauss A, Ehehalt R, Lorenzo Bermejo J, Burhenne J, et al. CYP3A activity in severe liver cirrhosis correlates with Child-Pugh and model for end-stage liver disease (MELD) scores. Br J Clin Pharmacol. 2014;77(1):160–9.PubMedCrossRef
35.
36.
Zurück zum Zitat Ceryak S, Bouscarel B, Fromm H. Comparative binding of bile acids to serum lipoproteins and albumin. J Lipid Res. 1993;34(10):1661–74.PubMedCrossRef Ceryak S, Bouscarel B, Fromm H. Comparative binding of bile acids to serum lipoproteins and albumin. J Lipid Res. 1993;34(10):1661–74.PubMedCrossRef
37.
Zurück zum Zitat Verbeeck RK. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 2008;64(12):1147–61.PubMedCrossRef Verbeeck RK. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 2008;64(12):1147–61.PubMedCrossRef
38.
Zurück zum Zitat Barre J, Houin G, Rosenbaum J, Zini R, Dhumeaux D, Tillement JP. Decreased alpha 1-acid glycoprotein in liver cirrhosis: consequences for drug protein binding. Br J Clin Pharmacol. 1984;18(4):652–3.PubMedPubMedCentralCrossRef Barre J, Houin G, Rosenbaum J, Zini R, Dhumeaux D, Tillement JP. Decreased alpha 1-acid glycoprotein in liver cirrhosis: consequences for drug protein binding. Br J Clin Pharmacol. 1984;18(4):652–3.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Barre J, Mallat A, Rosenbaum J, Deforges L, Houin G, Dhumeaux D, et al. Pharmacokinetics of erythromycin in patients with severe cirrhosis: respective influence of decreased serum binding and impaired liver metabolic capacity. Br J Clin Pharmacol. 1987;23(6):753–7.PubMedPubMedCentralCrossRef Barre J, Mallat A, Rosenbaum J, Deforges L, Houin G, Dhumeaux D, et al. Pharmacokinetics of erythromycin in patients with severe cirrhosis: respective influence of decreased serum binding and impaired liver metabolic capacity. Br J Clin Pharmacol. 1987;23(6):753–7.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Knott C, Williams CP, Reynolds F. Phenytoin kinetics during pregnancy and the puerperium. Br J Obstet Gynaecol. 1986;93(10):1030–7.PubMedCrossRef Knott C, Williams CP, Reynolds F. Phenytoin kinetics during pregnancy and the puerperium. Br J Obstet Gynaecol. 1986;93(10):1030–7.PubMedCrossRef
41.
Zurück zum Zitat Roberts JA, Pea F, Lipman J. The clinical relevance of plasma protein binding changes. Clin Pharmacokinet. 2013;52(1):1–8.PubMedCrossRef Roberts JA, Pea F, Lipman J. The clinical relevance of plasma protein binding changes. Clin Pharmacokinet. 2013;52(1):1–8.PubMedCrossRef
42.
Zurück zum Zitat Klotz U, Avant GR, Hoyumpa A, Schenker S, Wilkinson GR. The effects of age and liver disease on the disposition and elimination of diazepam in adult man. J Clin Invest. 1975;55(2):347–59.PubMedPubMedCentralCrossRef Klotz U, Avant GR, Hoyumpa A, Schenker S, Wilkinson GR. The effects of age and liver disease on the disposition and elimination of diazepam in adult man. J Clin Invest. 1975;55(2):347–59.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat George J, Murray M, Byth K, Farrell GC. Differential alterations of cytochrome P450 proteins in livers from patients with severe chronic liver disease. Hepatology. 1995;21(1):120–8.PubMed George J, Murray M, Byth K, Farrell GC. Differential alterations of cytochrome P450 proteins in livers from patients with severe chronic liver disease. Hepatology. 1995;21(1):120–8.PubMed
44.
Zurück zum Zitat Prasad B, Bhatt DK, Johnson K, Chapa R, Chu X, Salphati L, et al. Abundance of phase 1 and 2 drug-metabolizing enzymes in alcoholic and hepatitis C cirrhotic livers: a quantitative targeted proteomics study. Drug Metab Dispos. 2018;46(7):943–52.PubMedPubMedCentralCrossRef Prasad B, Bhatt DK, Johnson K, Chapa R, Chu X, Salphati L, et al. Abundance of phase 1 and 2 drug-metabolizing enzymes in alcoholic and hepatitis C cirrhotic livers: a quantitative targeted proteomics study. Drug Metab Dispos. 2018;46(7):943–52.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Yang LQ, Li SJ, Cao YF, Man XB, Yu WF, Wang HY, et al. Different alterations of cytochrome P450 3A4 isoform and its gene expression in livers of patients with chronic liver diseases. World J Gastroenterol. 2003;9(2):359–63.PubMedPubMedCentralCrossRef Yang LQ, Li SJ, Cao YF, Man XB, Yu WF, Wang HY, et al. Different alterations of cytochrome P450 3A4 isoform and its gene expression in livers of patients with chronic liver diseases. World J Gastroenterol. 2003;9(2):359–63.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat MacGilchrist AJ, Birnie GG, Cook A, Scobie G, Murray T, Watkinson G, et al. Pharmacokinetics and pharmacodynamics of intravenous midazolam in patients with severe alcoholic cirrhosis. Gut. 1986;27(2):190–5.PubMedPubMedCentralCrossRef MacGilchrist AJ, Birnie GG, Cook A, Scobie G, Murray T, Watkinson G, et al. Pharmacokinetics and pharmacodynamics of intravenous midazolam in patients with severe alcoholic cirrhosis. Gut. 1986;27(2):190–5.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat McConnell JB, Curry SH, Davis M, Williams R. Clinical effects and metabolism of diazepam in patients with chronic liver disease. Clin Sci (Lond). 1982;63(1):75–80.PubMedCrossRef McConnell JB, Curry SH, Davis M, Williams R. Clinical effects and metabolism of diazepam in patients with chronic liver disease. Clin Sci (Lond). 1982;63(1):75–80.PubMedCrossRef
Metadaten
Titel
Effect of Liver Cirrhosis on the Pharmacokinetics, Metabolism, and Tolerability of Daridorexant, A Novel Dual Orexin Receptor Antagonist
verfasst von
Benjamin Berger
Jasper Dingemanse
Giancarlo Sabattini
Stéphane Delahaye
Urs Duthaler
Clemens Muehlan
Stephan Krähenbühl
Publikationsdatum
18.05.2021
Verlag
Springer International Publishing
Erschienen in
Clinical Pharmacokinetics / Ausgabe 10/2021
Print ISSN: 0312-5963
Elektronische ISSN: 1179-1926
DOI
https://doi.org/10.1007/s40262-021-01028-8

Weitere Artikel der Ausgabe 10/2021

Clinical Pharmacokinetics 10/2021 Zur Ausgabe