12,570
Views
17
CrossRef citations to date
0
Altmetric
Review

Safe use of medication in patients with cirrhosis: pharmacokinetic and pharmacodynamic considerations

ORCID Icon, , , , &
Pages 45-57 | Received 14 Sep 2019, Accepted 04 Dec 2019, Published online: 11 Dec 2019

References

  • Asrani SK, Kouznetsova M, Ogola G, et al. Increasing health care burden of chronic liver disease compared with other chronic diseases, 2004–2013. Gastroenterology. 2018;155(3):719–729. e4.
  • Mokdad AA, Lopez AD, Shahraz S, et al. Liver cirrhosis mortality in 187 countries between 1980 and 2010: a systematic analysis. BMC Med. 2014;12:y.
  • Asrani SK, Devarbhavi H, Eaton J, et al. Burden of liver diseases in the world. J Hepatol. 2018;70(1):151–171.
  • Blachier M, Leleu H, Peck-Radosavljevic M, et al. The burden of liver disease in Europe: a review of available epidemiological data. J Hepatol. 2013;58(3):593–608.
  • Tsochatzis EA, Bosch J, Burroughs AK. Liver cirrhosis. Lancet. 2014;383(9930):1749–1761.
  • Ge PS, Runyon BA, Campion EW. Treatment of patients with cirrhosis. N Engl J Med. 2016;375(8):767–777.
  • Poordad FF. Presentation and complications associated with cirrhosis of the liver. Curr Med Res Opin. 2015;31(5):925–937.
  • Franz C, Egger S, Born C, et al. Potential drug-drug interactions and adverse drug reactions in patients with liver cirrhosis. Eur J Clin Pharmacol. 2012;68(2):179–188.
  • Franz C, Hildbrand C, Born C, et al. Dose adjustment in patients with liver cirrhosis: impact on adverse drug reactions and hospitalizations. Eur J Clin Pharmacol. 2013;69(8):1565–1573.
  • Johnson TN, Thomson AH. Pharmacokinetics of drugs in liver disease. In: North-Lewis P, editor. Drugs and the liver: a guide to drug handling in liver dysfunction. London: Pharmaceutical Press; 2008. p. 103–133.
  • Verbeeck RK. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 2008;64(12):1147–1161.
  • Morgan DJ, McLean AJ. Clinical pharmacokinetic and pharmacodynamic considerations in patients with liver disease. Clin Pharmacokinet. 1995;29(5):370–391.
  • Delco F, Tchambaz L, Schlienger R, et al. Dose adjustment in patients with liver disease. Drug Saf. 2005;28(6):529–545.
  • Breimer D. Pharmacokinetics in liver disease. Pharm Weekbl [Sci]. 1987;9(2):79–84.
  • Bertolotti M, Ferrari A, Vitale G, et al. Effect of liver cirrhosis on the systemic availability of naltrexone in humans. J Hepatol. 1997;27(3):505–511.
  • Weersink RA, Bouma M, Burger DM, et al. Safe use of proton pump inhibitors in patients with cirrhosis. Br J Clin Pharmacol. 2018;84(8):1806–1820.
  • Dalhoff K, Almdal TP, Bjerrum K, et al. Pharmacokinetics of paroxetine in patients with cirrhosis. Eur J Clin Pharmacol. 1991;41(4):351–354.
  • Gonzalez M, Goracci L, Cruciani G, et al. Some considerations on the predictions of pharmacokinetic alterations in subjects with liver disease. Expert Opin Drug Metab Toxicol. 2014;10(10):1397–1408.
  • Cotting J, Reichen J, Kutz K, et al. Pharmacokinetics of isradipine in patients with chronic liver disease. Eur J Clin Pharmacol. 1990;38(6):599–603.
  • Kleinbloesem C, Van Harten J, Wilson J, et al. Nifedipine: kinetics and hemodynamic effects in patients with liver cirrhosis after intravenous and oral administration. Clin Pharmacol Ther. 1986;40(1):21–28.
  • Turncliff RZ, Dunbar JL, Dong Q, et al. Pharmacokinetics of long‐acting naltrexone in subjects with mild to moderate hepatic impairment. J Clin Pharmacol. 2005;45(11):1259–1267.
  • Nasser AF, Heidbreder C, Liu Y, et al. Pharmacokinetics of sublingual buprenorphine and naloxone in subjects with mild to severe hepatic impairment (Child-Pugh classes A, B, and C), in hepatitis C virus-seropositive subjects, and in healthy volunteers. Clin Pharmacokinet. 2015;54(8):837–849.
  • Albarmawi A, Czock D, Gauss A, 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):160–169.
  • Orlando R, Piccoli P, De Martin S, et al. Cytochrome P450 1A2 is a major determinant of lidocaine metabolism in vivo: effects of liver function. Clin Pharmacol Ther. 2004;75(1):80–88.
  • Khatri A, Menon RM, Marbury TC, et al. Pharmacokinetics and safety of co-administered paritaprevir plus ritonavir, ombitasvir, and dasabuvir in hepatic impairment. J Hepatol. 2015;63(4):805–812.
  • Kaufmann P, Cruz HG, Krause A, et al. Pharmacokinetics of the novel oral prostacyclin receptor agonist selexipag in subjects with hepatic or renal impairment. Br J Clin Pharmacol. 2016;82(2):369–379.
  • Durand F, Valla D. Assessment of the prognosis of cirrhosis: Child–Pugh versus MELD. J Hepatol. 2005;42(1):S107.
  • Pugh RN, Murray-Lyon IM, Dawson JL, et al. Transection of the oesophagus for bleeding oesophageal varices. Br J Surg. 1973 Aug;60(8):646–649.
  • European Medicines Agency. Guideline on the evaluation of the pharmacokinetics of medicinal products in patients with impaired hepatic function. London. 2005. [cited 2019 Aug 9]. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-evaluation-pharmacokinetics-medicinal-products-patients-impaired-hepatic-function_en.pdf
  • Food and Drug Administration. Guidance for industry: pharmacokinetics in patients with impaired hepatic function: study design, data analysis, and impact on dosing and labeling. Rockville. 2003. [cited 2019 Aug 9]. Available from: https://www.fda.gov/media/71311/download
  • Spray JW, Willett K, Chase D, et al. Dosage adjustment for hepatic dysfunction based on Child-Pugh scores. Am J Health-system Pharm. 2007;64(7):692.
  • van Harten J, Duchier J, Devissaguet JP, et al. Pharmacokinetics of fluvoxamine maleate in patients with liver cirrhosis after single-dose oral administration. Clin Pharmacokinet. 1993;24(2):177–182.
  • Chalasani N, Gorski JC, Patel NH, et al. Hepatic and intestinal cytochrome P450 3A activity in cirrhosis: effects of transjugular intrahepatic portosystemic shunts. Hepatology. 2001;34(6):1103–1108.
  • De Winter S, Verelst S, Wauters J, et al. Pharmacokinetic changes after placement of a transjugular intrahepatic portosystemic shunt. Eur J Clin Pharmacol. 2014;70(3):377.
  • Spriet I, Meyfroidt G, Maleux G, et al. The impact of a transjugular intrahepatic portosystemic shunt on the pharmacokinetics of caspofungin in a critically ill patient. Pharmacology. 2012;90(5–6):247–250.
  • Hasler JA, Estabrook R, Murray M, et al. Human cytochromes P450. Mol Aspects Med. 1999;20(1–2):55–65.
  • Prasad B, Bhatt DK, Johnson K, 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):943–952.
  • Gilmore IT, Thompson RP. Kinetics of 14C-glycocholic acid clearance in normal man and in patients with liver disease. Gut. 1978;19(12):1110–1115.
  • Hayward KL, Powell EE, Irvine KM, et al. Can paracetamol (acetaminophen) be administered to patients with liver impairment? Br J Clin Pharmacol. 2016;81(2):210–222.
  • Weersink RA, Borgsteede SD, Okel E, et al. Paracetamol use in patients with liver cirrhosis and the risk of hepatotoxicity. Pharm Weekbl. 2016;151(11):22–30.
  • Benson GD, Koff RS, Tolman KG. The therapeutic use of acetaminophen in patients with liver disease. Am J Ther. 2005;12(2):133–141.
  • Ohnishi A, Murakami S, Akizuki S, et al. In vivo metabolic activity of CYP2C19 and CYP3A in relation to CYP2C19 genetic polymorphism in chronic liver disease. J Clin Pharmacol. 2005;45(11):1221–1229.
  • Ferron GM, Preston RA, Noveck RJ, et al. Pharmacokinetics of pantoprazole in patients with moderate and severe hepatic dysfunction. Clin Ther. 2001;23(8):1180–1192.
  • Parusov A, Loranskaya I, Ryzhikova K, et al. Polymorphic marker cyp2d6* 4 and the response to propranolol in Russian patients with liver cirrhosis. Eur J Clin Pharmacol. 2019.75_Supplement 1 (S31-).
  • Zhang F, Duan X, Zhang M, et al. Influence of cyp2d6 and β2‐adrenergic receptor gene polymorphisms on the hemodynamic response to propranolol in chinese han patients with cirrhosis. J Gastroenterol Hepatol. 2016;31(4):829–834.
  • Villeneuve JP, Verbeeck RK, Wilkinson GR, et al. Furosemide kinetics and dynamics in patients with cirrhosis. Clin Pharmacol Ther. 1986;40(1):14–20.
  • Gentilini P, La Villa G, Marra F, et al. Pharmacokinetics and pharmacodynamics of torasemide and furosemide in patients with diuretic resistant ascites. J Hepatol. 1996;25(4):481–490.
  • Schwartz S, Brater DC, Pound D, et al. Bioavailability, pharmacokinetics, and pharmacodynamics of torsemide in patients with cirrhosis. Clin Pharmacol Ther. 1993;54(1):90–97.
  • Keller E, Hoppe-Seyler G, Mumm R, et al. Influence of hepatic cirrhosis and end-stage renal disease on pharmacokinetics and pharmacodynamics of furosemide. Eur J Clin Pharmacol. 1981;20(1):27–33.
  • Janků I, Perlik F, Tkaczykova M, et al. Disposition kinetics and concentration-effect relationship of metipranolol in patients with cirrhosis and healthy subjects. Eur J Clin Pharmacol. 1992;42(3):337–340.
  • Taegtmeyer AB, Haschke M, Tchambaz L, 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.
  • d’Honneur G, Khalil M, Dominique C, et al. Pharmacokinetics and pharmacodynamics of pipecuronium in patients with cirrhosis. Anesth Analg. 1993;77(6):1203–1206.
  • Khalil M, D’honneur G, Duvaldestin P, et al. Pharmacokinetics and pharmacodynamics of rocuronium in patients with cirrhosis. Anesthesiology. 1994;80(6):1241–1247.
  • Duvaldestin P, Slavov V, Rebufat Y. Pharmacokinetics and pharmacodynamics of rapacuronium in patients with cirrhosis. Anesthesiology. 1999;91(5):1305–1310.
  • Lebrault C, Berger J, d’Hollander A, et al. Pharmacokinetics and pharmacodynamics of vecuronium (ORG NC 45) in patients with cirrhosis. Anesthesiology. 1985;62(5):601–605.
  • Van Miert M, Eastwood N, Boyd A, et al. The pharmacokinetics and pharmacodynamics of rocuronium in patients with hepatic cirrhosis. Br J Clin Pharmacol. 1997;44(2):139–144.
  • Devlin J, Head-Rapson A, Parker C, et al. Pharmacodynamics of mivacurium chloride in patients with hepatic cirrhosis. Br J Anaesth. 1993;71(2):227–231.
  • Head-Rapson A, Devlin J, Parker C, et al. Pharmacokinetics of the three isomers of mivacurium and pharmacodynamics of the chiral mixture in hepatic cirrhosis. Br J Anaesth. 1994;73(5):613–618.
  • Levy G. Effect of hepatic cirrhosis on the pharmacodynamics and pharmacokinetics of mivacurium in humans [journal article]. Pharm Res. 1994 May 01;11(5):772–773.
  • Baba T, Murabayashi S, Tomiyama T, et al. The pharmacokinetics of enalapril in patients with compensated liver cirrhosis. Br J Clin Pharmacol. 1990;29(6):766–769.
  • Ohnishi A, Tsuboi Y, Ishizaki T, et al. Kinetics and dynamics of enalapril in patients with liver cirrhosis. Clin Pharmacol Ther. 1989;45(6):657–665.
  • Thiollet M, Funck‐Brentano C, Grange J, et al. The pharmacokinetics of perindopril in patients with liver cirrhosis. Br J Clin Pharmacol. 1992;33(3):326–328.
  • Tsai H, Lees K, Howden C, et al. The pharmacokinetics and pharmacodynamics of perindopril in patients with hepatic cirrhosis. Br J Clin Pharmacol. 1989;28(1):53–59.
  • Croxen F. Undesirable side effects. In: North-Lewis P, editor. Drugs and the liver: a guide to drug handling in liver dysfunction. London: Pharmaceutical Press; 2008. p. 135–143.
  • Clària J, Kent JD, López-Parra M, et al. Effects of celecoxib and naproxen on renal function in nonazotemic patients with cirrhosis and ascites. Hepatology. 2005;41(3):579–587.
  • Wong F, Massie D, Hsu P, et al. Indomethacin-induced renal dysfunction in patients with well-compensated cirrhosis. Gastroenterology. 1993;104(3):869–876.
  • Laffi G, Daskalopoulos G, Kronborg I, et al. Effects of sulindac and ibuprofen in patients with cirrhosis and ascites: an explanation for the renal-sparing effect of sulindac. Gastroenterology. 1986;90(1):182–187.
  • Tandon P, Abraldes JG, Berzigotti A, et al. Renin–angiotensin–aldosterone inhibitors in the reduction of portal pressure: a systematic review and meta-analysis. J Hepatol. 2010;53(2):273–282.
  • Schepke M, Werner E, Biecker E, et al. Hemodynamic effects of the angiotensin II receptor antagonist irbesartan in patients with cirrhosis and portal hypertension. Gastroenterology. 2001;121(2):389–395.
  • Salerno F, Badalamenti S. Drug-induced renal failure in cirrhosis. In: Ginès P, Arroyo V, Rodés J, editors. Ascites and renal dysfunction in liver disease. Oxford: Blackwell Publishing Ltd; 2005. p. 372–382.
  • Bernardi M, Domenicali M. The renin-angiotensin-aldosterone system in cirrhosis. In: Ginès P, Arroyo V, Rodés J, editors. Ascites and renal dysfunction in liver disease. Oxford: Blackwell Publishing Ltd; 2005. p. 41–53.
  • Moore RD, Smith CR, Lietman PS. Increased risk of renal dysfunction due to interaction of liver disease and aminoglycosides. Am J Med. 1986;80(6):1093–1097.
  • Hampel H, Bynum GD, Zamora E, et al. Risk factors for the development of renal dysfunction in hospitalized patients with cirrhosis. Am J Gastroenterol. 2001;96(7):2206–2210.
  • Bajaj JS, O’Leary JG, Tandon P, et al. Targets to improve quality of care for patients with hepatic encephalopathy: data from a multi-centre cohort. Aliment Pharmacol Ther. 2019 Apr 29;49:1518–1527.
  • Grønbæk L, Watson H, Vilstrup H, et al. Benzodiazepines and risk for hepatic encephalopathy in patients with cirrhosis and ascites. United European Gastroenterol J. 2018;6(3):407–412.
  • Weiss N, Saint Hilaire PB, Colsch B, et al. Cerebrospinal fluid metabolomics highlights dysregulation of energy metabolism in overt hepatic encephalopathy. J Hepatol. 2016;65(6):1120–1130.
  • Schentag JJ, Cerra FB, Calleri GM, et al. Age, disease, and cimetidine disposition in healthy subjects and chronically ill patients. Clin Pharmacol Ther. 1981;29(6):737–743.
  • Bakti G, Fisch HU, Karlaganis G, et al. Mechanism of the excessive sedative response of cirrhotics to benzodiazepines: model experiments with triazolam. Hepatology. 1987;7(4):629–638.
  • Branch RA, Morgan MH, James J, et al. Intravenous administration of diazepam in patients with chronic liver disease. Gut. 1976;17:975–983.
  • Jochemsen R, Joeres RP, Wesselman JG, et al. Pharmacokinetics of oral brotizolam in patients with liver cirrhosis. Br J Clin Pharmacol. 1983;16(Suppl 2):322S.
  • Perney P, Butterworth RF, Mousseau DD, et al. Plasma and CSF benzodiazepine receptor ligand concentrations in cirrhotic patients with hepatic encephalopathy: relationship to severity of encephalopathy and to pharmaceutical benzodiazepine intake. Metab Brain Dis. 1998;13(3):201–210.
  • Lavoie J, Layrargues GP, Butterworth RF. Increased densities of peripheral‐type benzodiazepine receptors in brain autopsy samples from cirrhotic patients with hepatic encephalopathy. Hepatology. 1990;11(5):874–878.
  • Prakash R, Mullen KD. Mechanisms, diagnosis and management of hepatic encephalopathy. Nat Rev Gastroenterol Hepatol. 2010 Sep;7(9):515–525.
  • Butterworth RF. The liver-brain axis in liver failure: neuroinflammation and encephalopathy. Nat Rev Gastroenterol Hepatol. 2013;10(9):522–528.
  • Posner JB, Plum F. The toxic effects of carbon dioxide and acetazolamide in hepatic encephalopathy. J Clin Invest. 1960;39(8):1246–1258.
  • Margo CE. Acetazolamide and advanced liver disease. Am J Ophthalmol. 1986;101(5):611.
  • Maren TH. Acetazolamide and advanced liver disease. Am J Ophthalmol. 1986 Nov 15;102(5):672–673.
  • Wijdicks EFM, Longo DL. Hepatic encephalopathy. N Engl J Med. 2016;375(17):1660–1670.
  • Bunchorntavakul C, Chamroonkul N, Chavalitdhamrong D. Bacterial infections in cirrhosis: A critical review and practical guidance. World J Hepatol. 2016;8(6):307–321.
  • Bonnel AR, Bunchorntavakul C, Reddy KR. Immune dysfunction and infections in patients with cirrhosis. Clin Gastroenterol Hepatol. 2011;9(9):727–738.
  • Perrillo RR, Tamburro C, Regenstein F. Low-dose, titratable interferon alfa in decompensated liver disease caused by chronic infection with hepatitis B virus. Gastroenterology. 1995;109(3):908–916.
  • Hoofnagle JHJ, Di Bisceglie AM, Waggoner JG. Interferon alfa for patients with clinically apparent cirrhosis due to chronic hepatitis B. Gastroenterology. 1993;104(4):1116–1121.
  • Bajaj JS, Zadvornova Y, Heuman DM, et al. Association of proton pump inhibitor therapy with spontaneous bacterial peritonitis in cirrhotic patients with ascites. Am J Gastroenterol. 2009;104(5):1130–1134.
  • Gonzalez-Casas R, Jones EA, Moreno-Otero R. Spectrum of anemia associated with chronic liver disease. World J Gastroenterol. 2009 Oct 7;15(37):4653–4658.
  • Mitchell O, Feldman DM, Diakow M, et al. The pathophysiology of thrombocytopenia in chronic liver disease. Hepat Med. 2016;8:39–50.
  • Lisman T, Kamphuisen PW, Northup PG, et al. Established and new-generation antithrombotic drugs in patients with cirrhosis–possibilities and caveats. J Hepatol. 2013;59(2):358–366.
  • Marks PW. Hematologic manifestations of liver disease. Semin Hematol. 2013 07 01;50(3):216–221.
  • Peck‐Radosavljevic M, Wichlas M, Pidlich J, et al. Blunted thrombopoietin response to interferon alfa–induced thrombocytopenia during treatment for hepatitis C. Hepatology. 1998;28(5):1424–1429.
  • Heneghan MA, Allan ML, Bornstein JD, et al. Utility of thiopurine methyltransferase genotyping and phenotyping, and measurement of azathioprine metabolites in the management of patients with autoimmune hepatitis. J Hepatol. 2006 10 01;45(4):584–591.
  • Czaja AJ, Carpenter HA. Thiopurine methyltransferase deficiency and azathioprine intolerance in autoimmune hepatitis. Dig Dis Sci. 2006 05 01;51(5):968–975.
  • Aspen Pharma Trading Limited. Summary of product characteristics imuran 25 mg: medicines evaluation board; [updated 2018 December 4; cited 2019 May 28]. Available from: https://www.geneesmiddeleninformatiebank.nl/smpc/h05565_smpc.pdf
  • Pardo M, Marriott E, Moliner MC, et al. Risks and benefits of interferon-α in the treatment of hepatitis [journal article]. Drug Saf. 1995 November 01;13(5):304–316.
  • Causse X, Godinot H, Chevallier M, et al. Comparison of 1 or 3 MU of interferon alfa-2b and placebo in patients with chronic non-A, non-B hepatitis. Gastroenterology. 1991;101(2):497–502.
  • Rakela J, Wood JR, Czaja AJ, et al. Long-term versus short-term treatment with recombinant interferon alfa-2a in patients with chronic hepatitis B: a prospective, randomized treatment trial. Mayo Clin Proc. 1990 10 01;65(10):1330–1335.
  • Van Vlierberghe H, Delanghe JR, De Vos M, et al. Factors influencing ribavirin-induced hemolysis. J Hepatol. 2001 06 01;34(6):911–916.
  • Gupta NK, Lewis JH. The use of potentially hepatotoxic drugs in patients with liver disease. Aliment Pharmacol Ther. 2008;28(9):1021–1041.
  • Chalasani N, Bonkovsky HL, Fontana R, et al. Features and outcomes of 899 patients with drug-induced liver injury: the DILIN prospective study. Gastroenterology. 2015;148(7):1352. e7.
  • Food and Drug Administration. FDA warns about serious liver injury with ocaliva (obeticholic acid) for rare chronic liver disease. 2017. [cited 2019 Sep 2]. Available from: https://www.fda.gov/media/107517/download
  • Food and Drug Administration. FDA drug safety communication: FDA warns of serious liver injury risk with hepatitis C treatments viekira pak and technivie. 2015. [cited 2019 Sep 2]. Available from: https://www.fda.gov/media/94262/download
  • Health NIo. Drug record - viekira pak: dasabur, ombitasvir, paritraprevir and ritonavir. [updated 2018 Jan 10]. Available from: https://livertox.nih.gov/ViekiraPak.htm
  • Intercept statement regarding ocaliva® (obeticholic acid) safety and dosing in Primary Biliary Cholangitis (PBC) patients [Internet]. New York: Globe Newswire; 2017; Sept. 25, [4]. Available from: http://ir.interceptpharma.com/static-files/8013669f-4f8c-4543-90eb-8b479682a1e8
  • Failings in treatment advice, SPCs and black triangles. Drug Ther Bull. 2001;39(4):25–27.
  • Chang Y, Burckart GJ, Lesko LJ, et al. Evaluation of hepatic impairment dosing recommendations in FDA‐approved product labels. J Clin Pharmacol. 2013;53(9):962–966.
  • Bjornsson ES, Jacobsen EI, Einarsdottir R, et al. Discrepancies in liver disease labeling in the package inserts of commonly prescribed medications. Gastroenterology. 2015;148(2):269–273.
  • Schlatter C, Egger SS, Tchambaz L, et al. Pharmacokinetic changes of psychotropic drugs in patients with liver disease. Drug Saf. 2009;32(7):561–578.
  • Lewis JH, Stine JG. Review article: prescribing medications in patients with cirrhosis – a practical guide. Aliment Pharmacol Ther. 2013;37(12):1132–1156.
  • Rodighiero V. Effects of liver disease on pharmacokinetics. Clin Pharmacokinet. 1999;37(5):399–431.
  • Scheen AJ. Pharmacokinetic and toxicological considerations for the treatment of diabetes in patients with liver disease. Expert Opin Drug Metab Toxicol. 2014;10(6):839–857.
  • Weersink RA, Bouma M, Burger DM, et al. Evaluating the safety and dosing of drugs in patients with liver cirrhosis by literature review and expert opinion. BMJ Open. 2016;6(10):012991.
  • Weersink RA, Bouma M, Burger DM, et al. Evidence-based recommendations to improve the safe use of drugs in patients with liver cirrhosis. Drug Saf. 2018;41(6):603–613.
  • Salvà P, Costa J. Clinical pharmacokinetics and pharmacodynamics of zolpidem. Therapeutic implications. Clin Pharmacokinet. 1995;29(3):142–153.
  • Meda Pharmaceuticals Inc. FDA label edluar - zolpidem tartrate tablet 2013. [cited 2019 Sep 2]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/021997s005lbl.pdf
  • Nevo ON, Brinker AD, Diak I-L, et al. Response to: improvement of sleep architecture parameters in cirrhotic patients with recurrent hepatic encephalopathy with the use of rifaximin: hepatic encephalopathy in association with zolpidem. Eur J Gastroenterol Hepatol. 2017;29(9):1102–1103.
  • Clark A. Worsening hepatic encephalopathy secondary to zolpidem. J Pharm Technol. 1999;15:139–141.
  • Silva V, Bittencourt PL, Pinho S, et al. Delayed-onset hepatic encephalopathy induced by zolpidem: a case report. Clinics. 2008;63(4):565–566.
  • Parker G, Roberts CJ. Plasma concentrations and central nervous system effects of the new hypnotic agent zopiclone in patients with chronic liver disease. Br J Clin Pharmacol. 1983;16(3):259–265.
  • Gaillot J, Le Roux Y, Houghton GW, et al. Critical factors for pharmacokinetics of zopiclone in the elderly and in patients with liver and renal insufficiency. Sleep. 1987;10(Suppl. 1):7–21.
  • Hayward KL, Patel PJ, Valery PC, et al. Medication-related problems in outpatients with decompensated cirrhosis: opportunities for harm prevention. Hepatol Commun. 2019 May;3(5):620–631.
  • Polis S, Zang L, Mainali B, et al. Factors associated with medication adherence in patients living with cirrhosis. J Clin Nurs. 2016;25(1–2):204–212.
  • Kuo SZ, Haftek M, Lai JC. Factors associated with medication non-adherence in patients with end-stage liver disease. Dig Dis Sci. 2017;62(2):543–549.
  • Thomson MJ, Lok AS, Tapper EB. Optimizing medication management for patients with cirrhosis: evidence‐based strategies and their outcomes. Liver Int. 2018;38(11):1882–1890.
  • Smolders EJ, Berden FA, de Kanter CT, et al. The majority of hepatitis C patients treated with direct acting antivirals are at risk for relevant drug-drug interactions. United European Gastroenterol J. 2017;5(5):648–657.
  • Wouters H, Scheper J, Koning H, et al. Discontinuing inappropriate medication use in nursing home residents: a cluster randomized controlled trial. Ann Intern Med. 2017;167(9):609–617.
  • Weersink RA, Timmermans L, Monster-Simons M, et al. Evaluation of information in Summaries of Product Characteristics (SmPCs) on the use of a medicine in patients with hepatic impairment. Front Pharmacol. 2019;10. DOI:10.3389/fphar.2019.01031
  • Ferner RE, Aronson JK. Communicating information about drug safety. BMJ. 2006;333(7559):143–145.
  • Singh N, Victor LY, Mieles LA, et al. β-Lactam antibiotic-induced leukopenia in severe hepatic dysfunction: risk factors and implications for dosing in patients with liver disease. Am J Med. 1993;94(3):251–256.