Skip to main content
Erschienen in: Clinical Pharmacokinetics 11/2006

01.11.2006 | Current Opinion

Fatal Respiratory Depression after Multiple Intravenous Morphine Injections

verfasst von: Prof. Jörn Lötsch, Rafael Dudziak, Rainer Freynhagen, Jürgen Marschner, Gerd Geisslinger

Erschienen in: Clinical Pharmacokinetics | Ausgabe 11/2006

Einloggen, um Zugang zu erhalten

Abstract

A 26-year-old female was treated with morphine within the first 2 hours after knee surgery, in an attempt to titrate analgesia. The patient received a total of four intravenous injections of morphine 35mg in total. Soon after the last injection the patient had adequate pain relief, was in a good clinical state and had adequate blood oxygenation. However, 40 minutes later, the patient had a deep respiratory depression followed by a fatal cardiac arrest. Solving the case in a medico-legal context was possible by applying results of clinical pharmacokinetic research on opioid analgesics, most importantly morphine, to this particular clinical case. This knowledge made it possible to estimate the probable concentrations of morphine at the site of its effect, the brain, during the time of the fatal event, and to show that these concentrations could have produced respiratory depression. We mainly attribute the fatal intoxication of morphine to the lag period needed for the transfer of morphine across the blood-brain barrier. Because of its slow transfer between plasma and the effect site, the CNS effects of morphine are delayed from its plasma concentrations to a clinically relevant degree. Successive injections at short intervals of relatively high amounts of morphine increase the clinical relevance of this delay. The present report demonstrates an important application of clinical pharmacokinetics for explaining clinical observations at a scientific level and transferring theoretical knowledge from clinical pharmacokinetics into daily clinical practice as a basis for rational opioid selection.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Lötsch J, Skarke C, Schmidt H, et al. Pharmacokinetic modeling to predict morphine and morphine-6-glucuronide plasma concentrations in healthy young volunteers. Clin Pharmacol Ther 2002; 72(2): 151–62PubMedCrossRef Lötsch J, Skarke C, Schmidt H, et al. Pharmacokinetic modeling to predict morphine and morphine-6-glucuronide plasma concentrations in healthy young volunteers. Clin Pharmacol Ther 2002; 72(2): 151–62PubMedCrossRef
2.
Zurück zum Zitat Scott JC, Stanski DR. Decreased fentanyl and alfentanil dose requirements with age: a simultaneous pharmacokinetic and pharmacodynamic evaluation. J Pharmacol Exp Ther 1987; 240(1): 159–66PubMed Scott JC, Stanski DR. Decreased fentanyl and alfentanil dose requirements with age: a simultaneous pharmacokinetic and pharmacodynamic evaluation. J Pharmacol Exp Ther 1987; 240(1): 159–66PubMed
3.
Zurück zum Zitat Lötsch J, Skarke C, Schmidt H, et al. The transfer half-life of morphine-6-beta-glucuronide from plasma to effect site assessed by pupil size measurement in healthy volunteers. Anesthesiology 2001; 95(6): 1329–38PubMedCrossRef Lötsch J, Skarke C, Schmidt H, et al. The transfer half-life of morphine-6-beta-glucuronide from plasma to effect site assessed by pupil size measurement in healthy volunteers. Anesthesiology 2001; 95(6): 1329–38PubMedCrossRef
4.
Zurück zum Zitat Lötsch J, Skarke C, Schneider A, et al. The 5 HT4 receptor agonist mosapride does not antagonize morphine-induced respiratory depression. Clin Pharmacol Ther 2005; 78(3): 278–87PubMedCrossRef Lötsch J, Skarke C, Schneider A, et al. The 5 HT4 receptor agonist mosapride does not antagonize morphine-induced respiratory depression. Clin Pharmacol Ther 2005; 78(3): 278–87PubMedCrossRef
5.
Zurück zum Zitat Read DJ, Simon H, Brandi G, et al. Regulation of ventilation during rebreathing at imposed respiratory frequencies. Respir Physiol 1966; 2(1): 88–98PubMedCrossRef Read DJ, Simon H, Brandi G, et al. Regulation of ventilation during rebreathing at imposed respiratory frequencies. Respir Physiol 1966; 2(1): 88–98PubMedCrossRef
6.
Zurück zum Zitat Sheiner LB, Stanski DR, Vozeh S, et al. Simultaneous modeling of pharmacokinetics and pharmacodynamics: application to dtubocurarine. Clin Pharmacol Ther 1979; 25: 358–71PubMed Sheiner LB, Stanski DR, Vozeh S, et al. Simultaneous modeling of pharmacokinetics and pharmacodynamics: application to dtubocurarine. Clin Pharmacol Ther 1979; 25: 358–71PubMed
7.
Zurück zum Zitat Hull CJ, Van Beem HB, McLeod K, et al. A pharmacodynamic model for pancuronium. Br J Anaesth 1978; 50(11): 1113–23PubMedCrossRef Hull CJ, Van Beem HB, McLeod K, et al. A pharmacodynamic model for pancuronium. Br J Anaesth 1978; 50(11): 1113–23PubMedCrossRef
8.
Zurück zum Zitat Dershwitz M, Walsh JL, Morishige RJ, et al. Pharmacokinetics and pharmacodynamics of inhaled versus intravenous morphine in healthy volunteers. Anesthesiology 2000; 93(3): 619–28PubMedCrossRef Dershwitz M, Walsh JL, Morishige RJ, et al. Pharmacokinetics and pharmacodynamics of inhaled versus intravenous morphine in healthy volunteers. Anesthesiology 2000; 93(3): 619–28PubMedCrossRef
9.
Zurück zum Zitat Sarton E, Olofsen E, Romberg R, et al. Sex differences in morphine analgesia: an experimental study in healthy volunteers. Anesthesiology 2000; 93(5): 1245–54PubMedCrossRef Sarton E, Olofsen E, Romberg R, et al. Sex differences in morphine analgesia: an experimental study in healthy volunteers. Anesthesiology 2000; 93(5): 1245–54PubMedCrossRef
10.
Zurück zum Zitat Skarke C, Darimont J, Schmidt H, et al. Analgesic effects of morphine and morphine-6-glucuronide in a transcutaneous electrical pain model in healthy volunteers. Clin Pharmacol Ther 2003; 73(1): 107–21PubMedCrossRef Skarke C, Darimont J, Schmidt H, et al. Analgesic effects of morphine and morphine-6-glucuronide in a transcutaneous electrical pain model in healthy volunteers. Clin Pharmacol Ther 2003; 73(1): 107–21PubMedCrossRef
11.
Zurück zum Zitat Romberg R, Olofsen E, Sarton E, et al. Pharmacokinetic-pharmacodynamic modeling of morphine-6-glucuronide-induced analgesia in healthy volunteers: absence of sex differences. Anesthesiology 2004; 100(1): 120–33PubMedCrossRef Romberg R, Olofsen E, Sarton E, et al. Pharmacokinetic-pharmacodynamic modeling of morphine-6-glucuronide-induced analgesia in healthy volunteers: absence of sex differences. Anesthesiology 2004; 100(1): 120–33PubMedCrossRef
12.
Zurück zum Zitat Angst MS, Drover DR, Lötsch J, et al. Pharmacodynamics of orally administered sustained-release hydromorphone in humans. Anesthesiology 2001; 94(1): 63–73PubMedCrossRef Angst MS, Drover DR, Lötsch J, et al. Pharmacodynamics of orally administered sustained-release hydromorphone in humans. Anesthesiology 2001; 94(1): 63–73PubMedCrossRef
13.
Zurück zum Zitat Lalovic B, Kharasch E, Hoffer C, et al. Pharmacokinetics and pharmacodynamics of oral oxycodone in healthy human subjects: role of circulating active metabolites. Clin Pharmacol Ther 2006; 79(5): 461–79PubMedCrossRef Lalovic B, Kharasch E, Hoffer C, et al. Pharmacokinetics and pharmacodynamics of oral oxycodone in healthy human subjects: role of circulating active metabolites. Clin Pharmacol Ther 2006; 79(5): 461–79PubMedCrossRef
14.
Zurück zum Zitat Scott JC, Ponganis KV, Stanski DR. EEG quantitation of narcotic effect: the comparative pharmacodynamics of fentanyl and alfentanil. Anesthesiology 1985; 62: 234–41PubMedCrossRef Scott JC, Ponganis KV, Stanski DR. EEG quantitation of narcotic effect: the comparative pharmacodynamics of fentanyl and alfentanil. Anesthesiology 1985; 62: 234–41PubMedCrossRef
15.
Zurück zum Zitat Scott JC, Cooke JE, Stanski DR. Electroencephalographic quantitation of opioid effect: comparative pharmacodynamics of fentanyl and sufentanil. Anesthesiology 1991; 74(1): 34–42PubMedCrossRef Scott JC, Cooke JE, Stanski DR. Electroencephalographic quantitation of opioid effect: comparative pharmacodynamics of fentanyl and sufentanil. Anesthesiology 1991; 74(1): 34–42PubMedCrossRef
16.
Zurück zum Zitat Lemmens HJ, Dyck JB, Shafer SL, et al. Pharmacokineticpharmacodynamic modeling in drug development: application to the investigational opioid trefentanil. Clin Pharmacol Ther 1994; 56(3): 261–71PubMedCrossRef Lemmens HJ, Dyck JB, Shafer SL, et al. Pharmacokineticpharmacodynamic modeling in drug development: application to the investigational opioid trefentanil. Clin Pharmacol Ther 1994; 56(3): 261–71PubMedCrossRef
17.
Zurück zum Zitat Billard V, Gambus PL, Chamoun N, et al. A comparison of spectral edge, delta power, and bispectral index as EEG measures of alfentanil, propofol, and midazolam drag effect. Clin Pharmacol Ther 1997; 61(1): 45–58PubMedCrossRef Billard V, Gambus PL, Chamoun N, et al. A comparison of spectral edge, delta power, and bispectral index as EEG measures of alfentanil, propofol, and midazolam drag effect. Clin Pharmacol Ther 1997; 61(1): 45–58PubMedCrossRef
18.
Zurück zum Zitat Egan TD, Minto CF, Hermann DJ, et al. Remifentanil versus alfentanil: comparative pharmacokinetics and pharmacodynamics in healthy adult male volunteers [published erratum appears in Anesthesiology 1996 Sep; 85 (3): 695]. Anesthesiology 1996; 84(4): 821–33PubMedCrossRef Egan TD, Minto CF, Hermann DJ, et al. Remifentanil versus alfentanil: comparative pharmacokinetics and pharmacodynamics in healthy adult male volunteers [published erratum appears in Anesthesiology 1996 Sep; 85 (3): 695]. Anesthesiology 1996; 84(4): 821–33PubMedCrossRef
19.
Zurück zum Zitat Minto CF, Schnider TW, Egan TD, et al. Influence of age and gender on the pharmacokinetics and pharmacodynamics of remifentanil: I. Model development. Anesthesiology 1997; 86(1): 10–23PubMedCrossRef Minto CF, Schnider TW, Egan TD, et al. Influence of age and gender on the pharmacokinetics and pharmacodynamics of remifentanil: I. Model development. Anesthesiology 1997; 86(1): 10–23PubMedCrossRef
20.
Zurück zum Zitat Glass PS, Hardman D, Kamiyama Y, et al. Preliminary pharmacokinetics and pharmacodynamics of an ultra-shortacting opioid: remifentanil (GI87084B). Anesth Analg 1993; 77: 1031–40PubMedCrossRef Glass PS, Hardman D, Kamiyama Y, et al. Preliminary pharmacokinetics and pharmacodynamics of an ultra-shortacting opioid: remifentanil (GI87084B). Anesth Analg 1993; 77: 1031–40PubMedCrossRef
21.
Zurück zum Zitat Gambus PL, Gregg KM, Shafer SL. Validation of the alfentanil canonical univariate parameter as a measure of opioid effect on the electroencephalogram. Anesthesiology 1995; 83(4): 747–56PubMedCrossRef Gambus PL, Gregg KM, Shafer SL. Validation of the alfentanil canonical univariate parameter as a measure of opioid effect on the electroencephalogram. Anesthesiology 1995; 83(4): 747–56PubMedCrossRef
22.
Zurück zum Zitat Hermann DJ, Egan TD, Muir KT. Influence of arteriovenous sampling on remifentanil pharmacokinetics and pharmacodynamics. Clin Pharmacol Ther 1999; 65(5): 511–8PubMedCrossRef Hermann DJ, Egan TD, Muir KT. Influence of arteriovenous sampling on remifentanil pharmacokinetics and pharmacodynamics. Clin Pharmacol Ther 1999; 65(5): 511–8PubMedCrossRef
23.
Zurück zum Zitat Inturrisi CE, Colburn WA, Kaiko RF, et al. Pharmacokinetics and pharmacodynamics of methadone in patients with chronic pain. Clin Pharmacol Ther 1987; 41(4): 392–401PubMedCrossRef Inturrisi CE, Colburn WA, Kaiko RF, et al. Pharmacokinetics and pharmacodynamics of methadone in patients with chronic pain. Clin Pharmacol Ther 1987; 41(4): 392–401PubMedCrossRef
24.
Zurück zum Zitat Lötsch J, Skarke C, Wieting J, et al. Modulation of the central nervous effects of levomethadone by genetic polymorphisms potentially affecting its metabolism, distribution, and drug action. Clin Pharmacol Ther 2006; 79(1): 72–89PubMedCrossRef Lötsch J, Skarke C, Wieting J, et al. Modulation of the central nervous effects of levomethadone by genetic polymorphisms potentially affecting its metabolism, distribution, and drug action. Clin Pharmacol Ther 2006; 79(1): 72–89PubMedCrossRef
25.
Zurück zum Zitat Bouillon T, Schmidt C, Garstka G, et al. Pharmacokineticpharmacodynamic modeling of the respiratory depressant effect of alfentanil. Anesthesiology 1999; 91(1): 144–55PubMedCrossRef Bouillon T, Schmidt C, Garstka G, et al. Pharmacokineticpharmacodynamic modeling of the respiratory depressant effect of alfentanil. Anesthesiology 1999; 91(1): 144–55PubMedCrossRef
26.
Zurück zum Zitat Eap CB, Buclin T, Baumann P. Interindividual variability of the clinical pharmacokinetics of methadone: implications for the treatment of opioid dependence. Clin Pharmacokinet 2002; 41(14): 1153–93PubMedCrossRef Eap CB, Buclin T, Baumann P. Interindividual variability of the clinical pharmacokinetics of methadone: implications for the treatment of opioid dependence. Clin Pharmacokinet 2002; 41(14): 1153–93PubMedCrossRef
27.
Zurück zum Zitat Egan TD, Lemmens HJ, Fiset P, et al. The pharmacokinetics of the new short-acting opioid remifentanil (GI87084B) in healthy adult male volunteers. Anesthesiology 1993; 79: 881–92PubMedCrossRef Egan TD, Lemmens HJ, Fiset P, et al. The pharmacokinetics of the new short-acting opioid remifentanil (GI87084B) in healthy adult male volunteers. Anesthesiology 1993; 79: 881–92PubMedCrossRef
28.
Zurück zum Zitat Shafer SL, Varvel JR. Pharmacokinetics, pharmacodynamics, and rational opioid selection. Anesthesiology 1991; 74: 53–63PubMedCrossRef Shafer SL, Varvel JR. Pharmacokinetics, pharmacodynamics, and rational opioid selection. Anesthesiology 1991; 74: 53–63PubMedCrossRef
29.
Zurück zum Zitat Schraag S, Mohl U, Hirsch M, et al. Recovery from opioid anesthesia: the clinical implication of context-sensitive half-times. Anesth Analg 1998; 86(1): 184–90PubMed Schraag S, Mohl U, Hirsch M, et al. Recovery from opioid anesthesia: the clinical implication of context-sensitive half-times. Anesth Analg 1998; 86(1): 184–90PubMed
30.
Zurück zum Zitat Hughes MA, Glass PS, Jacobs JR. Context-sensitive half-time in multicompartment pharmacokinetic models for intravenous anesthetic drags. Anesthesiology 1992; 76(3): 334–41PubMedCrossRef Hughes MA, Glass PS, Jacobs JR. Context-sensitive half-time in multicompartment pharmacokinetic models for intravenous anesthetic drags. Anesthesiology 1992; 76(3): 334–41PubMedCrossRef
31.
Zurück zum Zitat Heidegger T, Minto CF, Schnider TW. Modern concepts in pharmacokinetics of intravenous anesthetics. Anaesthesist 2004; 53(1): 95–110PubMedCrossRef Heidegger T, Minto CF, Schnider TW. Modern concepts in pharmacokinetics of intravenous anesthetics. Anaesthesist 2004; 53(1): 95–110PubMedCrossRef
32.
Zurück zum Zitat Latasch L, Teichmuller T, Dudziak R, et al. Antagonisation of fentanyl-induced respiratory depression by nalbuphine. Acta Anaesthesiol Belg 1989; 40(1): 35–40PubMed Latasch L, Teichmuller T, Dudziak R, et al. Antagonisation of fentanyl-induced respiratory depression by nalbuphine. Acta Anaesthesiol Belg 1989; 40(1): 35–40PubMed
33.
Zurück zum Zitat Lehmann KA, Ribbert N, Horrichs-Haermeyer G. Postoperative patient-controlled analgesia with alfentanil: analgesic efficacy and minimum effective concentrations. J Pain Symptom Manage 1990; 5(4): 249–58PubMedCrossRef Lehmann KA, Ribbert N, Horrichs-Haermeyer G. Postoperative patient-controlled analgesia with alfentanil: analgesic efficacy and minimum effective concentrations. J Pain Symptom Manage 1990; 5(4): 249–58PubMedCrossRef
34.
Zurück zum Zitat Youngs EJ, Shafer SL. Pharmacokinetic parameters relevant to recovery from opioids. Anesthesiology 1994; 81(4): 833–42PubMedCrossRef Youngs EJ, Shafer SL. Pharmacokinetic parameters relevant to recovery from opioids. Anesthesiology 1994; 81(4): 833–42PubMedCrossRef
35.
Zurück zum Zitat Chaturvedi K, Shahrestanifar M, Howells RD. mu Opioid receptor: role for the amino terminus as a determinant of ligand binding affinity. Brain Res Mol Brain Res 2000; 76(1): 64–72PubMedCrossRef Chaturvedi K, Shahrestanifar M, Howells RD. mu Opioid receptor: role for the amino terminus as a determinant of ligand binding affinity. Brain Res Mol Brain Res 2000; 76(1): 64–72PubMedCrossRef
36.
Zurück zum Zitat Selley DE, Sim LJ, Xiao R, et al. mu-Opioid receptor-stimulated guanosine-5′-O-(gamma-thio)-triphosphate binding in rat thalamus and cultured cell lines: signal transduction mechanisms underlying agonist efficacy. Mol Pharmacol 1997; 51(1): 87–96PubMed Selley DE, Sim LJ, Xiao R, et al. mu-Opioid receptor-stimulated guanosine-5′-O-(gamma-thio)-triphosphate binding in rat thalamus and cultured cell lines: signal transduction mechanisms underlying agonist efficacy. Mol Pharmacol 1997; 51(1): 87–96PubMed
Metadaten
Titel
Fatal Respiratory Depression after Multiple Intravenous Morphine Injections
verfasst von
Prof. Jörn Lötsch
Rafael Dudziak
Rainer Freynhagen
Jürgen Marschner
Gerd Geisslinger
Publikationsdatum
01.11.2006
Verlag
Springer International Publishing
Erschienen in
Clinical Pharmacokinetics / Ausgabe 11/2006
Print ISSN: 0312-5963
Elektronische ISSN: 1179-1926
DOI
https://doi.org/10.2165/00003088-200645110-00001

Weitere Artikel der Ausgabe 11/2006

Clinical Pharmacokinetics 11/2006 Zur Ausgabe