Skip to main content

Opioids, an Integrative Part in Perioperative Medicine

  • Chapter
Opioids in Medicine
  • 1944 Accesses

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 189.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 249.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 249.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Geddes, I.C. and T.C. Gray, Hyperventilation for the maintenance of anaesthesia. Lancet, 1959, 2: pp. 4–6.

    PubMed  CAS  Google Scholar 

  2. Laborit, H. and P. Huguenard, L’hibernation artificielle par moyens pharmacodynamiques et physiques. Presse Med, 1951, 59: pp. 1359–1365.

    Google Scholar 

  3. De Castro, G. and P. Mundeler, Anesthésie sans sommeil: “Neurolpetanalgésie”. Acta Chir Belg, 1959, 58: pp. 689–693.

    Google Scholar 

  4. De Castro, J., P. Viars, and J.C. Leleu, Utilisation de la Pentazocine comme antimorphinique et analgesique dans une technique d’anesthesie analgesique sequentielle, in Utilisation de la Pentazocine en anesthesie-reanimation, J. De Castro, P. Viars, and J.C. Leleu, Editors, 1969, Ars Medici: Bruxelles. pp. 33–36.

    Google Scholar 

  5. Flacke, J.W., W.E. Flacke, and G.D. Williams, Acute pulmonary edema following naloxone reversal of high dose morphine anesthesia. Anesthesiology, 1977, 47: pp. 376–378.

    PubMed  CAS  Google Scholar 

  6. Gasparetto, A. and G. Giron, Clinical value of vitamin anesthesia: Thiamine algosynaptolysis according to De Castro and Mundeleer. Acta Anaesthesiol, 1964, 15: pp. 699–710.

    PubMed  CAS  Google Scholar 

  7. Freye, E. and H. Agoutin, The action of vitamin B1 (thiamine) on the cardiovascular system of the cat. Biomedicine, 1978, 28: pp. 315–319.

    PubMed  CAS  Google Scholar 

  8. Freye, E. and E. Hartung, The potential use of thiamine (Vit.B1) in patients with cardiac insufficiency. Acta Vtaminol Enzymol, 1982, 4: pp. 285–290.

    CAS  Google Scholar 

  9. Lobera, A. and J.L. Renaud-Salis, General anesthesia in major cancer surgery of the upper respiratory and digestive tracts. Significance of a fentanyl-thiamine combination. Ann Anesthesiol Fr, 1976, 17: pp. 621–627.

    PubMed  CAS  Google Scholar 

  10. Lagler, F., et al., Toxikologische Untersuchungen mit Tramadol, einem neuen Analgetikum. Drug Res, 1978, 28 (I): pp. 164–172.

    CAS  Google Scholar 

  11. Cookson, R.F., Carfentanil and lofentanil. Clin & Anaesthesiol, 1983, 1: pp. 156–158.

    CAS  Google Scholar 

  12. Leysen, J.E., W. Gommeren, and C.J.E. Niemegeers, 3H-sufentanil, a superior ligand for the mu-opiate receptor: binding properties and regional distribution in rat brain and spinal cord. Eur J Pharmacol, 1983, 87: pp. 209–225.

    PubMed  CAS  Google Scholar 

  13. Niemegeers, C.J.E. and P.A.J. Janssen, Alfentanil (R 39 209) – a particularly short-acting narcotic analgesic in rats. Drug Dev Res, 1981, 1: pp. 83–88.

    CAS  Google Scholar 

  14. De Castro, J., Association des analgésiques centraux et des neuroleptiques en cours d’intervention, in Les analgésiques et la douleur. Influences pharmacologiques diverses exercées sur morphiniques, G. Vourch, et al., Editors, 1971, Masson: Paris. pp. 185–194; 383–403.

    Google Scholar 

  15. De Castro, J., S. Andrieu, and J. Boogaerts, Buprenorphine: a review of its pharmacological properties and therapeutical uses. New Drug Series, ed. J. De Castro (Eds) Vol. 1. 1982, Kluwer: Antwerpen NVM & ISA. 180.

    Google Scholar 

  16. Bovill, J.G., et al., The pharmacokinetics of alfentanil (R 39209): a new opioid analgesic. Anesthesiology, 1982, 57: pp. 439–443.

    PubMed  CAS  Google Scholar 

  17. Sebel, P.S., et al., Effects of high dose fentanyl anesthesia on the electroencephalogram. Anesthesiology, 1981, 55: pp. 203–211.

    PubMed  CAS  Google Scholar 

  18. Stanley, T.H. and S. de Lange, Comparison of sufentanil-oxygen and fentanyl-oxygen anesthesia for mitral and aortic valvular surgery. J Cardiothoracic Anesth, 1988, 2: pp. 6–11.

    CAS  Google Scholar 

  19. De Lange, S., et al., Comparison of sufentanil-02 and fentanyl-02 for coronary artery surgery. Anesthesiology, 1982, 56: pp. 112–118.

    PubMed  Google Scholar 

  20. Tolksdorf, W., et al., Adrenalin-, Noradrenalin-, Blutdruck- und Herzfreqenzverhalten während der Intubation in Abhängigkeit unterschiedlicher Fentanyl-Dosen. Anästh Intenivther Notfallmed, 1987, 22: pp. 171–176.

    CAS  Google Scholar 

  21. Stark, R.D., et al., A review of the safety and tolerance of propofol (Diprivan ). Postgrad Med J, 1985, 61: pp. 152–156.

    PubMed  Google Scholar 

  22. Grant, I.S. and N. MacKenzie, Recovery following propofol (Diprivan) anaesthesia-a review of three different anaesthetic techniques. Postgrad Med J, 1985, 61:pp. 133–137.

    PubMed  Google Scholar 

  23. Cockshott, I.D., Propofol (Diprivan) pharmacokinetics and metabolism-an overview. Postgrad Med J, 1985, 61: pp. 45–50.

    PubMed  CAS  Google Scholar 

  24. Becker, L.D., et al., Biphasic respiratory depression after fentanyl-droperidol or fentanyl alone used to supplement nitrous oxide anesthesia. Anesthesiology, 1976, 44: pp. 291–296.

    PubMed  CAS  Google Scholar 

  25. Habib, A.S. and T.J. Gan, Food and Drug Administration black box warning on the perioperative use of deoperidol: a review of the cases. Anesth Analg, 2003, 96: pp. 1377–1379.

    PubMed  Google Scholar 

  26. Kissin, I., et al., Alfentanil potentiates midazolam-induced unconsciousness in subanalgesic doses. Anesth Analg, 1990, 71: pp. 65–69.

    PubMed  CAS  Google Scholar 

  27. Oldendorf, H., et al., Clinical pharmacokinetics of midazolam in intensive care patients, a wide interpatient variability? Clin Pharmacol Ther, 1988, 43: pp. 263–269.

    Google Scholar 

  28. Geller, E., et al., Risk and benefits of therapy with flumazenil (Anexate ) in mixed drug intoxications. Eur Neurol, 1991, 31: pp. 241–250.

    PubMed  CAS  Google Scholar 

  29. Freye, E. and A. Fournell, Postoperative Demaskierung einer überhängenden Vigilanzminderung nach Midazolameinleitung durch den Antagonisten Flumazenil (Ro 15–1788). Anaesthesist, 1988, 37: pp. 162–166.

    PubMed  CAS  Google Scholar 

  30. Freye, E., B. Neruda, and K. Falke, Flumazenil (Anexate®) for the reversal of residual benzodiazepine activity. Drugs Today, 1989, 25: pp. 119–124.

    Google Scholar 

  31. Bergmann, S.A., et al., GABA agonists enhancer morphine and fentanyl antinociception in rabbit tooth pulp and mouse hot plate test. Drug Dev Res, 1988, 14: pp. 111–122.

    Google Scholar 

  32. Tejwani, G.A., et al., Inhibition of morphine-induced tolerance and dependence by a benzodiazepine receptor agonist midazolam in the rat. Anesth Analg, 1993, 76: pp. 1052–1060.

    PubMed  CAS  Google Scholar 

  33. Luger, T.J., et al., The spinal potentiation effect and the supraspinal inhibitory effect of midazolam on opioid-induced analgesia in rats. Eur J Pharmacol, 1995, 275: pp. 153–162.

    PubMed  CAS  Google Scholar 

  34. Rosland, J.H., S. Hunskaar, and K. Hole, Diazepam attenuates morphine antinociception test-dependently in mice. Pharmacol Toxicol, 1990, 66: pp. 382–386.

    PubMed  CAS  Google Scholar 

  35. Luger, T.J., et al., Mechanisms of the influence of midazolam on morphine antinociception at spinal and supraspinal levels in rats. Eur J Pharmacol, 1994, 271:pp. 421–431.

    PubMed  CAS  Google Scholar 

  36. Luger, T.J., H.F. Hill, and A. Schlager, Can midazolam diminish sufentanil analgesia in patients with major trauma? A retrospective study with 43 patients. Drug Metab Drug Interact, 1992, 10: pp. 177–184.

    CAS  Google Scholar 

  37. Michaels, I. and P.C. Barash, Does nitrous oxide or a reduced FI02 alter the hemodanymic function during high dose sufentanil anesthesia? Anesth Analg, 1983, 62: p. 275.

    Google Scholar 

  38. Murphy, M.R. and C.C. Hug, The enflurane sparing effect of morphine, butorphanol, and nalbuphine. Anesthesiology, 1982, 57: pp. 489–492.

    PubMed  CAS  Google Scholar 

  39. Hall, R.I., M.R. Murphy, and C.C. Hug, The enflurane sparing effect of sufentanil in dogs. Anesthesiology, 1987, 67: pp. 518–525.

    PubMed  CAS  Google Scholar 

  40. Murphy, M.R. and C.C. Hug, The anesthetic potency of fentanyl in terms of its reduction of enflurane MAC. Anesthesiology, 1982, 57: pp. 485–488.

    PubMed  CAS  Google Scholar 

  41. Hecker, B.R., et al., The decrease of the minimum alveolar anesthetic concentration produced by sufentanil in rats. Anesth Analg, 1983, 62: pp. 987–990.

    PubMed  CAS  Google Scholar 

  42. Hartung, E., E. Freye, and H. Dehnen-Seipel, Enflurane in cardiac surgery. Acta Anaesth Belg, 1982, 33: pp. 141–145.

    PubMed  CAS  Google Scholar 

  43. Hartung, E. and E. Freye, An open comparison of propofol and enflurane for prolonged abdominal operations. Anaesthesia, 1988, 43: pp. 105–107.

    PubMed  Google Scholar 

  44. Ismaily, A.J., et al., Die Auswirkungen einer Kombinationsanästhesie mit Fentanyl und Enfluran auf den Kreislauf und die unmittelbare postoperative Phase. Anäst Intensivmed, 1987, 28: pp. 216–220.

    Google Scholar 

  45. Hartung, H.J., Klinische Erfahrungen mit Alfentanil zur “balanced anesthesia” bei Oberbauch-Eingriffen. Anaesthesist, 1988, 37: pp. 620–624.

    PubMed  CAS  Google Scholar 

  46. Stanley, T.H., Comparison of alfentanil with thiopental sodium for induction of anesthesia. 1982, Janssen Pharmazeutika, Beerse, Belgien.

    Google Scholar 

  47. Smith, N.T., et al., An electroencephalographic comparison of alfentanil with other narcotics and with thiopental. J Clin Monit, 1985, 1: pp. 236–244.

    PubMed  CAS  Google Scholar 

  48. Redmond, D.E. and Y.H. Hwang, The primate locus coeruleus and affects of clonidine on opiate withdrawal. J Clin Psychiatr, 1982, 43: pp. 25–31.

    CAS  Google Scholar 

  49. Eisenach, J.C., S.Z. Lysak, and C.M. Viscomi, Epidural clonidine analgesia following sugery. Phase I. Anesthesiology, 1989, 71: pp. 640–646.

    PubMed  CAS  Google Scholar 

  50. Eisenach, J.C., et al., Epidural clonidine analgesia for intractable cancer pain. Phase I. Anesthesiology, 1989, 71: pp. 647–652.

    PubMed  CAS  Google Scholar 

  51. Eisenach, J.C., et al., The epidural clonidine study group: epidural clonidine analgesia for intractable cancer pain. Pain, 1995, 61: pp. 391–399.

    PubMed  CAS  Google Scholar 

  52. Flacke, J.W., et al., Reduced narcotic requirement by clonidine with improved hemodynamic and adrenergic stability in patients undergoing coronary bypass surgery. Anesthesiology, 1987, 67: pp. 11–19.

    PubMed  CAS  Google Scholar 

  53. Ghignone, M., Effects of clonidine on narcotic requirements and hemodynamic response during induction of fentanyl anesthesia and endotracheal intubation. Anesthesiology, 1987, 67: pp. 3–10.

    PubMed  CAS  Google Scholar 

  54. Aantaa, R., A. Kallio, and R. Virtanen, Dexmedetomidine, a novel alpha2-adrenergic agonist: A review of its pharmacodynamic profile. Drugs Future, 1993, 18: pp. 49–56.

    Google Scholar 

  55. Noyer, M., et al., Mivazerol, a novel compound with high binding specificity for alpha2 adrenergic receptor: binding studies on different human and rat membrane preparations. Neurochem Int, 1994, 24: pp. 221–229.

    PubMed  CAS  Google Scholar 

  56. McSPI-Europe, Perioperative sympathicolysis. Beneficial effects of the alpha2-agonist mivazerol on hemodynamic stability and myocardial ischemia. Anesthesiology, 1997, 86: pp. 346–363.

    Google Scholar 

  57. Carabine, U.A., K.R. Milligan, and J. Moore, Extradural clonidine and bupivacaine for postoperative analgesia. Br J Anaesthesia, 1992, 68 (2): pp. 132–135.

    CAS  Google Scholar 

  58. Motsch, J., E. Gräber, and K. Ludwig, Addition of clonidine enhances postoperative analgesia from epidural morphine; a double blind study. Anesthesiology, 1990, 73: pp. 1067–1073.

    PubMed  CAS  Google Scholar 

  59. Gabriel, A.H., et al., Clonidine: an adjunct in isoflurane N20/02 relaxant anaesthesia. Anaesthesia, 1995, 50: pp. 290–296.

    PubMed  CAS  Google Scholar 

  60. Engelman, E., et al., Effects of clonidine on anesthetic requirements and hemodynamic response during aortic surgery. Anesthesiolgy, 1989, 71: pp. 178–187.

    CAS  Google Scholar 

  61. Hoffmann, W.E., et al., Dexmedetomidine improves neurologic outcome from incomplete ischemia in the rat: reversal by the alpha2 antagonist atipamezole. Anesthesiology, 1991, 75: pp. 328–332.

    Google Scholar 

  62. Jarrot, B., et al., Clonidine: understanding its disposition, sites and mechanism of action. Clin Exp Pharmacol Physiol, 1987, 14: pp. 471–479.

    Google Scholar 

  63. Bjoerkqvist, S.E., Clonidine in alcohol withdrawal. Acta Psychiat Scand, 1975, 52: p. 256.

    Google Scholar 

  64. Kleber, H.D., et al., Clonidine and naltrexone in the outpatient treatment of heroin withdrawal. Am J Drug Alcohol Abuse, 1987, 13 (1, 2): pp. 1–17.

    PubMed  CAS  Google Scholar 

  65. Vining, D.H., T.R. Kosten, and H.D. Kleber, Clinical utility of rapid clonidine-naltrexone detoxification for opioid abusers. Br J Addict, 1988, 63: pp. 567–575.

    Google Scholar 

  66. Striebel, H.W., D. Koenigs, and T. Heil, Clonidin – Stellenwert in der Anästhesie. Anaesthesist, 1993, 42: pp. 131–141.

    PubMed  CAS  Google Scholar 

  67. Maze, M. and W. Tranquilli, Alpha-2 adrenoreceptor agonists: defining the role in clinical anesthesisa. Anesthesiology, 1991, 74: pp. 581–605.

    PubMed  CAS  Google Scholar 

  68. Cube von, B., et al., Permeation morphinartiger Substanzen an den Ort der antinociceptiven Wirkung im Gehirn in Abhängigkeit von ihrer Lipoidlöslichkeit nach intravenöser und nach intraventrikulärer Applikation. Naunyn-Schmiedebergs Arch Pharmacol, 1970, 265: pp. 455–473.

    Google Scholar 

  69. Prys-Roberts, C. and C.E. Hug, Pharmacokinetics of Anaesthesia. 1984, Blackwell: Oxford Scientific Publications.

    Google Scholar 

  70. Hug, C.C.J., Pharmacokinetics of new synthetic narcotic analgesics, in Opioids in Anesthesia, F.G. Estafanous, Editor, 1984, Butterworth: Boston. pp. 50–60.

    Google Scholar 

  71. Westmoreland, C., et al., Pharmacokinetics of remifentanil (GI87084B). Anesthesiology, 1993, 79 (3A): p. A372.

    Google Scholar 

  72. Hermann, D.J., et al., Pharmacokinetic comparison of GI87084B, a novel ultra-short acting opioid, and alfentanil. Anesthesiology, 1991, 75: p. A379.

    Google Scholar 

  73. Taeger, K., et al., Uptake of fentanyl by human lung. Anesthesiology, 1984, 61: p. A246.

    Google Scholar 

  74. Cube von, B., et al., Permeation of morphine-like substances to their site of antinociceptive action in the brain after intravenous and intraventricular application and dependence on lipid solubility. Arch Pharmacol, 1970, 265: pp. 455–502.

    Google Scholar 

  75. Stanski, D.R. and C.C. Hug, Alfentanil-a kinetically predictable narcotic analgesic. Anesthesiology, 1982, 57: pp. 435–438.

    PubMed  CAS  Google Scholar 

  76. Schenk, H.D., F.B.M. Ensink, and M. Rhönisch, Alfentanil – Porträt eines Opioids zur Anästhesie. 1993, Urban & Schwarzenberg: München-Wien-Baltimore. 1–144.

    Google Scholar 

  77. Scott, J.G., J.E. Cooke, and D.R. Stanski, Electroencephalographic quantitation of opioid effects: comparative pharmacodnamics of fentanyl and sufentanil. Anesthesiology, 1991, 74: pp. 34–42.

    PubMed  CAS  Google Scholar 

  78. Michiels, M., R. Hendricks, and J. Heykants, A sensitive radioimmunassay for fentanyl: plasma levels in dogs and man. Eur J Pharmacol, 1974, 12: p. 153.

    Google Scholar 

  79. Miller, R.D., IV drug delivery systems, in Anesthesia. 2000, Churchill Livingstone: Oxford. pp. 377–408.

    Google Scholar 

  80. Shafer, S.L. and S.R. Varvel, Pharmacokinetics, pharmacodynamics, and rational opioid selection. Anesthesiology, 1991, 74: pp. 1136–1138.

    Google Scholar 

  81. Scott, J.C., K.V. Ponganis, and D.R. Stanski, EEG quantification of narcotic effect: the comparative pharmacodynamics of fentanyl and alfentanil. Anesthesiology, 1985, 62: pp. 234–241.

    PubMed  CAS  Google Scholar 

  82. Lemmens, H.J.M., et al., The application of pharmacokinetics dynamics and computer simulations to drug development: A-3665 versus fentanyl and alfentanil. Anesthesiology, 1992, 77 (3A): p. A456.

    Google Scholar 

  83. Ausems, M.E., D.R. Stanski, and C.C.J. Hug, An evaluation of the accuracy of pharmacokinetic data for the computer assisted infusion of alfentanil. Br J Anaesth, 1985, 57: pp. 1217–1225.

    PubMed  CAS  Google Scholar 

  84. Egan, T.D., et al., The pharmacokinetics of the new short-acting opioid remifentanil (GI87084B) in healthy adult male volunteers. Anesthesiology, 1993, 79: pp. 881–892.

    PubMed  CAS  Google Scholar 

  85. McDonnell, T.E., R.R. Bartowski, and J.J. Williams, ED50 of alfentanil for induction of anesthesia in unpremedicated young adults. Anesthesiology, 1982, 57: p. A362.

    Google Scholar 

  86. Minto, C.F., T.W. Schnider, and S.L. Shafer, Pharmacokinetics and pharmacodynamics of remifentanil. II-Model application. Anesthesiology, 1997, 86: pp. 24–33.

    PubMed  CAS  Google Scholar 

  87. Hughes, M.A., P.S.A. Glass, and J.R. Jacobs, Context-sensitive half-time in multicompartment pharmacokinetic models for intravenous anesthetic drugs. Anesthesiology, 1992, 76: pp. 334–341.

    PubMed  CAS  Google Scholar 

  88. Egan, T.D., Remifentanil pharmacokinetics and pharmacodynamics. Clin Pharmacokinet, 1995, 29: pp. 80–94.

    PubMed  CAS  Google Scholar 

  89. Read, D.J.C., A clinical method for assessing the ventilatory response to carbon dioxide. Aust Ann Med, 1979, 16: pp. 20–32.

    Google Scholar 

  90. Suttmann, H. and A. Doenicke, Interim report on dose-establishment with alfentanil. 1983, Janssen Pharmaceutica, Beerse, Belgium.

    Google Scholar 

  91. Freye, E., E. Hartung, and R. Buhl, Alfentanil als letzte Dosis (on-top) in der Neuroleptanalgesie mit Fentanyl. Anaesthesist, 1986, 35: pp. 231–237.

    PubMed  CAS  Google Scholar 

  92. Nauta, J., et al., Anesthetic induction with alfentanil: a new short-acting narcotic analgesic. Anest Analg, 1982, 61: pp. 267–272.

    CAS  Google Scholar 

  93. Sebel, P.S., N. De Brujin, and W.K. Neville, Median nerve somatosensory evoked potentials during anesthesia with sufentanil or fentanyl. Anesthesiology, 1988, 69 (A312).

    Google Scholar 

  94. Kochs, E., et al., Modulation of pain-related somatosensory evoked potentials by general anesthesia. Anesth Analg, 1990, 71: pp. 225–2230.

    PubMed  CAS  Google Scholar 

  95. Freye, E. and B. Neruda, Averaged somatosensory evoked potentials for intraoperative evaluation of analgesia in patients. J Clin Monit, 1988, 4: pp. 138–139.

    Google Scholar 

  96. Rundshagen, I., E. Kochs, and J. Schulte am Esch, Surgical stimulation increases median nerve somatosensory evoked responses during isoflurane-nitrous oxide anaesthesia. Br J Anaesth, 1995 (75).

    Google Scholar 

  97. Freye, E., Somatosensorisch evozierte Potentiale (SEP) zur Algesiemetrie, in Alfentanil – Ein neues, ultrakurzwirkendes Opioid, M. Zindler and E. Hartung, Editors, 1985, Urban & Schwarzenberg: München-Wien-Baltimore. pp. 17–23.

    Google Scholar 

  98. Minto, C.F., et al., Influence of age and gender on the pharmacokinetics and pharmacodynamics of remifentanil. I.Model development. Anesthesiology, 1997, 86: pp. 10–13.

    PubMed  CAS  Google Scholar 

  99. Crawford, M.W., et al., Development of acute opioid tolerance during infusion of remifentanil for pediatric scoliosis surgery. Anwesth Analg, 2006, 102: pp. 1662–1667.

    CAS  Google Scholar 

  100. Guignard, B., et al., Acute opioid tolerance: intraoperative remifentanil increases postoperative pain and morphine requirements. Anesthesiology, 2000, 93: pp. 409–417.

    PubMed  CAS  Google Scholar 

  101. Albrecht, S., et al., Postoperative pain control following remifentanil-based anaesthesia for major abdomional surgery. Anaesthesia, 2000, 55: pp. 315–322.

    PubMed  CAS  Google Scholar 

  102. De Castro, J., et al., Comparative study of cardiovascular, neurological, and metabolic side effects of eight narcotics in dogs. Acta Anaesth Belg, 1979, 30: pp. 5–99.

    PubMed  Google Scholar 

  103. Van Bever, W.F.M., et al., N-4-substituted 1-(2arylethyl)-4-piperidinyl-N-phenylpropanamides, a novel series of extremely poteut analgesics with unusually high safety margin. Drug Res/Arzneimittelforsch, 1978, 26: pp. 1548–1551.

    Google Scholar 

  104. Niemegeers, C.J.E., et al., Sufentanil, a very potent and extremely safe intravenous morphine-like compound in mice, rats and dogs. Drug Res/Arzneimittelforsch, 1976, 216: pp. 1551–1556.

    Google Scholar 

  105. Janssen, P.A.J., The development of new synthetic narcotics, in Opioids in Anesthesia, F.G. Estafanous, Editor, 1984, Butterworth: Boston. pp. 37–44.

    Google Scholar 

  106. Tollenaere, J.P. and H. Moereels, Atlas of the three-dimensional structure of drugs. 1979, Amsterdam, New York, Elsevier: Oxford/North Holland Biomedical Press.

    Google Scholar 

  107. Magnan, J., et al., The binding spectrum of narcotic analgesic drugs with different agonist and antagonist properties. Naunyn-Schmiedebergs Arch Pharmacol, 1982, 319: pp. 197–205.

    PubMed  CAS  Google Scholar 

  108. Flacke, J.W., et al., Comparison of meperidine, fentanyl and sufentanil in balanced anesthesia. Anesth Analg, 1985, 64: pp. 897–910.

    PubMed  CAS  Google Scholar 

  109. De Lange, S., et al., Antidiuretic and growth hormone responses during coronary artery surgery with sufentanil-oxygen and alfentanil-oxygen anesthesia in man. Anesth Analg, 1982, 61: p. 434.

    PubMed  Google Scholar 

  110. De Lange, S., et al., Catecholamine and cortisol responses to sufentanil-O2 and alfentanil-O2 anaesthesia during coronary artery surgery. Can Anaesth Soc J, 1983, 30: p. 248.

    PubMed  Google Scholar 

  111. Bailey, P.L., et al., Sufentanil produces shorter lasting respiratory depression and longer lasting analgesia than equipotent doses of fentanyl in human volunteers. Anesthesiology, 1986, 65: p. A493.

    Google Scholar 

  112. Bailey, P.L., et al., Differences in magnitude and duration of opioid induced respiratory depression and analgesia with fentanyl and sufentanil. Anesth Analg, 1990, 70: pp. 8–15.

    PubMed  CAS  Google Scholar 

  113. Spiegel, K. and G.W. Pasternack, Meptazinol: a novel mu-1 selective opioid analgesic. J Pharmacol Expt Ther, 1984, 228: pp. 414–419.

    CAS  Google Scholar 

  114. Jaffe, J.H. and W.R. Martin, Opioid analgesics and antagonists, in The Pharmacological Basis of Therapeutics, A.F. Gilman, et al., Editors, 1990, Pergamon Press: New York. pp. 485–531.

    Google Scholar 

  115. Bovill, J.G., et al., Electroencephalographic effects of sufentanil anaesthesia in man. Br J Anaesth, 1982, 54: pp. 45–52.

    PubMed  CAS  Google Scholar 

  116. Bowdle, T.A. and R.J. Ward, Induction of anesthesia with small doses of sufentanil or fentanyl: dose versus EEG response, speed of onset, and thiopental requirement. Anesthesiology, 1989, 70: pp. 26–30.

    PubMed  CAS  Google Scholar 

  117. Hilgenberg, J.C., Intraoperative awareness during high dose fentanyl-oxygen anesthesia. Anesthesiology, 1981, 54: pp. 341–343.

    PubMed  CAS  Google Scholar 

  118. Mummaneni, N., T.L.K. Tao, and A. Montoya, Awareness and recall during high-dose fentanyl-oxygen anesthesia. Anesth Analg, 1980, 59: pp. 948–949.

    PubMed  CAS  Google Scholar 

  119. Freye, E. and E. Hartung, Kardiovaskuläre und zentralnervöse Effekte unter Fentanyl versus Sufentanil bei der Intubation herzchirurgischer Patienten. Anästhesie Aktuell, 1993, 9: pp. 3–14.

    Google Scholar 

  120. Brian, S.E. and A.B. Seifen, Tonic-clonic activity after sufentanil. Anesth Analg, 1987, 66: p. 481.

    PubMed  Google Scholar 

  121. Freye, E., E. Hartung, and R. Buhl, Die Lungencompliance wird durch die rasche Injektion von Alfentanil beeinträchtigt. Anaesthesist, 1986, 35: pp. 543–546.

    PubMed  CAS  Google Scholar 

  122. Smith, N.T., et al., EEGs during high-dose fentanyl, sufentanil-, or morphine-oxygen anesthesia. Anesth Analg, 1984, 63: p. 386.

    PubMed  CAS  Google Scholar 

  123. Katz, R.I., et al., Two instances of seizure-like activity in the same patient associated with two different narcotics. Anesth Analg, 1988, 67: p. 289.

    PubMed  CAS  Google Scholar 

  124. Young, W.L., et al., The effect of sufentanil on cerebral hemodyamics during carotid endarterectomy. Anesthesiology, 1988, 69: p. A591.

    Google Scholar 

  125. Keykhah, M.M., D.S. Smith, and C. Carlson, Influence of sufentanil on cerebral metabolism and circulation in the rat. Anesthesiology, 1985, 63: pp. 274–277.

    PubMed  CAS  Google Scholar 

  126. Werner, C., Der Einfluβ von Sufentanil auf die regionale und globale Hirndurchblutung und den zerebralen Sauerstoffverbrauch beim Hund. Anaesthesist, 1992, 41: pp. 34–38.

    PubMed  CAS  Google Scholar 

  127. Milde, L.N., J.H. Milde, and W.J. Gallagiter, Effects of sufentanil on cerebral circulation and metabolism in dogs. Anesth Analg, 1990, 70 (2): pp. 138–146.

    PubMed  CAS  Google Scholar 

  128. Weinstabl, C., et al., Effects of sufentanil on intracranial pressure in neurological patients. Anaesthesia, 1991, 46: pp. 837–840.

    PubMed  CAS  Google Scholar 

  129. Sperry, R.J., et al., Fentanyl and sufentanil increase intracranial pressure in head trauma patients. Anesthesiology, 1992, 77 (3): pp. 416–420.

    PubMed  CAS  Google Scholar 

  130. Stephan, H., et al., Einfluβ von Sufentanil auf Hirndurchblutung, Hirnstoffwechsel und die C02-Reaktivität der menschlichen Hirngefäβe. Anaesthesist, 1991, 40: pp. 153–160.

    PubMed  CAS  Google Scholar 

  131. Freye, E., et al., Slow EEG-power spectra correlate with hemodynamic changes during laryngoscopy and intubation following induction with fentanyl and sufentanil. Acta Anaesth Belg, 1999, 50: pp. 71–76.

    PubMed  CAS  Google Scholar 

  132. Hull, R.I., R.I. Murphy, and C.C. Hug, The enflurane sparing effect of sufentanil in dogs. Anesthesiology, 1987, 67: pp. 518–525.

    Google Scholar 

  133. Monk, J.P., R. Beresford, and A. Ward, Sufentanil: A review of its pharmacological properties and therapeutic use. Drugs, 1988, 36: pp. 286–313.

    PubMed  CAS  Google Scholar 

  134. Gravlee, G.P., F.M. Ramsey, and R.C. Roy, Rapid administration of a narcotic and a neuromuscular blocker: a hemodynamic comparison of fentanyl, sufentanil, pancuronium, and vecuronium. Anesth Analg, 1988, 67: pp. 39–47.

    PubMed  CAS  Google Scholar 

  135. Brizgys, R.V., R. Morales, and B. Owens, Effects of thiopental requirements and hemodynamic response during induction and intubation. Anesthesiology, 1985, 63: p. A377.

    Google Scholar 

  136. Lappas, D.G., et al., Filling pressures of the heart and pulmonary circulation of the patient with coronary artery disease after large doses of morphine. Anesthesiology, 1975, 42: p. 153.

    PubMed  CAS  Google Scholar 

  137. Lappas, D.G., I. Placios, and C. Athanasiadis, Sufentanil dosage and myocardial blood flow and metabolism in patients with coronary artery disease. Anesthesiology, 1985, 63: p. A58.

    Google Scholar 

  138. Becker, C.E., et al., A quick guide to common drug interaction, in Patient Care, J. Bigelow, Editor, 1974, Miller & Fink: Philadelphia. pp. 1–32.

    Google Scholar 

  139. Sifton, D.W., Drug Interaction and Side Effects Index . 42 ed. Physicians Desk Reference (PDR), ed. M. Trelewicz. 1988, Medical Economics Company Inc.: Oradell, N.Y. 1–787.

    Google Scholar 

  140. Coté, D., R. Martin, and J.P. Tétrault, Haemodynamic interactions of muscle relaxants and sufentanil in coronary artery surgery. Can J Anaesth, 1991, 38(3): pp. 324–329.

    PubMed  Google Scholar 

  141. Maurer, P.M. and R.R. Bartkowski, Drug interactions of clinical significance with opioid analgesics. Druf Safety, 1993, 8: pp. 30–48.

    CAS  Google Scholar 

  142. Bovill, J.G., et al., Electroencephalographic effects of sufentanil anaesthesia in man. Br J Anaesth, 1982, 54: pp. 45–52.

    PubMed  CAS  Google Scholar 

  143. Helmers, J.H., L. Van Leuwen, and W. Zuurmond, Sufentanil-Dosierungsstudie bei allgemeinen chrirurgischen Eingriffen. Anaesthesist, 1989, 38: pp. 397–400.

    PubMed  CAS  Google Scholar 

  144. Gerwig, W.H., C.W. Thompson, and P. Blades, Pain control following upper abdominal operations. Arch Surg, 1951, 62: pp. 678–682.

    Google Scholar 

  145. Morris, T. and J. Tracey, Lignocaine: its effect on wound healing. Br J Surg, 1977, 64: pp. 902–903.

    PubMed  CAS  Google Scholar 

  146. Dundee, J.W., Problems associated with strong analgesics, in Pain. New Perspectives in Measurement and Management, A.W. Harcus, R. Smith, and B. Whittle, Editors, 1977, Churchill Livingstone: Edinburgh, London, New York. pp. 57–62.

    Google Scholar 

  147. Ferrari, H.A., R.L. Fuson, and S.J. Dent, The relationship of the anaesthetic agent to postoperative analgesic requirements. South Med J, 1969, 62: pp. 1201–1203.

    PubMed  CAS  Google Scholar 

  148. Clark, N.J., et al., Comparison of sufentanil-N2O and fentanyl-N2O in patients without cardiac disease undergoing general surgery. Anesthesiology, 1987, 66: pp. 130–135.

    PubMed  CAS  Google Scholar 

  149. Suwatakul, K., et al., Analysis of narcotic analgesic usage in the treatment of postoperative pain. JAMA, 1983, 250: pp. 926–929.

    Google Scholar 

  150. Koo, P.J., Postoperative pain management with a patient-controlled transdermal delivery system for fentanyl. Am J Health Syst Pharm, 2005, 62: pp. 1171–1176.

    PubMed  CAS  Google Scholar 

  151. Striebel, H., J. Hackenberger, and A. Wesel, Beurteilung der postoperativen Schmerzintensität. Selbst- versus Fremdbeurteilung. Schmerz, 1992, 6: pp. 199–203.

    Google Scholar 

  152. Ripamonti, C., Pain experienced by patients hospitalized at the National Cancer Institute of Milan: Research project “towards a pain-free hospital”. Tumori, 2000, 86: pp. 412–418.

    PubMed  CAS  Google Scholar 

  153. Strohbücker, B., H. Mayer, and G. Evers, Schmerzprävalenz an den Unikliniken Köln: Vorkommen und Intensität von Schmerzen bei stationären Patienten. Unveröffentlichte Masterarbeit, 2001, Institut für Pflegewissenschaft, Medizinische Fakultät, Universität Witten/Herdecke.

    Google Scholar 

  154. Marks, R.M. and E.J. Sachar, Undertreatment of medical inpatients with narcotic analgesics. Ann Int Med, 1973, 78: pp. 173–181.

    PubMed  CAS  Google Scholar 

  155. Angell, M., The quality of mercy. New Engl J Med, 1982, 306: pp. 98–99.

    PubMed  CAS  Google Scholar 

  156. Porter, J. and H. Hick, Addiction rare in patients treated with narcotics. New Engl J Med, 1980, 302: pp. 123–126.

    PubMed  CAS  Google Scholar 

  157. Taub, A., Opioid analgesics in the treatment of chronic intractable pain of non-neoplastic origin, in Narcotic Analgesics in Anesthesiology, L.M. Kitahata and J.G. Collins, Editors, 1982, Williams and Wilkins: Baltimore. pp. 199–208.

    Google Scholar 

  158. Portenoy, R.K. and K.M. Foley, Chronic use of opioid analgesics in non-malignant pain: Report of 38 cases. Pain, 1986, 25: pp. 171–186.

    PubMed  CAS  Google Scholar 

  159. Eide, W.K., Wind-up and the NMDA receptor complex from a clinical perspective. Eur J Pain, 2000, 4: pp. 5–17.

    PubMed  CAS  Google Scholar 

  160. Barsoum, M.W., Comparison of the efficacy and tolarability of tramadol, pethidine and nalbuphine in children with postoperative pain. Clin Drug Invest, 1995, 9: pp. 183–190.

    Google Scholar 

  161. Quiding, H., et al., Infants and young children metabolise codeine to morphine. A study after single and repeated rectal administration. Br J Clin Pharmacol, 1992, 33: pp. 45–49.

    PubMed  CAS  Google Scholar 

  162. Moore, R.A. and H.J. McQuay, Single-patient data meta-analysis of 3453 postoperative patients: oral tramadol versus placebo, codeine and combination analgesics. Pain, 1997, 69: pp. 287–294.

    PubMed  CAS  Google Scholar 

  163. Lewis, J.W., Buprenorphine. Drug Alcohol Depend, 1985, 14: pp. 363–372.

    PubMed  CAS  Google Scholar 

  164. Heel, R.C., et al., Buprenorphine: a review of its pharmacological properties and therapeutic efficacy. Drugs, 1979, 17: pp. 81–100.

    PubMed  CAS  Google Scholar 

  165. Lewis, J.W. Structure-activity relationships of opioids – a current perspective. in VIII. International Symposium on Medicinal Chemistry. 1985, Stockholm: Swedish Pharmaceutical Press.

    Google Scholar 

  166. Krizanits-Weine, F., et al., Präemptive Analgesie mit retardiertem Dihydrocodein bei Patienten mit elektiven Kniearthroskopien. Schmerz, 1996, 10: p. S54.

    Google Scholar 

  167. Steffen, P., et al., Nichtinvasive perioperative Analgesie nach Allgemeinanästhesien. Kombination von Dihydrocodein ret. mit den Nichtopioidanalgetika Diclofenac und Metamizol. Schmerz, 1996, 10: p. S51.

    Google Scholar 

  168. Lotsch, J., et al., Fatal respiratory depression after multiple intravenous morphine injections. Clin Pharmacokinet, 2006, 45: pp. 1051–1060.

    PubMed  Google Scholar 

  169. Wen-Ying, C., et al., Human opioid receptor A118G polymorphism affects intravenous patient-controlled analgesia morphine consumption after total abdominal hysterectomy. Anesthesiology, 2006, 105: pp. 334–337.

    Google Scholar 

  170. Good, P., et al., Prospective audit of short-term concurrent ketamine, opioid and anti-inflammatory (‘triple-agent’) therapy for episodes of acute and chronic pain. Intern Med J, 2005, 35: pp. 39–44.

    PubMed  CAS  Google Scholar 

  171. Saarne, A., Clinical evaluation of a new analgesic piritramide. Acta Anaesthesiol Scand, 1969, 13: pp. 11–19.

    PubMed  CAS  Google Scholar 

  172. Schmidt, W.K., et al., Nalbuphine. Drug Alcohol Depend, 1985, 14: pp. 339–362.

    PubMed  CAS  Google Scholar 

  173. Wood, P.L., Kappa agonists analgesics: evidence for μ2 and delta opioid receptor antagonism. Drug Dev Res, 1984, 4: pp. 429–435.

    CAS  Google Scholar 

  174. Wermeling, D.P., et al., Patient-controlled analgesia using butorphanol for postoperative pain control: an open label study, in Butorphanol Tartrate: Research Advances in Multiple Clinical Settings, C.E. Rosow, Editor, 1986, S. Karger: Basel, Paris, London, New York, Singapore, Sydney. pp. 31–39.

    Google Scholar 

  175. Freye, E., L. Azevedo, and E. Hartung, Reversal of fentanyl-related respiratory depression with nalbuphine; effects on the CO2-response curve of man. Acta Anaesth Belg, 1985, 36: pp. 365–374.

    PubMed  CAS  Google Scholar 

  176. Magruder, M.R., R.D. DeLaney, and C.A. DiFazio, Reversal of narcotic-induced respiratory depression with nalbuphine hydrochloride. Anesthesiol Rev, 1982, 9: pp. 34–37.

    Google Scholar 

  177. McCammon, R.L., R.K. Stoelting, and J.A. Madura, Effects of butorphanol, nalbuphine, and fentanyl on intrabiliary tract dynamics. Anesth Analg, 1984, 63: pp. 139–142.

    PubMed  CAS  Google Scholar 

  178. Romagnoli, A. and A.S. Keats, Ceiling effect for respiratory depression by nalbuphine. Clin Pharmacol Ther, 1980, 27: pp. 478–485.

    PubMed  CAS  Google Scholar 

  179. Herz, A., Opiat-Partialantagonisten, in Pentazocin im Spiegel der Erfahrungen, S. Kubicki and G.A. Neuhaus, Editors, 1981, Springer: Berlin-Heidelberg-New York. pp. 19–21.

    Google Scholar 

  180. Gal, T.J., C.A. Di Fazo, and J. Moscicki, Analgesic and respiratory depressant activity of nalbuphine: a comparison with morphine. Anesthesiology, 1982, 57: pp. 367–374.

    Google Scholar 

  181. Abboud, T.K., et al., Transnasal butorphanol: a new method for pain relief in post-cesarean section pain. Acta Anaesthesiol Scand, 1991, 35: pp. 14–18.

    PubMed  CAS  Google Scholar 

  182. Wetchler, B.V., C.D. Alexander, and M.A. Uhll, Transnasal butorphanol tartrate for pain control following ambulatory surgery. Curr Ther Res, 1992, 52 (4): pp. 571–580.

    Google Scholar 

  183. Joyce III, T.H., et al., Efficacy of transnasal butorphanol tartrate in postepisiotomy pain: a model to assess analgesia. Clin Therap, 1993, 15 (1): pp. 160–167.

    Google Scholar 

  184. Shyu, W.C., et al., Multiple-dose phase I study of transnasal butorphanol. Clin Pharmacol Ther, 1993, 54: pp. 34–41.

    PubMed  CAS  Google Scholar 

  185. Couch, J., et al. Evaluation of the efficacy and safety of Stadol® NS (transnasal butorphanol) in the treatment of acute migraine in outpatients. in 7th World Congress on Pain. 1993, Paris: ISAP Publications.

    Google Scholar 

  186. Diamond, S., et al., Transnasal butorphanol in the treatment of migraine headache pain. Headache Quat Curr Treat Res, 1992, 3 (2): pp. 164–171.

    Google Scholar 

  187. Morrison, C.E., et al., Pethidine compared with meptazinol during labour; a prospective randomised double-blind study in 1100 patients. Anaesthesia, 1987, 42: pp. 7–14.

    PubMed  CAS  Google Scholar 

  188. Jordan, C., A comparison of the respiratory effects of meptazinol, pentazocine and morphine. Br J Anaesth, 1979, 51: pp. 497–501.

    PubMed  CAS  Google Scholar 

  189. Keeri-Szanto, M., Drugs or drums: what relieves postoperative pain? Pain, 1979, 6: pp. 217–230.

    PubMed  CAS  Google Scholar 

  190. Eagen, K.J. and L. Ready, Patient satisfaction with intravenous PCA or epidural morphine. Can J Anaesth, 1994, 41: pp. 6–11.

    Google Scholar 

  191. White, P.F., Mishaps with patient-controlled analgesia. Anesthesiology, 1987, 66: pp. 81–83.

    PubMed  CAS  Google Scholar 

  192. Thomas, D.W. and H. Owen, Patient-controlled analgesia – the need for caution. Anaesthesia, 1988, 43: pp. 770–772.

    PubMed  CAS  Google Scholar 

  193. Parker, R.K., B. Holtmann, and P.F. White, Patient-controlled analgesia: does a concurrent opioid infusion improve pain mangement after surgery? JAMA, 1991, 266 (14): pp. 1947–1952.

    PubMed  CAS  Google Scholar 

  194. Lehmann, K.A., Neue Möglichkeiten zur Behandlung akuter Schmerzen. Drug Res Arzneimittelforsch, 1984, 34: pp. 1108–1114.

    CAS  Google Scholar 

  195. Ginsberg, B., et al., The influence of lockout intervals and drug selection on patient-controlled analgesia following gynecological surgery. Pain, 1995, 62: pp. 95–100.

    PubMed  CAS  Google Scholar 

  196. Butscher, K., J.Y. Mazoit, and K. Samii, Can immediate opioid requirements in the post-anaesthesia care unit be used to determine analgesic requirements on the ward? Can J Anaesth, 1995, 42: pp. 461–466.

    PubMed  CAS  Google Scholar 

  197. Krimmer, H., et al., Die kombinierte infusionsanalgesie – Ein alternatives Konzept zur postoperativen Schmerztherapie. Chirurg, 1986, 57: pp. 327–329.

    PubMed  CAS  Google Scholar 

  198. Viscusi, E.R., et al., Non-invasive, patient-controlled, fentanyl HCl analgesia: comparison of safety and efficacy to intravenous morphine pump for the treatment of postoperative pain after major surgery: a randomized, multi-center trial. Anesth Analg, 2002, 94: p. S. 224.

    Google Scholar 

  199. Friedman, J.D. and F.A. Dello Buono, Opioid antagonists in the treatment of opioid-induced constipation and pruritus. Am Pharmacother, 2003, 35: pp. 85–91.

    Google Scholar 

  200. Kurz, A. and D.I. Sessler, Opioid-induced bowel dysfunction; pathophysiology and potential new therapies. Drugs, 2003, 63: pp. 649–671.

    PubMed  CAS  Google Scholar 

  201. Liu, S.S., et al., ADL 8-2698, a trans-3, 4-dimethyl-4-(3-hydroxyphenyl) piperidine, prevents gastrointestinal effects of intravenous morphine without affecting analgesia. Clin Pharmacol Ther, 2001, 69: pp. 66–71.

    PubMed  CAS  Google Scholar 

  202. Holte, K. and H. Kehlet, Prevention of postoperativ ileus. Minerva Anesthesiol, 2002, 68: pp. 152–156.

    CAS  Google Scholar 

  203. Taguchi, A., et al., Selective postoperative inhiibition of gastrointestinal opioid receptors. N Engl J Med, 2001, 345: pp. 935–940.

    PubMed  CAS  Google Scholar 

  204. Schmidt, W.K., Alvimopan* (ADL 8-2698) is a novel peripheral opioid antagonist. Am J Surg, 2001, 182: pp. 27S–38S.

    PubMed  CAS  Google Scholar 

  205. Yuan, C.S. and J.F. Foss, Methylnaltrexone: investigation of clinical applications. Drug Dev Res, 2000, 50: pp. 133–141.

    CAS  Google Scholar 

  206. Yuan, C.S., et al., The safety and efficacy of oral methylnaltrexone in preventing morphine-induced delay in oral cecal transit time. Clin Pharmacol Ther, 1997, 61: pp. 1–9.

    Google Scholar 

  207. Yuan, C.S. and J.F. Foss, Oral methylnaltrexone for opioid-induced constipation. JAMA, 2000, 284: pp. 1383–1384.

    PubMed  CAS  Google Scholar 

  208. Yuan, C.S., et al., Methylnaltrexone prevents morphine-induced delay on oral-cecal transit time without affecting analgesia: a double-blind randomized placebo-controlled trial. Clin Pharmacol Ther, 1996, 59: pp. 469–475.

    PubMed  CAS  Google Scholar 

  209. Stevens, C.W., et al., Biochemical characterization and regional quantification of mu, delta, and kappa opioid binding sites in rat spinal cord. Brain Res, 1991, 550: pp. 77–85.

    PubMed  CAS  Google Scholar 

  210. Rawal, N., Klinischer Einsatz der rückenmarknahen Opioidanalgesie, Teil 1. Der Schmerz, 1996, 10: pp. 176–189.

    PubMed  CAS  Google Scholar 

  211. Börner, U., et al., Epidurale Opiatanalgesie – Gewebe und Liquorverträglichkeit der Opiate. Anaesthesist, 1980, 29: pp. 570–571.

    PubMed  Google Scholar 

  212. Bürkle, H., S. Dunbar, and H. van Aken, Remifentanil: a novel, short-acting, μ-opioid. Anesth Analg, 1996, 83: pp. 646–651.

    PubMed  Google Scholar 

  213. Rawal, N. and B. Tandon, Epidural and intrathecal morphine in intensive care units. Intensive Care Med, 1985, 11: pp. 129–135.

    PubMed  CAS  Google Scholar 

  214. De Castro, J. and L. Lecron, Peridurale Opiatanalgesie: Verschiedene Opiate – Komplikationen und Nebenwirkungen, in Peridurale Opiatanalgesie, M. Zenz, Editor, 1981, G. Fischer: Stuttgart, New York. pp. 103–107.

    Google Scholar 

  215. Moore, R.A., et al., Dural permeability to narcotics: in vitro determination and application to extradural administration. Br J Anaesth, 1982, 54: pp. 1117–1127.

    PubMed  CAS  Google Scholar 

  216. Leicht, C.H., et al., Evaluation and comparison of epidural sufentanil citrate and morphine sulfate for analgesia after cesarean section. Anesthesiology, 1986, 65: pp. A 365.

    Google Scholar 

  217. Camporesi, E.M., C.H. Nielsen, and P.R. Bromage, Ventilatory CO2 sensitivity after intravenous and epidural morphine in volunteers. Anesth Analg, 1983, 62: p. 633.

    PubMed  CAS  Google Scholar 

  218. Bromage, P.R., et al., Rostral spread of epidural morphine. Anesthesiology, 1982, 56: pp. 431–436.

    PubMed  CAS  Google Scholar 

  219. Johnson, A., et al., Influence of intrathecal morphine and naloxone intervention on postoperative ventilatory regulation in elderly patients. Acta Anaethesiol Scand, 1992, 36 (5): pp. 436–444.

    CAS  Google Scholar 

  220. McCaughey, W. and I.L. Graham, The respiratory depression of epidural morphine: time course and effect of posture. Anaesthesia, 1982, 37: pp. 990–994.

    PubMed  CAS  Google Scholar 

  221. Jaffe, J.H. and W.R. Martin, Opioid analgesics and antagonists, in The pharmacological Basis of Therapeutics, A.G. Gilman, et al., Editors, 1993, McGraw Hill: New York. pp. 485–531.

    Google Scholar 

  222. Glynn, C.I., L.E. Mather, and M.E. Cousins, Spinal narcotics and respiratory depression. Lancet, 1979, 2: p. 356.

    PubMed  CAS  Google Scholar 

  223. Cohen, S.E., S. Tan, and P.F. White, Sufentanil analgesia following cesarean section: epidural versus intravenous administration. Anesthesiology, 1988, 68: pp. 129–134.

    PubMed  CAS  Google Scholar 

  224. Davies, G.K., C.L. Tolhurst-Cleaver, and T.L. James, Respiratory depression after intrathecal opiates. Anaesthesia, 1980, 35: p. 1080.

    PubMed  CAS  Google Scholar 

  225. Gourlay, G.K., et al., Pharmacokinetics of fentanyl in lumbar and cervical CSF following lumbar epidural and intravenous administration. Pain, 1989, 38:pp. 253–259.

    PubMed  CAS  Google Scholar 

  226. Chalmer, P.C., C.M. Lang, and B.B. Greenhouse, The use of nalbuphine in association with epidural narcotics. Anesthesiol Rev, 1988, 15(2): pp. 21–27.

    Google Scholar 

  227. Cheng, E.Y. and J. May, Nalbuphine reversal of repiratory depression after epidural sufentanil. Crit Care Med, 1989, 17: pp. 378–379.

    PubMed  CAS  Google Scholar 

  228. Bromage, P.R., Camporesi, E.M., and P.A.C. Durant, Nonrespiratory side effects of epidural morphine. Anesth Analg, 1982, 61: p. 490.

    PubMed  CAS  Google Scholar 

  229. Rawal, N., K. Möllefors, and K. Axelsson, Naloxone reversal of urinary retention after epidural morphine. Lancet, 1981, 2: p. 1411.

    PubMed  CAS  Google Scholar 

  230. Cousins, M.J. and L.E. Mather, Intrathecal and epidural administration of opioids. Anesthesiology, 1984, 61: pp. 276–310.

    PubMed  CAS  Google Scholar 

  231. Rawal, N. and M. Wattwil, Respiratory depression after epidural morphine – an experimental and clinical study. Anesth Analg, 1982, 63: p. 8.

    Google Scholar 

  232. Eisenach, J.C., Epidural and spinal narcotics, in ASA Refresher Courses in Anesthesiology, P.G. Barash, Editor, 1992, Lippincott: Philadelphia. pp. 1–4.

    Google Scholar 

  233. Bailey, D.R. and B.E. Smith, Continuous epidural infuson of fentanyl for postoperative analgesia. Anesthesiology, 1980, 42: p. 538.

    Google Scholar 

  234. Gjessing, J. and P.J. Tomlin, Postoperative pain control with intrathecal morphine. Anaesthesia, 1981, 36: p. 268.

    PubMed  CAS  Google Scholar 

  235. Kitahata, L.M. and J.G. Collins, Spinal action of narcotic analgesics. Anesthesiology, 1981, 54: p. 153.

    PubMed  CAS  Google Scholar 

  236. Rutter, D.V., D.G. Skewes, and M. Morgan, Extradural opioids for postoperative analgesia: a double blind comparison of pethidine, fentanyl and morphine. Br J Anaesth, 1981, 53: p. 915.

    PubMed  CAS  Google Scholar 

  237. Weightman, W.M., Respiratory arrest during epidural infusion of bupivacaine and fentanyl. Anaesth Int Care, 1991, 19: pp. 282–284.

    CAS  Google Scholar 

  238. Waldvogel, H.H., and M. Fasano, Extradural administration of lofentanyl for balanced postoperative pain. Anaesthesist, 1983, 32: pp. 256–257.

    Google Scholar 

  239. Herz, A. and H.J. Teschemacher, Activities and site of antinociceptive action of morphine-like analgesics. Adv Drug Res, 1971, 6: pp. 79–119.

    CAS  Google Scholar 

  240. Howell, S.B., Clinical application of a novel sustained-release injectable drug delivery system DepoFoam technology. Cancer J, 2001, 7: pp. 219–227.

    PubMed  CAS  Google Scholar 

  241. Viscusi, E.R., et al., EREM Study Group: fourty-eight hours of postoperativ pain relief following total hip arthroplasty with a novel, extend-release epidural morphine formulation. Anesthesiology, 2005, 102: pp. 1014–1022.

    PubMed  CAS  Google Scholar 

  242. Carvallo, B., E. Riley, and G. Manvelian, Phase II study of long-acting encapsulated epidural morphine for postoperative pain after cesarean section. Reg Anesth Pain Med, 2004, 29: p. A37.

    Google Scholar 

  243. Gambling, D.R., et al., A compararive study of patient controllled epidural analgsia (PCEA) and continuous infusion epeidural analgesia (CIEA) during labour. Can J Anaesth, 1988, 35: pp. 249–254.

    PubMed  CAS  Google Scholar 

  244. Velickovic, I. and G. Leicht, Patient-controlled epidural analgesia for labor and delivery in parturient with chronic inflammatory demyelating polyneuropathy. Reg Anesth and Pain Med, 2001, 27: pp. 217–219.

    Google Scholar 

  245. Zenz, M., S. Piepenbrock, and M. Tryba, Epidural opiates: long-term experiences in cancer pain. Klin Wochenschr, 1985, 63: pp. 225–229.

    PubMed  CAS  Google Scholar 

  246. Durant, P.A.C. and T.L. Yaksh, Epidural injections of bupivacaine, morphine, fentanyl, lofentanil, and DADL in chronically implanted rats: a pharmacologic and pathologic study. Anesthesiology, 1986, 64: pp. 43–53.

    PubMed  CAS  Google Scholar 

  247. Camann, W.R., et al., A comparison of intrathecal, epidural, and intravenous sufentanil for labor analgesia. Anesthesiology, 1992, 77: pp. 884–892.

    PubMed  CAS  Google Scholar 

  248. McQuay, H.J., et al., Intrathecal opioids, potency, and lipophilicity. Pain, 1989, 36: pp. 111–115.

    PubMed  CAS  Google Scholar 

  249. Hansdottir, V., et al., The CSF and plasma pharmacokinetics of sufentanil after intrathecal administration. Anesthesiology, 1991, 74: pp. 264–269.

    PubMed  CAS  Google Scholar 

  250. D’Angelo, R., et al., Intrathecal sufentanil compared to epidural bupivacaine for labor analgesia. Anesthesiology, 1994, 80: pp. 1209–1215.

    PubMed  CAS  Google Scholar 

  251. Jones, R.D.M. and J.G. Jones, Intrathecal morphine: naloxone reverses respiratory depression but not analgesia. Br Med J, 1980, 281: pp. 645–646.

    PubMed  CAS  Google Scholar 

  252. Meignier, M., et al., Continuous intrathecal opioids and bupivcaine for the management of intractable cancer pain in children. Anesth Analg, 1993, 76: p. S259.

    Google Scholar 

  253. Brown, D.V. and R.J. McCarthy, Epidural and spinal opioids. Curr Opinion Anaesth, 1995, 8: pp. 337–341.

    Google Scholar 

  254. Mok, M.S., et al. Analgesic effect of intrathecal stadol, nubain, meperidine, morphine and fentanyl, a comparative study. in VIII. World Congress of Anaesthesiologists. 1984, Manila/Philippines.

    Google Scholar 

  255. Kalia, P.K., et al., Epidural pentazocine for postoperative pain relief. Anesth Analg, 1983, 62: p. 949.

    PubMed  CAS  Google Scholar 

  256. Malinovsky, J.M., et al., Ketamine and midazolam neurotoxicity in the rabbit. Anesthesiology, 1991, 75: pp. 91–97.

    PubMed  CAS  Google Scholar 

  257. Wilcox, G.L., et al., Mutual potentiation of antinociceptive effects of morphine and clonidine on motor and sensory responses in rat spinal cord. Brain Res, 1987, 405: pp. 84–93.

    PubMed  CAS  Google Scholar 

  258. Ossipov, M.H., L.J. Suarez, and T.C. Spaulding, Antinociceptive interactions between alpha2-adrenergic and opiate agonists at the spinal level of rodents. Anesth Analg, 1989, 68: pp. 194–200.

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

Copyright information

© 2008 Springer Science+Business Media B.V.

About this chapter

Cite this chapter

Freye, E., Levy, J.V. (2008). Opioids, an Integrative Part in Perioperative Medicine. In: Opioids in Medicine. Springer, Dordrecht. https://doi.org/10.1007/978-1-4020-5947-6_3

Download citation

  • DOI: https://doi.org/10.1007/978-1-4020-5947-6_3

  • Publisher Name: Springer, Dordrecht

  • Print ISBN: 978-1-4020-5946-9

  • Online ISBN: 978-1-4020-5947-6

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics