Skip to main content

Opioide im Rahmen der Allgemeinanästhesie

  • Chapter
Opioide in der Medizin
  • 71 Accesses

Zusammenfassung

Das Gebiet der Medizin, in dem sich der Einsatz der Opioide in immer größerem Rahmen durchgesetzt hat, ist die Anästhesiologie. Speziell für den intraoperativen Einsatz hat die Verwendung starkwirkender, zentraler Analgetika dazu geführt, dass die Sicherheit der Narkose zugenommen hat und störende Nebenwirkungen von Seiten des kardiovaskulären Systems, wie sie von anderen Narkoseverfahren mit Barbituraten und oder volatilen Anästhetika (Halothan, Enfluran, Isofluran, Desfluran, Sevofluran) bekannt sind (Abb. 19-1), kaum nachzuweisen sind.

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 54.99
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever

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.

Literatur

  1. Craig DB (1981) Postoperative recovery of pulmonary function. Anesth Anaig 60: 46–52

    CAS  Google Scholar 

  2. Spence AA (1980) Postoperative pulmonary complications in general anesthesia. In: Gray TC, Nunn JF, Utting JE (eds), vol 1. Butterworth, London, pp 591–608

    Google Scholar 

  3. Bonica JJ (1983) Current status of postoperative pain therapy. In: Yokota T, Dubner R (eds) Current topics in pain research and therapy. Exerpta Medica, Tokio, pp 169–189

    Google Scholar 

  4. Modig J (1982) Thromembolism and blood loss: Continuous epidural vs. general anesthesia with v-controlled ventilation. Reg Anesth 7: S84 - S88

    Google Scholar 

  5. Cohen FL (1980) Postsurgical pain relief. Patients status and nurses’ medication. Pain 9: 265–274

    Article  PubMed  CAS  Google Scholar 

  6. Marks RM, Sachar EJ (1973) Undertreatment of medical inpatients with narcotic analgesics. Ann Int Med 78: 173–181

    PubMed  CAS  Google Scholar 

  7. Angell M (1982) The quality of mercy. N Engl J Med 306: 98–99

    Article  PubMed  CAS  Google Scholar 

  8. Sehrt U (1985) Fragwürdiger Analgetikamissbrauch. MMW 127: 825–826

    Google Scholar 

  9. Mindach M (2000) Keine Opioidabhängigkeit bei Schmerzpatienten? Schmerz 14: 186–191

    Article  PubMed  CAS  Google Scholar 

  10. Zimmermann M, Handwerker HO (1984) Schmerz, Konzepte und Ärztliches Handeln. Springer, Berlin Heidelberg New York Tokio

    Google Scholar 

  11. Jurna I (1992) Zentrale Schmerzdämpfung durch peripher wirkende Analgetika. Der Schmerz 6: 61–66

    Article  PubMed  CAS  Google Scholar 

  12. Rexed B (1964) Some aspects of the cytoarchitectonics and synaptology of the spinal cord. Brain Res 11: 58–92

    Article  CAS  Google Scholar 

  13. Yaksh T (1999) Spinal systems and pain processing: development of novel analgesic drugs with mechanistically defined models. Trends Pharmacol Sei 20: 329–337

    Article  CAS  Google Scholar 

  14. Goodman RR, Snyder SH (1982) Autoradiographic localization of kappa opiate receptors to deep layers of the cerebral cortex may explain unique sedative and analgesic effects. Life Sei 31: 1291–1294

    Article  CAS  Google Scholar 

  15. Yaksh TL (1981) Spinal opiate analgesics. Characteristics and principles of action. Anesthesiology 11: 293–346

    CAS  Google Scholar 

  16. Yaksh TL, Howe JR (1982) Opiate receptors and their definition by antagonists. Anesthesiology 56: 246–249

    Article  PubMed  CAS  Google Scholar 

  17. Yaksh TL (1983) In vivo studies on the spinal opiate receptor systems mediating antinociception. I. ju and 5 receptor profiles in the primate. J Pharmacol Exp Ther 226: 303–316

    PubMed  CAS  Google Scholar 

  18. Cousins MJ, Cherry DA, Gourlay GK (1988) Acute and chronc pain: use of spinal opioids. In: Cousins MJ, Briden-baugh PO (eds) neural blockade in clinical anesthesia and management of pain. Lippincott, Philadelphia, pp 955–1029

    Google Scholar 

  19. Chattipakorn SC, Light AR, Willockson HH, Närhi M, Meix-ner W (1999) The effect of fentanyl on c-fos expression on the trigeminal brainstem complex produced by pulpal heat stimulation in the ferret. Pain 82: 207–215

    Article  PubMed  CAS  Google Scholar 

  20. Wall PD (1988) The prevention of postoperative pain. Pain 33: 289–290

    Article  PubMed  CAS  Google Scholar 

  21. Wall PD, Woolf CJ (1986) The brief and prolonged facila-tory effect of unmyelinated afferent input on the rat spinal cord are independently influenced by peripheral nerve section. Neuroscience 17: 1199–11205

    Article  PubMed  CAS  Google Scholar 

  22. Katz J, Kavanagh BP, Sandler AN et al. (1992) Preemptive analgesia-clinical evidence of neuroplasticity contributing to postoperative pain. Anesthesiology 77: 439–446

    Article  PubMed  CAS  Google Scholar 

  23. McQuay HJ (1992) Pre-emptive analgesia. Br J Anaesth 66: 1–2

    Article  Google Scholar 

  24. Kleckner NW, Dingledine R (1988) Requirement of glycine in ativation of NMDA receptors expressed in Xenoptus oocytes. Science 241: 835–837

    Article  PubMed  CAS  Google Scholar 

  25. Maurset A, Moberg ER, Oye I (1990) The analgesic effect of ketamine: Evidence for a non-opioid, PCP receptor mediated mechanism. In: Domino EF (ed) Status of ketamine in anesthesiology. NPP Books, Ann Arbor, pp 239–246

    Google Scholar 

  26. Smith DJ, Bouchal RL (1981) Ketamine interacts with dysphoric sigma opiate receptors. Anesthesiology 55: A234

    Article  Google Scholar 

  27. Meiler ST, Gebhart GF (1993) Nitric oxide ( NO) and nociceptive processing in the spinal cord. Pain 52: 127–136

    Google Scholar 

  28. Przewlocki R, Machelska H, Przewlocka B (1993) Inhibition of nitric oxide synthase enhances morphine antinociception in the rat spinal cord. Life Sei 53: 1–5

    Article  Google Scholar 

  29. Kolesnikov YA, Pick CG, Ciszewska G, Pasternack GW (1993) Blockade of tolerance to morphine but not to k opioids by a nitric oxide synthese inhibitor. Proc Natl Acad Sei USA 90: 5162–5166

    Article  CAS  Google Scholar 

  30. Basbaum AI, Fields HL (1984) Endogenous pain control systems: Brainstem spinal pathways and endorphin circuitry. Annu Rev Neurosci 7: 309–338

    Google Scholar 

  31. Fields KL, Anderson SD (1978) Evidence that raphe-spinal neurons mediate opiate and midbrain stimulation produced analgesia. Pain 5: 333–349

    Article  PubMed  CAS  Google Scholar 

  32. Wiklund L, Behzadi G, Kahlen P, Headley PM, Nicolopoulos LS (1988) Autoradiographic and electrophysiological evidence for exicitatory amino acid transmission in the periaqueductal gray projection to nucleus raphe magnus in the rat. Neurosci Lett 93: 158–163

    Article  PubMed  CAS  Google Scholar 

  33. Urban MO, Smith DJ (1993) Role of neurotensin in the nucleus raphe magnus in opioid-induced antinociception from the periaqueductal gray. J Pharmacol Expt Ther 265: 580–586

    CAS  Google Scholar 

  34. Melzack R, Wall PC (1965) Pain mechanisms: A new theory. Science 150: 971

    Google Scholar 

  35. Zimmermann M (1979) Neurophysiology of nociception, pain and pain therapy. In: Bonica JJ, Ventafridda V (eds) Advances in pain research, vol 2. Raven, New York, pp 13–23

    Google Scholar 

  36. Tölle TR, Schadrack J, Castro-Lopes JM, Evan G, Roques BP, Zieglgänsberger W (1994) Effects of kelartorphan and morphine before and after noxious stimulation on immediate-early gene expression in rat spinal cord neurons. Pain 56: 103–112

    Article  PubMed  Google Scholar 

  37. Ebersberger A, Anton F, Tolle TR, Zieglgansberger W (1995) Morphine, 5-HT2 and 5-HT3 receptor antagonists reduce c-fos expression in the trigeminal nuclear complex following noxious chemical stmulation of the rat nasal mucosa. Brain Res 676: 336–342

    Article  PubMed  CAS  Google Scholar 

  38. Kayser V, Fournie-Zaluski MC, Guilbaud G, Roques BP (1989) Potent antinociceptive effects of kelatorphan, a highly efficient inhibitor of multiple enkaphalin-degra-ding enzymes, systemically administered in normal and arthritic rats. Brain Res 497: 94–101

    Article  PubMed  CAS  Google Scholar 

  39. Hammond DL, Preslez R, Gogas KR, Basaum Al (1992) Morphine or U-50,488H suppresses fos proteine-like immuno-reactivity in the spinal cord and the nucleus tractus solita-rii evoked by noxious visceral stimulus in the rat. J Comp Neurol 315: 244–253

    Article  PubMed  CAS  Google Scholar 

  40. Higuchi T, Yamazaki O, Takazawa A et al. (1986) Effects of carbamazepine and valporic acid on brain immunore-active somatostatin and gamma-aminobutyric acid in amygdaloid-kindled rats. Eur J Pharmacol 125: 169–175

    Article  PubMed  CAS  Google Scholar 

  41. Tolle TR, Castro-Lopes JM, Schadrack J, Evan G, Zieglgansberger W (1996) Anticonvulsants suppress c-fos protein expression in spinal cord neurons following noxious thermal stimulation. Expt Neurol 132: 271–278

    Article  Google Scholar 

  42. Sawynok J, La Bella FS (1982) On the involvement of GABA in the analgesia produced by baclofen, muscimol and morphine. Neuropharmacology 21: 397–404

    Article  PubMed  CAS  Google Scholar 

  43. Castro-Lopes JM, Tolle TR, Pan NB, Ziegelgansberger W (1994) Expression of GRAD mRNA in spinal cord neurons of normal and monoarthritic rats. Mol Brain Res 26: 169–176

    Article  PubMed  CAS  Google Scholar 

  44. Hammond DL, Drowner EJ (1984) Effects of intrathecal administerd THIP, baclofen and muscimol on nociceptive threshold. Eur J Pharmacol 103: 121–125

    Article  PubMed  CAS  Google Scholar 

  45. Hao JX, Xu XJ, Yu YX, Seiger A, Wisenfeld-Hallin Z (1992) Baclofen reverses the hypersensitivity of dorsal horn dynamic range neurons to mechanical stimulation after transient spinal cord ischemia: implication for a tonic GABAergic inhibitory control of myelinated fiber input. J Neurophysiol 68: 392–396

    Google Scholar 

  46. Aigouy I, Fondras IC, Pajot J, Schoeffler P, Woda A (1992) Intrathecal midazolam vs. intrathecal morphine in orofacial nociception-an experimental study in rats. Neurosci Lett 139: 97–99

    Article  PubMed  CAS  Google Scholar 

  47. Guilbaud G, Benoist JM, Levante A, Gautron M, Wilier JC (1992) Primary somatosensory cortex in rats with pain-re-lated behaviors due to peripheral mononeuropathy after moderate ligation of one sciatic nerve: neuronal responsi-vity to somatic stimulation. Exp Brain Res 92: 227–245

    Article  PubMed  CAS  Google Scholar 

  48. Jensen TS, Yaksh TL (1992) Brainstem excitatory amino acid receptors in nociception: mircroinjection mapping and pharmacological characterization of glutamate-sensi-tive sites in brainstem associated with algogenic behavior. Neuroscience 442: 513–526

    Google Scholar 

  49. Mc Kenzie JS, Beechy NR (1962) The effects of morphine and pethidine on somatic evoked responses in the midbrain of the cat, and their relevance to analgesia. Electro-enceph Clin Neurophysiol 14: 501–519

    Article  Google Scholar 

  50. Simantov R, Snowman AM, Snyder SH (1976) A morphinelike factor »enkephalin« in rat brain: subcellular localization. Brain Res 107: 650–655

    Article  PubMed  CAS  Google Scholar 

  51. Hong JS, Yang HY, Fratta W, Costa E (1977) Determination of methionine enkephalin in discrete regions of rat brain. Brain Res 134: 383

    Article  PubMed  CAS  Google Scholar 

  52. Hassler R (1976) Über die antagonistischen Systeme der Schmerzempfindung und des Schmerzgefühls im peripheren und zentralen Nervensystem. In: Kubicki S, Neuhaus GA (Hrsg) Pentazocin im Spiegel der Entwöhnung. Springer, Berlin Heidelberg New York, pp 1–17

    Google Scholar 

  53. Hassler R (1976) Wechselwirkungen zwischen dem System der schnellen Schmerzempfindung und dem des langsamen, nachhaltigen Schmerzgefühl. Arch Klin Chir 342: 47

    CAS  Google Scholar 

  54. Pert CB, Snyder SH (1973) Opiate receptor: Demonstration in nervous tissue. Science 179: 1011–1014

    Google Scholar 

  55. Kuhar MJ, Pert CB, Snyder SH (1973) Regional distribution of opiate receptor binding in monkey and human brain. Nature 245: 447–450

    Article  PubMed  CAS  Google Scholar 

  56. Delia Bella D, Casacci F, Sassi A (1978) Opiate receptors: Different ligand affinity in various brain regions. Adv Bio-chem Psychopharmacol 18: 271–277

    Google Scholar 

  57. Pasternak GW, Wood PJ (1986) Minireview: Multiple mu opiate receptors. Life Sei 38: 1889–1898

    Google Scholar 

  58. Pasternak GW (1988) Multiple morphine and enkephalin receptors and the relief of pain. JAMA 259: 1362–1367

    Article  PubMed  CAS  Google Scholar 

  59. Wood PL, Charlson SE, Laue D, Hudgin RL (1981) Multiple opiate receptors: Differential binding of jn, k and 6 agonists. Neuropharmacology 20: 1215–1220

    Article  PubMed  CAS  Google Scholar 

  60. Wood PL (1982) Multiple opiate receptors: Support for unique jj, ö and k sites. Neuropharmacology 21: 487–497

    Article  PubMed  CAS  Google Scholar 

  61. Corbett D, Paterson SJ, Kosterlitz HW (1993) Selectivity of ligands for opioid receptors. In: Herz A (ed) Opioids I. Handbook of experimental pharmacology, vol 104/1. Springer, Berlin Heidelberg New York, pp 645–680

    Google Scholar 

  62. Perry DC, Rosenbaum JS, Kursowski M, Sadee W (1982) 3H-Etorphine receptor binding in vivo: Small fractional occupancy elicits analgesia. J Pharmacol Exp Ther 21: 272–279

    Google Scholar 

  63. Rosenbaum JS, Holford NHG, Sadee W (1984) Opiate receptor binding-effect relationship: Sufentanil and etor-phine produce analgesia at the ju-site with low fractional receptor occupancy. Brain Res 291: 317–324

    Google Scholar 

  64. Sadee W, Richards ML, Grevel J, Rosenbaum SJ (1983) In vivo characterization of four types of opioid binding sites in rat brain. Life Sei 33: 187–189

    Article  CAS  Google Scholar 

  65. Freye E (1987) Opioide in der Anästhesiologie. Wirkeffekte und klinische Anwendung. Urban and Schwarzenberg, München Wien Baltimore

    Google Scholar 

  66. Raffa RB, Friedrichs E, Reimann W, Shank RP, Codd EE, Vaught JL (1992) Opioid and nonopioid components independently contribute to the mechanism of action of tramadol, an »atypical« opioid analgesic. J Pharmacol Exp Ther 260 /1: 275–285

    PubMed  CAS  Google Scholar 

  67. Yanagita T (1978) Drug dependence potential of 1-(m-methoxyphenyl)-2-(-dimethylaminomethyl-cyclohe-xan-1)-hydrochlorid (tramadol) tested in monkeys. Drug Res 28: 159–163

    Google Scholar 

  68. Martin WR, Eades GG, Thompson JA, Huppier RE, Gilbert PE (1976) The effects of morphine and nalorphine-like drugs in the non-dependant and morphine-dependant chronic spinal dog. J Pharmacol Exp Ther 197: 517–532

    PubMed  CAS  Google Scholar 

  69. Sircar R, Zukin SR (1983) Characterization of specific sigma opiate/phencyclidine ( PCP)-binding sites in the human brain. Life Sei 33: 259–262

    Google Scholar 

  70. Finck AD, Nagai SH (1981) Ketamine interacts with opiate receptors in vivo. Anesthesiology 55: A241

    Google Scholar 

  71. Finck AD, Nagai SH (1981) Ketamine effects in opiate receptor bioassay. Anesthesiology 55: A242

    Google Scholar 

  72. Schmidt WK, Tarn SW, Shotzberger GS, Smith DH, Clark R, Vernier VG (1985) Nalbuphine. Drug Alcohol Depend 14: 339–362

    Article  CAS  Google Scholar 

  73. Schaal M, Freye E, Windelschmidt R (1986) Tifluadom, ein Benzodiazepin mit opioidartigem Wirkcharakter: Eine Untersuchung zum zentralen Wirkmechanismus an wachen Hunden. Z EEG-EMG 17: 27–31

    Google Scholar 

  74. De Castro J, Andrieu S, Boogaerts J (1982) Buprenorphine. A review of its pharmacological properties and therapeutical uses. De Castro J (ed) New Drug Series, vol 1. Kluwer NVM and ISA, Antwerpen

    Google Scholar 

  75. Freye E (1987) Opioid Agonists, Antagonists and Mixed Narcotic Analgesics. Theoretical background and considerations for practical use. Springer, Berlin Heidelberg New York Tokio

    Google Scholar 

  76. Romagnoli A, Keats AS (1980) Ceiling effect for respiratory depression by nalbuphine. Clin Pharmacol Ther 27: 478–485

    Article  PubMed  CAS  Google Scholar 

  77. Dhawan BN, Cesselin F, Raghubir R et al. (1996) International Union of Pharmacology. XII. Classification of opioid receptors. Pharmacol Rev 48: 567–592

    PubMed  CAS  Google Scholar 

  78. Noble F, Cox BM (1995) Differential regulation of D1 dopamine receptor and A2a adenosine receptor-stimulated adenyl-cyclase by ¡j, ö1; 52-opioid agonists in rat caudate putamen. J Neurochem 65: 125–133

    Article  PubMed  CAS  Google Scholar 

  79. Rossi GC, Leventhal L, Pan Y-X et al. (1997) Antisense mapping of MOR-1 in rats: distinguishing between morphine and morphine-6ß-glucuronide antinociception. J Parmacol Expt Ther 281: 109–114

    CAS  Google Scholar 

  80. Pan YX, Xu J, Bolan E et al. (1999) Indentification and characterization of three new alternatively spliced ju-opioid receptor isoforms. Mol Pharm 56: 396–403

    CAS  Google Scholar 

  81. Blake AD, Bot G, Reisine T (1996) Structure-function analysis of the cloned opiate receptors. Peptide and small molecule interaction. Chem Biol 3: 967–972

    Google Scholar 

  82. Henderson C, McKnight AT (1997) The orphan opioid receptor and its endogenous ligand-nociceptin/orphanin FQ. Trends Pharmacol Sei 18: 293–300

    Article  CAS  Google Scholar 

  83. Walker JR, Koob GF (1997) Commentary: orphan anxiety. Proc Natl Acad Sei USA 94: 14217–14219

    Article  CAS  Google Scholar 

  84. Nicolson JR, Paterson SJ, Menzies JRW, Corbett AD, McKnight AT (1998) Pharmacological studies on the »orphan« opioid receptor in central and peripheral sites. Can J Physiol Pharmacol 76: 304–313

    Article  Google Scholar 

  85. Jhamandas KH, Sutak M, Henderson G (1998) Antinocicep-tine and morphine modulatory actions of spinal orphanin FQ. Can J Physiol Pharmacol 76: 314–324

    Article  PubMed  CAS  Google Scholar 

  86. Foote RW, Maurer R (1982) Autoradiographic localization of opiate lat. K-receptors in the guinea pig brain. Eur J Pharmacol 85: 99–103

    Google Scholar 

  87. Freye E (1987) Neue Aspekte in der Schmerztherapie mit Opioiden. Anaesthesiol Reanimat 12: 3–17

    CAS  Google Scholar 

  88. Woolf CJ, Mannion RJ (1999) Neuropathie pain: aetiology, symptoms, mechanisms, and management. Lancet 353: 1959–1964

    Article  PubMed  CAS  Google Scholar 

  89. Ferreira SH (1981) Prostaglandins, aspirin-like drugs and analgesics. Nature New Biol 240: 200–203

    Google Scholar 

  90. Ventafridda V, Tamburini M, Caraceni A, De Conno F, Naldi F. A validation study of the WHO method of cancer pain relief. Cancer 1987 59: 851–856

    Article  Google Scholar 

  91. Wesselmann U, Burnett AL, Heinberg LJ. The urogenital and rectal pain syndromes. Pain 1997 73: 269–294

    Article  PubMed  CAS  Google Scholar 

  92. Maier C, Hildebrandt J (1991) Leserbrief zu dem Editorial von M. Zimmermann: Opioide für nicht tumorbedingte chronische Schmerzen. Schmerz 5: 90–91

    Google Scholar 

  93. Zech D (1991) Leserbrief zu dem Editorial von M. Zimmermann: Opioide für nicht tumorbedingte chronische Schmerzen? Schmerz 5: 89–90

    Article  PubMed  CAS  Google Scholar 

  94. Zenz M, Strumpf M, Willweber-Strumpf A (1991) Erwiederungen zu den vorstehenden Leserbriefen von D. Zech, C. Maier und J. Hildebrandt. Schmerz 5: 91–94

    Google Scholar 

  95. Zenz M, Strumpf M, Willweber-Strumpf A (1990) Orale Opiattherapie bei Patienten mit »nicht-malignen« Schmerzen. Schmerz 4: 14–21

    Article  PubMed  CAS  Google Scholar 

  96. Sorge J, Steffmann B, Lehmkuhl C, Pichlmayr I (1991) Opioidanalgetika bei »nichtmalignen «Schmerzen–Lang-zeitbehandlungsergebnisse bei Patienten mit rheumatischen Beschwerden. Schmerz 5: 60–66

    Article  PubMed  CAS  Google Scholar 

  97. Portenoy RK (1996) Opioid therapy for chronic nonmalig-nant pain: A review of the critical issues. J Pain Symptom Manage 11: 203–217

    Article  PubMed  CAS  Google Scholar 

  98. Dertwinkel R, Wiebalck A, Zenz M, Strumpf M (1996) Orale Opioide zur Therapie chronischer Nicht-Tumorschmerzen. Anästhesist 45: 495–505

    Article  CAS  Google Scholar 

  99. Florez J, Mediavilla A (1978) Respiratory and cardiovascular effects of met-enkephalin applied to the ventral surface of the brain stem. Brain Res 138: 585–590

    Article  Google Scholar 

  100. Ngai SH (1961) The effects of morphine and meperidine on the central respiratory mechanisms in the cat: The action of levallorphan in antagonizing these effects. J Pharmacol Exp Ther 131: 91–102

    PubMed  CAS  Google Scholar 

  101. Freye E, Härtung E (1981) Fentanyl in the fourth cerebral ventricle causes respiratory depression in the anesthetized but not in the awake dog. Acta Anaesthesiol Scand 25: 171–173

    Article  PubMed  CAS  Google Scholar 

  102. Freye E, Härtung E, Kalibe S (1983) Prevention of late fen-tanyl-induced respiratory depression after the injection of the opiate antagonists naltrexone and S-20682 as compared to naloxone. Br J Anaesth 55: 71–77

    Article  PubMed  CAS  Google Scholar 

  103. Stoeckel H, Hengstmann JH, Schüttler J (1979) Pharmacokinetics of fentanyl as a possible explanation for recurrence of respiratory depression. Br J Anaesth 51: 741

    Article  PubMed  CAS  Google Scholar 

  104. Freye E, Azevedo L, Härtung E (1985) Reversal of fentanyl-related respiratory depression with nalbuphine; effects on the C02-response curve of man. Acta Anaesthesiol Belg 36: 365–374

    PubMed  CAS  Google Scholar 

  105. Magruder MR, DeLaney RD, DiFazio CA (1982) Reversal of narcotic-induced respiratory depression with nalbuphine hydrochloride. Anesthesiol Rev 9: 34–37

    Google Scholar 

  106. Clark NJ, Meulemann T, Liu WS, Zwaniken P, Pace NL, Stanley TH (1987) Comparison of sufentanil-N20 and fen-tanyl-N20 in patients without cardiac disease undergoing general surgery. Anesthesiology 66: 130–135

    Article  PubMed  CAS  Google Scholar 

  107. Bailey PL, Streisand JB, East KA et al. (1990) Differences in magnitude and duration of opioid induced respiratory depression and analgesia with fentanyl and sufentanil. Anesth Analg 70: 8–15

    Article  PubMed  CAS  Google Scholar 

  108. Meert TF (1995) Mögliche funktionelle Rolle verschiedener Opioidrezeptoren. In: Henschel WF (Hrsg) II. Europäisches Analgesieforum. Urban and Schwarzenberg, München, Wien, Baltimore, pp 18–29

    Google Scholar 

  109. Vaught JL, Rothman RB, Westfall TC (1982) Mu and delta receptors: their role in analgesia and in the differential effects of opioid peptides on analgesia. Life Sei 30: 1443–1455

    Article  CAS  Google Scholar 

  110. Freye E, Schnitzler M, Schenk G (1991) Opioid-induced respiratory depression and analgesia may be mediated by different subreceptors. Pharm Res 8: 196–199

    Article  PubMed  CAS  Google Scholar 

  111. Freye E, Latasch L (1992) The opioid ö-reeeptor is involved in respiratory depression. Proceedings International Narcotics Research Conference. Keystone, Colorado, USA, p 147

    Google Scholar 

  112. Magnan J, Paterson SJ, Tavani A, Kosterlitz HW (1982) The binding spectrum of narcotic analgesic drugs with different agonist and antagonist properties. Naunyn-Schmie-debergs Arch Pharmacol 319: 197–205

    Article  CAS  Google Scholar 

  113. Sofuoglu M, Portoghese PS, Takemori AE (1990) Differential antagonism of delta receptor antagonists by naltrin-dole and its benzufuran analog ( NTB) in mice. Pharmacol Exp Ther 257: 676–680

    Google Scholar 

  114. Takemori AE, Portoghese PS (1992) Selective naltrexone-derived opioid receptor antagonists. Annu Rev Pharmacol Toxicol 32: 239–269

    Article  PubMed  CAS  Google Scholar 

  115. He L, Lee N (1998) Delta opioid receptor enhancement of mu opioid receptor-induced antinociception in spinal cord. J Pharmacol Exp Ther 285: 1181–1186

    PubMed  CAS  Google Scholar 

  116. Hughes MA, Glass PSA, Jacobs JR (1992) Context-sensitive half-time in multicompartment pharmacokinetic models for intravenous anesthetic drugs. Anesthesiology 76: 334–341

    Article  PubMed  CAS  Google Scholar 

  117. Egan TD, Lemmens HJM, Fiset P et al. (1992) The pharmacokinetics and pharmacodynamics of GI87084B. Anesthesiology 77/3 A: A369

    Google Scholar 

  118. Fink BR (1961) Influence of cerebral activity in wakefulness on regulation of breathing. J Appl Physiol 16: 15–23

    PubMed  CAS  Google Scholar 

  119. Sullivan CE, Murphy E, Kozar IE, Philipson EA (1978) Waking and ventilatory responses to laryngeal stimulation in sleeping dogs. J Appl Physiol 45: 681–688

    PubMed  CAS  Google Scholar 

  120. Latasch L, Christ R (1991) Respiratory safety. In: Lehmann K, Zech D (eds) Transdermal fentanyl. Springer, Berlin Heidellberg New York Tokio, pp 149–157

    Chapter  Google Scholar 

  121. De Castro J (1971) Association des analgésiques centraux et des neuroleptiques en cours d’intervention. In: Vourch G, De Castro J, Gauthier-Lafaye P, Guidicelli JF, Viars P (éds) Les analgésiques et la douleur. Influences pharma-cologiques diverses exercées sur morphiniques. Masson, Paris, pp 185–194

    Google Scholar 

  122. Becker LD, Paulson BA, Miller RD, Severinghaus FU, Eger El (1976) Biphasic respiratory depression after fentanyl-droperidol or fentanyl alone used to supplement nitrous oxide anesthesia. Anesthesiology 44: 291–296

    Article  PubMed  CAS  Google Scholar 

  123. Corail IM, Moore AR, Strunin L (1980) Plasma concentrations of fentanyl in normal surgical patients and those with severe renal and hepatic disease. Br J Anaesth 52: 101–110

    Google Scholar 

  124. Harper MH, Hickey RF, Cromwell TH, Linword S (1976) The magnitude and the duration of respiratory depression produced by fentanyl and fentanyl plus droperidol in man. J Pharmacol Exp Ther 199: 464–455

    PubMed  CAS  Google Scholar 

  125. Lehmann KA, Weski C, Hunger L, Heinrich C, Daub D (1982) Biotransformation von Fentanyl. II. Akute Arzneimittelinteraktion - Untersuchungen bei Ratte und Mensch. Anästhesist 31: 221

    Google Scholar 

  126. Schaer H, Baasch K, Reist F (1978) Die Atemdepression mit Fentanyl und ihre Antagonisierung mit Naloxon. Anästhesist 27: 259

    CAS  Google Scholar 

  127. Elstrom J (1977) Plasma protein binding of Phenytoin after cholecystectomy and neurosurgical operations. Acta Neur Scand 55: 455

    Article  Google Scholar 

  128. Gibaldi M, Perrier D (1975) Pharmacokinetics. Marcel Dekker, New York

    Google Scholar 

  129. MacClain DA, Hug CCJ (1980) Intravenous fentanyl kinetics. Clin Pharmacol Ther 28: 106

    Article  Google Scholar 

  130. Olson GD, Bennett WM, Porter GA (1975) Morphine and Phenytoin binding to human plasma protein in renal and hepatic failure. Clin Pharmacol Ther 17: 677

    Google Scholar 

  131. Lehmann KA, Freier J, Daub D (1982) Fentanyl-Pharma-kokinetik und postoperative Atemdepression. Anästhesist 31: 111

    CAS  Google Scholar 

  132. De Castro J, Viars P (1968) Utilisation pratique des analgésiques centraux en anesthésie et réanimation. Ars Med 23: 1–228

    Google Scholar 

  133. Freye E, Härtung E (1993) Kardiovaskuläre und zentralnervöse Effekte unter Fentanyl vs. Sufentanil bei der Intubation herzchirurgischer Patienten. Anästhesie Aktuell 9: 3–14

    Google Scholar 

  134. Moruzzi G, Magoun HW (1949) Brain stem reticular formation and activation of the EEG. Electroenceph Clin Neuro-physiol 1: 455–473

    CAS  Google Scholar 

  135. Akimoto H, Yamaguchi N, Okabe Ki et al. (1956) On the sleep induced through electrical stimulation in dog thalamus. Folia Psychiatr Neurol Jpn 10: 117–146

    Google Scholar 

  136. Quock RM, Mueller J (1991) Protection by U-50,488H against ß-chlornaltrexamine antagonism of nitrous oxide antinociception in mice. Brain Res 549: 162–164

    Article  PubMed  CAS  Google Scholar 

  137. Fink AD, Ngai SH, Berkowitz BA (1977) Antagonism of general anesthesia by naloxone in the rat. Anesthesiology 46: 241–245

    Article  Google Scholar 

  138. Freye E, Härtung E, Schenk GK (1983) Naloxone reverses the hypnotic effect and the depressed baroreceptor reflex of halothane-anaesthesia in the dog. Can Anaesth Soc J 8: 235–241

    Article  Google Scholar 

  139. Römer D, Büscher H, Hill RC et al. (1980) Bremazocine: A potent, long-acting opiate kappa-agonist. Life Sei 27: 971–978

    Article  Google Scholar 

  140. Freye E, Härtung E, Schenk GK (1983) Bremazocine: An opiate which induces sedation and analgesia but no respiratory depression. Anesth Analg 62: 483–488

    Google Scholar 

  141. Freye E, Boeck A, Ciramelli F (1986) The benzodiazepine (+)tifluadom (KC-6128) and not its optical isomer (KC-5911) induces opioid kappa-receptor related EEG power spectra and evoked potential changes. Pharmacology 33: 241–248

    Article  PubMed  CAS  Google Scholar 

  142. Von Voigtlander PF, Lahti RA, Ludeus JH (1983) U-50,488H: A seletive and structurally novel non-mu (kappa) agonist. J Pharmacol Exp Ther 224: 7–12

    Google Scholar 

  143. Maurer R, Cortes R, Probst A, Palacios JM (1983) Multiple opiate receptors in human brain. Life Sei 17: 231–234

    Article  Google Scholar 

  144. Wevers A, Schmidt P, Cserpan E et al. (1995) Cellular distribution of the mRNA for the K-opiod receptor in the human neocortex: a non-isotopic in situ hybridization study. Neurosci Lett 195: 1–4

    Article  Google Scholar 

  145. Wood PL (1984) k Agonists Analgesics: Evidence for \\2 and 6 opioid receptor antagonism. Drug Dev Res 4: 429–435

    Google Scholar 

  146. Wood PL, Sanschagrin D, Richard JW, Thakur M (1983) Multiple opiate receptor affinities of kappa and agonist/ antagonist analgesics: in vivo assessment. J Pharmacol Expt Ther 226: 545–550

    CAS  Google Scholar 

  147. Cheng EY, May J (1989) Nalbuphine reversal of respiratory depression after epidural sufentanil. Crit Care Med 17: 378–379

    Article  PubMed  CAS  Google Scholar 

  148. Pfeiffer A, Pasi A, Mehrain P, Herz A (1982) Opiate receptor binding sites in human brain. Brain Res 248: 87–96

    Article  PubMed  CAS  Google Scholar 

  149. Di Fiazo CA, Moscicki JC, Magruder MR (1981) Anesthetic potency of nalbuphine and interaction with morphine in rats. Anesth Analg 60: 629–622

    Google Scholar 

  150. Dumas PA (1984) MAC reduction of enflurane and isoflu-rane and postoperative findings with nalbuphine HCl and fentanyl: A retrospective study. In: Gomez QJ (ed) VI Ith World Congress of Anaesthesiologists. Manila/Philippines: Exerpta Medica, Amsterdam, pp 43–53

    Google Scholar 

  151. Murphy MR, Hug CC (1982) The enflurane sparing effect of morphine, butorphanol, and nalbuphine. Anesthesiology 57: 489–492

    Article  PubMed  CAS  Google Scholar 

  152. Ghoneim MM, Dhanarrj J, Choi WW (1984) Comparison of four opiod analgesics as supplements to nitrous oxide analgesia. Anesth Analg 63: 405–412

    Article  PubMed  CAS  Google Scholar 

  153. Bovill JG, Sebel PS, Wauquier A, Rog P (1982) Electroence-phalographic effects of sufentanil anaesthesia in man. Br J Anaesth 54: 45–52

    Article  PubMed  CAS  Google Scholar 

  154. Smith NT, Dec-Siilver H, Sanford TJ et al. (1984) EEGs during high-dose fentanyl, sufentanil-, or morphine-oxygen anesthesia. Anesth Analg 63: 386

    PubMed  CAS  Google Scholar 

  155. Freye E, Schinagel A, Härtung E (1985) EEG-Powerspektren und Plasmakonzen trationen von Alfentanil. In: Zindler M, Härtung E (Hrsg) Alfentanil–Ein neues, ultra kurzwirkendes Opioid. Urban and Schwarzenberg, München Wien Baltimore, pp 45–51

    Google Scholar 

  156. Bovill JG, Sebel S, Wauquier A, Rog P (1982) Electroence-phalographic effects of sufentanil anaesthesia in man. Br J Anaesth 54: 45–52

    Article  PubMed  CAS  Google Scholar 

  157. Bovill JG, Sebel S, Wauquier A, Rog P, Schuyt HC (1983) The influence of high dose alfentanil anaesthesia on the electroencephalogram: Correlation with plasma concentrations. Br J Anaesth 55: 199 S-209 S

    Google Scholar 

  158. Scott JC, Ponganis KV, Stanski DR (1985) EEG quantification of narcotic effect: The comparative pharmacodynamics of fentanyl and alfentanil. Anesthesiology 62: 234–241

    Google Scholar 

  159. Souza EB, Schmidt WK, Kuhar MJ (1987) Nalbuphine: an autoradiographic opioid receptor binding profile in the central nervous system on an agonist/antagonist analgesic. J Pharmacol Expt Ther 244: 391–402

    Google Scholar 

  160. Herz A, Teschemacher HJ (1971) Activities and site of antinociceptive action of morphine-like analgesics. Adv Drug Res 6: 79–119

    CAS  Google Scholar 

  161. Awouters F, Niemegeers CJE, Megens AAHP, Meert TF, Janssen PAJ (1988) Pharmacological profile of ritanerin: a very specific central serotonin S2-antagonist. Drug Dev Res 15: 61–73

    Article  CAS  Google Scholar 

  162. Leysen JE, Gommeren W, Niemegeers CJE (1983) 3H-su-fentanil, a superior ligand for the mu-opiate receptor: Binding properties and regional distribution in rat brain and spinal cord. Eur J Pharmacol 87: 209–225

    Google Scholar 

  163. Niemegeers CJE, Janssen PAJ (1981) Alfentanil (R 39 209)-a particularly short-acting narcotic analgesic in rats. Drug Dev Res 1: 83–88

    Article  CAS  Google Scholar 

  164. Van Bever WFM, Niemegeers C, Janssen P, Van Ree J (1978) N-4-substituted1 -(2arylethyl)-4-piperidinyl-N-phe-nylpropanamides, a novel series of extremely potent analgesics with unusually high safety margin. Arzneimittel-forsch/Drug Res 26: 1548–1551

    Google Scholar 

  165. Nilsson E, Ingvar DH (1967) EEG findings in neuroleptanalgesia. Acta Anaesth Scand 11: 121–127

    Article  PubMed  CAS  Google Scholar 

  166. Ingvar DH, Nilsson E (1961) Central nervous effects of neuroleptanalgesia as induced by haloperidol and phen-operidine. Acta anaesth Scand 5: 85–88

    Article  PubMed  CAS  Google Scholar 

  167. Kubicki S, P. Z. (1966) EEG-Veränderungen durch Neuro-leptanalgesie. Anästh Wiederbelebg 9: 44–49

    Google Scholar 

  168. Kubicki S, Stölzel R, Otten J, Hass J (1970) Exzitatorische und inhibitorische Phänomene am Zentralnervensystem, verursacht durch Fentanyl. In: Henschel WF (Hrsg) Neue Klinische Aspekte der Neuroleptanalgesie. Schattauer, Stuttgart New York, pp 21–30

    Google Scholar 

  169. Hassler R (1972) Über die Zweiteilung der Schmerzempfindung und des Schmerzgefühl. In: Janzen R, Keidel WD, Herz A, Steichele C (Hrsg) Schmerz. Thieme, Stuttgart, p 105

    Google Scholar 

  170. Freye E (1982) Opiatrezeptoren im Gehirn. Perimed Fachbuchverlagsgesellschaft, Erlangen

    Google Scholar 

  171. Freye E, Arndt JO (1979) Perfusion of the fourth cerebral ventricle with fentanyl induces naloxone reversible hypotension, bradycardia, baroreflex depression and sleep in unanaesthetized dogs. Naunyn-Schmiedebergs Arch Pharmacol 307: 123–128

    Article  PubMed  CAS  Google Scholar 

  172. Lord JAH, Waterfield AS, Hughes J, Kosterlitz HW (1977) Endogenous opioid peptides: multiple agonists and receptors. Nature 267: 495–499

    Article  PubMed  CAS  Google Scholar 

  173. Smith TW, Hughes J, Kosterlitz HW, Sosa RP (1976) Enkephalins: Isolation, distribution and function. In: Kosterlitz HW (Ed) Opiates and Endogenous Opioid Peptides. Elsevier, Amsterdam, pp 57–62

    Google Scholar 

  174. Czontos DL, Helmne RD (1983) Enkephalins and endorphins; clinical pharmacological and therapeutic implications. Drugs 26: 503–519

    Article  Google Scholar 

  175. Freye E, Schenk GK (1982) Die praktische Bedeutung endogener Opioide ( Endorphine ). Anästhesiol Intensiv-med 23: 280–290

    Google Scholar 

  176. Holaday JW, D’Amatao RJ (1983) Multiple opioid receptors: evidence for mu-delta binding site interactions in endotoxic shock. Life Sei 33: 703–706

    Article  CAS  Google Scholar 

  177. Long JB, Ruvio BA, Holaday JW (1984) ICI 174,864, a novel delta antagonist, reverses endotoxic shock: Pretreatment with dynorphin (1–13) a kappa agonist, blocks this therapeutic effect. Neuropeptides 5: 292–294

    Article  Google Scholar 

  178. Loh HH, Smith AP (1990) Molecular characterzation of opioid receptors. Annu Rev Pharmacol Toxicol 30: 123–147

    Article  PubMed  CAS  Google Scholar 

  179. Uhl GR, Childers S, Pasternack GW (1994) An opiate receptor gene family reunion. Trends Neurosci 17: 89–93

    Article  PubMed  CAS  Google Scholar 

  180. Freye E (1999) Opioide in der Medizin, 4. Aufl. Springer, Berlin Heidelberg New York Tokio

    Google Scholar 

  181. Tam S (1985) (+)-[3H]SKF 10,047, (+)-[3H]ethylketocycla-zocine, jli, k, 5 and phencyclidine binding sites in guinea pig brain membranes. Eur J Pharmacol 109: 33–41

    Article  Google Scholar 

  182. Cookson RF (1983) Carfentanil and Lofentanil. Clin Anaesthesin 1: 156–158

    CAS  Google Scholar 

  183. Ricaurte GA, Matello AL, Katz JL, Martello MB (1992) Long lasting effects of (±)-3,4-methylenedioxyamphetamine (MDMA) on central serotonergic neurons in nonhuman primates: neurochemical observations. J Pharmacol Exp Ther 261: 616–622

    PubMed  CAS  Google Scholar 

  184. Sanford TJ, Smith NT, Dec-Silver H, Harrison WK (1986) A comparison of morphine, fentanyl, and sufentanil anesthesia for cardiac surgery: Intubation emergence, and extubation. Anesth Analg 65: 259–266

    Google Scholar 

  185. Smith NT, Dec-Silver H, Harrison WK, Sanford TJ, Gilling JA (1982) A comparison among morphine, fentanyl and sufentanil anesthesia for open-heart surgery: Induction, emergence and extubation. Anesthesiology 57: A291

    Google Scholar 

  186. De Castro J, Van de Water A, Wouters L, Xhonneux R, Reneman R, Kay B (1979) Comparative study of cardiovascular, neurological, and metabolic side effects of eight narcotics in dogs. Acta Anaesth Belg 30: 5–99

    PubMed  Google Scholar 

  187. Hoten AO (1983) Another case of grand mal seizure after fentanyl aministration. Anesthesiology 60: 387–388

    Google Scholar 

  188. Goroszeniuk I, Malagosia A, Jones RM (1986) Genralized grand mal seizure after recovery from uncomplicated fentanyl-etomidate anesthesia. Anesth Analg 65: 979–981

    Article  PubMed  CAS  Google Scholar 

  189. Brian JE, Seiten AB (1987) Tonic-clonic activity after sufentanil. Anesth Analg 56: 329

    Google Scholar 

  190. Scott JC, Sarnquist FH (1983) Seizure-like movements during fentanyl infusion with absence of seizure activity in a simultaneous EEG recording. Anesthesiology 62: 812–814

    Article  Google Scholar 

  191. Carlsson C, D.S. S„ Kevkhah M, Engelbach I, Harp IR (1982) The effects of high-dose fentanyl on cerebral circulation and metabolism in rats. Anesthesiology 57: 375–380

    CAS  Google Scholar 

  192. Jaffe JH, Martin WR (1990) Opioid analgesics and antagonists. In: Gilman AF, Goodman LS, Rail TW, Murad F (eds) Goodman and Gilman’s. »The pharmacological basis of therapeutics«, vol 7. Macmillan, New York, pp 491–531

    Google Scholar 

  193. Kugler J, Grote B, Laub M, Doenicke A, Dick E (1977) Die hypnotische Wirkung von Fentanyl und Sufentanil. Anästhesist 26: 343–348

    CAS  Google Scholar 

  194. Chau TT, Carter FE, Harris LS (1982) 3H-codeine binding in the guinea pig lower brain stem. Pharmacology 25: 12–17

    Google Scholar 

  195. Heel RC, Brodgen RN, Speight TM, Avery GS (1979) Buprenorphine: A review of its pharmacological properties and therapeutic efficacy. Drugs 17: 81–100

    Google Scholar 

  196. Pircio AW, Gylys JA, Cavanagh RL (1976) The pharmacology of butorphanol, a 3,14-dihydroxymorphinan narcotic antagonist analgesic. Arch Int Pharmacodyn Ther 220: 231–257

    PubMed  CAS  Google Scholar 

  197. Christiani E, Stübing G (1972) Drogenmissbrauch und Drogenabhängigkeit. Deutscher Ärzteverlag, Köln

    Google Scholar 

  198. Jasinski DR (1977) Assessment of the abuse potentiality of morphine-like drugs. In: Martin WR (ed) Drug addiction, vol 1. Springer, Berlin Heidelberg New York, pp 197–258

    Chapter  Google Scholar 

  199. Jasinski DR, Pevnik JS, Griffith JD (1978) Human pharmacology and the abuse potential of the analgesic bupre-norphine. Arch Gen Psychiatry 35: 501–516

    Article  PubMed  CAS  Google Scholar 

  200. Lewis JW, Walter D (1992) Buprenorphine-background to its development as a treatment for opioid dependence. NIDA Res Monogr 121: 5–11

    PubMed  CAS  Google Scholar 

  201. Abbott A (1992) Neurobiological perspectives on drugs of abuse. Trends Pharmacol Sei 13: 169

    Article  CAS  Google Scholar 

  202. DiChiara G, Imperato A (1988) Opposite effects of ¡j and k opiate agonists on dopamine release in the nucleus accumbens and in the dorsal caudate of freely moving rats. J Pharmacol Exp Ther 244 /3: 1067–1080

    CAS  Google Scholar 

  203. Ramsey NF, van Ree JM (1992) Reward and abuse of opiates. Pharmacol Toxicol 71 /2: 81–94

    Article  PubMed  CAS  Google Scholar 

  204. Cherny Nl, Portenoy RK, Raber M, Zenz M (1994) Medikamentöse Therapie von Tumorschmerzen. Teil 1: Eigenschaften von Nichtopioiden und Opioiden. Schmerz 8: 195–209

    Google Scholar 

  205. Rommelspacher H (1981) Zur Frage des Abhängigkeitspotentials und des Mißbrauchs von Pentazocin. In: Kubi-cki S, Neuhaus GA (Hrsg) Pentazocin im Spiegel der Erfahrungen. Springer, Berlin Heidelberg New York, pp 58–63

    Google Scholar 

  206. Twycross RG (1988) Opioid analgesics in cancer pain: current practice and controversies. Cancer Surg 7: 29–53

    CAS  Google Scholar 

  207. Lehmann KA (1988) Analgosedierung mit Opioiden. In: Schulte am Esch J, Benzer H (Hrsg) Analgosedierung des Intensivpatienten. Springer, Berlin Heidelberg New York Tokio, pp 14–34 ( Reihe: Anaesthesiologie und Intensivmedizin, Bd 200 )

    Google Scholar 

  208. Martin WR, Jasinski DR (1977) Assessment of the abuse potential of narcotic analgesics in animal. In: Martin WR (ed) Drug addiction. I: Morphine, sedative/hypnotic and alcohol dependence. Springer, Berlin Heidelberg New York, p 159 (Handbook of experimental pharmacology, vol 45/I)

    Google Scholar 

  209. Houde RW, Wallenstein SL (1955) Report of analgesic studies at Memorial Center: Chlorpromazine potentiation, nalorphine-morphine combinations, dihydro-hydroxy- morphine. National Academy of Sciences, Washington. Bulletin of the 15th Meeting N. A. S. NCR Committee on Drug Addiction and Narcotics, pp 1134–1149

    Google Scholar 

  210. Dole VP, Nyswander ME (1965) A medical treatment for diacethylmorphine (heroin-)addiction. JAMA 193: 646–650

    Article  PubMed  CAS  Google Scholar 

  211. Benos J (1983) Ein Fall von sekundärer Buprenorphin- (Temgesic-)Abhängigkeit. Nervenarzt 54: 259–261

    PubMed  CAS  Google Scholar 

  212. Rosow CE (1984) Sufentanil citrate: A new opioid analgesic for use in anesthesia. Pharmacotherapy 4: 111–119

    Google Scholar 

  213. Sadee W, Wang Z (1993) Constitutive mu opioid receptor activation can account for narcotic tolerance and dependence. International Narcotics Research Conference (INRC). Uppsala Universitet Reprocentralen HSC, Skövde Sweden, p 102

    Google Scholar 

  214. Piepenbrock S, Hempelmann G, Peters H (1977) Veränderungen der Hämodynamik der Herzinotropie und des myocardialen Sauerstoffverbrauchs nach Antagonisie rung von hohen Dosen von Fentanyl mit Naloxon. Prakt Anästh 12: 275

    Google Scholar 

  215. Freye E, Härtung E (1982) Naloxone induces excitation of the cardiovascular system and a rise in myocardial oxygen consumption in fentanyl and meperidine-anesthetized dogs. Acta Anaesth Belg 33: 89–97

    PubMed  CAS  Google Scholar 

  216. Freye E (1976) Tyrosine hydroxylation in the rat striatum after fentanyl and droperidol in vivo. Expt Brain Res 26: 541–545

    CAS  Google Scholar 

  217. Goldstein DB, Goldstein A (1961) Possible role of enzyme inhibition and repression in drug tolerance and addiction. Biochem Pharmac 8: 48–53

    Article  Google Scholar 

  218. Scuderi P, Wetchler B, Sung Y-F et al. (1993) Treatment of postoperative nausea and vomiting after outpatient surgery with the 5-HT3 antagonist ondansetron. Anesthesiology 78 /1: 15–20

    Article  PubMed  CAS  Google Scholar 

  219. McKenzie R, Kovac A, O’Connor T et al. (1993) Comparison of ondansetron vs. placebo to prevent postoperative nausea and vomiting in women undergoing ambulatory gynecologic surgery. Anesthesiology 78: 21–28

    Article  PubMed  CAS  Google Scholar 

  220. Apfel CC, Greim A, Goepfert C et al. (1998) Postoperatives Erbrechen-Ein Score zur Voraussage des Erbrechensrisikos nach Inhalationsanästhesien. Anästhesist 47: 732–740

    Article  CAS  Google Scholar 

  221. Sneyed JR, Carr A, Byrom WD, Bilski AJT (1998) A metaanalysis of nausea and vomiting following maintenance of anaesthesia with propofol or inhational agents. Eur J Anaesth 15: 433–445

    Google Scholar 

  222. Freund FG, Martin WE, Wong KC, Hornbein TF (1973) Abdominal muscular rigidity induced by morphine and nitrous oxide. Anesthesiology 38: 358

    Article  PubMed  CAS  Google Scholar 

  223. Sokoll MD, Hoyt JL, Georgids SD (1972) Studies in mucular rigidity, nitrous oxide and narcotic analgesic agents. Anesth Analg 51: 16

    Article  PubMed  CAS  Google Scholar 

  224. Kuschinsky K, Hornykiewicz O (1972) Morphine katalepsy in the rat: relation to striatal dopamine metabolism. Eur J Pharmacol 19: 119

    Article  PubMed  CAS  Google Scholar 

  225. Freye E, Kuschinsky K (1976) The effect of fentanyl and droperidol on the dopamine metabolism of the rat striatum. Pharmacology 14: 1–7

    Article  PubMed  CAS  Google Scholar 

  226. Amalric M, Blasco TA, Smith NT, Lee DL, Swerdlow NR, Koob GF (1986) »Catatonia« produced by alfentanil is reversed by methylnaloxonium microinjections into the brain. Brain Res 386: 287–295

    Google Scholar 

  227. Jaffe TB, Ramsey FM (1983) Attenuation of fentanyl-induced truncal rigidity. Anesthesiology 58: 562

    Article  PubMed  CAS  Google Scholar 

  228. Freye E, Härtung E, Buhl R (1986) Die Lungencompliance wird durch die rasche Injektion von Alfentanil beeinträchtigt. Anästhesist 35: 543–546

    CAS  Google Scholar 

  229. Weinger MB, Segal IS, Maze M (1991) Dexmedetomidine, acting through central alpha2-adrenoceptors, prevents opiate-induced muscle rigidity in the rat. Anesthesiology 71: 242–249

    Article  Google Scholar 

  230. Paakkari P, Feuerstein G (1988) Antagonism of der-morphin-induced catalepsy with naloxone, TRH-analog CG3703 and the benzodiazepine antagonist, Ro 15–1788. Neuropharmacology 27 /10: 1007–1012

    Article  PubMed  CAS  Google Scholar 

  231. Illes P, Jurna I, Kaever V, Resch K (1996) Opioidanalgetika. In: Forth W, Henschler D, Rummel W, Starke K (Hrsg) Allgemeine und Spezielle Pharmakologie und Toxikologie, 7. Aufl. Akademischer Verlag, Heidelberg Berlin Oxford, pp 207–214

    Google Scholar 

  232. Kromer W (1993) Gastrointestinal effects of opioids. In: Herz A (ed) Opioids II,. Springer, Berlin Heidelberg New York, pp 163–190 (Handbook of experimental pharmacology, vol 104/11)

    Google Scholar 

  233. Champion SE, Sullivan SN, Chamberlain M, Vezina W (1982) Naloxone and morphine inhibit gastric emptying of solids. Can J Physiol Pharmacol 60: 732–734

    Article  PubMed  CAS  Google Scholar 

  234. Dingledine R, Goldstein A (1976) Effect of synaptic transmission blockade on morphine action in the guinea pig myenteric plexus. J Pharmacol Exp Ther 196: 97–106

    PubMed  CAS  Google Scholar 

  235. Polak JM, Sullivan SN, Bloom SR, Facer P, Pearse AGE (1977) Enkephalin-like immuno reactivity in the human gastrointestinal tract. Lancet 1: 972–974

    Article  PubMed  CAS  Google Scholar 

  236. Ambinder RF, Schuster MM (1979) Endorphins: new gut peptides with a familiar face. Gastroenterology 77: 1132

    PubMed  CAS  Google Scholar 

  237. Dashwood MR, Debnan ES, Bednall J, Thompson CS (1985) Autoradiographic localisation of opiate receptors in rat small intestine. Eur J Pharmacol 107: 267

    Article  PubMed  CAS  Google Scholar 

  238. North RA, Williams JT (1975) Enkephalines inhibit fireing of myenteric neurons. Nature 246: 460

    Google Scholar 

  239. Sahai KK, Hymson DL, Shapiro R (1978) The mode of actions of enkephalins in the guinea-pig ileum. Neurosci Lett 10: 317

    Article  Google Scholar 

  240. Nimmo WS (1984) Effect of anaesthesia on gastric motility and emptying. Br J Anaesth 56: 29

    Article  PubMed  CAS  Google Scholar 

  241. Ward SJ, Takemori AE (1983) Relative envolvement of receptor subtypes in opioid-induced inhibition of gastrointestinal transit in mice. J Pharmacol Exp Ther 224: 359–363

    PubMed  CAS  Google Scholar 

  242. Tavani A, Bianchi G, Ferreti P, Manara L (1980) Morphine is most effective on gastrointestinal propulsion in rats by intraperitoneal route: evidence for local action. Life Sei 27: 2211

    Article  CAS  Google Scholar 

  243. Manara L, Bianchi G, Ferretti P, Tavani A (1986) Inhibition of gastrointestinal transit by morphine in rats results pri-marely from direct drug action on gut opioid sites. J Pharmacol Exp Ther 237: 945–949

    PubMed  CAS  Google Scholar 

  244. Daniel EE, Sutherland WH, Bogoch A (1959) Effects of morphine and other drugs on motility of the terminal ileum. Gastroenterology 36: 510–523

    PubMed  CAS  Google Scholar 

  245. Hirning LD, Porreca F, Burks TF (1985) Mu, but not kappa opioid agonists induce contractions of the canine small intestine in vivo. Eur J Pharmacol 109: 49–54

    Article  PubMed  CAS  Google Scholar 

  246. Monferini E, Strada D, Manara L (1981) Evidence for opiate receptor binding in rat small intestine. Life Sci 29: 595–602

    Article  PubMed  CAS  Google Scholar 

  247. Terenius L (1972) Specific uptake of narcotic analgesics by subcellular fractions of the guinea pig ileum. Acta Pharmacol Toxicol 31: 50

    Google Scholar 

  248. Vater M, Aitkenhead AR (1985) Effect of morphine on gastric emptying. Anaesthesia 40: 81–82

    Article  PubMed  CAS  Google Scholar 

  249. Park GR, Weir DA (1985) A comparison of the effect of oral controlled release morphine and intramuscular morphine on gastric emptying. Anaesthesia 39: 645–648

    Google Scholar 

  250. Ingram DM, Sheiner HJ (1981) Postoperative gastric emptying. Br J Surg 68: 572–576

    Article  PubMed  CAS  Google Scholar 

  251. Yuan CS (1996) Gastric effects of ju-7 5 and K-opioid receptor agonists on brainstem unitary responses in the neonatal rat. Eur J Pharmacol 314: 27–32

    Article  PubMed  CAS  Google Scholar 

  252. Folwaczny C, Hundegger K, Volger C et al. (1995) Measurement of transit disorders in different gastrointestinal segments of patients with diabetis mellitus in relation to duration and severity of the disease by use of metal-detector test. Z. Gastroenterol 33: 517–526

    PubMed  CAS  Google Scholar 

  253. Wienbeck M, Korner MM (1981) Influence of opiates on colonic motility. Clin Res Rev 1: 199–204

    CAS  Google Scholar 

  254. Wienbeck M, Blasberg M (1986) Effect of an enkephalin analog on motility of the small and the large intestine in the cat. Z Gastroenterology 24: 179–187

    CAS  Google Scholar 

  255. Yee MK, Evans WD, Facey PE, Hayward WJ, Rosen M (1991) Gastric emptying and small bowel transit in male vol-unters after i.m. ketorolac and morphine. Br J Anaesth 67: 426–431

    Article  PubMed  CAS  Google Scholar 

  256. Nimmo WS, Wilson J, Prescott LF (1975) Narcotic analgesics and delayed gastric emptying during labour. Lancet I: 890–893

    Google Scholar 

  257. Yukioka H, Rosen M, Evans KT, Leach KG, Hayward MWJ, Saggu GS (1987) Gastric emptying and small bowel transit time in volunteers after intravenous morphine and nalbuphine. Anaesthesia 42: 704–710

    Article  PubMed  CAS  Google Scholar 

  258. Portenoy RK (1987) Constipation in the cancer patients: Causes and management. Med Clin North Am 71: 303–311

    PubMed  CAS  Google Scholar 

  259. Feritti P, Tavani A, Manara L (1981) Inhibition of gastrointestinal transit and antinociceptive effects of morphine and FK 33–824 in rats are differently prevented by naloxone and the N-methyl quarternary analog. Res Commun Subst Abuse 2: 1–11

    Google Scholar 

  260. Culpepper-Morgan JA, Inturrisi CE, Portenoy RK et al. (1992) Treatment of opioid-induced constipation with oral naloxone: A pilot study. Clin Pharmacol Ther 52: 90–95

    Google Scholar 

  261. Yuan CS, Foss JF, O’Connor M, Toledano A, Roizen MF, Moss J (1996) Methylnaltrexone prevents morphine-induced delay on oral-cecal transit time without affecting analgesia: A double-blind randomized placebo-controlled trial. Clin Pharmacol Ther 59: 469–475

    Google Scholar 

  262. Latasch L, Jurna I, Zimmermann M (1996) Opioid constipation: The antagonization of opioid-induced obstipation by naloxone. Dolor 11 (Suppl 1): 47

    Google Scholar 

  263. Robinson BA, Johansson L, Shaw J (1991) Oral naloxone in opioid-associated constipation. Lancet 338: 581–582

    Article  PubMed  CAS  Google Scholar 

  264. Jurna I, Jurna K, Baldauf J, Zenz M (1996) Orales Papave-rin reduziert die morphinbedingte Obstipation ohne Abschwachung der Analgesie nach oralem Morphin. Schmerz 10: 27–35

    Article  PubMed  CAS  Google Scholar 

  265. Rüssel J, Bass P, Goldberg LI, Schuster CR, Herz H (1982) Antagonism of gut, but not central effects of morphine with quarternary narcotic antagonists. Eur J Pharmacol 78: 255–261

    Article  Google Scholar 

  266. Dragonetti M, Bianchetti A, Sacilotto R et al. (1983) Leval-loprhan methyl iodide (SR 58002), a potent narcotic antgonist with peripheral selectivity superior to that of other quarternary compounds. Life Sci 33 (Suppl 1): 477–480

    Article  PubMed  CAS  Google Scholar 

  267. Yuan CS, Foss JF, Osinski J, Toledano A, Roizen MF, Moss J (1997) The safety and efficacy of oral methalnaltrexone in preventing morphine-induced delay in oral cecal transit time. Clin Pharmacol Ther 61: 1–9

    Article  Google Scholar 

  268. Yuan CS, Foss JF, Osinski J, Toledano A, Roizen MF, Moss J (2000) Methylnaltrexone for reversal of constipation due to chronic methadone use: a randomisesd controlled trial. JAMA 283: 367–372

    Article  PubMed  CAS  Google Scholar 

  269. Freye E, Rosenkranz B, Neruda B (1996) Die obstipierende Wirkung der Opioide Tilidin-N und Tramadol im Vergleich zu Codein. Eine vergleichende Gegenüberstellung zweier Wirkstoffkonzentrationen bei Probanden. Schmerz 10: 254–2. 60

    Google Scholar 

  270. Wilder-Smith CH, Bettiga A (1997) The analgesic tramadol has minimal effect on gastrointestinal motor function. Br J Clin Pharmacol 43: 1–5

    Article  Google Scholar 

  271. Hennies HH, Friedrichs E, Schneider J (1988) Receptor binding, analgesic and antitussive potency of tramadol and other selected opioids. Drug Res 38 /7: 877–880

    CAS  Google Scholar 

  272. Jage J, Jurna I (1993) Opioidanalgetika. In: Zenz M, I. J. (Hrsg) Lehrbuch der Schmerztherapie. Wiss. Verlagsges., Stuttgart, pp 137–153

    Google Scholar 

  273. Burks TF (1978) Gastrointestinal pharmacology. Ann Rev Pharmacol Toxicol 16: 15–31

    Google Scholar 

  274. Shook JE, Pelton JT, Hruby VJ, Burks TF (1987) Peptide opioid antagonist seperates peripheral and central opioid antitransit effects. J Pharmacol Exp Ther 243: 492–500

    PubMed  CAS  Google Scholar 

  275. Porreca F, Mosberg HI, Hurst R, Hruby VJ, Burks TF (1984) Roles of mu, delta and kappa opioid receptors in spinal and supraspinal mediation of gastrointestnal transit effects and hot-plate analgesia in the mouse. J Pharmacol Exp Ther 230: 341–348

    PubMed  CAS  Google Scholar 

  276. Culpepper-Morgan J, Kreek MJ, Holt PR, LaRoche D, Zhang J, O’Bryan L (1988) Orally administered kappa as well as mu opiate agonists delay gastrointestinal transit time in the guinea pig. Life Sci 42: 2073–2077

    Article  PubMed  CAS  Google Scholar 

  277. Wong CL (1984) The effects of morphine and nalbuphine on intestinal transit in mice. Meth and Find Exptl Clin Pharmacol 6: 685–689

    CAS  Google Scholar 

  278. Freye E, Knüfermann V (1991) Die gastro-coekale Transitzeit nach Fentanyl/Midazolam-im Vergleich zur Enfluran-und Ketamin/Midazolam-Narkose. Anästhesist 40 (Suppl 2): S 264

    Google Scholar 

  279. Niemegeers CJE, Schellenkens KHL, van Bever WFM, Janssen PAJ (1976) Sufentanil, a very potent and extremely safe intravenous morphine-like compound in mice, rats and dogs. Arzneimittelforsch/Drug Res 216: 1551–1556

    Google Scholar 

  280. Lagler F, Helm F, Etzel V, Kiel H (1978) Toxikologische Untersuchungen mit Tramadol, einem neuen Analgetikum. Arzneimittelforsch/Drug Res 28/I: 164–172

    Google Scholar 

  281. Janssen PAJ (1984) The development of new synthetic narcotics. In: Estafanous FG (ed) Opioids in anesthesia. Butterworth, Boston, pp 37–44

    Google Scholar 

  282. Janssen PAJ, Niemegeers CJE, Schellekens KHL, Lenarts FM (1971) Etomidate (R 16659) a potent short acting and relatively atoxic intravenous hypnotic agent in rats. Drug Res 21: 1234

    CAS  Google Scholar 

  283. Lewis JW, Ranee MJ, Sanger DJ (1983) The pharmacology and the abuse potential of buprenorphine. JAI Press, pp 103–154 (Advances in Substance Abuse, vol 3 )

    Google Scholar 

  284. Meert TF (1989) Pharmacological effects of epidural opioids. Therapeutics Today: Epidural Use of a new opioid: Sufentanil. Adis Press, Hong Kong, pp 1–9

    Google Scholar 

  285. De Castro J (1975) Nouveaux morphiniques, morphinoi-des, potentialisateurs et antidotes des morphiniques. Edition Academica, Bruxelles

    Google Scholar 

  286. Laubie M, Schmitt H, Vincent M, Remond G (1977) Central cardiovascular effects of morphinominetic peptides in dogs. Eur J Pharmacol 46: 76–71

    Article  Google Scholar 

  287. Freye E (1974) Cardiovascular effects of high doses of fentanyl, meperidine and naloxone in dogs. Anesth Analg 53: 40–47

    Article  PubMed  CAS  Google Scholar 

  288. Lappas DG, Geha D, Fischer JE, Laver MB, Lowenstein E (1975) Filling pressures of the heart and pulmonary circulation of the patient with coronary artery disease after large doses of morphine. Anesthesiology 42: 153

    Article  PubMed  CAS  Google Scholar 

  289. Braunwald E (1971) Control of myocardial oxygen consumption. Am J Cardiol 27: 416

    Article  PubMed  CAS  Google Scholar 

  290. Stahl KD, Simon EJ (1977) Receptor affinity and pharmacological potency of a series of narcotic analgesics, anti-diarrheal and neuroleptic drugs. Eur J Pharmacol 46: 199–205

    Article  PubMed  CAS  Google Scholar 

  291. De Lange S, Boscoe MJ, Stanley TH, Pace N (1982) Comparison of sufentanil-02 and fentanyl-02 for coronary artery surgery. Anesthesiology 56: 112–118

    Article  PubMed  Google Scholar 

  292. Stephan H, Sonntag H, Lange H, Lüpke K (1989) Die Wirkungen von Sufentanil in hohen Dosen auf die Hämodynamik und die elektroenezephalographische Aktivität von Koronarkranken. Anästhesist 38: 510–518

    CAS  Google Scholar 

  293. Zola EM, MacLeod DC (1983) Comparative effects and analgesic efficacy of the agonist-antagonist opioids. Drug Intel! Clin Pharm 17: 411–417

    CAS  Google Scholar 

  294. Dick W (1981) Möglichkeiten und Probleme der postoperativen Schmerzbekämpfung. Anästhesiol Intensivmed 2: 38–44

    Google Scholar 

  295. Boldt J, Kling D, von Bormann B, Knoblauch K, Görlach G, Hempelmann G (1987) Meptazinol, ein neuartiges Analgetikum. Anästhesist 36: 622–628

    CAS  Google Scholar 

  296. Strauer BE (1972) Contractile responses to morphine, piri-tramid and fentanyl: a comparative study of effects on the isolated myocardium. Anesthesiology 37: 304

    Article  PubMed  CAS  Google Scholar 

  297. Vargish T, Beamer KC, Daly T, Head R (1987) Myocardial opiate receptor activity is stereospecific, independent of muscarinic receptor antagonism, and may play a role in depressing myocardial function. Surgery 102: 171–177

    PubMed  CAS  Google Scholar 

  298. De Castro J, Viars P, Leleu JCL (1969) Utilisation de la pentazocine comme analgésique pour le traitement des douleurs post-opératoires. Etude comparative entre le pethidine, la piritramide et la pentazocine. In: De Castro J (ed) Utilisation de la pentazocine en anesthésie et réanimation. Ars Medici, Bruxelles, pp 99–109

    Google Scholar 

  299. Michaels I, Trout JR, Barash PG (1984) Nitrous oxide as an adjunct to narcotic anesthesia. In: Estafanous FG (ed) Opioids in anesthesia. Butterworth, Boston London, pp 256–260

    Google Scholar 

  300. Michaels I, Barash PC (1983) Does nitrous oxide or a reduced FI02 alter the hemodynamic function during high dose sufentanil anesthesia? Anesth Analg 62: 275

    Article  Google Scholar 

  301. Freye E (1975) Effects of high doses of fentanyl on myocardial infarction and cardiogenic shock in the dog. Resuscitation 3: 105–113

    Article  Google Scholar 

  302. Hess L, Vrana M, Vranova Z, Fejfar Z (1989) The antifibril-latory effect of fentanyl, sufentanil, and carfentanil in the acute phase of local myocardial ischemia in the dog. Acta Cardiol 150: 303–31 1

    Google Scholar 

  303. Saini V, Carr DB, Hagestad EL, Lown B, Verreir L (1988) Antifibrillatory action of the narcotic agent fentanyl. Am Heart J 115: 508–514

    Article  Google Scholar 

  304. Freye E, Avril G, Härtung E (1981) Les effets anti-arrythmi-ques des opiacés. Comparaison avec un beta-bloqueur chez le chien. Cahiers d’Anesthésiol 29: 591–598

    Google Scholar 

  305. DeSilva RA, Verrier RL, Lown B (1978) Protective effect of the vagotonic action of morphine sulfate on ventricular vulnerability. Cardiovasc Res 12: 167–181

    Article  CAS  Google Scholar 

  306. Suwatakul K, Weis OF, Alloza JL, Kelvie W, Weintraub K, Lasagna L (1983) Analysis of narcotic analgesic usage in the treatment of postoperative pain. JAMA 250: 926–929

    Article  Google Scholar 

  307. Gerwig WH, Thompson CW, Blades P (1951) Pain control following upper abdominal operations. Arch Surg 62: 678–682

    Article  Google Scholar 

  308. Morris T, Tracey J (1977) Lignocaine: its effect on wound healing. Br J Surg 64: 902–903

    Article  PubMed  CAS  Google Scholar 

  309. Dundee JW (1977) Problems associated with strong analgesics. In: Harcus AW, Smith R, Whittle B (eds) Pain. New perspectives in measurement and management. Churchill Livingstone, Edinburgh London New York, pp 57–62

    Google Scholar 

  310. Ferrari HA, Fuson RL, Dent SJ (1969) The relationship of the anaesthetic agent to postoperative analgesic requirements. South Med J 62: 1201–1203

    Article  PubMed  CAS  Google Scholar 

  311. Bonica JJ (1983) Pain research and therapy: achievements of the past and challenges of the future. In: Bonica JJ, Lindblom U, Iggo A (eds) Advances in pain research and therapy, vol 5. Raven, New York, pp 1–36

    Google Scholar 

  312. Porter J, Hick H (1980) Addiction rate in patients treated with narcotics. N Engl J Med 302: 123–126

    PubMed  CAS  Google Scholar 

  313. Taub A (1982) Opioid analgesics in the treatment of chronic intractable pain of non-neoplastic origin. In: Kitahata LM, Collins JG (eds) Narcotic analgesics in anesthesiology. Williams and Wilkins, Baltimore, pp 199–208

    Google Scholar 

  314. Portenoy RK, Foley KM (1986) Chronic use of opioid analgesics in non-malignant pain. Report of 38 cases. Pain 25: 171–186

    Article  PubMed  CAS  Google Scholar 

  315. Donald I (1977) Pain-a patients view. In: Harcus AW, Smith AWR, Whittle B (eds) Pain. New perspectives in measurement and management. Churchill Livingstone, Edinburgh London New York, pp 1–4

    Google Scholar 

  316. Lewis JW (1985) Buprenorphine. Drug Alcohol Depend 14: 363–372

    Article  CAS  Google Scholar 

  317. rwin JM (1977) The effect of Doxapram on buprenor-phine induced respiratory depression. Act Anaesth Belg 2: 93–106

    Google Scholar 

  318. Steffen P, Engelke J, Kutter B, Seeling W (1996) Nichtinvasive perioperative Analgesie nach Allgemeinanästhesien. Kombination von Dihydrocodein ret. mit den Nichtopio-idanalgetika Diclofenac und Metamizol. Schmerz 10: S51

    Google Scholar 

  319. Krizanits-Weine F, Bichler I, Müller A, Pietsch K, Frank M (1996) Präemptive Analgesie mit retardiertem Dihydrocodein bei Patienten mit elektiven Kniearthroskopien. Schmerz 10: S54

    Google Scholar 

  320. Freye E (1986) Die Wirkeffekte von Opioiden werden durch Subpopulationen von Rezeptoren vermittelt–Theoretische Grundlagen und praktische Folgerungen. Schmerz Pain Douleur 1: 3–9

    Google Scholar 

  321. Freye E (1986) Klinische Indikationsbereiche der Opioid- Agonisten, Agonisten-Antagonisten und der reinen Antagonisten. Schmerz Pain Douleur 2: 44–54

    Google Scholar 

  322. Herz A (1981) Opiat-Partialantagonisten. In: Kubicki S, Neuhaus GA (Hrsg) Pentazocin im Spiegel der Erfahrungen. Springer, Berlin Heidelberg New York, pp 19–21

    Google Scholar 

  323. Abboud TK, Zhu J, Gangolly J et al. (1991) Transnasal butorphanol: a new method for pain relief in postcesarean section pain. Acta Anaesthesiol Scand 35: 14–18

    Article  PubMed  CAS  Google Scholar 

  324. Wetchler BV, Alexander CD, Uhll MA (1992) Transnasal butorphanol tartrate for pain control following ambulatory surgery. Curr Ther Res 52 /4: 571–580

    Article  Google Scholar 

  325. Shyu WC, Pittmann KS, Robinson D, Barbhairya RH (1993) Multiple-dose phase I study of transnasal butorphanol. Clin Pharmacol Ther 54: 34–41

    Article  PubMed  CAS  Google Scholar 

  326. Joyce III TH, Kubicek MF, Skjonsky BS, Jones MM (1993) Efficacy of transnasal butorphanol tartrate in postepi-siotomy pain: a model to assess analgesia. Clin Therap 15 /1: 160–167

    Google Scholar 

  327. Couch J, Diamond D, Elkind A et al. (1993) Evaluation of the efficacy and safety of Stadol NS (transnasal butorphanol) in the treatment of acute migraine in outpatients. 7th World Congress on Pain. ISAP, Paris

    Google Scholar 

  328. Diamond S, Freitag FG, Diamond ML, Urban G (1992) Transnasal butorphanol in the treatment of migraine headache pain. Headache Quat Curr Treat Res 3 /2: 164–171

    Google Scholar 

  329. Houde RW (1979) Analgesic effectiveness of the narcotic agonist-antagonists. Br J Clin Pharmacol 7: 297S-308 S

    Google Scholar 

  330. Freye E, Helle G (1988) Der Agonist-Antagonist Nalbuphin verlängert die gastro-coekale Transitzeit und induziert kurzfristig Schmerzen nach Neurolepanästhesie mit Fentanyl. Anästhesist 37: 440–445

    CAS  Google Scholar 

  331. Arend I, von Arnim B, Nijssen J, Scheele J, Flohe L (1978) Tramadol und Pentazocin im klinischen Doppelblind- Crossover Vergleich. Arzneimittelforsch/Drug Res 28 /1: 199–208

    CAS  Google Scholar 

  332. Wermeling DP, Foster TS, Farrington EA et al. (1986) Patient-controlled analgesia using butorphanol for postoperative pain control: an open label study. In: Rosow CE (ed) Butorphanol tartrate: Research advances in multiple clinical settings, vol 12. Karger, Basel, pp 31–39

    Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  334. Watson GS, Edmond P (1977) Analgesics and urethral function. In: Harcus AW, Smith R, Whittle B (eds) Pain. New perspectives in measurement and management. Churchill Livingstone, Edinburgh London New York, pp 27–33

    Google Scholar 

  335. Saarne A (1969) Clinical evaluation of a new analgesic piritramide. Acta Anaesthesiol Scand 13: 11–19

    Article  PubMed  CAS  Google Scholar 

  336. Hermans B, Gommeren W, De Potter WP, Leysen JE (1983) Interaction of peptides and morphine-like narcotic analgesics with specifically labeled ju-and 5-opiate receptor binding. Arch Int Pharmacodyn 263: 317–319

    PubMed  CAS  Google Scholar 

  337. Martin WR (1981) Mini-Symposium II. Multiple opioid receptors. Life Sei 28: 1547–1554

    Article  CAS  Google Scholar 

  338. Keeri-Szanto M (1979) Drugs or drums: what relives postoperative pain? Pain 6: 217–230

    Article  PubMed  CAS  Google Scholar 

  339. Tamsen A, Hartwig P, Fagerlund C, Dahlstrom B, Bondes-sun U (1982) Patient-controlled analgesia therapy: clinical experience. Acta Anaesthesiol Scand 74: 157–160

    Article  CAS  Google Scholar 

  340. Thomas DW, Owen H (1988) Patient-controlled analgesia-the need for caution. Anaesthesia 43: 770–772

    Article  PubMed  CAS  Google Scholar 

  341. White PF (1987) Mishaps with patient-controlled analgesia. Anesthesiology 66: 81–83

    Article  PubMed  CAS  Google Scholar 

  342. Parker RK, Holtmann B, White PF (1991) Patient-controlled analgesia. Does a concurrent opioid infusion improve pain mangement after surgery? JAMA 266 /14: 1947–1952

    Article  PubMed  CAS  Google Scholar 

  343. Coyle N, Mauskopf A, Maggard J, Foley KM (1986) Continuous subcutaneous infusion of opiates in cancer pain patients. ONF 13 /4: 53–57

    CAS  Google Scholar 

  344. Lehmann KA (1984) Neue Möglichkeiten zur Behandlung akuter Schmerzen. Arzneimittelforsch/Drug Res 34: 1108–1114

    CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  346. Krimmer H, Pfeiffer H, Arbogast R, Sprotte G (1986) Die kombinierte Infusionsanalgesie–Ein alternatives Konzept zur postoperativen Schmerztherapie. Chirurg 57: 327–329

    PubMed  CAS  Google Scholar 

  347. Foley KM (1986) Current controversies in opioid therapy. In: Foley FM, Inturisse CE (eds) Advances in pain research and therapy, vol 8. Raven, New York, pp 3–11

    Google Scholar 

  348. Nielsson Ml, Groenbladh L, Widerloev E, nggard E (1983) Pharmacokinetic of methadone maintenance treatment: Characterization of therapeutic failures. Eur J Clin Pharmacol 25: 497–501

    Google Scholar 

  349. Taeger K (1981) Pharmakokinetik der Opiate Dolantin, Morphin und Fentanyl. Anästh Intensivmed 2 /28

    Google Scholar 

  350. Wörz R, Berlin J (1989) Behandlung chronischer Schmerzsyndrome mit Antidepressiva. Schmerz 3: 1–7

    Article  PubMed  Google Scholar 

  351. Tao P-L, Law P-Y, Loh HH (1986) Decrease in delta und mu opioid receptor binding capacity oin rat brain after chronic treatment. J Pharmacol Expt Ther 240: 809–816

    Google Scholar 

  352. Tao P-L, Chang L-R, Law PY, Loh HH (1988) Decrease in 5-opioid receptor density in rat brain after chronic (D-Ala2,D-Leu5) enkephalin treatment. Brain Res 462: 313–320

    Article  PubMed  CAS  Google Scholar 

  353. Elliott TE, Elliott BA (1992) Physicians attitudes and beliefs about use of morphine for cancer pain. J Pain Sympt Manag 3 /7: 141–148

    Article  Google Scholar 

  354. Robbins LN, Davis DH, Nurco DM (1974) How permament was Vietnam drug addiction. Am J Public Health 64: 38–43

    Article  Google Scholar 

  355. Maruto T, Swanson DW, Finlayson RE (1979) Drug abuse and dependency in patients with chronic pain. Mayo Clin Proc 54: 241–244

    Google Scholar 

  356. Tennant FS, Kelman GF (1983) Narcotic maintenance for chronc pain: medical and legal guidelines. Postgrad Med 73: 81–94

    Google Scholar 

  357. Twycross RG, Lack SA (1983) Symptom control in far-advanced cancer. Pain relief. Pittman, London

    Google Scholar 

  358. Bohn LM, Jefkowitz RJ, Gainetdinov RG, Peppel K, Caron MG, Lin FT (1999) Enhanced morphine analgesia in mice lacking ß-arrestin 2. Science 286: 2495–2498

    Article  PubMed  CAS  Google Scholar 

  359. Kanner RM, Foley KM (1981) Pattern of narcotic drug use in cancer pain clinic. Ann NY Acad Sei 362: 162–172

    Article  Google Scholar 

  360. Smith GD, Smith MT (1995) Morphine-3-glucuronide: evidence to support the putative role in the development of tolerance to the antinociceptive effect in the rat. Pain 62: 51–60

    Article  PubMed  CAS  Google Scholar 

  361. Morley JS, Watt JWG, Wells JC, Miles JB, Finnegan MJ, Leng G (1993) Methadone in pain uncontrolled by morphine. Lancet 342: 1243

    Article  PubMed  CAS  Google Scholar 

  362. Ross FB, Smith MT (1997) The intrinsic antinociceptive effects of oxycodone appear to be K-opioid receptor mediated. Pain 73: 151–157

    Article  PubMed  CAS  Google Scholar 

  363. Grabinski PY, Kaiko RF, Rogers AG, Houde RW (1983) Plasma levels and anlgesia following deltoid and gluteal injections of methadone and morphine. J Clin Pharmacol 23 /48

    Google Scholar 

  364. Rutter P, Muprphy F, Dudley H (1980) Controlled trial of different methods of administration of postoperative pain relief. BMJ 1: 3–12

    Google Scholar 

  365. Bruera E, Chadwick S, Bacovsky R (1981) Continuous subcutaneous infusion of narcotics using a portable disposable pump. J Palliat Care 1: 45–47

    Google Scholar 

  366. Waldmann C, Eason J, Rambohul E (1984) Serum morphine levels. A comparison between continuous subcutaneous and intravenous infusion in postoperative patients. Anaesthesia 39: 768–771

    Google Scholar 

  367. Dickson R, Russell P (1982) Continuous subcutaneous analgesia for terminal care at home. Lancet I: 165

    Google Scholar 

  368. Goeke H, Herbst M, Frucht U (1992) Schmerztherapeutische Tumornachsorge. Klinikarzt 1: 1–4

    Google Scholar 

  369. Goecke H, Herbst M (1993) Ambulante kontinuierliche subkutane Opioidanalgesie als PCAO (patient controlled analgesia in outpatients) bei schweren Tumorschmerzen. Schmerz 7: 31–39

    Article  Google Scholar 

  370. Duthie DJR, Rowbotham DJ, Wyld R, Henderson PD, Nimmo WS (1988) Plasma fentanyl concentrations during transdermal delivery of fentanyl to surgical patients. Br J Anaesth 60: 614–618

    Article  PubMed  CAS  Google Scholar 

  371. Plezia PM, Kramer TH, Linford J, Hameroff SR (1989) Transdermal fentanyl pharmacokinetics and preliminary clinical evaluation. Pharmacotherapy 9: 2–9

    PubMed  CAS  Google Scholar 

  372. Rowbotham DJ, Wyld R, Peacock JE, Duthie DJR, Nimmo WS (1989) Transdermal fentanyl for the relief of pain after upper abdominal surgery. Br J Anaesth 63: 56–59

    Article  PubMed  CAS  Google Scholar 

  373. Varvel JR, Shafer SL, Hwang SS, Coen PA, Stanski DR (1989) Absorption characteristics of transdermal administered fentanyl. Anesthesiology 70: 928–934

    Article  PubMed  CAS  Google Scholar 

  374. Larijani GE, Brell SD, Goldberg ME, Lessin JB (1988) Pharmacokinetics of fentanyl following transdermal application. Anesthesiology 69: A 363

    Google Scholar 

  375. Southam M, Gupta S, Knowles M, Hwang SS (1991) Transdermal fentanyl: an overview of pharmacokinetics, efficacy and safety. In: Lehmann KA, Zech D (eds) Transdermal fentanyl. Springer, Berlin Heidelberg New York, pp 107–116

    Chapter  Google Scholar 

  376. Hill HF (1990) Clinical pharmacology of transdermal fentanyl. Eur J Pain 11: 81–91

    Google Scholar 

  377. Payne R (1990) Experience with transdermal fentanyl in advanced cancer pain. Eur J Pain 11: 98–101

    Google Scholar 

  378. Nimmo WS (1990) Clinical summary: Transdermal fentanyl. Eur J Pain 11: 102–103

    Google Scholar 

  379. Duthie DJR, Nimmo WS (1987) Adverse effects of opioid analgesic drugs. Br J Aneasth 60: 61–77

    Article  Google Scholar 

  380. Hoiley PO, van Steenis C (1988) Postoperartive analgesia with fentanyl: pharmacokinetics and pharmakodynamics of constant rate iv and transdermal delivery. Br J Anaesth 60: 608–613

    Article  Google Scholar 

  381. Twycross RG (1984) Relief of pain. In: Saunders C (ed) The mangemement of terminal malignant disease. Edward Arnold, London, pp 64–90

    Google Scholar 

  382. Portenoy RK, Hagen NA (1990) Breakthrough pain: Definition, prevelance and characteristics. Pain 41: 273–281

    Google Scholar 

  383. Ahmedzai S, Brooks D (1997) Transdermal fentanyl vs. sustained-release oral morphine in cancer pain: preference, efficacy, and quality of life. J Pain Symptom Manage 13: 254–261

    Article  PubMed  CAS  Google Scholar 

  384. Allen L, Hays H, Jensen NH et al. (1998) Evidence for better analgesia with transdermal fentanyl in chronic pain treatment. Comparison with sustained release morphine in a cross-over efficacy, safety and quality of life trial. Proceedings 10th European Congress of Anesthesiology. Springer, Frankfurt am Main

    Google Scholar 

  385. McLesky CH (1990) Fentanyl TTS for postoperative analgesia. Eur J Pain 11: 92–97

    Google Scholar 

  386. Haak R, Gupta S (1993) Pulsatile drug delivery from elec-trotransport therapeutic systems. In: Fumy R, Junginger H, Peppas N (eds) Pulsatile drug delivery. Current applica-tions and future trends. Wiss. Verlagsges., Stuttgart, pp 99–112

    Google Scholar 

  387. Maier C (1996) Ganglionäre lokale Opioidanalgesie (GLOA). Thieme, Stuttgart New York

    Google Scholar 

  388. Wulf H, Maier C, Scheie HA (1991) Die Behandlung von Zoster-Neuralgien. Anästhesist 40: 523–529

    CAS  Google Scholar 

  389. Stein C, Comisel K, Haimerl E et al. (1991) Analgesic effect of intraarticular morphine after arthroscopic knee surgery. N Eng J Med 325: 1123–1126

    Article  CAS  Google Scholar 

  390. Glynn CJ, Casale R (1993) Morphine injected around the stellate ganglion does not modulate the sympathetic nevous system nor does it provide pain relief. Pain 53: 33–38

    Article  PubMed  CAS  Google Scholar 

  391. Laduron PH, Janssen FMH (1982) Axoplasmatic transport and possible recycling of opiate receptors labeled with 3H-lofentanil. Life Sei 31: 457–462

    Article  CAS  Google Scholar 

  392. Stein C, Millan MJ, Yassouridis A, Herz A (1988) Antinociceptive effects of jj-and k-agonists in inflammation are enhanced by a peripheral opioid receptor-specific mechanism. Eur J Pharmacol 155: 255–264

    Article  PubMed  CAS  Google Scholar 

  393. Stein C, Hassan A, Przewlocki R, Gramsch C, Peter K, Herz A (1990) Opioids from immunocytes interact with receptors on sensory nerves to inhibit nociception in inflammation. Physiol Pharmacol 87: 5934–5939

    Google Scholar 

  394. Ashburn MA, Streisand JB (1994) Oral transmucosal fen-tanyl-help or hindrance? Drug Safety 11: 295–300

    Article  PubMed  CAS  Google Scholar 

  395. Chien YW, Chang SF (1987) Intranasal drug delivery for systemic medications. Crit Rev Ther Drug Carr Syst 4: 67–194

    CAS  Google Scholar 

  396. Seymour D, Freitag FG, Diamond ML, Urban G (1992) Transnasal butorphanol in the treatment of migraine headache pain. Headache Quat Curr Treat Res 3: 164–171

    Google Scholar 

  397. Eagen KJ, Ready L (1994) Patient satisfaction with intravenous PCA or epidural morphine. Can J Anaesth 41: 6–11

    Article  Google Scholar 

  398. Striebel HW, Koenigs D, Krämer J (1992) Postoperative pain management by intranasal demand-adapted fen-tanyl titration. Anesthesiology 77: 281–285

    Article  PubMed  CAS  Google Scholar 

  399. Abrams R, Morrison JE, Villasenor A, Hencmann D, Da Fon-seca M, Mueller W (1993) Saftey and effectiveness of intranasal administration of sedative medications (ketamine, midazolam, or sufentanil) for urgent brief pediatric dental procedures. Anesth Prog 40: 63–66

    PubMed  CAS  Google Scholar 

  400. Helmers JH, Noorduin H, van Peer A, Van Leuwen L, Zuurmond WW (1989) Comparison of intravenous and intranasal sufentanil adsorption and sedation. Can J Anaesth 36: 494–497

    Article  PubMed  CAS  Google Scholar 

  401. Striebel WH, Malewicz J, Hermanns K (1993) Intranasal meperidine titration for postoperative pain relief. Anesth Analg 76: 1047–1051

    Article  PubMed  CAS  Google Scholar 

  402. Oral U, Isik G, Özbek H, Güler T (1995) Intranasal alfentanil for postoperative pain relief. BrJ Anaesth 74: 139

    Google Scholar 

  403. Schwagmeier R, Boerger N, Meissner W, Striebel HW (1995) Pharmacokinetics of intranasal alfentanil. J Clin Anesth 7: 109–113

    Article  PubMed  CAS  Google Scholar 

  404. Striebel WH, Wessel A, Rieger A (1993) Intranasales Fentanyl zur Therapie akuter Schmerzspitzen bei Karzinompatienten. Eine Pilotstudie. Schmerz 7: 174–178

    Google Scholar 

  405. Striebel HW, Olmann T, Spies C, Brummer G (1996) Patient-controlled intranasal analgesia (PCINA) for the management of postoperative pain: a pilot study. J Clin Anaesth 8: 4–8

    Article  CAS  Google Scholar 

  406. Farncombe M, Chater S (1994) Clinical application of nebulized opioids for treatment of dyspnoe in patients with malignant disease. Sup Care Cancer 2: 184–187

    Article  CAS  Google Scholar 

  407. Masters N, Heap G, Wedley MJ, Moore A (1988) Inhaled nebulized morphine and diamorphine: useful in general practice? Practitioner 232: 910–914

    PubMed  CAS  Google Scholar 

  408. Chrubasik J, Wüst H, Friedrich G, Geller E (1988) Absorption and bioavailability of nebulized morphine. Br J Anaesth 61: 228–230

    Article  PubMed  CAS  Google Scholar 

  409. Otulana BA, Morishige RJ, Beckman RA et al. (1999) Oral inhalation system for delivery of morphine produces an iv-like profile. In: 9th World Congress on Pain. IASP Press, Vienna, p 337

    Google Scholar 

  410. Stanley TH, de Lange S (1988) Comparison of sufentanil-oxygen and fentanyl-oxygen anesthesia for mitral and aortic valvular surgery. J Cardiothoracic Anesth 2: 6–11

    Article  CAS  Google Scholar 

  411. Sebel PS, Bovill JG, Wauquier A, Rog P (1981) Effects of high dose fentanyl anesthesia on the electroencephalogram. Anesthesiology 55: 203–211

    Article  PubMed  CAS  Google Scholar 

  412. Bovill JG, Sebel PS, Wauquire A, Rog P, Schuytt HC (1983) Influence of high-dose alfentanil in anesthesia on the electroencephalogram: Correlation with plasma concentrations. Br J Anaesth 55: 199S - 209S

    PubMed  Google Scholar 

  413. Tolksdorf W, Schäfer E, Pfeiffer J, von Mittelstaedt G (1987) Adrenalin-, Noradrenalin-, Blutdruck-und Herz-freqenzverhalten während der Intubation in Abhängigkeit unterschiedlicher Fentanyl-Dosen. Anästh Intensivther Notfallmed 22: 171–176

    Google Scholar 

  414. Podiesch I (1988) Disoprivan (Propofol)-ein neues intravenöses Hypnotikum. Fortschr Anaesth 2: 1–31

    Google Scholar 

  415. Cockshott ID (1985) Propofol (Diprivan) pharmacokinetics and metabolism–an overview. Postgrad Med J 61: 45–50

    PubMed  CAS  Google Scholar 

  416. Härtung E, Freye E (1988) An open comparison of propofol and enflurane for prolonged abdominal operations. Anaesthesia 43: 105–107

    Article  PubMed  Google Scholar 

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

    Google Scholar 

  418. Stark RD, Binks SM, Dutka VN, O’Connor KM, Armstein MJA, Glen JB (1985) A review of the safety and tolerance of propofol ( Diprivan ). Postgrad Med J 61: 152–156

    Google Scholar 

  419. Nauta J, de Lange S, Koopman D, Speidijk J, van Kleef J, Stanley T (1982) Anesthetic induction with alfentanil: A new short-acting narcotic analgesic. Anesth Analg 61: 267–272

    Google Scholar 

  420. Freye E, Härtung E (1988) Der spezifische Antagonist »Flumazenil« bei Benzodiazepin-lntoxikationen. Dtsch Ärztebl 42: 2909–2914

    Google Scholar 

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

    Google Scholar 

  422. Bergmann SA, Wynn RL, Peterson MD, Rudo FG (1988) GABA agonists enhance morphine and fentanyl antinoci-ception in rabbit tooth pulp and mouse hot plate test. Drug Dev Res 14: 111–122

    Article  Google Scholar 

  423. Tejwani GA, Rattan AK, Sribanditmongkol P, Sheu M-J, Zuniga J, McDonald JS (1993) Inhibition of morphine-induced tolerance and dependence by a benzodiazepine receptor agonist midazolam in the rat. Anesth Analg 76: 1052–1060

    Article  PubMed  CAS  Google Scholar 

  424. Rosland JH, Kole K (1990) 1,4-Benzodiazepines antagonize opiate-induced antinociception in mice. Anesth Analg 71: 242–248

    Google Scholar 

  425. Rosland JH, Hunskaar S, Hole K (1990) Diazepam attenuates morphine antinociception test-dependently in mice. Pharmacol Toxicol 66: 382–386

    Article  PubMed  CAS  Google Scholar 

  426. Luger TJ, Hill HF, Hayashi T, Zech C, Yaksh TL (1993) Spinal potentiating and supraspinal inhibitory effect of midazolam on morphine analgesia in rats. In: Proceedings 7th World Congress on Pain. IASP Publ, Paris, pp 201–202

    Google Scholar 

  427. McLesky CH (1984) Comparison of three infusion rates of alfentanil and incremental fentanyl as adjunct to nitrous oxide anesthesia for general surgery. Janssen Clin Res Rep

    Google Scholar 

  428. Stanley TH (1982) Comparison of alfentanil with thiopental sodium for induction of anesthesia. Janssen Pharma-zeutika, Beerse/Belgium

    Google Scholar 

  429. Murphy MR, Hug CC (1982) The anesthetic potency of fentanyl in terms of its reduction of enflurane MAC. Anesthesiology 57: 485–488

    Article  PubMed  CAS  Google Scholar 

  430. Ismaily AJ, Mötsch J, Altmayer P, Bleser W, Hutschenreuter K (1987) Die Auswirkungen einer Kombinationsanästhesie mit Fentanyl und Enfluran auf den Kreislauf und die unmittelbare postoperative Phase. Anäst Intensivmed 28: 216–220

    Google Scholar 

  431. Härtung HJ (1988) Klinische Erfahrungen mit Alfentanil zur »balanced anesthesia« bei Oberbauch-Eingriffen. Anästhesist 37: 620–624

    Google Scholar 

  432. Skubella U, Hucke H (1989) Alfentanil-Kombinationsnar-kose bei der Adeno-Tonsillektomie im Kindesalter–ein Vergleich mit der Enfluran-Inhalationsanästhesie. Anästh Intensivther Notfallmed 24: 362–367

    Article  PubMed  CAS  Google Scholar 

  433. Brizgys RV, Morales R, Owens B (1985) Effects of thiopental requirements and hemodynamic response during induction and intubation. Anesthesiology 63: A377

    Article  Google Scholar 

  434. Smith NT, Westover CJ, Qinn M, Benthuysen L, Silver DH, Sanford TJ (1985) An electroencephalographic comparison of alfentanil with other narcotics and with thiopental. J Clin Monit 1: 236–244

    Article  PubMed  CAS  Google Scholar 

  435. Eisenach JC, DuPen S, Dubois M, Miguel R, Allin D (1995) The epidural Clonidine study group. Epidural Clonidine analgesia for intractable cancer pain. Pain 61: 391–399

    Google Scholar 

  436. Aantaa R, Kanto J, Scheinin M, Kallio A, Scheinin H (1990) Dexmedetomidine, an alpha 2-adrenoreceptor agonist, reduces anesthetic requirements for patients under going minor gynecologic surgery. Anesthesiology 73: 230–235

    Article  PubMed  CAS  Google Scholar 

  437. Flacke JW, Bloor BC, Flacke WE et al. (1987) Reduced narcotic requirement by Clonidine with improved hemodynamic and adrenergic stability in patients undergoing coronary bypass surgery. Anesthesiology 67: 11–19

    Article  PubMed  CAS  Google Scholar 

  438. Engelman E, Lipzyc M, Gilbert E et al. (1989) Effects of Clonidine on anesthetic requirements and hemodynamic response during aortic surgery. Anesthesiology 71: 178–187

    Article  PubMed  CAS  Google Scholar 

  439. Noyer M, De Laveleye F, Vauquelin G, Gobert J, Wülfert E (1994) Mivazerol, a novel compound with high binding specificity for alpha2 adrenergic receptor: Binding studies on different human and rat membrane preparations. Neu-rochem Int 24: 221–229

    Google Scholar 

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

    Article  Google Scholar 

  441. Hoffmann WE, Kochs E, Werner C, Thomas C, A. RF (1991) Dexmedetomidine improves neurologic outcome from incomplete ischemia in the rat: reversal by the alpha2 antagonist atipamezole. Anesthesiology 75: 328–332

    Article  Google Scholar 

  442. Mötsch J, Gräber E, Ludwig K (1990) Addition of Clonidine enhances postoperative analgesia from epidural morphine a double blind study. Anesthesiology 73: 1067–1073

    Article  PubMed  Google Scholar 

  443. Carabine UA, Milligan KR, Moore J (1992) Extradural Clonidine and bupivacaine for postoperative analgesia. Br J Anaesth 68 /2: 132–135

    Article  PubMed  CAS  Google Scholar 

  444. Maze M, Tranquilli W (1991) Alpha-2 adrenoreceptor agonists: Defining the role in clinical anesthesisa. Anesthesiology 74: 581–605

    Google Scholar 

  445. Striebel HW, Koenigs D, Heil T (1993) Clonidin-Stellenwert in der Anästhesie. Anästhesist 42: 131–141

    CAS  Google Scholar 

  446. Blum K (1980) Alcohol and opiates. A review of common mechanism. In: Lacasse L, Levy M, Manzo L (eds) Neuro-toxikology. Pergamon, Oxford, pp 71–90

    Google Scholar 

  447. Haber H, Melzig M (1992) Tetrahydroisoqinoline-endo-gene Produkte nach chronischem Alkoholabusus. Pharmazie 47: 3–7

    PubMed  CAS  Google Scholar 

  448. Cohen G (1979) Interaction of catecholamines with acetal-dehyde to form tetrahydroisoquinoline neurotransmitters. In: Sharp CW, Abood L (eds) Membrane Mechanisms of drugs of abuse. Progr Clin Biol Res 27: 73–90

    Google Scholar 

  449. Melchior CL, Collins MA (1982) The route and significance of endogenous synthesis of alkaloids in animals. Crit Rev Tox 9: 313–356

    Article  CAS  Google Scholar 

  450. Collins MA (1980) Neuroamine condensation in human subjects. In: Begleiter H (ed) Biological effects of alcohol. Plenum Press, New York, pp 87–102

    Google Scholar 

  451. Hoffman PL, Urwyler S, Tabakoff B (1982) Alterations in opiate receptor function after chronic ethanol exposure. J Pharmacol Exp Ther 222: 182–189

    PubMed  CAS  Google Scholar 

  452. Herz A (1980) Pharmacological modulation of opiate-like peptide systems. Pharmacol Biochem Behav 1 (Suppl): 165–268

    Google Scholar 

  453. Schulz R, Wüster M, Duka T, Herz A (1980) Acute and chronic ethanol treatment changes endorphin levels in brain and pituitary. Psychopharmacology 68: 221–227

    Article  PubMed  CAS  Google Scholar 

  454. Hug CCJ (1984) Pharmacokinetics of new synthetic narcotic analgesics. In: Estafanous FG (ed) Opioids in anesthesia. Butterworth, Boston, pp 50–60

    Google Scholar 

  455. McDonnell TE, Bartowski RR, Williams JJ (1982) ED50 of alfentanil for induction of anesthesia in unpremedicated young adults. Anesthesiology 57: A 362

    Google Scholar 

  456. Stanski DR, Hug CC (1982) Alfentanil-a kinetically predictable narcotic analgesic. Anesthesiology 57: 435–438

    Article  PubMed  CAS  Google Scholar 

  457. Suttmann H, Doenicke A (1983) Interim report on dose-establishment with alfentanil. Janssen Pharmaceutica, Beerse/Belgium

    Google Scholar 

  458. Freye E, Härtung E, Buhl R (1986) Alfentanil als letzte Dosis (on-top) in der Neuroleptanalgesie mit Fentanyl. Anästhesist 35: 231–237

    CAS  Google Scholar 

  459. Shafer SL, Varvel JR (1991) Pharmacokinetics, pharmacodynamics, and rational opioid selection. Anesthesiology 74 /1: 53–63

    Article  PubMed  CAS  Google Scholar 

  460. Chapmann CR, Colpitts YM, Benedetti C, Butler S (1982) Event-related potential correlates of analgesia comparison of fentanyl, acupuncture and nitrous oxide. Pain 14: 327–337

    Article  Google Scholar 

  461. Freye E, Härtung E, Schenk GK (1989) Somatosensory-evoked potentials during block of surgical stimulation with propofol. Br J Anaesth 63: 357–359

    Article  PubMed  CAS  Google Scholar 

  462. Kochs E, Treede RD, Schulte am Esch J, Bromm B (1990) Modulation of pain-related somatosensory evoked potentials by general anesthesia. Anesth Analg 71: 225–2230

    Article  PubMed  CAS  Google Scholar 

  463. Freye E (1985) Somatosensorisch evozierte Potentiale (SEP) zur Algesiemetrie. In: Zindler M, Härtung E (eds) Alfentanil-Ein neues, ultrakurzwirkendes Opioid. Urban and Schwarzenberg, München Wien Baltimore, pp 17–23

    Google Scholar 

  464. Freye E (1991) Opiate und Opiatantagonisten. II. Der praktische Einsatz der Opioide. DAZ 49: 2605–2615

    Google Scholar 

  465. Egan TD (1995) Remifentanil pharmacokinetics and pharmacodynamics. Clin Pharmacokinet 29: 80–94

    Article  PubMed  CAS  Google Scholar 

  466. Hermann DJ, Marton JP, Donn KH et al. (1991) Pharmacokinetic comparison of GI87084B, a novel ultra-short acting opioid, and alfentanil. Anesthesiology 75: A 379

    Google Scholar 

  467. Flacke JW, Bloor BC, Kripke BJ, Flacke WE, Warneck CM (1985) Comparison of meperidine, fentanyl and sufentanil in balanced anesthesia. Anesth Analg 64: 897–910

    PubMed  CAS  Google Scholar 

  468. De Lange S, Boscoe MJ, Stanley TH, De Bruijn N, Philbin DM, Coggins CH (1982) Antidiuretic and growth hormone responses during coronary artery surgery with sufentanil-oxygen and alfentanil-oxygen anesthesia in man. Anesth Analg 61: 434

    Article  PubMed  Google Scholar 

  469. De Lange S, Stanley TH, Boscoe MJ et al. (1983) Catecholamine and Cortisol responses to sufentanil-02 and alfentanil-02 anaesthesia during coronary artery surgery. Can Anaesth Soc J 30: 248–255

    Article  PubMed  Google Scholar 

  470. Bailey PL, Streisand JB, Pace NL, Bayless J, Stanley TH (1986) Sufentanil produces shorter lasting respiratory depression and longer lasting analgesia than equipotent doses of fentanyl in human volunteers. Anesthesiology 65: A 493

    Google Scholar 

  471. Freye E (1991) Opioide–Zentrale Analgetika. Ihre Wirkweise beim Menschen. DIA-GM 12: 1105–1118

    Google Scholar 

  472. Bowdle TA, Ward RJ (1989) Induction of anesthesia with small doses of sufentanil or fentanyl: Dose vs. EEG response, speed of onset, and thiopental requirement. Anesthesiology 70: 26–30

    Google Scholar 

  473. Brian SE, Seifen AB. Tonic-clonic activity after sufentanil. Anesth Analg 1987 66: 481

    Article  PubMed  Google Scholar 

  474. Katz Rl, Eide TR, Hartman A, Poppers PJ (1988) Two instances of seizure-like activity in the same patient associated with two different narcotics. Anesth Analg 67: 289

    PubMed  CAS  Google Scholar 

  475. Young WL, Prohovnik I, Carrell JW, Ornstein E (1988) The effect of sufentanil on cerebral hemodyamics during carotid endarterectomy. Anesthesiology 69: A 591

    Google Scholar 

  476. Keykhah MM, Smith DS, Carlson C (1985) Influence of sufentanil on cerebral metabolism and circulation in the rat. Anesthesiology 63: 274–277

    Article  PubMed  CAS  Google Scholar 

  477. Werner C (1992) Der Einfluß von Sufentanil auf die regionale und globale Hirndurchblutung und den zerebralen Sauerstoffverbrauch beim Hund. Anästhesist 41: 34–38

    CAS  Google Scholar 

  478. Milde LN, Milde JH, Gallagiter WJ (1990) Effects of sufentanil on cerebral circulation and metabolism in dogs. Anesth Analg 70 /2: 138–146

    Article  PubMed  CAS  Google Scholar 

  479. Weinstabl C, Mayer N, Richling B, Czech T, Spiss CK (1991) Effects of sufentanil on intracranial pressure in neurological patients. Anaesthesia 46: 837–840

    Article  PubMed  CAS  Google Scholar 

  480. Sperry RJ, Bailey PL, Reichman MV, Peterson JC, Peterson PB, Pace NL (1992) Fentanyl und sufentanil increase intracranial pressure in head trauma patients. Anesthesiology 77 /3: 416–420

    Article  PubMed  CAS  Google Scholar 

  481. Stephan H, Gröger P, Weyland A, Hoeft A, Sonntag H (1991) Einfluß von Sufentanil auf Hirndurchblutung, Hirnstoffwechsel und die C02-Reaktivität der menschlichen Hirngefäße. Anästhesist 40: 153–160

    CAS  Google Scholar 

  482. Mathews HML, Furness G, Carson IW (1988) Comparison of sufentanil-oxygen and fentanyl-oxygen anaestheia for coronary artery bypass grafting. B J Anaesth 60: 530–535

    Article  CAS  Google Scholar 

  483. Hecker BR, Lake CL, DiFazio CA, Moscicki JC, Engle JS. The decrease of the minimum alveolar anesthetic concentration produced by sufentanil in rats. Anesth Analg 1983 62: 987–990

    Article  PubMed  CAS  Google Scholar 

  484. Hall Rl, Murphy MR, Hug CC (1987) The enflurane sparing effect of sufentanil in dogs. Anesthesiology 67: 518–525

    Article  PubMed  CAS  Google Scholar 

  485. Gravlee GP, Ramsey FM, Roy RC (1987) Rapid administration of a narcotic and a neuromuscular blocker: A hemodynamic comparison of fentanyl, sufentanil, pancuronium, and vecuronium. Anesth Analg 67: 39–47

    Google Scholar 

  486. Freye E (1994) Opioide und NSAIDs. In: Doenicke A (Hrsg) Zentraleuropäischer Anästhesiekongress, Wien. Anästhesist 44/2: S149

    Google Scholar 

  487. Helmers JH, Van Leuwen L, Zuurmond W (1989) Sufen-tanil-Dosierungsstudie bei allgemeinen chrirurgischen Eingriffen. Anästhesist 38: 397–400

    CAS  Google Scholar 

  488. Monk JP, Beresford R, Ward A (1988) Sufentanil. A review of its pharmacological properties and therapeutic use. Drugs 36: 286–313

    Google Scholar 

  489. Morton JP, Hardman HD, Kamiyama Y, Donn KH, Glass PSA (1991) Analgesic efficacy of single escalating doses of Gl 87084B administered intravenously to healthy adult male volunteers. Anesthesiology 75/3 A: A378

    Google Scholar 

  490. Schuster SV, Bilotta JM, Lutz MW, James MK (1991) Analgesic activity of the ultrashort acting opioid, Gl 87804B. FASEB Journal 5 /4: 2846

    Google Scholar 

  491. Westmoreland C, Sebel PS, Hug Jr CC, Hoke JF, Muir KT (1993) Histamine levels and hemodynamic responses following remifentanil. Anesthesiology 79 /3: A111

    Google Scholar 

  492. Westmoreland C, Hoke JF, Sebel PS, Hug Jr CC, Muir LT (1993) Pharmacokinetics of remifentanil (GI87084B). Anesthesiology 79 /3: A372

    Google Scholar 

  493. Egan TD, Lemmens HJM, Fiset P et al. (1993) The pharmacokinetics of the new short-acting opioid remifentanil (GI87084B) in healthy adult male volunteers. Anesthesiology 79: 881–892

    Article  PubMed  CAS  Google Scholar 

  494. Anand KJS, Brown MJ, Canson RC, Christofides ND, Bloom SR, Aynsley-Green A (1985) Can the human neonate mount an endocrine and metabolic response to surgery? J Pediatr Surg 20: 41–48

    Article  PubMed  CAS  Google Scholar 

  495. Anand KJS, Hansen DD, Hickey RP (1990) Hormonal-meta-bolic stress response in neonates undergoing cardiac surgery. Anesthesiology 73: 661–670

    Article  PubMed  CAS  Google Scholar 

  496. Kanwal JS, Anand MB (1986) The stress response to surgical trauma: From physiological basis to therapeutic implications. Prog Food Nutr Sei 10: 67–123

    Google Scholar 

  497. Hickey PR, Hansen DD, Wessel DT, Lang P, Jonas RA (1985) Pulmonary and systemic hemodynamic responses to fentanyl in infants. Anesth Analg 64: 483–486

    PubMed  CAS  Google Scholar 

  498. Anand KJS, Sippe WG, Aynsley-Green A (1987) Randomised trial of fentanyl anaesthesia in preterm babies undergoing surgery: effects on the stress response. Lancet I: 243–248

    Google Scholar 

  499. Anand KJS (1995) Analgesia and sedation in ventilated neonates. Neon Resp Dis 5: 1–10

    Google Scholar 

  500. Pinto-Martin JA, Riolo S, Cnaan A, Holzman C, Süsser MW, Paneth N (1995) Cranial ultrasound prediction of disabling and nondisabling cerebral palsy at age two in a low birth weight population. Pediatrics 95: 249–254

    PubMed  CAS  Google Scholar 

  501. Low JA, Froese AB, Smith JT, Galbraith RS, Sauerbrei EE, Karchmar EJ (1992) Hypotension and hypoxemia in the preterm newborn during the four days following delivery identifies infants at risk of echosonographically demonstrable cerebral lesions. Clin Invest Med 15: 60–65

    PubMed  CAS  Google Scholar 

  502. Anand KJS (1993) Relationships between stress responses and clinical outcome in newborns, infants, and children. Crit Care Med 21: S358 - S359

    Article  PubMed  CAS  Google Scholar 

  503. Roizen MF, Lampe GH, Benefiel DJ (1987) Is increased operative stress associated with worse outcome? Anesthesiology 67: A1

    Article  Google Scholar 

  504. Fitzgerald M (1987) Neurobiology of fetal and neonatal pain. In: Wall PD, Melzack R (eds) The textbook of pain. Chirchill Livingstone, Edinburgh, pp 153–163

    Google Scholar 

  505. McGrath PJ, Johnson G, Goodman JT, Schillinger J, Dunn J, Chapmanm JA (1985) CHEOPS: A behavioral scale for rating postoperative pain in children. Adv Pain Res Ther 9: 395–402

    Google Scholar 

  506. Anand KJS, Craig D (1966) New perspectives on the definition of pain. Pain 67: 3–6

    Google Scholar 

  507. Taddio A, Goldbach M, Ipp M, Stevens B, Koren G (1995) Effect of neonatal circumcision on pain responses during vaccination of boys. Lancet 345: 291–292

    Article  PubMed  CAS  Google Scholar 

  508. Johnston CC, Stevens B, Craig KD, Grunau RVE (1993) Developmental changes in pain expression in preterm, full-term, two-and four-month-old children. Pain 52: 201–208

    Article  PubMed  CAS  Google Scholar 

  509. Reynold ML, Fitzgerald M (1995) Long-term sensory hyperinnervation following neonatal skin wounds. J Comp Neurol 9: 89–95

    Google Scholar 

  510. Fitzgerald M, Anand KJS. Developmental neuroanatomy and neurophysiology of pain. In: Schechter NL, Berde CB, Yasrter M (eds) (1994) Pain in infants, children and adolescents. Williams and Wilkins, Baltimore, pp 11–31

    Google Scholar 

  511. Grunau VE, Whitfield MF, Petrie JH (1994) Pain sensitivity and treatment in extremely low-birth-weight premature toddlers and pre-term and full-term controls. Pain 58: 341–346

    Article  PubMed  CAS  Google Scholar 

  512. McGrath PA (1990) Pain in children: Nature, asssessment and management. Guilford, New York

    Google Scholar 

  513. Caspi A, Henry B, McGee RO, Moffitt TE, Silva PA (1995) Temperamental origins of child and adolescent behavior problems: from age three to age fifteen. Child Develop 66: 55–68

    Article  PubMed  CAS  Google Scholar 

  514. Grunau RVE, Whitfield MF, Petrie JH, Fryer EL (1994) Early pain experience, child and family factors as precursors of somatization: a prospective study of extremely premature and fullterm children. Pain 56: 353–359

    Article  PubMed  CAS  Google Scholar 

  515. Plotsky PM, Meaney MJ (1993) Early postnatal experience alters hypothalamic corticotropin-releasing factor ( CRF), mRNA, median eminence CRF content and stress-induced release in adult rats. Mol Brain Res 18: 195–200

    Google Scholar 

  516. Landfield PW, McEwan BS, Sapolsky RM, Meaney MJ (1996) Hippocampal cell death. Science 272: 1249–1251

    Article  PubMed  CAS  Google Scholar 

  517. Anand KJS, McGrath PJ. Future directions. In: Anand KJS, McGrath PJ (eds) (1993) Pain in neonates. Elsevier, Amsterdam, pp 321–333

    Google Scholar 

  518. Meaney MJ, Sharma S, Sarrieau S, Plotsky PM (1988) Postnatal development and environmental regulation of hippocampal glucocorticoid and mineralocoticoid receptors in the rat. Dev Brain Res 43: 158–162

    Article  Google Scholar 

  519. McEwen BS (1994) The plasticity of the hippocampus is the reason for its vulnerability. Neurosciences 6: 239–246

    Google Scholar 

  520. Jacobson B, Eklund G, Hamberger L, Linnarsson D, Sevdall G, Vlaverius M (1987) Perinatal origin of adult self-destructive behavior. Acta Psychiat Scand 76: 364–371

    Article  PubMed  CAS  Google Scholar 

  521. Schade JP, Ford H (1972) Basic Neurology, 2nd Edn. Elsevier, Amsterdam

    Google Scholar 

  522. Bauer-Miettingen U (1984) Probleme der postoperativen Schmerzbekämpfung bei Kindern. Orthopädie 13: 244–248

    Google Scholar 

  523. Williamson PS, Williamson ML (1983) Physiologie stress reduction by a local anesthetic during newborn circumcision. J Pediatr 71: 36–40

    CAS  Google Scholar 

  524. Coyle JT, Pert CB (1976) Ontogenetic development of (3H)-naloxone binding in rat brain. Neuropharmacology 15: 555–560

    Article  PubMed  CAS  Google Scholar 

  525. Jacobsen M (1970) Developmental neurobiology. Holt, Rinehard and Winston, New York

    Google Scholar 

  526. Zhang AZ, Pasternak GW (1981) Ontogeny of opioid pharmacology and receptors: high and low affinity site differences. Eur J Pharmacol 73: 2940

    Article  Google Scholar 

  527. Purcell-Jones G, Dormon F, Sumner E (1987) The use of opioids in neonates. A retrospective study of 933 cases. Anaesthesia 42: 1320–1323

    Article  Google Scholar 

  528. Woltmann M, Roth BL, Coscia CJ (1982) Differential postnatal development of mu and delta opiate receptores. Dev Brain Res 3: 679–684

    Article  Google Scholar 

  529. Rosenbaum JS, Holford NHG, Sadee W (1985) In vivo receptor binding of opioid drugs at the mu site. J Pharmacol Exp Ther 233: 735–740

    PubMed  CAS  Google Scholar 

  530. Holaday JW, Porreca F, Rothmann RB (1990) Functional coupling among opioid receptor types. In: Estafanous FG (ed) Opioids in anesthesia, vol II. Butterworth-Heine-mann, Boston London, pp 50–71

    Google Scholar 

  531. Goldmann A, Lloyd-Thomas AR (1991) Pain management in children. Br Med Bull 47: 676–689

    Google Scholar 

  532. Leslie FM, Tso S, Harlbutt DE (1982) Differential appearance of opiate receptor subtypes in neonatal rat brain. Life Sei 31: 1393–1396

    Article  CAS  Google Scholar 

  533. Gal TJ, Di Fazo CA, Moscicki J (1982) Analgesic and respiratory depressant activity of nalbuphine: a comparison with morphine. Anesthesiolog 57: 367–374

    Article  CAS  Google Scholar 

  534. Navarro G, Garcia-Flores S (1984) The use of nalbuphine vs. pethidine in women with labour pain. Mex J Gyn Obstet 5: 20–25

    Google Scholar 

  535. Pasternak GW, Zhang A, Tecott L (1980) Developmential differences between high and low affinity opiate binding sites: their relationship to analgesia and respiratory depression. Life Sei 27: 1185–1190

    Article  CAS  Google Scholar 

  536. Sircar R, Zukin SR (1983) Ontogenety of sigma opiate/ phencyclidine-binding sites in rat brain. Life Sei 33: 255–258

    Article  CAS  Google Scholar 

  537. Smith DJ, Bouchal RL, DeSanctis CA (1987) Properties of the interaction between ket amine and opiate binding sites in vivo and in vitro. Neuropharmacology 26: 1253–1260

    Article  PubMed  CAS  Google Scholar 

  538. Freye E, Dähn H, Engel M (1989) Die zentralnervösen exzi-tatorischen Effekte von Ketamin werden durch den Zusatz von Midazolam verringert–Eine postnarkotische Untersuchung bei Patienten nach kieferchirurgischen Eingriffen in Spontanatmung. Anästh Intensivther Notfallmed 6: 368–372

    Google Scholar 

  539. Piepenbrock S (1987) Prinzipien der Schmerzbehandlung bei Kindern in der peri operativen Phase. In: Meier H (Hrsg) Analgesie bei Kindern. Perimed, Erlangen, pp 59–66

    Google Scholar 

  540. Ikkola KT, Hamunen K, Maunuksela EL (1995) Clinical pharmacokinetics and pharmacodynamics of opioid analgesics in infants and children. Clin Pharmacokinet 28: 385–404

    Article  Google Scholar 

  541. Roth B, Houben F, Hartwig S, Theisohn M, Schlünder C (1991) Erfahrungen zur Analgesie und Sedierung in der pädiatrischen Intensivmedizin. In: Henschel WF (Hrsg) I. Europäisches Analgesieforum–Die Analgesie im Mittelpunkt der Anästhesie. Urban and Schwarzenberg, München, Wien, Baltimore, pp 235–247

    Google Scholar 

  542. Greeley WJ, de Bruijn NP, Davis DP (1986) Pharmacokinetics of sufentanil in pediatric patients. Anesthesiology 65: A 422

    Google Scholar 

  543. Akil H, Watson SJ, Young E (1984) Endogenous opioids. Biology and function. Ann Rev Neurosci 7: 223–255

    Google Scholar 

  544. Greeley WJ, de Bruijn NP, Davis DP (1987) Sufentanil pharmacokinetics in pediatric cardiovascular patients. Anesth Analg 66: 1067–1072

    Article  PubMed  CAS  Google Scholar 

  545. Cube von B, Teschemacher HJ, Herz A, Hess R (1970) 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 265: 455–473

    Article  Google Scholar 

  546. Taeger K, Weninger E, Franke N, Finsterer U, Peter K (1984) Uptake of fentanyl by human lung. Anesthesiology 61: A246

    Article  Google Scholar 

  547. Scott JG, Cooke JE, Stanski DR (1991) Electroencephalo-graphic quantitation of opioid effects: Comparative phar-macodnamics of fentanyl and sufentanil. Anesthesiology 74: 34–42

    Google Scholar 

  548. Freye E (1989) Opioid agonists, antagonists and mixed narcotic analgesics: Their use in postoperative and chronic pain management. Drugs Today 25: 741–754

    Google Scholar 

  549. Okum G, Hauser A, Horrow J, Keykhah M, Begen S (1993) Sufentanil pharmaco kinetics during tourniquet use. Anesth Analg 76: S309

    Google Scholar 

  550. Schenk HD, Ensink FBM, Rhönisch M (1993) Alfentanil - Porträt eines Opioids zur Anästhesie. Urban and Schwarzenberg, München Wien Baltimore

    Google Scholar 

  551. Sifton DW (1988) Drug Interaction and Side Effects Index™ 42 Edn. Physicians Desk Reference (PDR) Medical Economics Company, Oradell/N.Y.

    Google Scholar 

  552. Leak JA (1999) Herbal medicine: Is it an alternative or an unknown? A brief review of popular herbals used iby patients in a pain and symptom managmement practice setting. Curr Rev Pain 3: 226–236

    Article  PubMed  Google Scholar 

  553. Gruenwald J, Brendler T, Jaenicke C (1998) PDR for herbal medicine, Vol 1. Medical Economics Company, Oradell/ N.Y.

    Google Scholar 

  554. Wood M (1986) Plasma drug binding: implications for anesthesiologists. Anesth Analg 65: 786–804

    PubMed  CAS  Google Scholar 

  555. Duthie DJR, Nimmo WS (1987) Adverse effects of opioid analgesics drugs. Br J Anaesth 59: 61–77

    Article  PubMed  CAS  Google Scholar 

  556. Maurer PM, Bartkowski RR (1993) Drug interactions of clinical significance with opioid analgesics. Druf Safety 8: 30–48

    Article  CAS  Google Scholar 

  557. Wilcox GL, Carlson GH, Joachim A, Jurna I (1987) Mutual potentiation of antinociceptive effects of morphine and Clonidine on motor and sensory responses in rat spinal cord. Brain Res 405: 84–93

    Article  PubMed  CAS  Google Scholar 

  558. Becker CE, Briggs AH, Fleckenstein L, Greenberg BL, Hausten PD, Hussar DA (1974) A quick guide to common drug interaction. In: Bigelow J (ed) Patient care. Miller and Fink, Philadelphia: pp 1–32

    Google Scholar 

  559. Hoffmann P (1987) Kombination von Benzodiazepinen und Opioiden. In: Schulte am Esch J, Benzer H (Hrsg) Anal-gosedierung bei Intensivpatienten. Springer, Berlin Heidelberg New York Tokio, pp 50–61 ( Reihe: Anaesthesio-logie und Intensivmedizin, Bd 200 )

    Google Scholar 

  560. Kapp W (1981) Pharmakolgische und toxikologische Aspekte zu Benzodiazepinen. Anästh Intensivther Notfallmed 16: 125–127

    Article  PubMed  CAS  Google Scholar 

  561. Kamp HD (1988) Langzeitsedierung mit Benzodiazepinen. In: Schulte am Esch J, Benzer H (Hrsg) Analgosedierung des Intensivpatienten. Springer, Berlin Heidelberg New York Tokio, pp 35–49 ( Reihe: Anaesthesiologie und Intensivmedizin, Bd 200 )

    Google Scholar 

  562. Borchard U (1981) Tranquillanzien aus pharmakologischer Sicht. Medica 9: 638–643

    Google Scholar 

  563. De Castro J, Parmentier P (1975) Antimorphinques et anesthésie analgésique séquentielle. Ill: Pharmacodynamic des principaux antidotes de la morphine. Academia S.A., Bruxelles

    Google Scholar 

  564. Blum R, Zsigmond EK, Winnie AP (1982) Potentiation of opioid analgesia by Hr and H2-antagonists. Life Sci 31: 1229–1232

    Article  Google Scholar 

  565. Freye E (1992) Die perioperative Behandlung des Arzneimittelabhängigen. Anästh Intensivmed 3: 59–67

    Google Scholar 

  566. Rawal N (1996) Klinischer Einsatz der rückenmarknahen Opioidanalgesie, Teil 1. Schmerz 10: 176–189

    Article  PubMed  CAS  Google Scholar 

  567. Börner U, Müller H, Stoyanov M, Hempelmann G (1980) Epidurale Opiatanalgesie–Gewebe und Liquorverträg-lichkeit der Opiate. Anästhesist 29: 570–571

    Google Scholar 

  568. Bürkle H, Dunbar S, van Aken H (1996) Remifentanil: a novel, short-acting, u-opioid. Anesth Analg 83: 646–651

    PubMed  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  570. Stevens CW, Lacey CB, Miller KE, Eide RP, Seybold VS (1991) Biochemical characterization and regional quantification of in, 5 and k opioid binding sites in rat spinal cord. Brain Res 550: 77–85

    Article  PubMed  CAS  Google Scholar 

  571. De Castro J, Lecron L (1981) Peridurale Opiatanalgesie. Verschiedene Opiate–Komplikationen und Nebenwirkungen. In: Zenz M (Hrsg) Peridurale Opiatanalgesie. G. Fischer, Stuttgart New York, pp 103–109

    Google Scholar 

  572. Moore RA, Bullingham RES, McQuay HJ, Hand CW, Aspel JB, Allen MC (1982) Dural permeability to narcotics: in vitro determination and application to extradural administration. Br J Anaesth 54: 1117–1127

    Article  PubMed  CAS  Google Scholar 

  573. Leicht CH, Rosen MA, Dailey PA et al. (1986) Evaluation and comparison of epidural sufentanil citrate and morphine sulfate for analgesia after cesarean section. Anesthesiology 65: A 365

    Google Scholar 

  574. Camporesi EM, Nielsen CH, Bromage PR (1983) Ventilatory C02 sensitivity after intra venous and epidural morphine in volunteers. Anesth Analg 62: 633

    Article  PubMed  CAS  Google Scholar 

  575. Bromage PR, Camporesi EM, Durant PAC, Nielsen CH (1982) Rostral spread of epidural morphine. Anesthesiology 56: 431–436

    Article  PubMed  CAS  Google Scholar 

  576. Johnson A, Bengtsson M, Söderlind K, Löfström JB (1992) Influence of intrathecal morphine and naloxone intervention on postoperative ventilatory regulation in elderly patients. Acta Anaesthesiol Scand 36 /5: 436–444

    Article  PubMed  CAS  Google Scholar 

  577. McCaughey W, Graham IL (1982) The respiratory depression of epidural morphine: time course and effect of posture. Anaesthesia 37: 990–994

    Article  PubMed  CAS  Google Scholar 

  578. Glynn CI, Mather LE, Cousins ME (1979) Spinal narcotics and respiratory depression. Lancet II: 356

    Google Scholar 

  579. Cohen SE, Tan S, White PF (1988) Sufentanil analgesia following cesarean section: epidural vs. intravenous administration. Anesthesiology 68: 129–134

    Article  PubMed  CAS  Google Scholar 

  580. Chalmer PC, Lang CM, Greenhouse BB (1988) The use of nalbuphine in association with epidural narcotics. Anes-thesiol Rev 15 /2: 21–27

    Google Scholar 

  581. Bromage PR, Camporesi EM, Durant PAC (1982) Nonrespiratory side effects of epidural morphine. Anesth Analg 61: 490

    PubMed  CAS  Google Scholar 

  582. Rawal N, Möllefors K, Axelsson K (1981) Naloxone reversal of urinary retention after epidural morphine. Lancet II: 1411

    Google Scholar 

  583. Cousins MJ, Mather LE (1984) Intrathecal and epidural administration of opioids. Anesthesiology 61: 276–310

    Article  PubMed  CAS  Google Scholar 

  584. Rawal N, Wattwil M (1982) Respiratory depression after epidural morphine - an experimental and clinical study. Anesth Analg 63: 8

    Google Scholar 

  585. Eisenach JC (1992) Epidural and spinal narcotics. In: Barash PG (ed) ASA Refresher courses in anesthesiology, vol 20. Lippincott, Philadelphia, pp 1–4

    Google Scholar 

  586. Bailey DR, Smith BE (1980) Continuous epidural infusion of fentanyl for postoperative analgesia. Anesthesiology 42: 538

    Article  Google Scholar 

  587. Davies GK, Tolhurst-Cleaver CL, James TL (1980) Respiratory depression after intrathecal opiates. Anaesthesia 35: 1080

    Article  PubMed  CAS  Google Scholar 

  588. Gjessing J, Tomlin PJ (1981) Postoperative pain control with intrathecal morphine. Anaesthesia 36: 268

    Article  PubMed  CAS  Google Scholar 

  589. Rutter DV, Skewes DG, Morgan M (1981) Extradural opioids for postoperative analgesia. A double blind comparison of pethidine, fentanyl and morphine. Br J Anaesth 53: 915

    Google Scholar 

  590. Kitahata LM, Collins JG (1981) Spinal action of narcotic analgesics. Anesthesiology 54: 153

    Article  PubMed  CAS  Google Scholar 

  591. Gourlay GK, Murphy TM, Plummer JL, Kowalski SR, Cherry DA, Cousins MJ (1989) Pharmacokinetics of fentanyl in lumbar and cervical CSF following lumbar epidural and intravenous administration. Pain 38: 253–259

    Article  PubMed  CAS  Google Scholar 

  592. Weightman WM (1991) Respiratory arrest during epidural infusion of bupivacaine and fentanyl. Anaesth Int Care 19: 282–284

    CAS  Google Scholar 

  593. Vertommen JD, Vandermeulen E, van Aken H et al. (1991) The effects of the addition of sufentanil to 0.125% bupivacaine on the quality of analgesia during labor and on the incidence of instrumental deliveries. Anesthesiology 74: 809–814

    Article  PubMed  CAS  Google Scholar 

  594. Justins DM, Knott C, Luthmann J, Reynolds P (1982) A controlled trial of extradural fentanyl in labour. Br J Anaesth 54: 409–414

    Article  PubMed  CAS  Google Scholar 

  595. Chesnut DH, Owen CL, Bates JN, Ostman LG, Choi WW, Geiger MW (1988) Continuous infusion epidural analgesia during labor: a randomized double-blind comparison of 0.0625% bupivacaine/0.002% fentanyl vs. 0.125% bupivacaine. Anesthesiology 68: 754–758

    Article  Google Scholar 

  596. Fischer RL, Lubenow TR, Liceaga A, McCarthy RJ, Ivanko-vich AD (1988) Comparison of continuous epidural fenta-nyl-bupivacaine and morphine-bupivacaine in management of postoperative pain. Anesth Analg 67: 559–563

    Article  PubMed  CAS  Google Scholar 

  597. Waldvogel HH, Fasano M (1983) Extradural administration of lofentanyl for balanced postoperative pain. Anästhesist 32: 256–257

    Google Scholar 

  598. Durant PAC, Yaksh TL (1986) Epidural injections of bupivacaine, morphine, fentanyl, lofentanil, and DADL in chronically implanted rats: a pharmacologic and pathologic study. Anesthesiology 64: 43–53

    Article  PubMed  CAS  Google Scholar 

  599. Meert TF, Lu HR, van Craenndonck H, Janssen PAJ (1988) Comparison between epidural fentanyl, sufentanil, carfen-tanil, lofentanil and alfentanil in rats: analgesia and other in vivo effects. Eur J Anaesth 5: 313–321

    CAS  Google Scholar 

  600. Vertomme JD, Lemmers E, van Aken H (1994) Comparison of the addition of three different doses of sufentanil to 0.125% bupivacaine given epidurally during lanbor. Anaesthesia 49: 678–681

    Article  Google Scholar 

  601. Vandermeulen EP, van Aken H, Vertommen JD (1995) Labor pain relief using bupi vacaine and sufentanil: patient controlled epidural analgesia vs. intermittent injections. Gynecol Reprod Biol 59: 35–38

    Article  Google Scholar 

  602. Browner G, Mertes N, Buerkle MAF, van Aken H (2000) Acute pain management: analysis implications and consequences after prospective experience with 6349 surgical patients. Eur J Anaesthesiol 17: 566

    Google Scholar 

  603. Camann WR, Denney RA, Holby ED, Datta S (1992) A comparison of intrathecal, epidural, and intravenous sufentanil for labor analgesia. Anesthesiology 77: 884–892

    Article  PubMed  CAS  Google Scholar 

  604. McQuay HJ, Sullivan AS, Smallmann K, Dikenson AH. Intrathecal opioids, potency, and lipophilicity. Pain 1989 36: 111–115

    Article  PubMed  CAS  Google Scholar 

  605. Hansdottir V, Hedner T, Woestenborghs R, Nordberg G (1991) The CSF and plasma pharmacokinetics of sufentanil after intrathecal administration. Anesthesiology 74: 264–269

    Article  PubMed  CAS  Google Scholar 

  606. D’Angelo R, Anderson MT, Philipp J, Eisenach JC (1994) Intrathecal sufentanil compared to epidural bupivacaine for labor analgesia. Anesthesiology 80: 1209–1215

    Article  PubMed  Google Scholar 

  607. Jones RDM, Jones JG (1980) Intrathecal morphine: naloxone reverses respiratory depression but not analgesia. Br Med J 281: 645–646

    Article  PubMed  CAS  Google Scholar 

  608. Meignier M, Canansia MF, Lejus C, Souron R (1993) Continuous intrathecal opioids and bupivcaine for the mange-ment of intractable cancer pain in children. Anesth Analg 76: S259

    Article  Google Scholar 

  609. Brown DV, McCarthy RJ. Epidural and spinal opioids. Curr Opinion Anaesth 1995 8: 337–341

    Article  Google Scholar 

  610. Cousins MJ (1988) The spinal route of analgesia. Acta Anaesth Belg 39: 71–82

    PubMed  CAS  Google Scholar 

  611. Mok MS, Lippmann M, Wang JJ, Chan K, H, Lee TY (1981) Efficacy of epidural nalbuphine in postoperative pain control. Anesthesiology 61: A187

    Google Scholar 

  612. Kalia PK, Madan S, Saksena R, Batra RK, Gode GR (1983) Epidural pentazocine for postoperative pain relief. Anesth Analg 62: 949

    Article  PubMed  CAS  Google Scholar 

  613. Mok MS, Tsai K, Chan KH, Lee TY, Lippman M (1984) Analgesic effect of intrathecal stadol, nubain, meperidine, morphine and fentanyl, a comparative study. VIII. World Congress of Anaesthesiologists. Manila/Philippines, A213

    Google Scholar 

  614. Malinovsky JM, Cozian A, Lepage JY, Mussini JM, Pinaud M, Souron R (1991) Ketamine and midazolam neurotoxicity in the rabbit. Anesthesiology 75: 91–97

    Article  PubMed  CAS  Google Scholar 

  615. Ossipov MH, Suarez LJ, Spaulding TC (1989) Antinociceptive interactions beween alpha2-adrenergic and opiate agonists at the spinal level of rodents. Anesth Analg 68: 194–200

    Article  PubMed  CAS  Google Scholar 

  616. Carabine UA, Milligan KR, Moor J (1992) Extradural cloni-dine and bupivacaine for postoperative analgesia. 68: 132–135

    CAS  Google Scholar 

  617. Eisenach JC, Lysak SZ, Viscomi CM (1989) Epidural cloni-dine analgesia following sugery. Phase I. Anesthesiology 71: 640–646

    Google Scholar 

  618. Eisenach JC, Rauck RL, Buzzanell C, Lysak SZ (1989) Epidural clonidine analgesia for intractable cancer pain. Phase I. Anesthesiology 71: 647–652

    Google Scholar 

  619. Zenz M, Piepenbrock S, Tryba M (1985) Epidural opiates: Long-term experiences in cancer pain. Klin Wochenschr 63: 225–229

    Google Scholar 

  620. Herz A (1996) Peripheral opioid analgesia–facts and mechanisms. Progr. Brain Res 110: 95–104

    Article  CAS  Google Scholar 

  621. Paton WD (1957) The action of morphine and related substances on the actylcholine output of coaxially stimulated guinea pig ileum. Br J Pharmacol 11: 119

    Google Scholar 

  622. Stein C (1993) Peripheral mechanism of opioid analgesia. Anesth Analg 76: 182–191

    Article  PubMed  CAS  Google Scholar 

  623. Stein C (1993) Periphere Opiatrezeptoren und ihre Bedeutung für die postoperative Schmerztherapie. Schmerz 7: 4–7

    Article  PubMed  CAS  Google Scholar 

  624. Picard PR, Tramèr MR, McQuay HJ, Moore RA (1997) Analgesie efficacy of peripheral opioids (all except intraarticular): A qualitative systemic review of randomized controlled trials. Pain 72: 309–318

    Google Scholar 

  625. Sefrin P (1986) Auswirkungen des Schmerzes als pathogenetischer Faktor in der Notfallmedizin am Beispiel des Polytraumatisierten. In: Sefrin P (Hrsg) Schmerz in der Notfallmedizin. Zuckschwerdt, München, pp 11–20 ( Reihe: Klinische und experimentelle Notfallmedizin, Bd 5 )

    Google Scholar 

  626. Jaättela A, Alha A, Avkainen V et al (1975) Plasma catecholamines in severely injured patients: a prospective study on 45 patients with multiple injuries. Br J Surg 62: 177

    Article  PubMed  Google Scholar 

  627. Dick W (1978) Schmerzbehandlung im Notfall und der Notsituation. Notfallmed 4: 52

    Google Scholar 

  628. Busse C (1987) Notfalltherapie mit Betäubungsmitteln und Analgetika. Notfallmedizin 13: 426–439

    Google Scholar 

  629. Sefrin P, Blumenberg D (1988) Präklinische Analgesie bei internistischen Notfall patienten. Notfallmedizin 32: 636–641

    Google Scholar 

  630. Pfenninger E (1992) Möglichkeiten der Schmerzbehandlung am Unfallort. Klinikmagazin 3: 20–23

    Google Scholar 

  631. Seferin W (1982) Indikationen zur Beatmung und Narkose im Rettungsdienst. In: Schildberg FW, de Pay AW (Hrsg) Atemstörungen im Rettungsdienst. Interdisziplinäre Aspekte. Perimed, Erlangen, pp 153–158

    Google Scholar 

  632. Jones J, Hoggart B, Withey J, Donaghue K, Ellis B (1979) What the patients say: A study of reactions to intensive care. Intensive Care Med 5: 89–92

    Google Scholar 

  633. Dennhardt R (1986) Fentanyl/Droperidol zur Analgesie und Sedierung bei beatmeten Intensivpatienten. Anästh Akt 11: 1–5

    Google Scholar 

  634. Hecht U, Lehmkuhl P, Pichlmayr I (1989) Sedierung zur postoperativen Beatmung: Midazolam vs. Propofol-Erste Ergebnisse. In: Link J, Eyrich K (Hrsg) Analgesie und Sedierung in der Intensivmedizion. Springer, Berlin Heidelberg New York Tokio, pp 99–104 ( Reihe: Anaesthesiologie und Intensivmedizin, Bd 212 )

    Google Scholar 

  635. Adams HA, Biscoping J, Thiel A, Hempelmann G (1989) Analgosedierung beatmungspflichtiger Intensivpatienten mit einer Ketamin-Midazolam-Kombination. In: Link J, Eyrich K (Hrsg) Analgesie und Sedierung in der Intensivmedizion. Sprin ger, Berlin Heidelberg New York Tokio, pp 105–112 ( Reihe: Anaesthesiologie und Intensiv medi-zin, Bd 212 )

    Google Scholar 

  636. Lenhart FP, Frey L, Wilm V, Taeger K (1989) Sedierung langzeitbeatmeter Patienten mit Methohexital und Opio- ¡den. In: Link J, Eyrich K (Hrsg) Analgesie und Sedierung in der Intensivmedizin. Springer, Berlin,Heidelberg, New York, pp 74–84 ( Reihe: Anaesthesiologie und Intensivmedizin, Bd 212 )

    Google Scholar 

  637. Brandl M, Braun J, Knoll GR, Schütz W (1989) Langzeit-sedierung neurochirurgischer Patienten mit Methohexital. In: Link J, Eyrich K (Hrsg) Analgesie und Sedierung in der Intensivmedizin. Springer, Berlin Heidelberg New York Tokio, pp 64–73 ( Reihe: Anaesthesiologie und Intensivmedizin, Bd 212 )

    Google Scholar 

  638. Hoffmann P (1987) Möglichkeiten individueller Analgose-dierung in der Intensivmedizin. In: Henschel WF (Hrsg) Anästhesiologie–Klinisches Fach auf drei Säulen. Zuck-schwerdt, München, pp 351–356

    Google Scholar 

  639. Kochs E, Bischoff P, Rust U, Schulte am Esch J (1988) Beeinflussung des Hypophysen-Nebennierenrinden-Systems durch Langzeitanalgosedierung. In: Schulte am Esch J, Benzer H (Hrsg) Analgosedierung des Intensivpatienten. Springer, Berlin Heidelberg New York Tokio, pp 62–68 ( Reihe: Anaesthesiologie und Intensivmedizin, Bd 200 )

    Google Scholar 

  640. Kurth M (1983) Anästhesie und Analgosedierung mit Keta-min bei Patienten einer Intensivstation. Anästh Intensiv-med 24: 270–272

    Google Scholar 

  641. Adams HA, Biscoping J, Russ W, Bachmann B, Ratthey K, Hempelmann G (1988) Untersuchungen zur sedativ-anal-getischen Medikation beatmungspflichtiger Patienten. Anästhesist 37: 2658–2676

    Google Scholar 

  642. Cohen AT (1987) Experience with alfentanil infusion as an intensive care sedative analgesie. Eur J Anaesthesiol 1: 63–66

    CAS  Google Scholar 

  643. Farina ML, Levati A, Tognoni GA (1981) A multicentre study of ICU drug utilization. Intensive Care Med 7: 125–131

    Article  PubMed  CAS  Google Scholar 

  644. Gast PH, Fischer A, Sear JW (1981) Intensive care sedation now. Lancet II: 863–864

    Google Scholar 

  645. Kochs E, Schulte am Esch J (1987) Hormone des Hypophy-sen-Nebennierenrindensystems bei Patienten unter Lang-zeitsedierung mit Etomidat und Fentanyl. Anästhesist 33: 402–407

    Google Scholar 

  646. Sear JW, Fischer A, Summerfield RJ (1987) Is alfentanil by infusion useful for sedation in the ICU? Eur J Anaesth Supp 1: 55–61

    CAS  Google Scholar 

  647. Sinclair ME, Sear JW, Summerfield RJ, Fisher A (1988) Alfentanil infusions on the intensive therapy unit. Intensive Care Med 14: 55–59

    Article  PubMed  CAS  Google Scholar 

  648. Uray E, Kosa CS (1969) Wirkung der bei Neuroleptanal-gesie verwendeten Medikamente auf die Druckwerte der Gallenwege. Anästhesist 18: 74–77

    CAS  Google Scholar 

  649. Kroesen G, Bodner E, Russe W, Troyer E (1978) Beeinflussung der intraoperativen Cholangiometrie durch Anästhesiemethoden. Anästhesist 27: 21–24

    CAS  Google Scholar 

  650. Nickel B, Schmickaly R, Kursawe HK et al. (1986) Beitrag zur Therapie der Delirium tremens. Z Klin Med 41: 1643–1646

    Google Scholar 

  651. Ohlendorf H, Jong Md, Steenhoek A, Janknegt R (1988) Clinical pharmacokinetics of midazolam in intensive care patients, a wide interpatient variability? Clin Pharmacol Ther 43: 263–269

    Article  Google Scholar 

  652. Striebel HW, Papadopoulos G, Heinemeyer G, Link J (1989) Langzeitanalgosedierung mit Flunitrazepam, Fentanyl und Dehydrobenzperidol bei schwerstkranken Intensivpatienten: Pharmakokinetik-Pharmakodynamik. In: Link J, Eyrich K (Hrsg) Analgesie und Sedierung in der Intensivmedizin. Springer, Berlin Heidelberg New York Tokio, pp 135–148 ( Reihe: Anaesthesiologie und Intensivmedizin, Bd 212 )

    Google Scholar 

  653. Bergmann H (1978) Rohypnol. Pharmakologische Grundlagen - Klinische Anwendung. In: Ahnefeld FW, Bergmann H, Burri H (Hrsg) Klinische Anästhesiologie und Intensivtherapie, Bd. 17. Springer, Berlin Heidelberg New York Tokio, S 130

    Google Scholar 

  654. Alazia M, Levron JC (1987) Etude pharmacocinetique d’une perfusion intraveineuse prolongée de fentanyl en réanimation. Ann Fr Anesth Réanim 6: 465–466

    Article  PubMed  CAS  Google Scholar 

  655. Ramsey MAE, Savage TM, Simpson BNRJ, Goodwin R (1974) Controlled sedation with alphaloxone-alphadone. Br Med J II: 656–659

    Google Scholar 

  656. Weiss M, Schneider M, Wernet P (1994) Einfluß von Hyp-notika und Sedativa auf die Immunabwehr. Jahrbuch der Anästhesiologie und Intensivmedizin. Biermann, München, pp 27–37

    Google Scholar 

  657. Moudgil GC, Singal DP, Gordon J, Forrest JB (1984) Fentanyl, sufenanil and alfentanil do not inhibit cell-mediated immune response. Anesthesiology 61/3: A 355

    Google Scholar 

  658. Kalenda Z, Scheijgrond JW (1976) Anaesthesia with sufen-tanil-analgesia in carotid and vertebral arteriography. A comparison with fentanyl. Anästhesist 25: 380–383

    Google Scholar 

  659. Cheour S (1988) Comparison of sufentanil-N20 and fenta-nyl-N20 in patients undergoing general surgery. A blind study. Anesthesiology 69: A213

    Article  Google Scholar 

  660. Wappler F, Scholz J, Prause A, Möllenberg O, Bause H, Schulte am Esch J (1998) Stufenkonzept zur Analgosedierung in der Intensivmedizin mit Sufentanil. Anästhesiol Intensivmed Notfallmed Schmerzther 33: 18–26

    Article  Google Scholar 

  661. Kessler P (1997) Industrieforum: Sufentanil in der klinischen Anwendung–Wo liegen die Vorteile? Anästhesiol Intensivmed 12: 647–652

    Google Scholar 

  662. Schwieger IA, Hall Rl, Hug CCH (1991) Less than additive antinociceptive interaction between midazolam and fentanyl in enflurane-anesthetized dogs. Anesthesiology 74: 1060–1066

    Article  PubMed  CAS  Google Scholar 

  663. Palaoglu O, Ayhan IH (1986) The possible role of benzodiazepine receptors in morphine analgesia. Pharmacol Biochem Behav 25: 215–217

    Article  PubMed  CAS  Google Scholar 

  664. Tonner PH, Scholz J (1996) Clinical perspectives of alpha-2 adrenoreceptor agonists. Curr Opinion Anaesth 9: 471–480

    Google Scholar 

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

    Google Scholar 

  666. Bredas P, Grounds M, Roedler G, Torres KM (1998) Optimizing analgesia in the intensive care unit. In: Maze M, Morrison P (eds) Redefining sedation. Royal Society of Medicine Press, London, pp 51–61

    Google Scholar 

  667. Talke P, Richardson CA, Scheinin M, Fischer DM (1997) Postoperative pharmacokinetics and sympatholytic effects of dexmedetomidine. Anesth Analg 85: 1136–1142

    PubMed  CAS  Google Scholar 

  668. Shafer A, White P, Schüttler J, Rosenthal MH (1983) Use of fentanyl infusion in the intensive care unit: Tolerance to its anesthetic effects? Anesthesiology 59: 245–248

    PubMed  CAS  Google Scholar 

  669. Gear RW, Miaskowski C, Heller PH, Paul SM, Gordon NC, Levine JD (1997) Benzodiazepine mediated antagonism of opioid analgesia. Pain 71: 25–29

    Article  PubMed  CAS  Google Scholar 

  670. Daghero AM, Bradley EL, Kissin I (1987) Midazolam antagonizes the analgesic effect of morphine in rats. Anesth Analg 66: 944–947

    Article  PubMed  CAS  Google Scholar 

  671. Rosland JH, Hole K (1990) Benzodiazepine-induced antagonism of opioid antinociception may be abolished by spinalization or blockade of the benzodiazepine receptor. Pharmacol Biochem Behav 37: 505–509

    Article  PubMed  CAS  Google Scholar 

  672. Rady JJ, Fujimoto JM (1993) Dynorphin A(1–17) mediates midazolam antagonism of morphine antinociception in mice. Pharmacol Biochem Behav 46: 331–339

    Article  PubMed  CAS  Google Scholar 

  673. Hofbauer R, Tesinsky P, Hammerschmidt V et al. (1999) No reduction in the sufentanil requirement of elderly patients undergoing ventilatory support in the medical intensive care unit. Eur J Anaesthesiol 16: 702–707

    PubMed  CAS  Google Scholar 

  674. Boulard G, Maurette P, Pouguet P et al (1983) Syndrome abstinence après arrêt de la sedation par fentanyl en neuro-reanimation. Ann Fr Anesth Réanim 2: 100–101

    Article  PubMed  CAS  Google Scholar 

  675. Freye E (1987) Opiate und ihre Antagonisten–Bedeutung der Rezeptorspezifität für die klinische Wirkung. Analgesie und Sedierung in der Intensivmedizin. Infomed Institut für Medizinische Kommunikation, München, pp 19–21

    Google Scholar 

  676. Gal TJ, DiFazio CA (1986) Prolonged antagonism of opioid action with intravenous nalmefene in man. Anesthesiology 64: 175–180

    Article  PubMed  CAS  Google Scholar 

  677. Martin WR, Jasinski DR, Mansky PA (1973) Naltrexone, an antagonist for the treatment of heroin dependence. Arch Gen Psychiatry 28: 784–791

    Article  PubMed  CAS  Google Scholar 

  678. Martin WR (1979) History and development of mixed opioid agonists, partial agonists and antagonists. Br J Clin Pharmacol 7: 732

    Article  Google Scholar 

  679. Moore LR, Bikhazi GB, Tuttle RR, Weidler DJ (1990) Antagonism of fentanyl-induced respiratory depression with nalmefene. Meth Find Expt Clin Pharmacol 12 /1: 29–35

    CAS  Google Scholar 

  680. Nagrajan R, Jeyashree S, Shanmugam M, Subramanian VB, Tuttle RR, Caldwell RW (1992) Reversal effects of morphine by nalmefene. J Clin Res Drug Dev 7: 25–29

    Google Scholar 

  681. Flacke JW, Flacke WE, Williams GD (1977) Acute pulmonary edema following naloxone reversal of high dose morphine anesthesia. Anesthesiology 47: 376–378

    Article  PubMed  CAS  Google Scholar 

  682. Smith TC (1979) Comparison of naloxone and naltrexone in man. Anesthesiology 51: S 573

    Article  Google Scholar 

  683. Goldfrank L, Flamenbaum N, Weismann RS (1981) General management of the poisened overdosed patient. Part 1. Patients in coma with altered mental status. Hosp Physician 17: 24–62

    Google Scholar 

  684. Volans GN, Henry JA (1983) Naloxon in der Notfallmedizin. Therapiewoche 33: 2095–2105

    Google Scholar 

  685. Freye E, Härtung E, Schenk GK (1983) Effects of the three narcotic antagonists (naltrexone, diprenorphine, S-20682) on blood pressure, heart rate and electrical cortical activity. Pharmacology 26: 110–116

    Article  PubMed  CAS  Google Scholar 

  686. Volavka J, James B, Reker D, Mallya A, Cho D, Pevnik J (1979) EEG and other effects of naltrexone and heroin in man. Pharmakopsychiat 12: 79–85

    Article  CAS  Google Scholar 

  687. Verebey K, Volavka J, Mule SJ, Resnik RB (1976) Naltrexone: Disposition, meta bolism and effects after acute and chronic dosing. Clin Pharmacol Ther 20: 315–328

    Google Scholar 

  688. Kleber HD, Topazian M, Gaspari J, Riordan CE, Kosten T (1987) Clonidine and naltrexone in the outpatient treatment of heroin withdrawal. Am J Drug Alcohol Abuse 13/1, 2: 1–17

    Article  Google Scholar 

  689. Volpicelli JR, Alterman AI, Hayashida M, O’Brien CP (1992) Naltrexone in the treatment of alcohol dependence. Arch Gen Psychiatry 49: 876–880

    Article  PubMed  CAS  Google Scholar 

  690. ’Malley SS, Jaffe AJ, Chang G, Schottenfeld RS, Meyer RE, Rounsaville B (1992) Naltrexone and coping skills therapy for alcohol dependence. Arch Gen Psychiatry 49: 881–887

    Article  Google Scholar 

  691. Greenstein RA, Evans BD, McLellan AT, O’Brien C (1983) Predictors of favorable outcome following naltrexone treatment. Drug Alcohol Depend 12: 173–180

    Article  PubMed  CAS  Google Scholar 

  692. Sagy M, Shavit G, Oron Y, Vidnre BA, Gitter S, Same Y (1987) Nonopiate effects of naloxone on cardiac muscle contractility. J Cardiovasc Phramacol 9: 682–685

    Article  CAS  Google Scholar 

  693. Sanky R (1985) Naloxone abolishes self injuring in a mentally retarded child. Ann Neurol 17: 520

    Article  Google Scholar 

  694. EgarterC (1987) Plasma-Beta-Endorphinspiegel und Nalo-xontest bei Hyperlaktin ämie. Gynäk Rdsch 27: 38–46

    Google Scholar 

  695. Faden AI, Jacobs TP, Holaday JW (1981) Opiate antagonist improves neurologic recovery after spinal injury. Science 211: 493

    Article  PubMed  CAS  Google Scholar 

  696. Stokes BT (1984) Improvement in injury-induced hypocalcemia by high dose naloxone intervention. Brain Res 290: 187

    Article  PubMed  CAS  Google Scholar 

  697. Barsan WG, Olinger CP, Adams HP et al. (1989) Use of high dose naloxone in acute stroke: Possible side-effects. Crit Care Med 17: 762–767

    Article  PubMed  CAS  Google Scholar 

  698. Katz J, Kavanagh BP, Sandler AN et al. (1992) Preemptive analgesia-clinical evidence of neuroplasticity contributing to postoperative pain. Anesthesiology 77: 439–446

    Article  PubMed  CAS  Google Scholar 

  699. Wall PD (1988) The prevention of postoperative pain. Pain 33: 289–290

    Article  PubMed  CAS  Google Scholar 

  700. Kehlet H (1989) The stress response to surgery: release mechanisms and the modifying effect of pain relief. Acta Chir Scand 550 (Suppl): 22–28

    CAS  Google Scholar 

  701. Kehlet H (1989) Surgical stress: the role of pain and analgesia. Br J Anaesth 63: 189–195

    Article  PubMed  CAS  Google Scholar 

  702. Scott NB, Kehlet H (1988) Regional anaesthesia and surgical mobidity. Br J Surg 75: 299–304

    PubMed  CAS  Google Scholar 

  703. Woolf CJ (1983) Evidence for a central component of post-injury hypersensitivity. Nature 306: 686–688

    Article  PubMed  CAS  Google Scholar 

  704. Coderre TJ, Melzack R (1987) Cutaneous hyperalgesia: contribution of the peripheral and central nervous system to the increase in pain sensitivity after injury. Brain Res 404: 95–106

    Article  PubMed  CAS  Google Scholar 

  705. Cook AJ, Woolf CJ, Wall PD, McMahon SB (1987) Dynamic receptive field plasticity in the rat spinal cord dorsal horn following C-primary afferent input. Nature 325: 151–153

    Article  PubMed  CAS  Google Scholar 

  706. Wall PD (1988) The prevention of postoperative pain. Pain 33: 289–290

    Article  PubMed  CAS  Google Scholar 

  707. Lombard MC, Besson JM (1989) Attempts to gauge the relative importance of pre-and postoperative effects of morphine on the transmission of noxious messages in the dorsal horn of the rat spinal cord. Pain 37: 335–345

    Article  PubMed  CAS  Google Scholar 

  708. Katz J, Kavanagh BP, Sandler AN (1992) Preemptive analgesia: clinical evidence of neuroplasticity contributing to postoperative pain. Anesthesiology 77: 439–446

    Article  PubMed  CAS  Google Scholar 

  709. Davies SN, Lodge D (1987) Evidence for involvement of N-methylaspertate receptors in »wind-up« of class 2 neurones in the dorsal horn of the rat. Brain Res 424: 402–406

    Article  PubMed  CAS  Google Scholar 

  710. Hong JS, Yang HY, Fratta W, Costa E (1977) Determination of methionine enkephalin in discrete regions of rat brain. Brain Res 134: 383

    Article  PubMed  CAS  Google Scholar 

  711. Snyder SH, U’Prichard DC, Greenberg DA (1978) Neurotransmitter receptor binding in the brain. In: Lipton MA, DiMascio A, Killam KF (eds) Psychopharmacology: A generation of progress. Raven, New York, pp 361–370

    Google Scholar 

  712. I.Wood PL (1982) Multiple opiate receptors: Support for unique ju, 5 and k sites. Neuropharmacology 21: 487–497

    Article  Google Scholar 

  713. Wood PL (1984) k agonists analgesics: Evidence for u2 and 5 opioid receptor antagonism. Drug Dev Res 4: 429–435

    Google Scholar 

  714. Kaiser C, Pontecorvo MJ, Mewshaw RE (1991) Sigma receptor ligands: function and activity. Neurotransmissions 7 /1: 1–5

    Google Scholar 

  715. McKnight AT, Rees DC (1991) Opioid receptors and their ligands. Neurotransmissions 7 /2: 1–6

    Google Scholar 

  716. Kosterlitz HW, Paterson SJ (1980) Characterization of opioid receptors in nervous tissue. Proc R Soc Lond 210: 113–122

    Article  PubMed  CAS  Google Scholar 

  717. Kockott G (1984) Chronisch, vorwiegend psychisch bedingter Schmerz. MMW 126: 667–672

    Google Scholar 

  718. Pinsky JJ (1978) Psychodynamics and psychotherapy in the treatment of patients with chronic pain. In: Crue BL (ed) Chronic Pain. Spectrum, New York

    Google Scholar 

  719. Pilowsky J (1981) Current views on the role of psychiatrists in the management of the chronic pain. In: Swerd-low (ed) The therapy of pain. MTI Press, Lancaster

    Google Scholar 

  720. Smith TW, Hughes J, Kosterlitz HW, Sosa RP (1976) Enkephalins: Isolation, distribution and function. Opiates and Endogenous Opioid Peptides. Elsevier, Amsterdam

    Google Scholar 

  721. Yeadon M, Kitchen I (1990) Multiple opioid receptors mediate the respiratory depressant effect of fentanyl-like drugs in the rat. Gen Pharmac 21: 655–664

    Article  CAS  Google Scholar 

  722. Ling GSF, Spiegel K, Lockhart SH, Pasternak GW (1985) Separation of opioid analgesia from respiratory depression: Evidence of different receptor mechanism. J Pharmacol Exp Ther 232: 149–155

    Google Scholar 

  723. Pasternak GW, Wood PJ (1986) Minireview: Multiple mu opiate receptors. Life Sci 38: 1889–1898

    Google Scholar 

  724. Pasternak GW (1988) Multiple morphine and enkephalin receptors and the relief of pain. JAMA 259: 1362–1367

    Article  PubMed  CAS  Google Scholar 

  725. Magnan J, Paterson SJ, Tavani A, Kosterlitz HW (1982) The binding spectrum of narcotic analgesic drugs with different agonist and antagonist properties. Naunyn-Schmie-debergs Arch Pharmacol 319: 197–205

    Article  CAS  Google Scholar 

  726. Portoghese PS, Sultana M, Takemori AE (1988) Naltrindole, a highly selective and potent non-peptide delta opioid receptor antagonist. Eur J Pharmacol 146: 185–186

    Article  PubMed  CAS  Google Scholar 

  727. Weissmann DE, Haddox JD (1989) Opioid pseudoaddic-tion-an iatrogenic syndrome. Pain 36: 363–366

    Article  Google Scholar 

  728. Willweber-Strumpf A, Zenz M, Strumpf M (1992) Medikamentenabhängigkeit bei der Therapie chronischer Schmerzen. Z Gesamt Inn Med 47: 312–317

    CAS  Google Scholar 

  729. Twycross RG, Lack SA (1983) Symptom control in far-advanced cancer. Pain relief. Pittman, London

    Google Scholar 

  730. Borrison HL, Wang SC (1953) Physiology and pharmacology of vomiting. Pharmacol Rev 5: 192–230

    Google Scholar 

  731. Havemann U, Turski L, Kuschinsky K (1982) Role of opioid receptors in the substantia nigra in morphine-induced muscular rigidity. Life Sci 31: 2319–2322

    Article  PubMed  CAS  Google Scholar 

  732. Paakkari P, Feuerstein G (1988) Antagonism of dermor-phin-induced catalepsy with naloxone, TRH-analog CG3703 and the benzodiazepine antagonist, Ro 15–1788. Neuropharmacology 27 /10: 1007–1012

    Article  PubMed  CAS  Google Scholar 

  733. De Vos V (1978) Immobilization of free-ranging wild animals using a new drug. Vet Ree 103: 64–68

    Article  Google Scholar 

  734. Wörz R, Berlin J (1989) Behandlung chronischer Schmerzsyndrome mit Antidepressiva. Schmerz 3: 1–7

    Article  PubMed  Google Scholar 

  735. Atkinson RE, Schofield P, Mellor P (1990) Opioids in the treatment of cancer pain. In: Doyle D (ed) London Int Congress and Symposium Series, London, vol 146, pp 81–87

    Google Scholar 

  736. Jick H, Miettinen OS, Shapiro S, Lewis GP, Siskind V, Slone D (1970) Comprehensive drug surveillance. JAMA 213: 1455–1460

    Article  PubMed  CAS  Google Scholar 

  737. Babayan EA, Lepakhin VK, Rudenko GM (1980) Opioid analgesics and narcotic antagonists. In: Dukes MNG (ed) Meyers’s side effects of drugs. Exerpta Medica, Amsterdam, pp 105

    Google Scholar 

  738. Schultheiss R, Schramm J, Neidhardt J (1992) Dose changes in long-and medium-term intrathecal morphine therapy of cancer pain. Neurosurgery 31 /4: 664–670

    Article  PubMed  CAS  Google Scholar 

  739. Bruera E, Brenneis C, Michaud M, Chadwick S, MacDonald RN (1987) Continuous sc infusion of narcotics using a portable disposable device in patients with advanced cancer. Cancer Treat Rep 71 /10: 953–957

    PubMed  CAS  Google Scholar 

  740. Schoon W, Erdmann H, Kleeberg UR (1987) Die kontinuierliche subkutane Opi oid infusion in der onkologischen Schmerztherapie. Med Klin 82 /23: 805–811

    CAS  Google Scholar 

  741. Goecke H, Herbst M (1993) Ambulante kontinuierliche subkutane Opioidanalgesie als PCAO (patient controlled analgesia in outpatients) bei schweren Tumorschmerzen. Schmerz 7: 31–39

    Article  Google Scholar 

  742. Moulin DE, Johnson NG, Murray-Parson N, Geoghegan MF, Goodwin VA, Chester MA (1992) Subcutaneous narcotic infusions for cancer pain: treatment outcome and giudelines for use. Can Med Assoc J 146 /6: 891–897

    CAS  Google Scholar 

  743. Herbst M, Goecke H (1991) Ambulante subkutane Infusionsanalgesie mit Opioiden. In: Henschel WF (Hrsg) I. Europäisches Analgesieforum–Die Analgesie im Mittelpunkt der Anästhesie. Urban and Schwarzenberg, München Wien Baltimore, pp 178–187

    Google Scholar 

  744. Westerling D, Andersson KE (1984) Rectal administration of morphine hydrogel: Absorption and bioavailability in women. Acta Anaesth Scand 28: 540–543

    Google Scholar 

  745. Danny WE (1929) Operative relief from pain in lesions of the mouth. Arch Surg 19: 143–148

    Article  Google Scholar 

  746. Sweet WH (1976) Controlled thermocoagulation of trigeminal ganglion and rootlets for differential destruction of pain fibers. Facial pain other than trigeminal neuralgia. Clin Neurosurg 23: 96–102

    PubMed  CAS  Google Scholar 

  747. Young RF, Brechner T (1986) Electrical stimulation of the brain for relief of intractable pain due to cancer. Cancer 57: 1266–1272

    Article  PubMed  CAS  Google Scholar 

  748. Lobato RD, Madrd JL, Fatela LV, Rivas JJ, Reig E, Lamas E (1983) Intraventricular morphine for control of pain in terminal cancer patients. J Neurosurg 59: 627–633

    Article  PubMed  CAS  Google Scholar 

  749. Obbens EAMT, Hill CS, Leavens ME, Ruthenbeck SS, Otis F (1987) Intraventricular morphine administration for control of chronic cancer pain. Pain 28: 61–68

    Article  PubMed  CAS  Google Scholar 

  750. Sandouk P, Serrie A, Urtizberra M, Debray M, Got P, Scherrmann JM (1991) Morphine pharmacokinetics and pain assessment after intracerebroventricular administration in patients with terminal cancer. Clin Pharmacol Ther 49: 442–448

    Article  PubMed  CAS  Google Scholar 

  751. Dennis GC, DeWitty RL (1990) Long-term intraventricular infusion of morphine for intractable pain in cancer of the head and neck. Neurosurgery 26 /3: 404–407

    Article  PubMed  CAS  Google Scholar 

  752. Lehmann KA, Einnolf C, Eberlein HJ, Nagel R (1991) Transdermal fentanyl for the treatment of pain after major uro-logical operations. A randmized double-blind comparison with pacebo using intravenous patient-controlled analgesia. Eur J Clin Pharmacol 41 /1: 17–21

    Article  PubMed  CAS  Google Scholar 

  753. De Castro J, Van de Water A, Wouters L, Xhonneux R, Reneman R, Kay B (1979) Comparative study of cardiovascular, neurological, and metabolic side effects of eight narcotics in dogs. Acta Anaesth Belg 30: 5–99

    PubMed  Google Scholar 

  754. Ghignone M (1987) Effects of clonidine on narcotic requirements and hemodynamic response during induction of fenanyl anesthesia and endotracheal intubation. Anesthesiology 67: 3–10

    Article  PubMed  CAS  Google Scholar 

  755. Fahmy NR, Sunder N, Scoter NA (1984) A comparison of histamine-releasing properties and hemodynamic effects of morphine and nalbuphine in humans. Anesth Analg 63: 210

    Google Scholar 

  756. Hermann DJ, Marton JP, Donn KH et al (1991) Pharmacokinetic comparison of GI87084B, a novel ultra-short acting opioid, and alfentanil. Anesthesiology 75: A 379

    Google Scholar 

  757. Herz A, Teschemacher HJ (1971) Activities and site of antinociceptive action of morphine-like analgesics. Adv Drug Res 6: 79–119

    CAS  Google Scholar 

  758. Ferreira SH, Nakamura M (1979) Prostaglandin hyperalgesia: the peripheral analgesic activity of morphine, enkephalin and opioid antagonists. Prostaglandins 18: 191–200

    Article  PubMed  CAS  Google Scholar 

  759. Joris JL, Dubner R, Hargreaves KM (1987) Opioid analgesia at peripheral sites: a target for opioid released during stress and inflammation. Anesth Analg 66: 1277–1281

    Article  PubMed  CAS  Google Scholar 

  760. Stein C, Comisel K, Haimerl E et al. (1991) Analgesic effect of intraarticular morphine after arthroscopic knee surgery. N Engl J Med 325: 1123–1 126

    Google Scholar 

  761. Raja SN, Dickstein RE, Johnson CA (1992) Comparison of postoperative analgesic effects of intraarticular bupiva-caine and morphine following arthroscopic knee surgery. Anesthesiology 77 /6: 1143–1147

    Article  PubMed  CAS  Google Scholar 

  762. Joshi GP, McCarroll SM, O’Brien TM, Lanane P (1993) Intraarticular analgesia following knee arthroscopy. Anest Analg 76: 333–336

    CAS  Google Scholar 

  763. Bailey PL, Streisand JB, East KA et al. (1990) Differences in magnitude and duration of opioid induced respiratory depression and analgesia with fentanyl and sufentanil. Anesth Analg 70: 8–15

    Article  PubMed  CAS  Google Scholar 

  764. Hughes MA, Glass PSA, Jacobs JR (1992) Context-sensitive half-time in multicompartment pharmacokinetic models for intravenous anesthetic drugs. Anesthesiology 76: 334–341

    Article  PubMed  CAS  Google Scholar 

  765. Herz A (1981) Opiat-Partialantagonisten. In: Kubicki S, Neuhaus GA (Hrsg) Pentazocin im Spiegel der Erfahrungen. Springer, Berlin Heidelberg New York Tokio, pp 19–21

    Google Scholar 

  766. Freye E, Latasch L, Portoghese PS (1992) The delta receptor is envolved in sufentanil-induced respiratory depression. Eur J Anaesthesiol 9: 457–462

    PubMed  CAS  Google Scholar 

  767. Eisenach JC (1992) Epidural and spinal narcotics. In: Barash PG (ed) ASA Refresher courses in anesthesiology, vol 20. Lippincott, Philadelphia, pp 1–4

    Google Scholar 

  768. Twycross RG (1988) Opioid analgesics in cancer pain: current practice and controversies. Cancer Sur 7: 29–53

    CAS  Google Scholar 

  769. Cookson RF (1983) Carfentanil and Lofentanil. Clin Anaesthesiol 1: 156–158

    CAS  Google Scholar 

  770. Janssen PAJ (1984) The development of new synthetic narcotics. In: Estafanous FG (ed) Opioids in anesthesia. Butterworth, Boston, pp 37–44

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

Copyright information

© 2004 Springer-Verlag Berlin Heidelberg

About this chapter

Cite this chapter

Freye, E. (2004). Opioide im Rahmen der Allgemeinanästhesie. In: Opioide in der Medizin. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-662-09096-1_19

Download citation

  • DOI: https://doi.org/10.1007/978-3-662-09096-1_19

  • Publisher Name: Springer, Berlin, Heidelberg

  • Print ISBN: 978-3-662-09097-8

  • Online ISBN: 978-3-662-09096-1

  • eBook Packages: Springer Book Archive

Publish with us

Policies and ethics