Skip to main content

Der opiatabhängige Patient

  • Chapter
Book cover Opioide in der Medizin

Zusammenfassung

Während die Zahl der Alkoholabhängigen in den alten und neuen Bundesländern auf etwa 5 Mio. der Gesamtbevölkerung geschätzt wird, beträgt die Zahl der Heroinabhängigen etwa 120.000. Wegen der Dunkelziffer kann die Zahl jedoch auch gut doppelt so hoch sein [1]. Letztlich muss mit einer Zunahme der von dieser Stoffklasse Abhängigen gerechnet werden. Dies ist aus der Tatsache abzuleiten, dass, trotz Abnahme der Zahl der Drogentoten in den letzten Jahren, die Zahl an Erstkonsumenten harter Drogen rapide zugenommen hat (□ Abb. 33–1). Hierfür sind neben dem akuten Preisverfall und einer größeren Reinheit des angebotenen Stoffs auch neuere Analoga schon bekannter Opioide, sog. »Designerdrogen« (Modifikationen bekannter Opioide) mitverantwortlich, die sich durch eine höhere Wirkpotenz auszeichnen [2, 3].

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.

Literaturverzeichnis

  1. Wahl C (1987) »Was Ihr wollt« - zur statistischen Einschätzung von Konsum, Mißbrauch und Abhängigkeit bei legalen und illegalen Drogen. In: Ness B, Wahkl C, Ziegler H (Hrsg) Jahrbuch zur Frage der Suchtgefahren. Neuland, Hamburg, pp 89

    Google Scholar 

  2. Henderson G (1988) Designer drugs: The new synthetic drugs of abuse. Clinical update in toxicology. Du Pont Pharmaceuticals, Delaware/WI

    Google Scholar 

  3. Sahihi A (1989) Designer-Drogen. Die neue Gefahr. Beltz, Weinheim Basel

    Google Scholar 

  4. Scheerer S (1989) Die Heroinszene. In: Scheerer S, Vogt I (Hrsg) Drogen und Drogenpolitik. Campus, Frankfurt am Main, pp 285–299

    Google Scholar 

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

    Google Scholar 

  6. 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 

  7. 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 

  8. 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 

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

    Article  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  11. DiChiara G, Imperato A (1988) Opposite effects of mu and kappa 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 

  12. Thomas DN, Fontana DJ, Post RM, Weiß SRB, Pert A (1993) The involvement of the mesocorticolimbic dopamine system in the conditioned effects of cocaine. In: Harris L (ed) Problems of drug dependence. Proceedings of the 54th Annual Scientific Meeting 1992. NIDA, Rockville. Vol 260, PP 132

    Google Scholar 

  13. Wang Y, Bilsky E, Porreca F, Sadee W (1994) Constitutive p opioid receptor activation as a regulatory mechanism underlying narcotic tolerance and dependence. Life Sci 54: 339–350

    Google Scholar 

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

    Google Scholar 

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

    Google Scholar 

  16. 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 

  17. Höllt V, Przewlocki R, Herz A (1978) ß-Endorphin-like immunoreactivity in plasma, pituitaries and hypothalamus of rats following treatment with opiates. Life Sci 23: 1057

    Article  PubMed  Google Scholar 

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

    Article  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  20. Cookson RF (1983) Carfentanil and lofentanil. Clin Anaesthesiol 1: 156–158

    CAS  Google Scholar 

  21. Langston JW, Ballard P, Tetrud JW, I (1983) I. Chronic parkinsonism in humans due to a product of meperidine-analog synthesis. Science 219: 979–980

    Google Scholar 

  22. Langston JW, Irwin J, E. B. L, Forno LSC (1984) Pargyline prevents MPTP-induced parkinsonism in primates. Science 225: 1480–1482

    Article  PubMed  CAS  Google Scholar 

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

    Google Scholar 

  24. Freye E (1992) Medikamenteninteraktion bei der Metha-dontherapie. Z Ärztl Fortbild 86: 731–737

    CAS  Google Scholar 

  25. Schuster S, Weilemann LS, Meyer J (1989) Drogennotfälle. Notarzt 5: 23–28

    Google Scholar 

  26. Kreek MJ (1973) Medical safety and side effects of methadone in tolerant individuals. JAMA 233: 665–668

    Article  Google Scholar 

  27. Kreek MJ, Khuri E, Flomenberg N, Albeck H, Ochshorn M (1989) Immune status of unselected methadone maintained former heroin addicts. In: Quiron R, Jhamandas K, Gianoulakis C (eds) The International Narcotics Research Conference (INRC) ‘89. Liss, New York. Progress in clinical and biological research, vol 328, pp 445–448

    Google Scholar 

  28. Latasch L, Schneider M, Steinau U (1986) Morphologische Befunde an Skelettmuskelbiopsien bei Heroinabhängigen. Klinikarzt 15: 252

    Google Scholar 

  29. McCammon RL (1986) Anesthesia for the chemically dependent patient. In: Review Course Lectures 1986. International Anesthesia Research Society (IARS), Cleveland, pp 47–55

    Google Scholar 

  30. Martin WR, Jasinski DR, Haertzen CA et al. (1973) Methadone–A réévaluation. Arch Gen Psychiatry 28: 286–295

    Article  PubMed  CAS  Google Scholar 

  31. Freye E, Schenk GK (1990) Methadon als Ersatztherapie beim Opiatabhängigen? Klinikarzt 2: 57–61

    Google Scholar 

  32. Mark LC (1966) Hypotension during anesthesia in narcotic addicts. N Y State J Med 66: 2685–2695

    Google Scholar 

  33. 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. Handbook of experimental pharmacology, vol 45/I. Springer, Berlin Heidelberg New York

    Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  35. Winkler KR (1980) Zur Strafbarkeit des Arztes gemäß § 11 Abs. 1 Nr. 9 Betäubungsmittelgesetz. Suchtgefahren 26: 28–37

    Google Scholar 

  36. Rösinger C, Gastpar M (1991) Methadonsubstitution in der Behandlung schwerkranker Opiatabhängiger. Dtsch Ärztebl 44: C-2079-C-2088

    Google Scholar 

  37. Franke M (1985) Übergeordnete Gesichtspunkte zur Bewertung von Methadonprogrammen. Suchtgefahren 31: 95–100

    Google Scholar 

  38. Hartmann H, Uchtenhagen A, Fuchs W, Pasi A (1984) Hausärztliche Methadonbehandlung bei Heroinabhängigen. Schweiz Rundschau Med 73: 299–304

    CAS  Google Scholar 

  39. Olsen GD, Wendel HA, Livermore JD, Leger RM, Lynn RK, Gerber N (1976) Clinical effects and pharmacokinetics of racemic methadone and its optical isomers. Clin Pharmacol Ther 21: 147–157

    Google Scholar 

  40. Keup W (1978) Methadon-Erhaltungsprogramme. Dtsch Ärztebl 18: 1179–1182

    Google Scholar 

  41. Beaver WT, Wallentein SL, Houde RW, Roger A (1976) A clinical comparison of the analgesic effects of methadone and morphine administered intramuscularly and of orally administered methadone. Clin Pharmacol Ther 8: 415–426

    Google Scholar 

  42. Olsen GD, Livermore JD, Wendel HA et al. (1976) Methadone induced respiratory depression in female methadone maintenance subjects. Clin Res 24: 388 A

    Google Scholar 

  43. Brune K, Dietzel K, Möller N (1986) Pharmakologie des Schmerzes-Abhängigkeit, Sucht. In: Wörz R (Hrsg) Pharmakologie bei Schmerz. Edition Medizin VCH, Weinheim, pp 120–134

    Google Scholar 

  44. Nielsson MI (1982) Clinical pharmacokinetics of methadone. University Press, Upp sala/Sweden (Thesis )

    Google Scholar 

  45. lnturrisi CE (1973) Disposition of methadone in man after single oral dose. Clin Pharmacol Ther 13: 923–930

    Google Scholar 

  46. Nielsson MJ, Widerloev E, Meresaar U, nggard E (1982) Effect of urinary ph on the disposition of methadone in man. Eur J Clin Pharmacol 22: 331–342

    Google Scholar 

  47. Romach MK, Piafsky KM, Abel JG, Khouw V, Seilers EM (1981) Methadone binding to orosomucoid (alphal-acid-glucoprotein). Determinant of free fraction in plasma. Clin Pharmacol Ther 21: 307–321

    Google Scholar 

  48. 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 

  49. Kreek MJ, Gutjahr CL, Garfield JN, Bowen DV, Field FH (1976) Drug interactions with methadone. Ann N Y Acad Sei 281: 350–370

    Article  CAS  Google Scholar 

  50. Novick DM, Kreek MJ, Fanizza AH, Yancovitz SR, Gelb AM, Stenger RM (1981) Methadone disposition in patients with chronic liver disease. Clin Pharmacol Ther 30: 353–362

    Article  PubMed  CAS  Google Scholar 

  51. Blinick G, Wallach RC, Jerez E (1969) Pregnancy in narcotic addicts treated by medical withdrawal. The methadone detoxification program. Am J Obst Gynecol 105: 997–1003

    Google Scholar 

  52. Rothstein P, Gould JB (1974) Born with a habit–Infants of drug addicted mothers. Pediatr Clin North Am 21: 307–321

    PubMed  CAS  Google Scholar 

  53. Geber WF, Schramm LC (1975) Congenital malformations of the central nervous system produced by narcotic analgesics in the hamster. Am J Obstet Gynecol 12: 705–713

    Google Scholar 

  54. Gourlay GK, Wilson PR, Glynn GJ (1982) Pharmacodynamics and pharmacokinetics of methadone during the operative period. Anesthesiology 57: 458–467

    Article  PubMed  CAS  Google Scholar 

  55. 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 

  56. De Castro J, Viars P (1968) Utilisation pratique des analgésiques centraux en anesthésie et réanimation. Ars Med 23: 74–74

    Google Scholar 

  57. Liebson I, Bigelow G, Flamer R (1971) Alcoholism among methadone patients; a specific treatment method. Am J Psychiat 130: 483–485

    Google Scholar 

  58. Stimmel B, Hanburg R, Stuniano V, Korts D, Jackson G, Cohen M (1982) Alcoholism as a risk factor in methadone maintenance. Am J Med 73: 631–636

    Article  PubMed  CAS  Google Scholar 

  59. Chabalko J, La Rosa J, du Pont RL (1973) Death of methadone users in the district of Columbia. Int J Addict 8: 897–908

    PubMed  CAS  Google Scholar 

  60. Schulte RM (1986) Medikamentenabhängigkeit und Poly-toxikomanie. Ergebnisse einer empirischen Untersuchung bei Drogenabhängigen und Alkoholikern. Dtsch Ärztebl 83: 3451

    Google Scholar 

  61. 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 

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

    Google Scholar 

  63. 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 (eds) Les analgésiques et la douleur. Influences pharma-cologiques diverses exercées sur morphiniques. Masson, Paris, pp 185–194

    Google Scholar 

  64. Freye E (1991) Opioide in der Medizin. Wirkung und Einsatzgebiete zentraler Anal getika, 2. Aufl. Springer, Berlin Heidelberg New York Tokio

    Google Scholar 

  65. Bewley TH, Ghodse AH (1984) Opiate analgesics and narcotic antagonists. In: Dukes MNG (ed) Meyler’s side effects of drugs, 10th Edn. Elsevier, Amsterdam, pp 120–134

    Google Scholar 

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

    Article  Google Scholar 

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

    Google Scholar 

  68. Keup W (1983) Clonidin–seine Möglichkeiten in der Pharmakotherapie der Heroin abhängigkeit. Dtsch Ärztebl 80 /3: 25–32

    Google Scholar 

  69. Gold MS, Pottash AC, Sweeney DR, Kleber HD (1980) Opiate withdrawal using Clonidin. A safe, effective, and rapid nonopiate treatment. JAMA 243: 343–348

    Google Scholar 

  70. Groß A, Soyka M (1999) Buprenorphin–ein neuer Ansatz in der Pharmakotherapie opiatabhängiger Patienten. Suchtmed 1: 5–14

    Google Scholar 

  71. Kosten TR, Morgan C, Kleber HD (1992) Phase II clinical trials of buprenorphine: Detoxification and induction into naltrexone. NIDA Res Monogr 121: 101–119

    Google Scholar 

  72. Kosten TR, Schottenfeld R, Ziedonis D, Falcioni J (1993) Buprenorphine versus methadone maintenance for opioid dependence. J Nev Ment Dis 181 /6: 358–364

    Article  CAS  Google Scholar 

  73. Guthrie SK (1990) Pharmacologic interventions for the treatment of opioid dependence and withdrawal. Ann Pharmacother 24/7–8: 721–734

    Google Scholar 

  74. Bickel WK, Stitzler ML, Bigelow GE, Liebson IA, Jasinski DR, Johnson RE (1988) Buprenorphine: dose-related blockade of opioid challenge effects in opioid dependent humans. J Pharmacol Exp Ther 247 /1: 47–53

    PubMed  CAS  Google Scholar 

  75. Woods JH, France CP, Winger GD (1992) Behavioral pharmacology of buprenorphine issues relevant to its potential in treating drug abuse. NIDA Res Monogr 121: 12–27

    PubMed  CAS  Google Scholar 

  76. Bickel WK, Amass L (1995) Buprenorphine treatment of opioid dependence. A review. Exp Clin Psychopharmacol 3: 477–489

    Google Scholar 

  77. Fudala PJ, Johnson RE, Bunker E (1989) Abrupt withdrawal of buprenorphine following chronic administration. Clin Pharmacol Ther 186/2: IVB-1

    Google Scholar 

  78. Mello NK, Mendelson JH (1980) Buprenorphine suppresses heroin use by heroin addicts. Science 207: 657–659

    Article  PubMed  CAS  Google Scholar 

  79. Mendelson JH, Mello NK, Teoh SK, Kuehnle J, Sintavana-rong P (1991) Buprenorphine treatment for concurrent heroin and cocaine dependence: phase I study. NIDA Research Monograph Series 105, pp 196–202

    Google Scholar 

  80. Mendelson JH, Mello NK (1992) Human laboratory studies of buprenorphine. NIDA Res Monogr 121: 38–60

    PubMed  CAS  Google Scholar 

  81. Auriacombe M, Franques P, Bertorelle V, Tignol J (1999) Use of buprenorphine for substitution treatment–a French experience in Bordeaux and Bayonne. Res Clin For 19: 47–50

    Google Scholar 

  82. Kosten TR, Morgan C, Kleber HD (1991) Treatment of heroin addicts using buprenorphine. Am J Drug Alcohol Abuse 17 /2: 119–128

    Article  PubMed  CAS  Google Scholar 

  83. Resnick RB, Galanter M, Pycha C, Cohen A, Grandison P, Flood N (1992) Buprenorphine: an alternative to methadone for heroin dependence treatment. Psychopharmacol Bull 28 /1: 109–113

    PubMed  CAS  Google Scholar 

  84. Strain EC, Preston KL, Liebson IA, Bigelow GE (1992) Acute effects of buprenorphine, hydromorphone and naloxone in methadone-maintained volunteers. J Pharmacol Exp Ther 261 /3: 985–993

    PubMed  CAS  Google Scholar 

  85. Blaine JD (1992) Buprenorphine: an alternative treatment for opioid dependence. NIDA Res Monogr 121: 1–4

    Google Scholar 

  86. Marion IJ (1955) LAAM in the treatment of opiate addiction. Treatment improvement protocol (TIP), vol 22, US Department of Health and Human Services Washington DC

    Google Scholar 

  87. Ling W, Rawson RA, Compton MA (1994) Substitution pharmacotherapies of opioid addiction from methadone to LAAM and buprenorphine. J Psycho Drugs 26: 119–128

    Article  CAS  Google Scholar 

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

    Google Scholar 

  89. Eberlein HJ (1982) Narkose nach Heroinentzug. Dtsch Med Wochenschr 107: 1819

    Google Scholar 

  90. Freye E (1992) Die perioperative Behandlung des Arzneimittelabhängigen. Anästh Intensiv med 3: 59–67

    Google Scholar 

  91. Giuffrida JG, Bizzari DV, Saure AC, Sharoff RL (1970) Anesthetic management of drug abusers. Anesth Analg 49: 272–278

    Article  PubMed  CAS  Google Scholar 

  92. Eismann B, Lam RC, Rush B (1964) Surgery in the narcotic addict. Ann Surg 159: 748–759

    Article  Google Scholar 

  93. Rapp SE, Ready LB, Nessly ML (1995) Acute pain management in patients with prior opioid consumption: a case.- controlled retrospective review. Pain 61: 195–201

    Article  PubMed  CAS  Google Scholar 

  94. Hughes JR, Bickel WK, Hggins ST (1991) Buprenorphine for pain relief in a patient with drug abuse. Am J Drug Alcohol Abuse 17 /4: 451–455

    Article  PubMed  CAS  Google Scholar 

  95. Gelfand ML, Hammer H, Hevizy J (1967) Asymptomatic pulmonary atelactasis in drug addicts. Dis Chest 53: 782–786

    Article  Google Scholar 

  96. Bschor F (1987) Zur Revision des Abstinenzparadisgmas in der Behandlung Sucht kranker. Ambulante, medikamen-tengestützte Therapie, ein möglicher ärztlicher Beitrag zur AIDS-Prävention. Dtsch Med Wochenschr 112: 907

    Article  PubMed  CAS  Google Scholar 

  97. Singarajah C, Lavies NG (1992) An overdose of ecstasy. Anaesthesia 47: 686–687

    Article  PubMed  CAS  Google Scholar 

  98. Henry JA, Jeffreys KJ, Dawling S (1992) Toxicity and deaths from 3,4-methylenedioxymethamphetamine (»ectasy«). Lancet 340: 384–387

    Article  PubMed  CAS  Google Scholar 

  99. Dowling GP, McDonough ET, Bost RO (1987) »Eve« and »ecstasy«: A report of five deaths with the use of MDEA and MDMA. JAMA 257: 1615–1617

    Google Scholar 

  100. Gouzoulis E, Borchardt D, Hermle L (1993) A case of toxic psychosis induced by »Eve« (3,4-methylenedioxyethyl-amphetamine). Arch Gen Psychiatry 50: 75

    Article  PubMed  CAS  Google Scholar 

  101. Manchada S, Conolly MJ (1993) Cerebral infraction in association with ecstasy. Postgrad Med J 69: 874–875

    Article  Google Scholar 

  102. Freye E (1997) Kokain, Ecstasy und verwandte Designerdrogen - Wirkungsweise, Überdosierung, therapeutische Notfallmaßnahmen. Barth, Heidelberg Leipzig

    Google Scholar 

  103. Täschner KL (1994) Drogen und Straßenverkehr. Dtsch Ärzte Z 35

    Google Scholar 

  104. Külpmann WR. Feststellung des Beigebrauchs bei der Methadon-Substitutions behandlung von i.v.-Heroinabhängigen. Dtsch Ärztebl 1994 2: C-149-C-151

    Google Scholar 

  105. Baselt RC (1982) Disposition of toxic drugs and chemicals in man, 2nd Edn. Davis Biomedical Publ, Philadelphia

    Google Scholar 

  106. Hawks RL, Chiang CN (1986) Urine testing for drugs of abuse. US Department of Health and Human Servivces (NIH), Rockville/MD Maryland, NIDA Research Monograph Series, vol 73

    Google Scholar 

  107. Blum K (1984) Handbook of abusable drugs. Gardner, New York

    Google Scholar 

  108. Ladewig D, Stohler L (1994) Das Opiatentzugssyndrom–Skalierung und medikamentöse Strategien. In: Tretter F, Busello-Spieth W, Bender WH (Hrsg) Therapie von Ent-zugssyndromen. Springer, Berlin Heidelberg New York Tokio, pp 144–168

    Google Scholar 

  109. Loimer N, Schmid RW, Presslich O, Lenz K (1989) Continuous naloxone administration suppresses opiate withdrawal symptoms in human opiate addicts during detoxification treatment. J Psychiatr Res 23: 81–86

    Article  PubMed  CAS  Google Scholar 

  110. Loimer N, Lenz K, Schmid R, Presslich O (1991) Technique for greatly shortening the transition from methadone to naltrexone maintenence of patients addicted to opiates. Am J Psychiat 148: 933–935

    PubMed  CAS  Google Scholar 

  111. Gossop M (1988) Clonidine and the treatment of the opiate withdrawal syndrome. Drug Alcohol Depend 21: 253–259

    Article  PubMed  CAS  Google Scholar 

  112. Legarda L, Gossop H (1994) A 24 hour inpatient detoxification treatment for heroin addicts: a preliminary investigation. Drug Alcohol Depend 35: 91–93

    Article  PubMed  CAS  Google Scholar 

  113. Tretter F (1996) Von der Phantasie, die Sucht auszuschlafen. MMW 138: 76–77

    Google Scholar 

  114. Tretter F, Burkhardt D, Buzello-Spieth S, Reiss J, Walcher S, Büchele W (1996) Anwendungsverfahren mit dem forcierten Opiatentzug in Narkose. MMW 138: 787–791

    Google Scholar 

  115. Reiss J (1996) Drogenentzug unter Narkose–Eine Konsequenz der Substitutionstherapie. Fortschr Med 24: 12–13

    Google Scholar 

  116. Klienbaum P, Thürauf N, Michel MC, Scherbaum N, Gaspar M, Peters J (1998) Profound increase in epinephrine in plasma and cardiovascular stimulation after p-opiate receptor blockade in opiate addicted patients during barbiturate-induced anesthesia for acute detoxyfication. Anesthesiology 88: 1154–1161

    Article  Google Scholar 

  117. Glen JB (1998) The development of »Diprifusor«: a TCI system for propofol. Anaesthesia S3: 13–21

    Article  Google Scholar 

  118. Charney DS, Heninger GR, Kleber HD (1986) The combined use of Clonidine and naltrexone a rapid, safe, and effective treatement of abrupt withdrawal from methadone. Am J Psychiatry 143 /7: 831–837

    PubMed  CAS  Google Scholar 

  119. Vining DH, Kosten TR, Kleber HD (1988) Clinical utility of rapid clonidine-naltrexone detoxification for opioid abusers. Br J Addict 63: 567–575

    Article  Google Scholar 

  120. 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 

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

    Article  PubMed  CAS  Google Scholar 

  122. 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 

  123. 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 

  124. Redmond DE, Hwang YH (1982) The primate locus coeru-leus and affects of Clonidine on opiate withdrawal. J Clin Psychiatry 43: 25–31

    PubMed  CAS  Google Scholar 

  125. Foote SL, Bloom FE, Aston-Jones G (1983) Nucleus locus ceruleus: New evidence for anatomical and physiological specificity. Physiol Rev 63: 844–895

    Google Scholar 

  126. Jarrot B, Conway EL, Maccarone C, Lewis SJ (1987) Clonidine: understanding its disposition, sites and mechanism of action. Clin Exp Pharmacol Physiol 14: 471–479

    Article  Google Scholar 

  127. Freye E (1999) Elektroenzphalograpie, spinal und morto-risch evozierte Potentiale. In: Jantzen JP, Löffler W (Hrsg) Neuroanästhesie. Thieme, Stuttgart, pp 155–191

    Google Scholar 

  128. Sigl JC, Chamoun NG (1994) An introduction to bispectral analysis for the EEG. J Clin Monit 10: 392–404

    Article  PubMed  CAS  Google Scholar 

  129. Freye E, Grabitz K, Sandmann W (1996) Was bringt die 3. Generation im Neuro monitoring für die klinische Praxis? Anästhesiol Intensivmed 3: 120–127

    Google Scholar 

  130. Blumberg H, Dayton HB (1974) Naloxone, naltrexone, and related noroxymorphones. In: Braude MC, Harris LS, Smith MJP, Villareal JE (eds) Narcotic antagonists. Raven, New York (Adv Biochem Psychopharm) vol 8, pp 33–43

    Google Scholar 

  131. 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 

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

    Article  Google Scholar 

  133. Jenkins SA (1996) Somatostatin and octreotide. Medi-media Asia, Hong Kong

    Google Scholar 

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

    Google Scholar 

  135. Takeda T, Taniyama K, Baba S, Tanaka C (1989) Putative mechanisms involved in excitatory and inhibitory effects of somatostatin on intestinal motility. Am J Physiol 257: G 532-G 538

    Google Scholar 

  136. Williams J, North RA (1978) Inhibition offirering of myenteric neurones by somatostatin. Brain Res 155: 165–168

    Article  PubMed  CAS  Google Scholar 

  137. Gagniella TS, O’Dorisio TM, Fassler JE, Mekhjian HS (1990) Treatment of endocrine and nonendocrine secretiory diarrheal states with Sandostatin®. Metabolism 39: 172–175

    Article  Google Scholar 

  138. Sehppard M, Shapairo B, Berelowitz M, Pimstone B (1979) Metabolic clearance and plasma half dissapearance time of exogenous somatostatin in man. J Clin Endocrinol Metab 48: 50–53

    Article  Google Scholar 

  139. Klepstad P, Maurset A, Moberg ER, Oye I (1990) Evidence of a role for NMDA receptors in pain perception. Eur J Pharmacol 187: 513–518

    Article  PubMed  CAS  Google Scholar 

  140. Irifune M, Shimizu M, Nomoto M, Fukuda T (1992) Keta-mine induced anesthesia involves the N-methyl-D-aspar-tate receptor-channel complex in mice. Brain Res 569: 1–9

    Article  Google Scholar 

  141. Monaghan DT, Bridges RJ, Cotman CW (1989) The excitatory amino acid receptors: their classes, pharmacology and distinct properties in the function of the central nervous system. Annu Rev Pharmacol Toxicol 29: 365–462

    Article  PubMed  CAS  Google Scholar 

  142. 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 

  143. Arner S, Meyerson BA (1988) Lack of analgesic effect of opioids on neuropathic and idiopathic forms of pain. Pain 33: 11–23

    Article  PubMed  CAS  Google Scholar 

  144. Dickenson AH. Neurophysiology of opioid poorly responsive pain. Cancer Surv 1995 21: 5–16

    Google Scholar 

  145. Brown JP, Boden P, Singh L, Gee NS (1996) Mechanimss of action of gabapentin. Rev Contemp Pharmacother 7: 203–214

    CAS  Google Scholar 

  146. Field MJ, Oles RJ, Lewis AS, McVlerary S, Hughes J, Singh L (1997) Gabapentin (neurontin) and S(f)-3-isobutylgaba represent a novel class of selective antihyperalgesic agents. Br J Pharmacol 121: 1513–1522

    Article  PubMed  CAS  Google Scholar 

  147. Mellik GA, Larry DO, Mellik B (1997) Reflex sympathetic dystrophy treated with gabapentin. Arch Phys Med Reha-bil 78: 98–105

    Article  Google Scholar 

  148. Rosner H, Rubin L, Kestenbaum A (1996) Gabapentin adjunctive therapy in neuropathic pain states. Clin J Pain 12: 56–58

    Article  PubMed  CAS  Google Scholar 

  149. Sandyk R (1992) L-tryptophan in neuropsychiatric disorders: a review. Intern J Neurosci 67: 127–144

    Article  CAS  Google Scholar 

  150. 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 

  151. Sandyk R, Fisher H (1988) Serotonin in involuntary movement disorders. Intern J Neurosci 42: 185–205

    Article  CAS  Google Scholar 

  152. Sandyk R (1986) L-tryptophan in the treatment of restless legs syndrome. Am J Psychiatry 143: 554–555

    PubMed  CAS  Google Scholar 

  153. Cucchia AT, Monnat M, Spagnoli J, Ferrero F, Bertschy G (1998) Ultra-rapid opiate detoxification using deep sedation with oral midazolam: short and long term results. Drug Alcohol Depend 52: 243–250

    Article  PubMed  CAS  Google Scholar 

  154. Yamaguchi K, Anderson JM (1992) Biocompatibility studies of naltrexone sustained release fomulations. J Cont Rel 19: 299–314

    Article  CAS  Google Scholar 

  155. Tefft JA, Roskos KV, Heller J (1992) The effect of lipase on release of naltrexone from triglycxeride-coated celllulose acetate phtalate micropheres. J Biomed Material Res 26: 713–724

    Article  CAS  Google Scholar 

  156. Holaday JW, Tortella FC (1984) Multiple opioid receptors: Possible physiological functions of m and 5 binding sites in vivo. In: Genazzini AR, Muller EE (eds) Central and peripheral endorphins: Basis and clinical aspects. Raven, New York, pp 237–250

    Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  158. Schmidhammer H, Smith CFC, Erlach D et al. (1990) Synthesis and biological evaluation of 14-Alkoxymorphinans. 3. Extensive study on cyprodime-related compounds. J Med Chem 33: 1200–1206

    Article  PubMed  CAS  Google Scholar 

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

    Google Scholar 

  160. Takemori AE, Sofuoglu M, Sultana M, Nagase H, Porto-ghese PS (1990) Pharmacology of highly selective, non-peptide delta opioid receptor antagonists. In: van Ree JM, Mulder AH, Wiegant VM, van Wimersma Greidanus TB (eds) New leads in opioid research. Excerpta Medica, Amsterdam New York (International congress series), vol 914, pp 277–278

    Google Scholar 

  161. Romer D, Buscher H, Hill RC et al. (1980) Bremazocine: A potent, long-acting opiate kappa-agonist. Life Sci 27: 971–978

    Article  PubMed  CAS  Google Scholar 

  162. Buzas B, Toth G, Cavagnero S, Hruby VJ, Borsodi A (1992) Synthesis and binding characteristics of the highly delta-specific new tritiated opioid peptide [3H]deltorphin II. Life Sci 50: PL 75-PL 77

    Google Scholar 

  163. Rossier JT, Vargo M, Minick S, Ling N, Bloom EF, Guillemin R (1977) Regional dissociation of (3-endorphin and enkephalin contents in rat brain and pituitary. Proc Natl Acad Sci USA 74: 5262–5165

    Article  Google Scholar 

  164. Frederickson RCA, Geary LE (1982) Endogenous opioid peptides: Review of physiological, pharmacologiocal and clinical aspects. Prog Neurobiol 19: 16–69

    Google Scholar 

  165. Smith TW, Hughes J, Kosterlitz HW, Sosa RP (1976) Enkephalins: Isolation, distribution and function. Opiates and endogenous opioid peptides. Elsevier, Amsterdam

    Google Scholar 

  166. Feurle GE, Helmstaetter V, Weber U (1982) Met-and Leu-enkephalin immuno-and bio-reactivity in human stomach and pancreas. Life Sci 31: 2961–2969

    Article  PubMed  CAS  Google Scholar 

  167. Kabayashi ST, Ushida T, Ohashi T et al. (1991) Met-eneke-phalin-Arg-Gly-Leu-like immunoractivity in adrenal chromaffin cells and carotid body chief cells of the dog and monkey. Biomed Res 4: 201–210

    Google Scholar 

  168. De Wald D, Lewis RV (1983) Enkephalin-containing polypeptide levels in normal tensive and SH rat adrenal glands. Peptides 4: 121–123

    Article  Google Scholar 

  169. Maysinger D, Hollt v, Seizinger BR, Mehraein P, Pasi A, Herz A (1982) Parallel distribution of immunoreactive alpha-neoendorphin and dynorphin in rat and human tissue. Neuropeptides 2: 211–225

    Article  CAS  Google Scholar 

  170. Czlonkowski A, Costa T, Przewlocki A, Pasi A, Herz A (1983) Opioid receptor binding in human spinal cord. Brain Res 267: 392–396

    Article  PubMed  CAS  Google Scholar 

  171. Watson SJ, Akil H, Ghazarsson VE, Goldstein A (1981) Dynorphin immunocytochemical localization in brain and peripheral nervous system: prelimary studies. Proc Natl Acad Sci USA 78: 1260–1263

    Article  PubMed  CAS  Google Scholar 

  172. Watson SJ, Khachaturian H, Akil H, Coy DH, Goldstein A (1982) Comparison of the distribution of dynorphin system and enkephalin system in brain. Science 218: 1134–1136

    Article  PubMed  CAS  Google Scholar 

  173. Feuerstein G, Molineaux CJ, Rosensberg JG, Faden AD, Cox BM (1983) Dynorphin and Leu-Enkephalin in brain nuclei and pituitary of WKY and SHR rats. Peptides 4: 225–229

    Article  PubMed  CAS  Google Scholar 

  174. Höckfelt T, Ljungdahl A, Terenius I, Eide R, Nilsson G (1977) Immunohistochemical analysis of peptide pathways possibly related to pain and analgesia: enkephalin and substance P. Proc Natl Acad Sci USA 74: 3081

    Article  Google Scholar 

  175. Bruni JF, van Vugt D, Marschall S, Meilers J (1977) Effect of naloxone, morphine, methionine-enkephalin on serum prolactin, lutenizing hormone, follicle stimulating hormone, thyroid stimulating hormone and growth hormone. Life Sci 21: 461

    Article  PubMed  CAS  Google Scholar 

  176. Dupont A, Cusan L, Garon L, Labrie F, Li H (1977) ß-Endor-phin. Stimulation of growth hormone release in vivo. Proc Natl Acad Sci USA 74: 358

    Google Scholar 

  177. Shaar CJ, Frederickson RCA, Dinninger NB, Jackson L (1977) Enkephalin analogues and naloxone modulate the release of growth hormone and prolactin. Evidence for regulation by an endgenous opioid peptide in brain. Life Sci 21: 853

    Google Scholar 

  178. Copolov DL, Helme RD (1983) Enkephalins and Endorphins Clinical, pharmacological and therapeutic implications. Drugs 26: 503–519

    Article  PubMed  CAS  Google Scholar 

  179. Akil H, Richardson DE, Barchas JD, Li CH (1978) Appearance of ß-enorphin-like immunoreactivity in human ventricular cerebropinal fluid upon electrical stimulation. Proc Natl Acad Sci USA 75: 5170–5172

    Article  PubMed  CAS  Google Scholar 

  180. Hosobuchi Y, Adams JE, Lichnitz R (1977) Pain relief by electrical stimulation of the central gray matter in humans and its reversal by naloxone. Science 197: 183

    Article  PubMed  CAS  Google Scholar 

  181. yama T, Jin T, Yamaha R (1980) Profound analgesic effects of ß-endorphin in man. Lancet I: 122–124

    Google Scholar 

  182. Genazzini AR, Nappi G, Facchinetti F et al. (1984) Progressive impairment of CSF ß- EP levels in migraine sufferers. Pain 18: 127–133

    Article  Google Scholar 

  183. Sicuteri F, Anselmi B, Curradi C, Michelacci S, Sassi A (1978) Morphine-like factors in CSF headache suffers. In: Costa E, Trabucchi M (eds) Adv Biochem Psychopharm, vol 18. Raven, New York, pp 363–366

    Google Scholar 

  184. Sicuteri F (1979) Headache as the most common disease of the antionociveptive system: analogies with morphine abstinence. In: Bonica JJ (ed) Advances in pain research and therapy, vol 3. Raven, New York, pp 359–365

    Google Scholar 

  185. Sicuteri F, Fanciullacci M, Michelacci S (1978) Decentratil-sation supersensitivity in headache and central panalge-sia. In: Friedman AP, Granger ME, Critchley M (eds) Clinical studies on headache, vol 6. Karger, Basel, pp 19–33

    Google Scholar 

  186. Seymour D (1993) Double-blind, placebo-controlled efficacy study of Stadol(HNS (transnasal butorphanol) and intramuscular methadone in the acute treatment of migraine. 7th World Congress of Pain. ISAP Publications, Paris

    Google Scholar 

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

    Google Scholar 

  188. Symons IE, Emons PC, Farman JV (1982) Endogenous opioid poisoning? Br Med J 284: 469–470

    Article  CAS  Google Scholar 

  189. Cohen M, Pickar D, Dubois M (1982) Role of the endogenous opioid system in the human stress response. Psychiatry Clin North Am 6: 457

    Google Scholar 

  190. Miralles FS, Olaso MJ, Fuentes T, Lopez T, Laorden ML, Puig MM (1983) Presurgical stress and plasma endorphin levels. Anesthesiology 59: 366–367

    Article  PubMed  CAS  Google Scholar 

  191. Laorden ML, Miralles F, Fuentes T, Lopez F, Cantera M (1984) Effects of stresstherapy on plasma beta-endorphin-like immunorectivity. Meth and Find Expl Clin Pharmacol 6: 671–674

    CAS  Google Scholar 

  192. Hynynen M, Lethinen A-M, Salmenperä M, Fyhrquist F, Takkunen O, Heinonmen J (1986) Continuous infusion of fentanyl or alfentanil for coronary artery surgery-Effects on plasma Cortisol concentration, ß-endorphin immuno-reactivity and arginine vasopressin. Br J Anaesth 58: 1260–1266

    Article  PubMed  CAS  Google Scholar 

  193. 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 

  194. Bloom F, Segal D, Ling N, Guillemin R (1976) Endorphins: profound behavioral effects in rats suggests new etiological factors in mental illness. Science 194: 630–632

    Article  PubMed  CAS  Google Scholar 

  195. Günne LM, Lindström L, Terenius L (1977) Naloxone-induced reversal of schizophrenic halluzinations. J Neural Transm 40: 15

    Article  Google Scholar 

  196. Schenk GK, Enderes P, Engelmeier MP et al. (1978) Application of morphine antagonist naloxone in psychic disorders. Arzneimittelforsch/Drug Res 28: 1274–1277

    CAS  Google Scholar 

  197. Kline NS, Li CH, Lehmann E, Lajtha A, Laski E, Copper T (1977) ß-Endorphin induced changes in schizophrenic and depressed patients. Arch Gen Psychiatry 34: 1111

    Article  PubMed  CAS  Google Scholar 

  198. Van Ree JM, Otte AP (1980) Effects of ( Des-Tyr)- y-endorphin and ß-endorphin as compared to haloperidol and amphetamine on nucleus accumbens self-stimulation. Neuropharmacology 19: 429–434

    Google Scholar 

  199. Van Ree JM (1986) Role of pituitary and related neuropeptides in alcoholism and pharmacodependence. Prog Neu-ropsychopharmacol Biol Psychiatr 10: 219–228

    Article  Google Scholar 

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

    Google Scholar 

  201. Kennedy SH, Goldbloom DS (1991) Current perspectives on drug therapies for anorexia nervosa and bulemia nervosa. Drugs 41 /3: 367–377

    Article  PubMed  CAS  Google Scholar 

  202. Spiegel TA, Stunkard AJ, Shrager WE, O’Brien CP, Morrison MF, Stellar E (1987) Effect of naltrexone on food intake, hunger, and satiety in obese men. Physiol Behav 40 /2: 135–141

    Article  PubMed  CAS  Google Scholar 

  203. de Zwaan M, Mitchell JE (1992) Opiate antagonists and eating behavior: a review. J Clin Pharmacol 32 /12: 1060–1072

    PubMed  Google Scholar 

  204. Shaw WN, Mitch CH, Leander JD, Mendelsohn LG, Zimmerman DM (1991) The effect of the opioid antagonist LY 255–582 on body weight of the obese Zucker rat. Int J Obes Relat Metab Disord 15 /6: 387–395

    CAS  Google Scholar 

  205. Sandyk R (1988) Naltrexone suppresses abormal sexual behaviour in tourette’s syndrome. Int J Neurosci 43/1–2: 107–110

    Google Scholar 

  206. Sandman CA (1988) (3-Endorphin dysregulation in autistic and self-injurious behavior. A neurodevelopmental hypothesis. Synapse 2/3: 93–199

    Google Scholar 

  207. Campbell M, Anderson LT, Small AM, Locascio JJ, Lynch NS, Choroco MC (1990) Naltrexon in autistic children: a double-blind and placebo-controlled study. Psycho-pharmacol Bull 26 /1: 130–135

    CAS  Google Scholar 

  208. Tseng LF, Loh HH, Li CH (1976) (3-Endorphin: Cross tolerance and cross physical dependance on morphine. Proc Natl Acad Sci USA 73: 4187

    Google Scholar 

  209. Wei E, Loh HH (1976) Physical dependance on opiate-like peptides. Science 193: 1262

    Article  PubMed  CAS  Google Scholar 

  210. Belluzzi JD, Stein L (1977) Enkephalin may mediate euphoria and drive-reduction reward. Nature 266: 556

    Article  PubMed  CAS  Google Scholar 

  211. Dingledine R, Valentino RJ, Bostock E, King ME, Chang KJ (1983) Down-regulation of the 5- but not (j-opioid receptors in the hippocampal slice aassiociated with loss of physiological response. Life Sci 33: 333–336

    Article  PubMed  CAS  Google Scholar 

  212. 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 

  213. Dole VP, Nyswander M (1967) Heroin addiction - a metabolic disease. Arch Intern Med 120: 19

    Article  PubMed  CAS  Google Scholar 

  214. Abdelhamid EE, Sultana PM, Portoghese PS, Takemori AE (1991) Selective blockade of the delta opioid receptor prevents the development of morphine tolerance and dependence in mice. J Pharmacol Exp Ther 258: 299–303

    PubMed  CAS  Google Scholar 

  215. Lee PHK, McNutt R, Chang K-J (1992) A nonpeptide delta-opioid receptor antagonist BW373U96, supppresses nalo-xone-precipitated morphine abstinence. Proceedings International Narcotic Research Conference (INRC), Key-stone/CO, USA

    Google Scholar 

  216. D’Amato RJ, Holaday JW (1984) Multiple opiate receptors in endotoxic shock: Evidence for 5 involvement and p-5 interactions in vivo. Proc Natl Acad Sci USA 81: 2898–2901

    Article  PubMed  Google Scholar 

  217. 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 

  218. Hamilton AJ, Carr DB, La Rovere JM, Black PM (1986) Endotoxic shock elicits greater endorphin secretion than hemorrhage. Circ Shock 19: 47–54

    PubMed  CAS  Google Scholar 

  219. Malcolm DS, Zaloga GP, Willey SC, Amir S, Holaday JW (1988) Naloxone potentiates epinephrine’s pressor action in endotoxemic shock. Circ Shock 25: 259–265

    PubMed  CAS  Google Scholar 

  220. Holaday JW, D’Amato RJ, Ruvio BA, Feuertein G, Faden Al (1983) Adrenalectomy blocks pressure responses to naloxone in endotoxic shock. Circ Shock 11: 201–210

    PubMed  CAS  Google Scholar 

  221. Malcolm DS, Zaloga GP, Willey SC, Amir S, Holaday JW (1988) Naloxone potentiates epinephrine’s pressure actions in endotoxemic rats. Circ Shock 25: 259–265

    PubMed  CAS  Google Scholar 

  222. Xu T, Wang T, Han J (1992) Involvement of opioid receptors in nucleus tractus solitarii in modulating endotoxic hypotension in rats. Neurosci Lett 146 /1: 72–74

    Article  PubMed  CAS  Google Scholar 

  223. Curtis MT, Lefer AM (1982) Beneficial action of a new opiate antagonist (Win 44,441–3) in hemorrhagic shock. Eur J Pharmacol 78: 307–313

    Article  PubMed  CAS  Google Scholar 

  224. Feuerstein G, Faden Al, Krumins SA (1984) Alteration in opiate receptors binding after hemorrhagic shock. Eur J Pharmacol 100: 245–246

    Article  PubMed  CAS  Google Scholar 

  225. Paciorek PM, Todd MH (1982) Comparison of the cardiovascular effects of meptazinol and naloxone following anaphylactic shock in anaesthetized rats. Br J Pharmacol 76: 245 P

    Article  Google Scholar 

  226. Amir S (1983) Antianaphylactic effect of naloxone in mice is mediated by increased central sympathetic outflow to sympathetic nerve endings and adrenal medulla. Brain Res 274: 180–183

    Article  PubMed  CAS  Google Scholar 

  227. Holaday JW, Faden Al (1980) Naloxone acts at central opiate receptors to reverse hypotension, hypothermia, and hypoventilation in spinal shock. Brain Res 75: 295–300

    Article  Google Scholar 

  228. Faden Al, Jacobs TP, Zivin JA. Comparison of naloxone and a delta-selective antagonist in experimental spinal »stroke«. Life Sci 1983 33: 707–710

    Article  PubMed  CAS  Google Scholar 

  229. Faden Al (1983) Opiate antagonist in the treatment of stroke. Current concepts in cerebrovascular disease. Stroke 18: 27–31

    Google Scholar 

  230. Groeger JS (1986) Opioid antagonists in circulatory shock. Crit Care Med 14: 170–171

    Article  PubMed  CAS  Google Scholar 

  231. Wybran J (1985) Enkephalins and endorphins as modifiers of the immune system: present and future. Fed Proc 44: 92–94

    PubMed  CAS  Google Scholar 

  232. Mathews PM, Froelich CJ, Sibbitt JWL, Brankhurst AD (1983) Enhancement of natural cytotoxicity by (3-endor-phin. J Immunol 130: 1658–1662

    PubMed  CAS  Google Scholar 

  233. Gilman SC, Schwartz JM, Milner RJ, Bloom FE, Feldman JD (1982) (3-Endorphin enhances lymphozyte proliferative responses. Proc Natl Acad Sci USA 79: 4226–4230

    Google Scholar 

  234. Bryant HU, Bernton EW, Holaday JW (1988) Immunosuppressive effects of chronic morphine treatment in mice. Life Sci 41: 1731–1738

    Article  Google Scholar 

  235. Yeager MY, Yu CT, Campbell AS, Moschella M, Guyre PM (1992) Effect of morphin and (3-endorphin on human Fc receptor-dependant and natural killer cell functions. Clin Immun Immunpath 62 /3: 336–343

    Article  CAS  Google Scholar 

  236. Fiatarone MA, Morley JE, Bloom EE, Benton D, Makinodan T, Salomon GF (1988) Endogenous opioids and the exercise -induced augmentation of natural killer cell activity. Lab Clin Med 112: 552–554

    Google Scholar 

  237. Palm S, Lehzen S, Mignat C, Steinmann J, Leimenstoll G, Maier G (1998) Does prolonged oral treatment with sustained-release morphine tablets influence immune function? Anesth Analg 86: 166–172

    PubMed  CAS  Google Scholar 

  238. Carr DJJ (1991) Minireview: The role of endogenous opioids and their receptors in the immune system. Soc Expt Biol Med 37: 710–720

    Google Scholar 

  239. Hamel U, Kielwein G, Teschemacher H (1985) p-Casomor-phin immunoreactive material in cows milk incubated with various bacterial species. J Dairy Res 52: 139–148

    Google Scholar 

  240. Teschemacher H, Brantl V, Henschen A, Lottspeich F (1990) p-Casomorphin-(3-casein fragments with opioid activity: Detection and structure. In: Nyberg F, Brantl V (eds) (3-Casomorphin and related peptides. Fyris-Trych AB, Uppsala, pp 9–14

    Google Scholar 

  241. Volterra A, Restani P, Brunello N, Galli CL, Racagni G (1986) Interaction of ß-casomorphins with multiple opioid receptors: In vitro and in vivo studies in the newborn rat brain. Dev Brain Res 30: 25–30

    Google Scholar 

  242. Ramabadran K (1988) Sudden infant death syndrome and opioid peptides. Am J Dis Child 142: 12–13

    PubMed  CAS  Google Scholar 

  243. Schusdziarra V, Schick A, de la Fuente A et al. (1983) Effect of ß-casomorphins and analogs on insulin release in dogs. Endocrinology 112: 885–889

    Article  PubMed  CAS  Google Scholar 

  244. Lindström LH, Lyrenäs S, Nyberg F, Terenius L (1990) ß-Casomorphins in postpartum psychosis. In: Nyberg F, Brantl V (eds) ß-Casomorphins and related peptides. Fyris- Tryck AB, Uppsala, pp 157–162

    Google Scholar 

  245. Kreis GJ (1986) Physiological role of somatostatin in the digestive tract: gastric secretion, intestinal absorption, and motility. Scand J Gastroenterol 21: 47–53

    Google Scholar 

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

    Article  Google Scholar 

  247. Teschemacher H (1987) Opioidrezeptoren und Opioid-peptide. Pharm Z 10: 556–567

    Google Scholar 

  248. Siegfried J (1988) Electrostimulation and neurosurgical measures in cancer pain. Springer, Berlin Heidelberg New York Tokio (Recent Results in Cancer Research), vol 108, pp 28–32

    CAS  Google Scholar 

  249. Meynadier J, Dalmas S, Lecomte JM, Gros CI, Schwartz JC (1989) Potent analgesic effect of inhibitors of enkephalin metabolism administered intrathecal to cancer patients. Pain Clin 4 /2: 201–205

    Google Scholar 

  250. Oye B (1985) Medikamentöse Schmerztherapie. Wiss. Buchverlag Dr. Peter Nietsch, Weiler/Bingen

    Google Scholar 

  251. Teschemacher H, Schweigerer L (1985) Opioid preptides: do they have immunological significance? Trends Pharmacol Sci 6: 368–370

    Article  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  253. 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 

  254. Lewis JW, Walter D (1992) Buprenorphine–background to its development as a treatment for opioid dependence. NIDA Research Monograph Series 121, pp 5–11

    CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

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

    CAS  Google Scholar 

  257. Keup W (1983) Clonidin–seine Möglichkeiten in der Pharmakotherapie der Heroin abhängigkeit. Dtsch Ärztebl 80 /3: 25–32

    Google Scholar 

  258. Seevers MH (1972) Characteristics of dependence on and abuse of psychoactive drugs. In: Mule SJ, Brill H (eds) Chemical and biological aspects of drug dependence, vol 2. CRC Press, Cleveland, pp 13–21

    Google Scholar 

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

    Google Scholar 

  260. Holaday JW, Tortella FC (1984) Multiple opioid receptors: Possible physiological functions of jj and 5 binding sites in vivo. In: Genazzini AR, Müller EE (eds) Central and peripheral endorphins: Basis and clinical aspects. Raven, New York, pp 237–250

    Google Scholar 

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

    Article  CAS  Google Scholar 

  262. Schmidhammer H, Smith CFC, Erlach D et al. (1990) Synthesis and biological evaluation of 14-Alkoxymorphinans. 3. Extensive study on cyprodime-related compounds. J Med Chem 33: 1200–1206

    Article  PubMed  CAS  Google Scholar 

  263. 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 

  264. Buzas B, Toth G, Cavagnero S, Hruby VJ, Borsodi A (1992) Synthesis and binding characteristics of the highly delta-specific new tritiated opioid peptide [3H]deltorphin II. Life Sei 50: PL 75-PL 77

    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). Der opiatabhängige Patient. In: Opioide in der Medizin. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-662-09096-1_33

Download citation

  • DOI: https://doi.org/10.1007/978-3-662-09096-1_33

  • 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