Skip to main content
Log in

Comparison of naltrexone, 6α-naltrexol, and 6β-naltrexol in morphine-dependent and in nondependent rhesus monkeys

  • Original Investigation
  • Published:
Psychopharmacology Aims and scope Submit manuscript

Abstract

Rationale

Some opioid receptor ligands that appear to be neutral antagonists can have inverse agonist activity under conditions of increased constitutive activity (e.g., agonist treatment).

Objectives

This study compared the opioid receptor antagonist naltrexone and its metabolites 6α-naltrexol and 6β-naltrexol in nondependent and morphine-dependent monkeys to see whether their potencies varied according to drug treatment and, presumably, to differences in constitutive activity of μ opioid receptors.

Results

In monkeys (n = 4) receiving 3.2 mg/kg per day of morphine and discriminating 0.0178 mg/kg naltrexone, naltrexone and each metabolite increased responding on the naltrexone lever in a dose-related manner with naltrexone being 8- and 71-fold more potent than 6α- and 6β-naltrexol, respectively. After 27 h of no-morphine treatment, monkeys responded on the naltrexone lever, and this effect was reversed by morphine. Naltrexone and each metabolite prevented morphine reversal of naltrexone-lever responding, and their rank order potency was the same as their substitution for naltrexone; however, the potency between naltrexone and each metabolite was slightly greater in morphine-dependent as compared to morphine-deprived monkeys. In a separate group (n = 3) of nondependent monkeys discriminating 1.78 mg/kg of morphine, all three compounds antagonized morphine with the same potency as in the reversal study (morphine-dependent monkeys), with Schild analyses showing no difference in apparent affinities (pA 2) between nondependent and morphine-dependent monkeys.

Conclusion

Naltrexone and 6α- and 6β-naltrexol have qualitatively similar effects, and their potencies do not vary markedly with opioid treatment, suggesting that under these conditions, they do not vary with regard to inverse agonism.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  • Anton RF (2001) Pharmacologic approaches to the management of alcoholism. J Clin Psychiatry 62(Suppl 20):11–17

    PubMed  CAS  Google Scholar 

  • Arunlakshana O, Schild HO (1959) Some quantitative uses of drug antagonists. Br J Pharmacol Chemother 14:48–58

    PubMed  CAS  Google Scholar 

  • Arvanitakis L, Geras-Raaka E, Varma A, Gershengorn MC, Cesarman E (1997) Human herpes virus KSHV encodes a constitutively active G-protein coupled receptor linked to cell proliferation. Nature (Lond) 385:347–350

    Article  CAS  Google Scholar 

  • Berg KA, Harvey JA, Spampinato U, Clarke WP (2005) Physiological relevance of constitutive activity of 5-HT2A and 5-HT2C receptors. Trends Pharmacol Sci 26:625–630

    Article  PubMed  CAS  Google Scholar 

  • Charpentier S, Jarvie KR, Severynse DM, Caron MG, Tiberi M (1996) Silencing of the constitutive activity of the dopamine D1B receptor. Reciprocal mutations between D1 receptor subtypes delineate residues underlying activation properties. J Biol Chem 271:28071–28076

    Article  PubMed  CAS  Google Scholar 

  • Costa T, Ogino Y, Munson PJ, Onaran O, Rodbard D (1992) Drug efficacy at guaninenucleotide-binding regulatory protein linked receptors: thermodynamic interpretations of negative antagonism and of receptor activity in the absence of ligand. Mol Pharmacol 41:549–560

    PubMed  CAS  Google Scholar 

  • De Deurwaerdère P, Navailles S, Berg KA, Clarke WP, Spampinato U (2004) Constitutive activity of the serotonin2C receptor inhibits in vivo dopamine release in the rat striatum and nucleus accumbens. J Neurosci 24:3235–3241

    Article  PubMed  Google Scholar 

  • France CP, Woods JH (1989) Discriminative stimulus effects of naltrexone in morphine-treated rhesus monkeys. J Pharmacol Exp Ther 250:937–943

    PubMed  CAS  Google Scholar 

  • France CP, de Costa BR, Jacobson AE, Rice KC, Woods JH (1990) Apparent affinity of opioid antagonists in morphine-treated rhesus monkeys discriminating between saline and naltrexone. J Pharmacol Exp Ther 252:600–604

    PubMed  CAS  Google Scholar 

  • Gerak LR, France CP (2007) Time-dependent decreases in apparent pA2 values for naltrexone studied in combination with morphine in rhesus monkeys. Psychopharmacology 193:315–321

    Google Scholar 

  • Gonzalez G, Oliverto A, Kosten TR (2004) Combating opiate dependence: a comparison among the available pharmacological options. Expert Opin Pharmacother 5:713–725

    Article  PubMed  CAS  Google Scholar 

  • Hamad MO, Kiptoo PK, Stinchbomb AL, Crooks PA (2006) Synthesis and hydrolytic behavior of two novel tripartite codrugs of naltrexone and 6β-naltrexol with hydroxybupropion as potential alcohol abuse and smoking cessation agents. Bioorg Med Chem 14:7051–7061

    Article  PubMed  CAS  Google Scholar 

  • Harvey JA, Welsh SE, Hood H, Romano AG (1999) effects of 5-HT2 receptor antagonists on a cranial nerve reflex in the rabbit: evidence for inverse agonism. Psychopharmacology 141:162–168

    Article  PubMed  CAS  Google Scholar 

  • Kiptoo PK, Hamad MO, Crooks PA, Stinchcomb AL (2006) Enhancement of transdermal delivery of 6β-naltrexol via a codrug linked to hydroxybupropion. J Control Release 113:137–145

    Article  PubMed  CAS  Google Scholar 

  • Ko MC, Divin MF, Lee H, Woods JH, Traynor JR (2006) Differential in vivo potencies of naltrexone and 6beta-naltrexol in the monkey. J Pharmacol Exp Ther 316:772–779

    Article  PubMed  CAS  Google Scholar 

  • Malspeis L, Bathala MS, Ludden TM, Bhat HB, Frank SG, Sokoloski TD, Morrison BE, Reuning RH (1975) Metabolic reduction of naltrexone. I. Synthesis, separation and characterization of naloxone and naltrexone reduction products and qualitative assay of urine and bile following administration of naltrexone, alpha-naltrexol, or beta-naltrexol. Res Commun Chem Pathol Pharmacol 12:43–65

    PubMed  CAS  Google Scholar 

  • Meyer MC, Straughn AB, Lo MW, Schary WL, Whitney CC (1984) Bioequivalence, dose-proportionality, and pharmacokinetics of naltrexone after oral administration. J Clin Psychiatry 45:15–19

    PubMed  CAS  Google Scholar 

  • Milligan G, Bond RA, Lee M (1995) Inverse agonism, pharmacological curiosity or potential therapeutic strategy. Trends Pharmacol Sci 16:10–13

    Article  PubMed  CAS  Google Scholar 

  • Misra AL, Bloch R, Vardy J, Mule SJ, Verebely K (1976) Disposition of (15,16-3H)naltrexone in the central nervous system of the rat. Drug Metab Dispos 4:276–280

    PubMed  CAS  Google Scholar 

  • Porter SJ, Somogyi AA, White JM (2002) In vivo and in vitro studies of 6β-naltrexol, the major human metabolite of naltrexone. Addict Biol 7:218–225

    Google Scholar 

  • Raehal KM, Lowery JJ, Bhamidipati CM, Paolino RM, Blair JR, Wang D, Sadee W, Bilsky EJ (2005) In vivo characterization of 6beta-naltrexol, an opioid ligand with less inverse agonist activity compared with naltrexone and naloxone in opioid-dependent mice. J Pharmacol Exp Ther 313:1150–1162

    Article  PubMed  CAS  Google Scholar 

  • Romano AG, Quinn JL, Liu R, Dave KD, Schwab D, Alexander G, Aloyo VJ, Harvey JA (2006) Effects of serotonin depletion on 5-HT2A-mediated learning in the rabbit: evidence for constitutive activity of the 5-HT2A receptor in vivo. Psychopharmacology 184:173–181

    Article  PubMed  CAS  Google Scholar 

  • Samama P, Contecchia S, Costa T, Lefkowitz RJ (1993) A mutation-induced activated state of the β2-adrenergic receptor: extending the ternary complex model. J Biol Chem 268:4625–4636

    PubMed  CAS  Google Scholar 

  • Tallarida RJ, Cowan A, Adler MW (1979) pA2 and receptor differentiation: a statistical analysis of competitive antagonism. Life Sci 25:637–654

    Article  PubMed  CAS  Google Scholar 

  • Walker EA, Sterious SN (2005) Opioid antagonists differ according to negative intrinsic efficacy in a mouse model of acute dependence. Br J Pharmacol 145:975–983

    Article  PubMed  CAS  Google Scholar 

  • Wang D, Raehal KM, Bilsky EJ, Sadee W (2001) Inverse agonists and neutral antagonists at . μ. opioid receptor (MOR): possible role of basal receptor signaling in narcotic dependence. J Neurochem 77:1590–1600

    Article  PubMed  CAS  Google Scholar 

  • Wang D, Raehal KM, Lin ET, Lowery JJ, Kieffer BL, Bilsky EJ, Sadee W (2004) Basal signaling activity of mu opioid receptor in mouse brain: role in narcotic dependence. J Pharmacol Exp Ther 308:512–520

    Article  PubMed  CAS  Google Scholar 

  • Wang D, Sun X, Sadee W (2007) Different effects of opioid antagonists on μ-, δ-, and κ-opioid receptors with and without agonist pretreatment. J Pharmacol Exp Ther 321:544–552

    Article  PubMed  CAS  Google Scholar 

  • Wang Z, Edward JB, Porreca F, Sadee W (1994) Constitutive μ-opioid receptor activation as a regulatory mechanism underlying narcotic tolerance and dependence. Life Sci 54:PL339–PL350

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

The authors thank Christopher Cruz, Blake Harrington, and Brandi Taylor for their expert technical assistance. This work was supported by USPHS Grant DA05018 (CPF) and a Senior Scientist Award to CPF (DA17918).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Charles P. France.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Li, JX., McMahon, L.R. & France, C.P. Comparison of naltrexone, 6α-naltrexol, and 6β-naltrexol in morphine-dependent and in nondependent rhesus monkeys. Psychopharmacology 195, 479–486 (2008). https://doi.org/10.1007/s00213-007-0914-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00213-007-0914-9

Keywords

Navigation