Skip to main content

Advertisement

Log in

Methylnaltrexone: its pharmacological effects alone and effects on morphine in healthy volunteers

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

Abstract

Rationale

Methylnaltrexone bromide (MTNX) is a peripherally acting mu-opioid receptor antagonist, prescribed for the treatment of opioid-induced constipation in patients with advanced illness who are receiving palliative care. Studies have used this drug to determine if other opioid-induced effects besides constipation are altered by MTNX in humans and have suggested, based on their results, that these other effects are altered by peripheral opioid actions.

Objective

The primary objective of this report is to present results that provide indirect evidence that MTNX has centrally mediated effects, albeit slight, and secondarily to describe the effects of MTNX on psychopharmacological effects of morphine.

Methods

In a crossover, randomized, placebo-controlled, double-blind study, 29 healthy volunteers received 0.45 mg/kg MTNX or saline subcutaneously, followed by saline intravenously. In three other conditions, 0.143 mg/kg of morphine sulfate administered intravenously was preceded by subcutaneous administration of 0, 0.225, or 0.45 mg/kg MTNX. Before and after drug administration, subjective and physiological measures, including pupil diameter, were assessed.

Results

Two separate analyses confirmed that 0.45 mg/kg MTNX alone induced a slight degree of miosis, a centrally mediated opioid agonist effect. This dose had minimal subjective effects. MTNX at either or both the 0.225 and 0.45 mg/kg dose reduced some subjective effects of morphine without altering miosis.

Conclusions

We present indirect evidence that MTNX crosses the blood–brain barrier in humans. Therefore, whether the reductions in subjective effects of morphine by MTNX that were observed in past studies and in this study can be attributed to peripheral mechanisms is open to question.

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
Fig. 4
Fig. 5

Similar content being viewed by others

References

  • American Psychiatric Association (2000) Diagnostic and statistical manual of mental disorders DSM-IV-TR. American Psychiatric Association, Washington, Text revision

    Google Scholar 

  • Bader S, Dürk T, Becker G (2013) Methylnaltrexone for the treatment of opioid-induced constipation. Expert Review of Gastroenterology & Hepatology 7:13–26

    Article  CAS  Google Scholar 

  • Beattie DT, Cheruvu M, Mai N, O’Keefe M, Johnson-Rabidoux S, Peterson C et al (2007) The in vitro pharmacology of the peripherally restricted opioid receptor antagonists, alvimopan, ADL 08–0011 and methylnaltrexone. Naunyn-Schmiedeberg’s Archives of Pharmacology 375:205–220

    Article  CAS  PubMed  Google Scholar 

  • Bigliardi PL, Stammer H, Jost G, Rufli T, Buchner S, Bigliardi-Qi M (2007) Treatment of pruritis with topically applied opiate receptor antagonist. Journal of the American Academy of Dermatology 56:979–988

    Article  PubMed  Google Scholar 

  • Brown DR, Goldberg LI (1985) The use of quaternary narcotic antagonists in opiate research. Neuropharmacology 24:181–191

    Article  CAS  PubMed  Google Scholar 

  • Butelman ER, Harris TJ, Kreek MJ (2004) Antiallodynic effects of loperamide and fentanyl against topical capsaicin-induced allodynia in unanesthetized primates. Journal of Pharmacology and Experimental Therapeutics 311:155–163

    Article  CAS  PubMed  Google Scholar 

  • Chamberlain BH, Cross K, Winston JL, Thomas J, Wang W, Su C et al (2009) Methylnaltrexone treatment of opioid-induced constipation in patients with advanced illness. Journal of Pain and Symptom Management 38:683–690

    Article  CAS  PubMed  Google Scholar 

  • Evans JM, Hogg MI, Lunn JN, Rosen M (1974) A comparative study of the narcotic against activity of naloxone and levallorphan. Anaesthesia 29:721–727

    Article  CAS  PubMed  Google Scholar 

  • Foss JF, O’Connor MF, Yuan CS, Murphy M, Moss J, Roizen MF (1997) Safety and tolerance of methylnaltrexone in healthy humans: a randomized, placebo-controlled, intravenous, ascending-dose, pharmacokinetic study. Journal of Clinical Pharmacology 37:25–30

    Article  CAS  PubMed  Google Scholar 

  • Fraser HF, Isbell H (1961) Human pharmacology and addictiveness of ethyl 1-(3-cyano-3,3-phenylpropyl)-4-phenyl-4-piperadine carboxylate hydrochloride. Bulletin on Narcotics 13:29–43

    CAS  Google Scholar 

  • Fraser HF, van Horn GD, Martin WR, Wolbach AB, Isbell H (1961) Methods for evaluating addiction liability. (a) “attitude” of opiate addicts toward opiate-like drugs, (b) a short-term “direct” addiction test. Journal of Pharmacology and Experimental Therapeutics 133:371–387

    CAS  PubMed  Google Scholar 

  • Garnock-Jones KP, McKeage K (2010) Methylnaltrexone. Drugs 70:919–928

    Article  CAS  PubMed  Google Scholar 

  • Gatti A, Sabato AF (2012) Management of opioid-induced constipation in cancer patients: focus on methylnaltrexone. Clinical Drug Investigation 32:293–301

    Article  CAS  PubMed  Google Scholar 

  • Haertzen CA (1966) Development of scales based on patterns of drug effects, using the Addiction Research Center Inventory (ARCI). Psychological Reports 18:163–194

    Article  CAS  PubMed  Google Scholar 

  • Hannington-Kiff JG (1970) Measurement of recovery from outpatient general anaesthesia with a simple ocular test. British Medical Journal 3:132–135

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Holzer P (2009) Opioid receptors in the gastrointestinal tract. Regulatory Peptides 155:11–17

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Jasinski DR, Martin WR, Haertzen CA (1967) The human pharmacology and abuse potential of N-allylnoroxymorphone (naloxone). Journal of Pharmacology and Experimental Therapeutics 157:420–426

    CAS  PubMed  Google Scholar 

  • Kast RE (2009) Use of FDA approved methamphetamine to allow adjunctive use of methylnaltrexone to mediate core anti-growth factor signaling effects in glioblastoma. Journal of Neuro-Oncology 94:163–167

    Article  CAS  PubMed  Google Scholar 

  • Korey A, Zilm DH, Sellers EM (1979) A comparison of the dependence liabilities of two antidiarrheal agents, nufenoxole and loperamide, in volunteer subjects (abstract). Clinical Pharmacology and Therapeutics 25:232

    Google Scholar 

  • Kumor KM, Haertzen CA, Jasinski DR, Johnson RE (1988) The psychopharmacologic and prolactin response after large doses of naloxone in man. Pharmacology, Biochemistry, and Behavior 30:967–975

    Article  CAS  PubMed  Google Scholar 

  • Lee HK, Wang SC (1975) Mechanism of morphine-induced miosis in the dog. Journal of Pharmacology and Experimental Therapeutics 192:415–431

    CAS  PubMed  Google Scholar 

  • Levy JH, Brister NW, Shearin A, Ziegler J, Hug CC, Adelson DM et al (1989) Wheal and flare responses to opioids in humans. Anesthesiology 70:756–760

    Article  CAS  PubMed  Google Scholar 

  • Loimer N, Schmid R, Grünberger J, Linzmayer L (1990) Naloxone induces miosis in normal subjects. Psychopharmacology 101(2):282–283

    Article  CAS  PubMed  Google Scholar 

  • Lötsch J, Skarke C, Grösch S, Darimont J, Schmidt H, Geisslinger G (2002) The polymorphism A118G of the human mu-opioid receptor gene decreases the pupil constrictory effect of morphine-6-glucuronide but not that of morphine. Pharmacogenetics 12:3–9

    Article  PubMed  Google Scholar 

  • Martin WR, Sloan JW, Sapira JD, Jasinski DR (1971) Physiologic, subjective and behavioral effects of amphetamine, methamphetamine, ephedrine, phenmetrazine, and methylphenidate in man. Clinical Pharmacology and Therapeutics 12:245–258

    CAS  PubMed  Google Scholar 

  • Murphy DB, Sutton JA, Prescott LF, Murphy MB (1997) Opioid-induced delay in gastric emptying: a peripheral mechanism in humans. Anesthesiology 87:765–770

    Article  CAS  PubMed  Google Scholar 

  • Murray RB, Adler MW, Korczyn AD (1983) The pupillary effects of opioids. Life Sciences 33:495–509

    Article  CAS  PubMed  Google Scholar 

  • Portenoy RK, Thomas J, Boatwright MLM, Tran D, Galasso FL, Stambler N et al (2008) Subcutaneous methylnaltrexone for the treatment of opioid-induced constipation in patients with advanced illness: a double-blind, randomized, parallel group, dose-ranging study. Journal of Pain and Symptom Management 35:458–468

    Article  CAS  PubMed  Google Scholar 

  • Preston KL, Bigelow GE, Bickel WK, Liebson IA (1989) Drug discrimination in human postaddicts: agonist–antagonist opioids. Journal of Pharmacology and Experimental Therapeutics 250:184–196

    CAS  PubMed  Google Scholar 

  • Ray R, Ruparel K, Newberg A, Wileyto EP, Loughead JW, Divgi C et al (2011) Human mu opioid receptor (OPRM1 A118G) polymorphism is associated with brain mu-opioid receptor binding potential in smokers. Proceedings of the National Academy of Sciences of the United States of America 108:9268–9273

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Reisine T, Pasternak G (1996) Opioid analgesics and antagonists. In: Hardman JG, Gilman AG, Limbird LE (eds) Goodman and Gilman’s The pharmacological basis of therapeutics. McGraw-Hill, New York, pp 521–555

    Google Scholar 

  • Rosow CE, Gomery P, Chen TY, Stefanovich P, Stambler N, Israel R (2007) Reversal of opioid-induced bladder dysfunction by intravenous naloxone and methylnaltrexone. Clinical Pharmacology and Therapeutics 82:48–53

    Article  CAS  PubMed  Google Scholar 

  • Rubin P, Blaschke TF, Guilleminault C (1981) Effect of naloxone, a specific opioid inhibitor, on blood pressure fall during sleep. Circulation 63:117–121

    Article  CAS  PubMed  Google Scholar 

  • Russell J, Bass P, Goldberg LI, Schuster CR, Merz H (1982) Antagonism of gut, but not central effects of morphine with quaternary opiate antagonists. European Journal of Pharmacology 78:255–261

    Article  CAS  PubMed  Google Scholar 

  • Slatkin N, Thomas J, Lipman AG, Wilson G, Boatwright ML, Wellman C et al (2009) Methylnaltrexone for treatment of opioid-induced constipation in advanced illness patients. Journal of Supportive Oncology 7:39–46

    CAS  PubMed  Google Scholar 

  • Thomas J, Karver S, Cooney GA, Chamberlain BH, Watt CK, Slatkin NE et al (2008) Methylnaltrexone for opioid-induced constipation in advanced illness. NEJM 358:2332–2343

    Article  CAS  PubMed  Google Scholar 

  • Trujillo KA, Belluzzi JD, Stein L (1989) Effects of opiate antagonists and their quaternary analogues on nucleus accumbens self-stimulation. Behavioural Brain Research 33:181–188

    Article  CAS  PubMed  Google Scholar 

  • Wechsler D (1958) The measurement and appraisal of adult intelligence. Williams and Wilkins, Baltimore

    Book  Google Scholar 

  • Weinstock LB, Chang AC (2011) Methylnaltrexone for treatment of acute colonic pseudo-obstruction. Journal of Clinical Gastroenterology 45:883–884

    Article  CAS  PubMed  Google Scholar 

  • Willer JC, Boreau F, Dauthier C, Bonora M (1979) Study of naloxone in normal awake man: effects on heart rate and respiration. Neuropharmacology 18:469–472

    Article  CAS  PubMed  Google Scholar 

  • Yamamoto A, Sugimoto Y (2010) Involvement of peripheral mu opioid receptors in scratching behavior in mice. European Journal of Pharmacology 649:336–341

    Article  CAS  PubMed  Google Scholar 

  • Yuan C-S, Foss JF (1999) Gastric effects of methylnaltrexone on mu, kappa, and delta opioid agonists induced brainstem unitary responses. Neuropharmacology 38:425–432

    Article  CAS  PubMed  Google Scholar 

  • Yuan C-S, Foss JF, O’Connor M, Toledano A, Roizen MF, Moss J (1996) Methylnaltrexone prevents morphine-induced delay in oral–cecal transit time without affecting analgesia: a double-blind, randomized placebo-controlled trial. Clinical Pharmacology and Therapeutics 59:469–475

    Article  CAS  PubMed  Google Scholar 

  • Yuan C-S, Foss JF, O’Connor M, Osinski J, Roizen MF, Moss J (1998) Efficacy of orally administered methylnaltrexone in decreasing subjective effects after intravenous morphine. Drug and Alcohol Dependence 52:161–165

    Article  CAS  PubMed  Google Scholar 

  • Yuan CS, Foss JF, O’Connor M, Osinski J, Karrison T, Moss J et al (2000) Methylnaltrexone for reversal of constipation due to chronic methadone use: a randomized controlled trial. JAMA 283:367–372

    Article  CAS  PubMed  Google Scholar 

  • Yuan C-S, Wei G, Foss JF, O’Connor M, Karrison T, Osinski J (2002) Effects of subcutaneous methylnaltrexone on morphine-induced peripherally mediated side effects: a double-blind randomized placebo-controlled trial. Journal of Pharmacology and Experimental Therapeutics 300:118–123

    Article  CAS  PubMed  Google Scholar 

  • Yuan CS, Doshan H, Charney MR, O’Connor M, Karrison T, Maleckar SA et al (2005) Tolerability, gut effects, and pharmacokinetics of methylnaltrexone following repeated intravenous administration in humans. Journal of Clinical Pharmacology 45:538–546

    Article  CAS  PubMed  Google Scholar 

  • Zand F, Amini A, Asadi S, Farbood A (2014) The effect of methylnaltrexone on the side effects of intrathecal morphine after orthopedic surgery under spinal anesthesia. Pain Practice. doi:10.1111/papr.12185

    PubMed  Google Scholar 

  • Zilm DH (1980) Naloxone response in non-dependent man: effect on six physiological variables. Neuropharmacology 19:591–595

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

Research was supported in part by grant R21 DA031318 from the National Institute on Drug Abuse. We wish to thank Drs. Ravi Singh, William (Scott) Jones, Joseph Enayati, Michael Esposito, Hasan Chowdhury, Kelly Eaton, Amy Gruber, Annie Amin, and Keith Rodriguez for administering the drugs and monitoring the physiological status of the research volunteers; Karin Kirulis for screening potential subjects and conducting the structured interviews; Katarzyna Zoszak, Sean M. Apfelbaum, and Sandra Gutierrez for assistance in conducting the experimental sessions; and for the research volunteers who participated in the study. We also wish to thank an anonymous reviewer who offered critical and valuable comments on earlier iterations of this manuscript that resulted in a more coherent and parsimonious representation of the findings of the study.

Conflicts of interest

The authors have no conflicts of interest to report.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to James P. Zacny.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zacny, J.P., Wroblewski, K. & Coalson, D.W. Methylnaltrexone: its pharmacological effects alone and effects on morphine in healthy volunteers. Psychopharmacology 232, 63–73 (2015). https://doi.org/10.1007/s00213-014-3637-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00213-014-3637-8

Keywords

Navigation