Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Translational Therapeutics

Possible mechanism for improving the endogenous immune system through the blockade of peripheral μ-opioid receptors by treatment with naldemedine

Abstract

Background

It has been considered that activation of peripheral μ-opioid receptors (MORs) induces side effects of opioids. In this study, we investigated the possible improvement of the immune system in tumour-bearing mice by systemic administration of the peripheral MOR antagonist naldemedine.

Methods

The inhibitory effect of naldemedine on MOR-mediated signalling was tested by cAMP inhibition and β-arrestin recruitment assays using cultured cells. We assessed possible changes in tumour progression and the number of splenic lymphocytes in tumour-bearing mice under the repeated oral administration of naldemedine.

Results

Treatment with naldemedine produced a dose-dependent inhibition of both the decrease in the cAMP level and the increase in β-arrestin recruitment induced by the MOR agonists. Repeated treatment with naldemedine at a dose that reversed the morphine-induced inhibition of gastrointestinal transport, but not antinociception, significantly decreased tumour volume and prolonged survival in tumour-transplanted mice. Naldemedine administration significantly decreased the increased expression of immune checkpoint-related genes and recovered the decreased level of toll-like receptor 4 in splenic lymphocytes in tumour-bearing mice.

Conclusions

The blockade of peripheral MOR may induce an anti-tumour effect through the recovery of T-cell exhaustion and promotion of the tumour-killing system.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Evaluation of the antagonistic activity of naldemedine for µ-opioid receptors.
Fig. 2: Evaluation of the pharmacological profiles of the peripheral µ-opioid receptor antagonist naldemedine.
Fig. 3: Changes in tumour growth by the repeated treatment with peripheral µ-opioid receptor agonists and antagonists.
Fig. 4: Changes in the number of spleen-derived immune cells by the repeated administration of naldemedine in LLC cell-transplanted mice.
Fig. 5: Changes in mRNA expression of immune checkpoint receptors and cytotoxicity-related genes in CD8+ T cells by the repeated administration of naldemedine in LLC cell-transplanted mice.
Fig. 6: Changes in mRNA expression of opioid receptors and pattern recognition receptors in lymphocytes by the repeated administration of naldemedine in LLC cell-transplanted mice.

Similar content being viewed by others

Data availability

All data presented within the article are available upon request from the corresponding author.

References

  1. Trang T, Al-Hasani R, Salvemini D, Salter MW, Gutstein H, Cahill CM. Pain and poppies: the good, the bad, and the ugly of opioid analgesics. J Neurosci. 2015;35:13879–88.

    Article  CAS  Google Scholar 

  2. Peng J, Sarkar S, Chang SL. Opioid receptor expression in human brain and peripheral tissues using absolute quantitative real-time RT-PCR. Drug Alcohol Depend. 2012;124:223–8.

    Article  CAS  Google Scholar 

  3. Machelska H, Celik MO. Opioid Receptors in immune and glial cells-implications for pain control. Front Immunol. 2020;11:300.

    Article  CAS  Google Scholar 

  4. Camilleri M, Drossman DA, Becker G, Webster LR, Davies AN, Mawe GM. Emerging treatments in neurogastroenterology: a multidisciplinary working group consensus statement on opioid-induced constipation. Neurogastroenterol Motil. 2014;26:1386–95.

    Article  CAS  Google Scholar 

  5. Corder G, Tawfik VL, Wang D, Sypek EI, Low SA, Dickinson JR, et al. Loss of mu opioid receptor signaling in nociceptors, but not microglia, abrogates morphine tolerance without disrupting analgesia. Nat Med. 2017;23:164–73.

    Article  CAS  Google Scholar 

  6. Mathew B, Lennon FE, Siegler J, Mirzapoiazova T, Mambetsariev N, Sammani S, et al. The novel role of the mu opioid receptor in lung cancer progression: a laboratory investigation. Anesth Analg. 2011;112:558–67.

    Article  CAS  Google Scholar 

  7. Janku F, Johnson LK, Karp DD, Atkins JT, Singleton PA, Moss J. Treatment with methylnaltrexone is associated with increased survival in patients with advanced cancer. Ann Oncol. 2018;29:1076.

    Article  CAS  Google Scholar 

  8. Kanemasa T, Koike K, Arai T, Ono H, Horita N, Chiba H, et al. Pharmacologic effects of naldemedine, a peripherally acting mu-opioid receptor antagonist, in in vitro and in vivo models of opioid-induced constipation. Neurogastroenterol Motil. 2019;31:e13563.

    Article  CAS  Google Scholar 

  9. Watari R, Matsuda A, Ohnishi S, Hasegawa H. Minimal contribution of P-gp on the low brain distribution of naldemedine, a peripherally acting mu-opioid receptor antagonist. Drug Metab Pharmacokinet. 2019;34:126–33.

    Article  CAS  Google Scholar 

  10. Doi, S, Mori, T, Uzawa, N, Arima, T, Takahashi, T, Uchida, M et al. Characterization of methadone as a beta-arrestin-biased mu-opioid receptor agonist. Mol Pain 2016;12:1744806916654146.

  11. Suda Y, Kuzumaki N, Sone T, Narita M, Tanaka K, Hamada Y, et al. Down-regulation of ghrelin receptors on dopaminergic neurons in the substantia nigra contributes to Parkinson’s disease-like motor dysfunction. Mol Brain. 2018;11:6.

    Article  Google Scholar 

  12. Watkins LR, Wang X, Mustafa S, Hutchinson MR. In vivo veritas: (+)-Naltrexone’s actions define translational importance: a letter in response to Skolnick et al. ‘Translational potential of naloxone and naltrexone as TLR4 antagonists’. Trends Pharm Sci. 2014;35:432–3.

    Article  CAS  Google Scholar 

  13. Chan HCS, McCarthy D, Li J, Palczewski K, Yuan S. Designing safer analgesics via mu-opioid receptor pathways. Trends Pharm Sci. 2017;38:1016–37.

    Article  CAS  Google Scholar 

  14. Zylla D, Gourley BL, Vang D, Jackson S, Boatman S, Lindgren B, et al. Opioid requirement, opioid receptor expression, and clinical outcomes in patients with advanced prostate cancer. Cancer. 2013;119:4103–10.

    Article  CAS  Google Scholar 

  15. Zhang H, Sun M, Zhou D, Gorur A, Sun Z, Zeng W, et al. Increased mu-opioid receptor expression is associated with reduced disease-free and overall survival in laryngeal squamous cell carcinoma. Br J Anaesth. 2020;125:722–9.

    Article  CAS  Google Scholar 

  16. Okazaki T, Chikuma S, Iwai Y, Fagarasan S, Honjo T. A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application. Nat Immunol. 2013;14:1212–8.

    Article  CAS  Google Scholar 

  17. Schietinger A, Greenberg PD. Tolerance and exhaustion: defining mechanisms of T cell dysfunction. Trends Immunol. 2014;35:51–60.

    Article  CAS  Google Scholar 

  18. Trapani JA, Smyth MJ. Functional significance of the perforin/granzyme cell death pathway. Nat Rev Immunol. 2002;2:735–47.

    Article  CAS  Google Scholar 

  19. Rossaint J, Zarbock A. Perioperative inflammation and its modulation by anesthetics. Anesth Analg. 2018;126:1058–67.

    Article  CAS  Google Scholar 

  20. Zhang P, Yang M, Chen C, Liu L, Wei X, Zeng S. Toll-like receptor 4 (TLR4)/opioid receptor pathway crosstalk and impact on opioid analgesia, immune function, and gastrointestinal motility. Front Immunol. 2020;11:1455.

    Article  CAS  Google Scholar 

  21. Franchi S, Moretti S, Castelli M, Lattuada D, Scavullo C, Panerai AE, et al. Mu opioid receptor activation modulates Toll like receptor 4 in murine macrophages. Brain Behav Immun. 2012;26:480–8.

    Article  CAS  Google Scholar 

  22. Yang X, Jiang X, Chen G, Xiao Y, Geng S, Kang C, et al. T cell Ig mucin-3 promotes homeostasis of sepsis by negatively regulating the TLR response. J Immunol. 2013;190:2068–79.

    Article  CAS  Google Scholar 

  23. Souza-Fonseca-Guimaraes F, Parlato M, Philippart F, Misset B, Cavaillon JM, Adib-Conquy M, et al. Toll-like receptors expression and interferon-gamma production by NK cells in human sepsis. Crit Care. 2012;16:R206.

    Article  Google Scholar 

Download references

Acknowledgements

The authors thank Ms. Kana Morita, Ms. Arisa Gina, Ms. Anna Hori, Mr. Shoki Matsumura, Mr. Takuya Kurimura, Ms. Natsumi Kawamura and Ms. Mayu Takagi for their help with the experiments.

Funding

This work was supported in part by grants from the MEXT-Supported Program for the Strategic Research Foundation at Private Universities, 2014–2018, S1411019. This research was also supported by Hoshi University.

Author information

Authors and Affiliations

Authors

Contributions

EG, YH, TM and Minoru N wrote the manuscript. NK and Minoru N edited the manuscript. Michiko N, YS, KM and NK conducted in vitro studies. YH, YI, AS, DS, MT, SY, KT and KY conducted in vivo studies. HT provided technical support. HA, TY and KH synthesised naldemedine. EG, KA, MI and EI provided medical comments. NK and Minoru N supervised the overall project. All of the authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Naoko Kuzumaki or Minoru Narita.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

All animal studies were conducted in accordance with the Guiding Principles for the Care and Use of Laboratory Animals at Hoshi University, as adopted by the Committee on Animal Research of Hoshi University.

Consent to publish

Not applicable.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gondoh, E., Hamada, Y., Mori, T. et al. Possible mechanism for improving the endogenous immune system through the blockade of peripheral μ-opioid receptors by treatment with naldemedine. Br J Cancer 127, 1565–1574 (2022). https://doi.org/10.1038/s41416-022-01928-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-022-01928-x

This article is cited by

Search

Quick links