Skip to main content
Top
Published in: Current Oncology Reports 10/2022

Open Access 01-06-2022 | Naltrexone | Anesthesiology and Critical Care (JP Cata, Section Editor)

Antagonists of the Mu-Opioid Receptor in the Cancer Patient: Fact or Fiction?

Authors: Amparo Belltall, Guido Mazzinari, Oscar Diaz-Cambronero, Pilar Eroles, María Pilar Argente Navarro

Published in: Current Oncology Reports | Issue 10/2022

Login to get access

Abstract

Purpose of Review

Antagonists of mu-opioid receptor role in cancer progression remains to be elucidated. The objective of this review was to summarize the available evidence on antagonists of mu-opioid receptor effect on tumor progression and prognosis in different types of cancers and an evaluation of the available findings on their mechanism of action.

Recent Findings

We have found studies related to methylnaltrexone (MNTX) and naltrexone (NTX) usage in cancer outcomes-related setting. We found consistent preclinical evidence of a potential action of MNTX and NTX on cancer growth and spread mediated mainly by effect on the opioid growth factor receptor (OGFr) axis, which results in depressed cell replication. However, clinical results are scarce and limited to poor-quality evidence.

Summary

Further high-quality studies are warranted to study antagonists of mu-opioid receptor role as a therapeutic option in different types of cancer, especially in patients where the classical treatment causes unacceptable side effects.
Literature
1.
go back to reference Montagna G, Gupta HV, Hannum M, Tan KS, Lee J, Scarpa JR, et al. Intraoperative opioids are associated with improved recurrence-free survival in triple-negative breast cancer. Br J Anaesth. 2021;126(2):367–76.CrossRef Montagna G, Gupta HV, Hannum M, Tan KS, Lee J, Scarpa JR, et al. Intraoperative opioids are associated with improved recurrence-free survival in triple-negative breast cancer. Br J Anaesth. 2021;126(2):367–76.CrossRef
2.
go back to reference Jorand R, Biswas S, Wakefield DL, Tobin SJ, Golfetto O, Hilton K, et al. Molecular signatures of mu opioid receptor and somatostatin receptor 2 in pancreatic cancer. Mol Biol Cell. 2016;27(22):3659–72.CrossRef Jorand R, Biswas S, Wakefield DL, Tobin SJ, Golfetto O, Hilton K, et al. Molecular signatures of mu opioid receptor and somatostatin receptor 2 in pancreatic cancer. Mol Biol Cell. 2016;27(22):3659–72.CrossRef
3.
go back to reference Nylund G, Pettersson A, Bengtsson C, Khorram-Manesh A, Nordgren S, Delbro DS. Functional expression of μ-opioid receptors in the human colon cancer cell line, HT-29, and their localization in human colon. Dig Dis Sci. 2008;53(2):461–6.CrossRef Nylund G, Pettersson A, Bengtsson C, Khorram-Manesh A, Nordgren S, Delbro DS. Functional expression of μ-opioid receptors in the human colon cancer cell line, HT-29, and their localization in human colon. Dig Dis Sci. 2008;53(2):461–6.CrossRef
4.
go back to reference Zhang YF, Xu QX, Liao LD, Xu XE, Wu JY, Wu ZY, et al. Association of mu-opioid receptor expression with lymph node metastasis in esophageal squamous cell carcinoma. Dis Esophagus. 2015;28(2):196–203.CrossRef Zhang YF, Xu QX, Liao LD, Xu XE, Wu JY, Wu ZY, et al. Association of mu-opioid receptor expression with lymph node metastasis in esophageal squamous cell carcinoma. Dis Esophagus. 2015;28(2):196–203.CrossRef
5.
go back to reference Yao Y, Sai Y, Yong Yao R, Kun Zhuang L, Wei Qi W, Lv J, Zhou F, et al. MOR1 expression in gastric cancer: A biomarker associated with poor outcome. Clin Transl Sci. 2015;8(2):137–42.CrossRef Yao Y, Sai Y, Yong Yao R, Kun Zhuang L, Wei Qi W, Lv J, Zhou F, et al. MOR1 expression in gastric cancer: A biomarker associated with poor outcome. Clin Transl Sci. 2015;8(2):137–42.CrossRef
6.
go back to reference Chen DT, Pan JH, Chen YH, Xing W, Yan Y, Yuan YF, et al. The mu-opioid receptor is a molecular marker for poor prognosis in hepatocellular carcinoma and represents a potential therapeutic target. Br J Anaesth. 2019;122(6):e157–67.CrossRef Chen DT, Pan JH, Chen YH, Xing W, Yan Y, Yuan YF, et al. The mu-opioid receptor is a molecular marker for poor prognosis in hepatocellular carcinoma and represents a potential therapeutic target. Br J Anaesth. 2019;122(6):e157–67.CrossRef
7.
go back to reference Singleton PA, Mirzapoiazova T, Hasina R, Salgia R, Moss J. Increased μ-opioid receptor expression in metastatic lung cancer. Br J Anaesth. 2014;113(SUPPL. 1):103–8.CrossRef Singleton PA, Mirzapoiazova T, Hasina R, Salgia R, Moss J. Increased μ-opioid receptor expression in metastatic lung cancer. Br J Anaesth. 2014;113(SUPPL. 1):103–8.CrossRef
8.
go back to reference 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(23):4103–10.CrossRef 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(23):4103–10.CrossRef
9.
go back to reference 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(5):722–9.CrossRef 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(5):722–9.CrossRef
10.
go back to reference Díaz-Cambronero O, Mazzinari G, Giner F, Belltall A, Ruiz-Boluda L, Marqués-Marí A, et al. Mu opioid receptor 1 (MOR-1) expression in colorectal cancer and oncological long-term outcomes: a five-year retrospective longitudinal cohort study. Cancers (Basel). 2020;12(1):134. https://doi.org/10.3390/cancers12010134. Díaz-Cambronero O, Mazzinari G, Giner F, Belltall A, Ruiz-Boluda L, Marqués-Marí A, et al. Mu opioid receptor 1 (MOR-1) expression in colorectal cancer and oncological long-term outcomes: a five-year retrospective longitudinal cohort study. Cancers (Basel). 2020;12(1):134. https://​doi.​org/​10.​3390/​cancers12010134.
11.
go back to reference Singleton PA, Moss J, Karp DD, Atkins JT, Janku F. The mu opioid receptor: A new target for cancer therapy? Cancer. 2015 Aug 15;121(16):2681–8. https://doi.org/10.1002/cncr.29460. Epub 2015 Jun 4. PMID: 26043235. The authors present preclinical and clinical data that support their hypothesis that the mu opioid receptor is a potential target for cancer therapy because of its plausible role in tumor progression. The authors also propose the hypothesis that PAMORAs can be used to target the mu opioid receptor. Singleton PA, Moss J, Karp DD, Atkins JT, Janku F. The mu opioid receptor: A new target for cancer therapy? Cancer. 2015 Aug 15;121(16):2681–8. https://​doi.​org/​10.​1002/​cncr.​29460. Epub 2015 Jun 4. PMID: 26043235. The authors present preclinical and clinical data that support their hypothesis that the mu opioid receptor is a potential target for cancer therapy because of its plausible role in tumor progression. The authors also propose the hypothesis that PAMORAs can be used to target the mu opioid receptor.
19.
go back to reference Ringerike T, Pike E, Nevjar J, Klemp M. The use of naltrexone in low doses beyond the approved indication [Internet]. Oslo, Norway: Knowledge Centre for the Health Services at The Norwegian Institute of Public Health (NIPH); 2015 Apr. Report from Norwegian Knowledge Centre for the Health Services (NOKC) No. 8-2015. Ringerike T, Pike E, Nevjar J, Klemp M. The use of naltrexone in low doses beyond the approved indication [Internet]. Oslo, Norway: Knowledge Centre for the Health Services at The Norwegian Institute of Public Health (NIPH); 2015 Apr. Report from Norwegian Knowledge Centre for the Health Services (NOKC) No. 8-2015.
22.
go back to reference Chamie K, Golla V, Lenis AT, Lec PM, Rahman S, Viscusi ER. Peripherally Acting μ-Opioid Receptor Antagonists in the Management of Postoperative Ileus: a Clinical Review. J Gastrointest Surg. 2021 Jan;25(1):293–302. https://doi.org/10.1007/s11605-020-04671-x. Epub 2020 Aug 10. PMID: 32779081; PMCID: PMC7851096. This review discuss recent clinical trials evaluating the safety and efficacy of PAMORAs, with a focus on alvimopan and methylnaltrexone in patients with POI or postoperative OIC. Chamie K, Golla V, Lenis AT, Lec PM, Rahman S, Viscusi ER. Peripherally Acting μ-Opioid Receptor Antagonists in the Management of Postoperative Ileus: a Clinical Review. J Gastrointest Surg. 2021 Jan;25(1):293–302. https://​doi.​org/​10.​1007/​s11605-020-04671-x. Epub 2020 Aug 10. PMID: 32779081; PMCID: PMC7851096. This review discuss recent clinical trials evaluating the safety and efficacy of PAMORAs, with a focus on alvimopan and methylnaltrexone in patients with POI or postoperative OIC.
24.
26.
go back to reference Libran Oriol A, Cruz-Sequeiros C, Luque-Blanco A, Porta-Sales J. Peri- pheral acting mu opioid receptor antagonists in the treatment of opioid- induced constipation: review. Rev Soc Esp Dolor. 2020;27(1):37–49. Libran Oriol A, Cruz-Sequeiros C, Luque-Blanco A, Porta-Sales J. Peri- pheral acting mu opioid receptor antagonists in the treatment of opioid- induced constipation: review. Rev Soc Esp Dolor. 2020;27(1):37–49.
28.
go back to reference Fernández-Montes A, de Velasco G, Aguín S, Farriols C, Guirado-Risueño M, Jerviz-Guía VG, Baeza-Nadal MV, Chicas-Sett R, Fírvida JL, García-Navalón F, Martín P, Perezagua-Marín C, Rodríguez D, Santamaría J, Saurí T, Cobo M. Insights into the Use of Peripherally Acting μ-Opioid Receptor Antagonists (PAMORAs) in Oncologic Patients: from Scientific Evidence to Real Clinical Practice. Curr Treat Options Oncol. 2021 Feb 26;22(3):26. https://doi.org/10.1007/s11864-021-00816-5. PMID: 33635493. A panel of experts pools their clinical experience with PAMORAs in cancer patients with OIC and highlights the importance of timing and choice of therapy in achieving prompt OIC management and benefitting patients. Fernández-Montes A, de Velasco G, Aguín S, Farriols C, Guirado-Risueño M, Jerviz-Guía VG, Baeza-Nadal MV, Chicas-Sett R, Fírvida JL, García-Navalón F, Martín P, Perezagua-Marín C, Rodríguez D, Santamaría J, Saurí T, Cobo M. Insights into the Use of Peripherally Acting μ-Opioid Receptor Antagonists (PAMORAs) in Oncologic Patients: from Scientific Evidence to Real Clinical Practice. Curr Treat Options Oncol. 2021 Feb 26;22(3):26. https://​doi.​org/​10.​1007/​s11864-021-00816-5. PMID: 33635493. A panel of experts pools their clinical experience with PAMORAs in cancer patients with OIC and highlights the importance of timing and choice of therapy in achieving prompt OIC management and benefitting patients.
30.
go back to reference Wang R, Zhang Y, Shan F. Interaction of opioid growth factor (OGF) and opioid antagonist and their significance in cancer therapy. Int Immunopharmacol. 2019 Oct;75:105785. https://doi.org/10.1016/j.intimp.2019.105785. Epub 2019 Aug 9. PMID: 31404891. This review provides insight into the interactions between OGF and OGFr in the treatment of cancers. Wang R, Zhang Y, Shan F. Interaction of opioid growth factor (OGF) and opioid antagonist and their significance in cancer therapy. Int Immunopharmacol. 2019 Oct;75:105785. https://​doi.​org/​10.​1016/​j.​intimp.​2019.​105785. Epub 2019 Aug 9. PMID: 31404891. This review provides insight into the interactions between OGF and OGFr in the treatment of cancers.
32.
go back to reference Buggy DJ, Freeman J, Johnson MZ, Leslie K, Riedel B, Sessler DI, Kurz A, Gottumukkala V, Short T, Pace N, Myles PS; StEP-COMPAC Group. Systematic review and consensus definitions for standardised endpoints in perioperative medicine: postoperative cancer outcomes. Br J Anaesth. 2018;121(1):38–44. https://doi.org/10.1016/j.bja.2018.03.020.CrossRef Buggy DJ, Freeman J, Johnson MZ, Leslie K, Riedel B, Sessler DI, Kurz A, Gottumukkala V, Short T, Pace N, Myles PS; StEP-COMPAC Group. Systematic review and consensus definitions for standardised endpoints in perioperative medicine: postoperative cancer outcomes. Br J Anaesth. 2018;121(1):38–44. https://​doi.​org/​10.​1016/​j.​bja.​2018.​03.​020.CrossRef
33.
34.
go back to reference Gorur A, Patiño M, Shi T, Corrales G, Takahashi H, Rangel R, Gleber-Netto FO, Pickering C, Myers JN, Cata JP. Low doses of methylnaltrexone inhibits head and neck squamous cell carcinoma growth in vitro and in vivo by acting on the mu-opioid receptor. J Cell Physiol. 2021;236(11):7698–710. https://doi.org/10.1002/jcp.30421 (Epub 2021 May 26 PMID: 34038587).CrossRefPubMed Gorur A, Patiño M, Shi T, Corrales G, Takahashi H, Rangel R, Gleber-Netto FO, Pickering C, Myers JN, Cata JP. Low doses of methylnaltrexone inhibits head and neck squamous cell carcinoma growth in vitro and in vivo by acting on the mu-opioid receptor. J Cell Physiol. 2021;236(11):7698–710. https://​doi.​org/​10.​1002/​jcp.​30421 (Epub 2021 May 26 PMID: 34038587).CrossRefPubMed
36.
go back to reference Qu N, Meng Y, Handley MK, Wang C, Shan F. Preclinical and clinical studies into the bioactivity of low-dose naltrexone (LDN) for oncotherapy. Int Immunopharmacol. 2021 Jul;96:107714. https://doi.org/10.1016/j.intimp.2021.107714. Epub 2021 May 11. PMID: 33989971. The authors summarize the associated studies on LDN oncotherapy to highlight the potential mechanisms and prospective clinical applications. Qu N, Meng Y, Handley MK, Wang C, Shan F. Preclinical and clinical studies into the bioactivity of low-dose naltrexone (LDN) for oncotherapy. Int Immunopharmacol. 2021 Jul;96:107714. https://​doi.​org/​10.​1016/​j.​intimp.​2021.​107714. Epub 2021 May 11. PMID: 33989971. The authors summarize the associated studies on LDN oncotherapy to highlight the potential mechanisms and prospective clinical applications.
37.
go back to reference Liubchenko K, Kordbacheh K, Khajehdehi N, Visnjevac T, Ma F, Khan JS, Storey M, Abd-Elsayed A, Visnjevac O. Naltrexone’s Impact on Cancer Progression and Mortality: A Systematic Review of Studies in Humans, Animal Models, and Cell Cultures. Adv Ther. 2021;38(2):904–24. https://doi.org/10.1007/s12325-020-01591-9 (Epub 2020 Dec 18 PMID: 33337537).CrossRefPubMed Liubchenko K, Kordbacheh K, Khajehdehi N, Visnjevac T, Ma F, Khan JS, Storey M, Abd-Elsayed A, Visnjevac O. Naltrexone’s Impact on Cancer Progression and Mortality: A Systematic Review of Studies in Humans, Animal Models, and Cell Cultures. Adv Ther. 2021;38(2):904–24. https://​doi.​org/​10.​1007/​s12325-020-01591-9 (Epub 2020 Dec 18 PMID: 33337537).CrossRefPubMed
40.
go back to reference Murugan S, Rousseau B, Sarkar DK. Beta 2 Adrenergic Receptor Antagonist Propranolol and Opioidergic Receptor Antagonist Naltrexone Produce Synergistic Effects on Breast Cancer Growth Prevention by Acting on Cancer Cells and Immune Environment in a Preclinical Model of Breast Cancer. Cancers (Basel). 2021;13(19):4858. https://doi.org/10.3390/cancers13194858.CrossRef Murugan S, Rousseau B, Sarkar DK. Beta 2 Adrenergic Receptor Antagonist Propranolol and Opioidergic Receptor Antagonist Naltrexone Produce Synergistic Effects on Breast Cancer Growth Prevention by Acting on Cancer Cells and Immune Environment in a Preclinical Model of Breast Cancer. Cancers (Basel). 2021;13(19):4858. https://​doi.​org/​10.​3390/​cancers13194858.CrossRef
44.
go back to reference Wang CZ, Li XL, Sun S, Xie JT, Aung HH, Tong R, McEntee E, Yuan CS. Methylnaltrexone, a peripherally acting opioid receptor antagonist, enhances tumoricidal effects of 5-Fu on human carcinoma cells. Anticancer Res. 2009;29(8):2927–32 (PMID: 19661297).PubMed Wang CZ, Li XL, Sun S, Xie JT, Aung HH, Tong R, McEntee E, Yuan CS. Methylnaltrexone, a peripherally acting opioid receptor antagonist, enhances tumoricidal effects of 5-Fu on human carcinoma cells. Anticancer Res. 2009;29(8):2927–32 (PMID: 19661297).PubMed
48.
49.
51.
go back to reference Wang X, Zhang R, Wu T, Shi Y, Zhou X, Tang D, Yu W, So EC, Wu X, Pan Z, Tian J. Successive treatment with naltrexone induces epithelial-mesenchymal transition and facilitates the malignant biological behaviors of bladder cancer cells. Acta Biochim Biophys Sin (Shanghai). 2021;53(2):238–48. https://doi.org/10.1093/abbs/gmaa169 (PMID: 33410473).CrossRef Wang X, Zhang R, Wu T, Shi Y, Zhou X, Tang D, Yu W, So EC, Wu X, Pan Z, Tian J. Successive treatment with naltrexone induces epithelial-mesenchymal transition and facilitates the malignant biological behaviors of bladder cancer cells. Acta Biochim Biophys Sin (Shanghai). 2021;53(2):238–48. https://​doi.​org/​10.​1093/​abbs/​gmaa169 (PMID: 33410473).CrossRef
52.
go back to reference 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. 2016 Nov;27(11):2032–2038. https://doi.org/10.1093/annonc/mdw317. Epub 2016 Aug 29. Erratum in: Ann Oncol. 2018 Apr 1;29(4):1076. PMID: 27573565; PMCID: PMC6267944. This unplanned post hoc analysis of two randomized trials demonstrates that treatment with MNTX and, even more so, response to MNTX are associated with increased OS, which supports the preclinical hypothesis that MOR can play a role in cancer progression. 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. 2016 Nov;27(11):2032–2038. https://​doi.​org/​10.​1093/​annonc/​mdw317. Epub 2016 Aug 29. Erratum in: Ann Oncol. 2018 Apr 1;29(4):1076. PMID: 27573565; PMCID: PMC6267944. This unplanned post hoc analysis of two randomized trials demonstrates that treatment with MNTX and, even more so, response to MNTX are associated with increased OS, which supports the preclinical hypothesis that MOR can play a role in cancer progression.
53.
go back to reference Lissoni P, Malugani F, Bordin V, Conti A, Maestroni G, Tancini G. A new neuroimmunotherapeutic strategy of subcutaneous low-dose interleukin-2 plus the long-acting opioid antagonist naltrexone in metastatic cancer patients progressing on interleukin-2 alone. Neuro Endocrinol Lett. 2002;23(3):255–8 (PMID: 12080288).PubMed Lissoni P, Malugani F, Bordin V, Conti A, Maestroni G, Tancini G. A new neuroimmunotherapeutic strategy of subcutaneous low-dose interleukin-2 plus the long-acting opioid antagonist naltrexone in metastatic cancer patients progressing on interleukin-2 alone. Neuro Endocrinol Lett. 2002;23(3):255–8 (PMID: 12080288).PubMed
55.
go back to reference Khan A. Long-term remission of adenoid cystic tongue carcinoma with low dose naltrexone and vitamin D3-a case report. Oral Health Dent Manag. 2014;13(3):721–4 (PMID: 25284545).PubMed Khan A. Long-term remission of adenoid cystic tongue carcinoma with low dose naltrexone and vitamin D3-a case report. Oral Health Dent Manag. 2014;13(3):721–4 (PMID: 25284545).PubMed
60.
go back to reference Schwartz L, Buhler L, Icard P, Lincet H, Steyaert JM. Metabolic treatment of cancer: intermediate results of a prospective case series. Anticancer Res. 2014;34(2):973–80 (PMID: 24511042).PubMed Schwartz L, Buhler L, Icard P, Lincet H, Steyaert JM. Metabolic treatment of cancer: intermediate results of a prospective case series. Anticancer Res. 2014;34(2):973–80 (PMID: 24511042).PubMed
Metadata
Title
Antagonists of the Mu-Opioid Receptor in the Cancer Patient: Fact or Fiction?
Authors
Amparo Belltall
Guido Mazzinari
Oscar Diaz-Cambronero
Pilar Eroles
María Pilar Argente Navarro
Publication date
01-06-2022
Publisher
Springer US
Published in
Current Oncology Reports / Issue 10/2022
Print ISSN: 1523-3790
Electronic ISSN: 1534-6269
DOI
https://doi.org/10.1007/s11912-022-01295-z

Other articles of this Issue 10/2022

Current Oncology Reports 10/2022 Go to the issue

Genitourinary Cancers (DP Petrylak and JW Kim, Section Editors)

Integration of Liquid Biopsies in Clinical Management of Metastatic Prostate Cancer

Melanoma (DB Johnson, Section Editor)

Evolving Treatment Approaches to Mucosal Melanoma

Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine