Skip to main content
Log in

Naltrexone’s Impact on Cancer Progression and Mortality: A Systematic Review of Studies in Humans, Animal Models, and Cell Cultures

  • Review
  • Published:
Advances in Therapy Aims and scope Submit manuscript

Abstract

Background

Naltrexone (NTX) is an opioid antagonist traditionally used as a treatment for alcohol and opioid use disorders, but various studies have documented its involvement in cancer progression, exploring possible anticancer potential, when administered at high doses or as low dose naltrexone (LDN). Herein we present a systematic review of cancer-related outcomes from case reports, clinical trials, and retrospective and prospective studies conducted using cell cultures, animal models, and human subjects receiving NTX/LDN.

Methods

A systematic search of NTX in cancer therapy was conducted. Outcomes including tumor size and number, latency to tumor development, survival duration, progression of disease, and scan results were assessed in clinical and animal studies, and cell number was used as the outcome measure of culture studies.

Results

Several case reports demonstrate notable survival durations and metastatic resolutions in patients with late stage cancer when administered an average LDN dose of 3–5 mg/day. Animal and cell culture studies suggest an overarching principle of NTX involvement in cancer pharmacophysiology, suggesting that high doses and continuous administration can foster cancer progression, whereas low doses and intermittent treatment may hinder cell proliferation, impede tumorigenesis, and have potential anticancer efficacy.

Conclusion

This review emphasizes the value of potential future research on NTX in cancer therapy, and warrants need for a better understanding of underlying mechanisms. Future controlled studies with more robust sample sizes, particularly in humans, are needed to fully elucidate its potential in cancer therapy.

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

Similar content being viewed by others

References

  1. Brown N, Panksepp J. Low-dose naltrexone for disease prevention and quality of life. Med Hypotheses. 2009;72(3):333–7. https://doi.org/10.1016/j.mehy.2008.06.048.

    Article  CAS  PubMed  Google Scholar 

  2. McLaughlin PJ, Zagon IS. Duration of opioid receptor blockade determines biotherapeutic response. Biochem Pharmacol. 2015;97(3):236–46. https://doi.org/10.1016/j.bcp.2015.06.016.

    Article  CAS  PubMed  Google Scholar 

  3. Zagon IS, McLaughlin PJ. Duration of opiate receptor blockade determines tumorigenic response in mice with neuroblastoma: a role for endogenous opioid systems in cancer. Life Sci. 1984;35(4):409–16. https://doi.org/10.1016/0024-3205(84)90651-9.

    Article  CAS  PubMed  Google Scholar 

  4. Li Z, You Y, Griffin N, Feng J, Shan F. Low-dose naltrexone (LDN): a promising treatment in immune-related diseases and cancer therapy. Int Immunopharmacol. 2018;61:178–84. https://doi.org/10.1016/j.intimp.2018.05.020.

    Article  CAS  PubMed  Google Scholar 

  5. Aylsworth CF, Hodson CA, Meites J. Opiate antagonists can inhibit mammary tumor growth in rats. Proc Soc Exp Biol Med. 1979;161(1):18–20.

    Article  CAS  Google Scholar 

  6. Zagon IS, McLaughlin PJ. Naltrexone modulates tumor response in mice with neuroblastoma. Science. 1983;221(4611):671–3. https://doi.org/10.1126/science.6867737.

    Article  CAS  PubMed  Google Scholar 

  7. Zagon IS, McLaughlin PJ. Opioid antagonists inhibit the growth of metastatic murine neuroblastoma. Cancer Lett. 1983;21(1):89–94. https://doi.org/10.1016/0304-3835(83)90087-3.

    Article  CAS  PubMed  Google Scholar 

  8. McLaughlin PJ, Zagon IS. Modulation of human neuroblastoma transplanted into nude mice by endogenous opioid systems. Life Sci. 1987;41(12):1465–72. https://doi.org/10.1016/0024-3205(87)90711-9.

    Article  CAS  PubMed  Google Scholar 

  9. Horn PT, Mirkin BL. Lack of effect of the opioid antagonist, naltrexone, on the in situ growth of C-1300, N1E–115 and NS206 murine neuroblastoma tumor cell lines. Life Sci. 1989;45(26):2539–45. https://doi.org/10.1016/0024-3205(89)90237-3.

    Article  CAS  PubMed  Google Scholar 

  10. Donahue RN, McLaughlin PJ, Zagon IS. Low-dose naltrexone suppresses ovarian cancer and exhibits enhanced inhibition in combination with cisplatin. Exp Biol Med (Maywood). 2011;236(7):883–95. https://doi.org/10.1258/ebm.2011.011096.

    Article  CAS  Google Scholar 

  11. Zagon IS, Porterfield NK, McLaughlin PJ. Opioid growth factor - opioid growth factor receptor axis inhibits proliferation of triple negative breast cancer. Exp Biol Med (Maywood). 2013;238(6):589–99. https://doi.org/10.1177/1535370213489492.

    Article  CAS  Google Scholar 

  12. Lee YS, Wurster RD. Differential effects of methionine enkephalin on the growth of brain tumor cells. J Neurooncol. 1994;19(1):11–5. https://doi.org/10.1007/BF01051044.

    Article  CAS  PubMed  Google Scholar 

  13. Zagon IS, McLaughlin PJ. Opioid antagonist (naltrexone) stimulation of cell proliferation in human and animal neuroblastoma and human fibrosarcoma cells in culture. Neuroscience. 1990;37(1):223–6. https://doi.org/10.1016/0306-4522(90)90207-K.

    Article  CAS  PubMed  Google Scholar 

  14. Berkson BM, Rubin DM, Berkson AJ. Revisiting the ALA/N (alpha-lipoic acid/low-dose naltrexone) protocol for people with metastatic and nonmetastatic pancreatic cancer: a report of 3 new cases. Integr Cancer Ther. 2009;8(4):416–22. https://doi.org/10.1177/1534735409352082.

    Article  CAS  PubMed  Google Scholar 

  15. Berkson BM, Rubin DM, Berkson AJ. The long-term survival of a patient with pancreatic cancer with metastases to the liver after treatment with the intravenous alpha-lipoic acid/low-dose naltrexone protocol. Integr Cancer Ther. 2006;5(1):83–9. https://doi.org/10.1177/1534735405285901.

    Article  PubMed  Google Scholar 

  16. Berkson BM, Calvo RF. The long-term survival of a patient with stage iv renal cell carcinoma following an integrative treatment approach including the intravenous α-lipoic acid/low-dose naltrexone protocol. Integr Cancer Ther. 2018;17(3):986–93. https://doi.org/10.1177/1534735417747984.

    Article  PubMed  Google Scholar 

  17. 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.

    PubMed  Google Scholar 

  18. 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.

    PubMed  Google Scholar 

  19. Friedman JD, Dello Buono FA. Opioid antagonists in the treatment of opioid-induced constipation and pruritus. Ann Pharmacother. 2001;35(1):85–91. https://doi.org/10.1345/aph.10121.

    Article  CAS  PubMed  Google Scholar 

  20. Singleton PA, Moss J, Karp DD, Atkins JT, Janku F. The mu opioid receptor: a new target for cancer therapy? Cancer. 2015;121(16):2681–8. https://doi.org/10.1002/cncr.29460.

    Article  CAS  PubMed  Google Scholar 

  21. Long L. Routine piloting in systematic reviews—a modified approach? Syst Rev. 2014;3(1):77. https://doi.org/10.1186/2046-4053-3-77.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Downs SH, Black N. The feasibility of creating a checklist for the assessment of the methodological quality both of randomised and non-randomised studies of health care interventions. J Epidemiol Community Health. 1998;52(6):377–84. https://doi.org/10.1136/jech.52.6.377.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lissoni P, Meregalli S, Fossati V, et al. Radioendocrine therapy of brain tumors with the long acting opioid antagonist naltrexone in association with radiotherapy. Tumori. 1993;79(3):198–201.

    Article  CAS  Google Scholar 

  24. Berkson BM, Rubin DM, Berkson AJ. Reversal of signs and symptoms of a B cell lymphoma in a patient using only low-dose naltrexone. Integr Cancer Ther. 2007;6(3):293–6. https://doi.org/10.1177/1534735407306358.

    Article  CAS  PubMed  Google Scholar 

  25. Miskoff JA, Chaudhri M. Low dose naltrexone and lung cancer: a case report and discussion. Cureus. 2018;10(7):e2924.

    PubMed  PubMed Central  Google Scholar 

  26. 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.

    CAS  PubMed  Google Scholar 

  27. Lissoni P, Malugani F, Malysheva O, et al. Neuroimmunotherapy of untreatable metastatic solid tumors with subcutaneous low-dose interleukin-2, melatonin and naltrexone: modulation of interleukin-2-induced antitumor immunity by blocking the opioid system. Neuro Endocrinol Lett. 2002;23(4):341–4.

    CAS  PubMed  Google Scholar 

  28. McLaughlin PJ, Stucki JK, Zagon IS. Modulation of the opioid growth factor ([Met(5)]-enkephalin)-opioid growth factor receptor axis: novel therapies for squamous cell carcinoma of the head and neck. Head Neck. 2012;34(4):513–9. https://doi.org/10.1002/hed.21759.

    Article  PubMed  Google Scholar 

  29. McLaughlin PJ, Levin RJ, Zagon IS. Opioid growth factor (OGF) inhibits the progression of human squamous cell carcinoma of the head and neck transplanted into nude mice. Cancer Lett. 2003;199(2):209–17.

    Article  CAS  Google Scholar 

  30. Koo KL, Tejwani GA, Abou-Issa H. Relative efficacy of the opioid antagonist, naltrexone, on the initiation and promotion phases of rat mammary carcinogenesis. Anticancer Res. 1996;16(4A):1893–8. http://ar.iiarjournals.org/. Accessed 25 Jan 2018.

  31. Donahue RN, McLaughlin PJ, Zagon IS. The opioid growth factor (OGF) and low dose naltrexone (LDN) suppress human ovarian cancer progression in mice. Gynecol Oncol. 2011;122(2):382–8. https://doi.org/10.1016/j.ygyno.2011.04.009.

    Article  CAS  PubMed  Google Scholar 

  32. Abou-Issa H, Tejwani GA. Antitumor activity of naltrexone and correlation with steroid hormone receptors. Biochem Biophys Res Commun. 1991;175(2):625–30. https://doi.org/10.1016/0006-291X(91)91611-F.

    Article  CAS  PubMed  Google Scholar 

  33. Aboalsoud A, El-Ghaiesh SH, Abd Elmonem FF, Salem ML, Abdel Rahman MN. The effect of low-dose naltrexone on solid Ehrlich carcinoma in mice: the role of OGFr, BCL2, and immune response. Int Immunopharmacol. 2020;78:106068.

    Article  CAS  Google Scholar 

  34. Hytrek SD, McLaughlin PJ, Lang CM, Zagon IS. Inhibition of human colon cancer by intermittent opioid receptor blockade with naltrexone. Cancer Lett. 1996;101(2):159–64. https://doi.org/10.1016/0304-3835(96)04119-5.

    Article  CAS  PubMed  Google Scholar 

  35. Tejwani GA, Gudehithlu KP, Hanissian SH, Gienapp IE, Whitacre CC, Malarkey WB. Facilitation of dimethylbenz[a]anthracene-induced rat mammary tumorigenesis by restraint stress: role of beta-endorphin, prolactin and naltrexone. Carcinogenesis. 1991;12(4):637–41. https://doi.org/10.1093/carcin/12.4.637.

    Article  CAS  PubMed  Google Scholar 

  36. Zagon IS, McLaughlin PJ. Modulation of murine neuroblastoma in nude mice by opioid antagonists. J Natl Cancer Inst. 1987;78(1):141–7. https://doi.org/10.1093/jnci/78.1.141.

    Article  CAS  PubMed  Google Scholar 

  37. Ebrahimpour S, Tabari MA, Youssefi MR, Aghajanzadeh H, Behzadi MY. Synergistic effect of aged garlic extract and naltrexone on improving immune responses to experimentally induced fibrosarcoma tumor in BALB/c mice. Pharmacognosy Res. 2013;5(3):189–94. https://doi.org/10.4103/0974-8490.112426.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lewis JW, Shavit Y, Terman GW, Nelson LR, Gale RP, Liebeskind JC. Apparent involvement of opioid peptides in stress-induced enhancement of tumor growth. Peptides. 1983;4(5):635–8. https://doi.org/10.1016/0196-9781(83)90010-4.

    Article  CAS  PubMed  Google Scholar 

  39. Machado MC, da Costa-Neto JM, Portela RD, et al. The effect of naltrexone as a carboplatin chemotherapy-associated drug on the immune response, quality of life and survival of dogs with mammary carcinoma. PLoS One. 2018;13(10):e0204830.

    Article  Google Scholar 

  40. Donahue RN, McLaughlin PJ, Zagon IS. Low-dose naltrexone targets the opioid growth factor-opioid growth factor receptor pathway to inhibit cell proliferation: mechanistic evidence from a tissue culture model. Exp Biol Med (Maywood). 2011;236(9):1036–50. https://doi.org/10.1258/ebm.2011.011121.

    Article  CAS  Google Scholar 

  41. Zagon IS, Donahue RN, McLaughlin PJ. Opioid growth factor-opioid growth factor receptor axis is a physiological determinant of cell proliferation in diverse human cancers. Am J Physiol Regul Integr Comp Physiol. 2009;297(4):R1154–61. https://doi.org/10.1152/ajpregu.00414.2009.

    Article  CAS  PubMed  Google Scholar 

  42. Zagon IS. Endogenous opioid systems and neural cancer: transmission and scanning electron microscopic studies of murine neuroblastoma in tissue culture. Brain Res Bull. 1988;21(5):777–84. https://doi.org/10.1016/0361-9230(88)90046-9.

    Article  CAS  PubMed  Google Scholar 

  43. Zagon IS, Hytrek SD, McLaughlin PJ. Opioid growth factor tonically inhibits human colon cancer cell proliferation in tissue culture. Am J Physiol. 1996;271(3 Pt 2):R511–8.

    CAS  PubMed  Google Scholar 

  44. Donahue RN, McLaughlin PJ, Zagon IS. Cell proliferation of human ovarian cancer is regulated by the opioid growth factor-opioid growth factor receptor axis. Am J Physiol Regul Integr Comp Physiol. 2009;296(6):R1716–25. https://doi.org/10.1152/ajpregu.00075.2009.

    Article  CAS  PubMed  Google Scholar 

  45. Zagon IS, Roesener CD, Verderame MF, Ohlsson-Wilhelm BM, Levin RJ, McLaughlin PJ. Opioid growth factor regulates the cell cycle of human neoplasias. Int J Oncol. 2000;17(5):1053–61. https://doi.org/10.3892/ijo.17.5.1053.

    Article  CAS  PubMed  Google Scholar 

  46. McLaughlin PJ, Levin RJ, Zagon IS. Regulation of human head and neck squamous cell carcinoma growth in tissue culture by opioid growth factor. Int J Oncol. 1999;14(5):991–8. https://doi.org/10.3892/ijo.14.5.991.

    Article  CAS  PubMed  Google Scholar 

  47. McLaughlin PJ, Zagon IS, Park SS, Conway A, Donahue RN, Goldenberg D. Growth inhibition of thyroid follicular cell-derived cancers by the opioid growth factor (OGF)—opioid growth factor receptor (OGFr) axis. BMC Cancer. 2009;9:369. https://doi.org/10.1186/1471-2407-9-369.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Donahue RN, McLaughlin PJ, Zagon IS. Under-expression of the opioid growth factor receptor promotes progression of human ovarian cancer. Exp Biol Med (Maywood). 2012;237(2):167–77. https://doi.org/10.1258/ebm.2011.011321.

    Article  CAS  Google Scholar 

  49. Ma M, Wang X, Liu N, Shan F, Feng Y. Low-dose naltrexone inhibits colorectal cancer progression and promotes apoptosis by increasing M1-type macrophages and activating the Bax/Bcl-2/caspase-3/PARP pathway. Int immunopharmacology. 2020;83:106388.

    Article  CAS  Google Scholar 

  50. Liu WM, Scott KA, Dennis JL, Kaminska E, Levett AJ, Dalgleish AG. Naltrexone at low doses upregulates a unique gene expression not seen with normal doses: implications for its use in cancer therapy. Int J Oncol. 2016;49(2):793–802. https://doi.org/10.3892/ijo.2016.3567.

    Article  CAS  PubMed  Google Scholar 

  51. Zagon IS, Donahue R, McLaughlin PJ. Targeting the opioid growth factor: opioid growth factor receptor axis for treatment of human ovarian cancer. Exp Biol Med (Maywood). 2013;238(5):579–87. https://doi.org/10.1177/1535370213488483.

    Article  CAS  Google Scholar 

  52. Zagon IS, McLaughlin PJ. Gene-peptide relationships in the developing rat brain: the response of preproenkephalin mRNA and [Met5]-enkephalin to acute opioid antagonist (naltrexone) exposure. Brain Res Mol Brain Res. 1995;33(1):111–20. https://doi.org/10.1016/0169-328X(95)00119-D.

    Article  CAS  PubMed  Google Scholar 

  53. Zagon IS, McLaughlin PJ. Opioid growth factor and the treatment of human pancreatic cancer: a review. World J Gastroenterol. 2014;20(9):2218–23. https://doi.org/10.3748/wjg.v20.i9.2218.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Chia BK, Tey HL. Metastatic hepatocellular carcinoma with paraneoplastic itch: effective treatment with naltrexone. J Drugs Dermatol. 2014;13(12):1440. http://jddonline.com/articles/dermatology/S1545961614P1440X/1. Accessed 25 Jan 2018.

  55. Valentine AD, Meyers CA, Talpaz M. Treatment of neurotoxic side effects of interferon-alpha with naltrexone. Cancer Invest. 1995;13(6):561–6. https://doi.org/10.3109/07357909509024923.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Funding

No funding or sponsorship was received for this study or publication of this article.

Authorship

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Authorship Contributions

Karina Liubchenko: data curation, investigation, project administration, visualization, writing – original draft. Kevin Kordbacheh: investigation, writing – original draft, visualization. Nika Khajehdehi: writing – original draft. Tanja Visnjevac: writing – review and editing. Frederick Ma: writing – review and editing. James Khan: writing – review and editing. Myles Storey: data curation, writing – review and editing. Alaa-Abd Elsayed: writing – review and editing. Ognjen Visnjevac: conceptualization, data curation, investigation, project administration, visualization, supervision, writing – review and editing

Disclosures

Dr. Abd-Elsayed is a consultant for Medtronic and Avanos. The remaining authors have nothing to disclose (Karina Liubchenko, Kevin Kordbacheh, Nika Khajehdehi, Tanja Visnjevac, Frederick Ma, James S. Khan, Myles Storey and Ognjen Visnjevac).

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not contain any new studies with human participants or animals performed by any of the authors.

Data Availability

All data generated or analyzed during this study are included in this published article.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alaa Abd-Elsayed.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liubchenko, K., Kordbacheh, K., Khajehdehi, N. et al. Naltrexone’s Impact on Cancer Progression and Mortality: A Systematic Review of Studies in Humans, Animal Models, and Cell Cultures. Adv Ther 38, 904–924 (2021). https://doi.org/10.1007/s12325-020-01591-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12325-020-01591-9

Keywords

Navigation