Abstract

BackgroundThere is increasing evidence that opioid analgesics may interfere with tumour growth. It is currently thought that these effects are mediated by transactivation of receptor tyrosine kinase (RTK)-controlled ERK1/2 and Akt signalling. The growth of many breast cancer cells is dependent on hyperactive ErbB receptor networks and one of the most successful approaches in antineoplastic therapy during the last decade was the development of ErbB-targeted therapies. However, the response rates of single therapies are often poor and resistance mechanisms evolve rapidly. To date there is no information about the ability of opioid analgesics to interfere with the growth of ErbB-driven cancers.Methods and Principal FindingsHere we demonstrate that ErbB2 overexpressing BT474 human breast cancer cells carry fully functional endogenous µ-opioid receptors. Most interestingly, the acute opioid effects on basal and Heregulin-stimulated ERK1/2 and Akt phosphorylation changed considerably during chronic Morphine treatment. Investigation of the underlying mechanism by the use of protein kinase inhibitors and co-immunoprecipitation studies revealed that chronic Morphine treatment results in rearrangement of the ErbB signalling network leading to dissociation of ERK1/2 from Akt signalling and a switch from ErbB1/ErbB3 to ErbB1/ErbB2-dependent cell growth. In chronically Morphine-treated cells Heregulin-stimulated ERK1/2 signalling is redirected via a newly established PI3K- and metalloproteinase-dependent feedback loop. Together, these alterations result in apoptosis of BT474 cells. A similar switch in Heregulin-stimulated ERK1/2 signalling from an ErbB2-independent to an ErbB2-, PI3K- and metalloproteinase-dependent mechanism was also observed in κ-opioid receptor expressing SKBR3 human mammary adenocarcinoma cells.Conclusions and SignificanceThe present data demonstrate that the ErbB receptor network of human breast cancer cells represents a target for chronic Morphine treatment. Rearrangement of ErbB signalling by chronic Morphine may provide a promising strategy to enhance the sensitivity of breast cancer cells to ErbB-directed therapies and to prevent the development of escape mechanisms.

Highlights

  • Opioids are potent analgesics and widely used for anaesthetic pre-medication and management of cancer pain

  • The present data demonstrate that the ErbB receptor network of human breast cancer cells represents a target for chronic Morphine treatment

  • The results show that BT474 cells contain about 7.362 fmol of m-opioid receptors per mg of membrane protein (n = 5) and that chronic Morphine treatment does not affect receptor abundance (7.462 fmol per mg of membrane protein; n = 3)

Read more

Summary

Introduction

Opioids are potent analgesics and widely used for anaesthetic pre-medication and management of cancer pain. Opioid receptors may regulate the activity of a variety of mitogen-activated protein (MAP) kinases, including Extracellular Signal-Regulated Kinases 1 and 2 (ERK1/ 2), c-Jun N-terminal Kinase (JNK), p38, Signal Transducer and Activator of Transcription 5 (STAT5) and Protein Kinase B (PKB/Akt) [2,3]. Activation of these ‘‘non classical’’ opioid effector systems is mediated via transactivation of receptor tyrosine kinase (RTK)-associated ERK1/2 and Akt signalling pathways [4,5]. To date there is no information about the ability of opioid analgesics to interfere with the growth of ErbB-driven cancers

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call