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.

  • Opinion
  • Published:

Sympathetic nervous system regulation of the tumour microenvironment

Abstract

The peripheral autonomic nervous system (ANS) is known to regulate gene expression in primary tumours and their surrounding microenvironment. Activation of the sympathetic division of the ANS in particular modulates gene expression programmes that promote metastasis of solid tumours by stimulating macrophage infiltration, inflammation, angiogenesis, epithelial–mesenchymal transition and tumour invasion, and by inhibiting cellular immune responses and programmed cell death. Haematological cancers are modulated by sympathetic nervous system (SNS) regulation of stem cell biology and haematopoietic differentiation programmes. In addition to identifying a molecular basis for physiologic stress effects on cancer, these findings have also identified new pharmacological strategies to inhibit cancer progression in vivo.

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

Figure 1: Sympathetic nervous system regulation of the tumour microenvironment.
Figure 2: Molecular mechanisms for sympathetic nervous system regulation of tumour progression.

Similar content being viewed by others

References

  1. Weiner, H. Perturbing the Organism: The Biology of Stressful Experience (Univ. of Chicago Press, 1992).

    Google Scholar 

  2. Sapolsky, R. M. Why Zebras Don't Get Ulcers: A Guide To Stress, Stress-Related Diseases, And Coping (Freeman, 1994).

    Google Scholar 

  3. Sherwood, L. Human Physiology: From Cells to Systems (Cengage Learning, 2015).

    Google Scholar 

  4. Katayama, Y. et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell 124, 407–421 (2006).

    Article  CAS  PubMed  Google Scholar 

  5. Sloan, E. K. et al. Social stress enhances sympathetic innervation of primate lymph nodes: mechanisms and implications for viral pathogenesis. J. Neurosci. 27, 8857–8865 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lutgendorf, S. K. et al. Depression, social support, and β-adrenergic transcription control in human ovarian cancer. Brain Behav. Immun. 23, 176–183 (2009).

    Article  CAS  PubMed  Google Scholar 

  7. Powell, N. D. et al. Social stress up-regulates inflammatory gene expression in the leukocyte transcriptome via β-adrenergic induction of myelopoiesis. Proc. Natl Acad. Sci. USA 110, 16574–16579 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Scheiermann, C., Kunisaki, Y. & Frenette, P. S. Circadian control of the immune system. Nat. Rev. Immunol. 13, 190–198 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cole, S. W. Social regulation of human gene expression: mechanisms and implications for public health. Am. J. Publ. Health 103 (Suppl. 1), S84–S92 (2013).

    Article  Google Scholar 

  10. Cole, S. W. Human social genomics. PLoS Genet. 10, e1004601 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hanoun, M., Maryanovich, M., Arnal-Estape, A. & Frenette, P. S. Neural regulation of hematopoiesis, inflammation, and cancer. Neuron 86, 360–373 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Irwin, M. R. & Cole, S. W. Reciprocal regulation of the neural and innate immune systems. Nat. Rev. Immunol. 11, 625–632 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Antoni, M. H. et al. The influence of bio-behavioural factors on tumour biology: pathways and mechanisms. Nat. Rev. Cancer. 6, 240–248 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Cole, S. W. & Sood, A. K. Molecular pathways: β-adrenergic signaling in cancer. Clin. Cancer Res. 18, 1201–1206 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Armaiz-Pena, G. N., Cole, S. W., Lutgendorf, S. K. & Sood, A. K. Neuroendocrine influences on cancer progression. Brain Behav. Immun. 30, S19–S25 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Cole, S. W. Nervous system regulation of the cancer genome. Brain Behav. Immun. 30 (Suppl.), S10–S18 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Powe, D. G. & Entschladen, F. Targeted therapies: using β-blockers to inhibit breast cancer progression. Nat. Rev. Clin. Oncol. 8, 511–512 (2011).

    Article  PubMed  Google Scholar 

  18. Richter, S. D. et al. Time kinetics of the endocrine response to acute psychological stress. J. Clin. Endocrinol. Metab. 81, 1956–1960 (1996).

    CAS  PubMed  Google Scholar 

  19. Schommer, N. C., Hellhammer, D. H. & Kirschbaum, C. Dissociation between reactivity of the hypothalamus–pituitary–adrenal axis and the sympathetic–adrenal–medullary system to repeated psychosocial stress. Psychosom. Med. 65, 450–460 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Wingenfeld, K., Whooley, M. A., Neylan, T. C., Otte, C. & Cohen, B. E. Effect of current and lifetime posttraumatic stress disorder on 24-h urinary catecholamines and cortisol: results from the Mind Your Heart Study. Psychoneuroendocrinology 52, 83–91 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Sloan, E. K., Capitanio, J. P., Tarara, R. P. & Cole, S. W. Social temperament and lymph node innervation. Brain Behav. Immun. 22, 717–726 (2008).

    Article  PubMed  Google Scholar 

  22. Schofl, C., Becker, C., Prank, K., von zur Muhlen, A. & Brabant, G. Twenty-four-hour rhythms of plasma catecholamines and their relation to cardiovascular parameters in healthy young men. Eur. J. Endocrinol. 137, 675–683 (1997).

    Article  CAS  PubMed  Google Scholar 

  23. Dimitrov, S. et al. Cortisol and epinephrine control opposing circadian rhythms in T cell subsets. Blood 113, 5134–5143 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Eng, J. W. et al. Housing temperature-induced stress drives therapeutic resistance in murine tumour models through β2-adrenergic receptor activation. Nat. Commun. 6, 6426 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Mendez-Ferrer, S., Lucas, D., Battista, M. & Frenette, P. S. Haematopoietic stem cell release is regulated by circadian oscillations. Nature 452, 442–447 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. Dutta, P. et al. Myocardial infarction accelerates atherosclerosis. Nature 487, 325–329 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Heidt, T. et al. Chronic variable stress activates hematopoietic stem cells. Nat. Med. 20, 754–758 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Nakai, A., Hayano, Y., Furuta, F., Noda, M. & Suzuki, K. Control of lymphocyte egress from lymph nodes through β2-adrenergic receptors. J. Exp. Med. 211, 2583–2598 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sloan, E. K., Capitanio, J. P. & Cole, S. W. Stress-induced remodeling of lymphoid innervation. Brain Behav. Immun. 22, 15–21 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Elenkov, I. J., Wilder, R. L., Chrousos, G. P. & Vizi, E. S. The sympathetic nerve — an integrative interface between two supersystems: the brain and the immune system. Pharmacol. Rev. 52, 595–638 (2000).

    CAS  PubMed  Google Scholar 

  31. Kohm, A. P. & Sanders, V. M. Norepinephrine and β 2-adrenergic receptor stimulation regulate CD4+ T and B lymphocyte function in vitro and in vivo. Pharmacol. Rev. 53, 487–525 (2001).

    CAS  PubMed  Google Scholar 

  32. Cole, S. et al. Computational identification of gene-social environment interaction at the human IL6 locus. Proc. Natl Acad. Sci. USA 107, 5681–5686 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Hori, Y. et al. Naftopidil, a selective α1-adrenoceptor antagonist, suppresses human prostate tumor growth by altering interactions between tumor cells and stroma. Cancer Prev. Res. 4, 87–96 (2011).

    Article  CAS  Google Scholar 

  34. Calvani, M. et al. Norepinephrine promotes tumor microenvironment reactivity through β3-adrenoreceptors during melanoma progression. Oncotarget 6, 4615–4632 (2015).

    Article  PubMed  Google Scholar 

  35. Thaker, P. H. et al. Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma. Nat. Med. 12, 939–944 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Dal Monte, M. et al. Functional involvement of β3-adrenergic receptors in melanoma growth and vascularization. J. Mol. Med. 91, 1407–1419 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Sterling, P. in Allostasis, Homeostasis, and the Costs of Physiological Adaptation (ed. Schulkin, J.) 17–64 (Cambridge Univ. Press, 2004).

    Book  Google Scholar 

  38. Chida, Y., Hamer, M., Wardle, J. & Steptoe, A. Do stress-related psychosocial factors contribute to cancer incidence and survival? Nat. Clin. Pract. Oncol. 5, 466–475 (2008).

    Article  PubMed  Google Scholar 

  39. Powe, D. G. et al. β-blocker drug therapy reduces secondary cancer formation in breast cancer and improves cancer specific survival. Oncotarget 1, 628–638 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Barron, T. I., Connolly, R. M., Sharp, L., Bennett, K. & Visvanathan, K. β blockers and breast cancer mortality: a population-based study. J. Clin. Oncol. 29, 2635–2644 (2011).

    Article  CAS  PubMed  Google Scholar 

  41. Melhem-Bertrandt, A. et al. β-blocker use is associated with improved relapse-free survival in patients with triple-negative breast cancer. J. Clin. Oncol. 29, 2645–2652 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. De Giorgi, V. et al. Treatment with β-blockers and reduced disease progression in patients with thick melanoma. Arch. Intern. Med. 171, 779–781 (2011).

    PubMed  Google Scholar 

  43. Lemeshow, S. et al. β-blockers and survival among Danish patients with malignant melanoma: a population-based cohort study. Cancer Epidemiol. Biomarkers Prev. 20, 2273–2279 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Aydiner, A., Ciftci, R., Karabulut, S. & Kilic, L. Does β-blocker therapy improve the survival of patients with metastatic non-small cell lung cancer? Asian Pac. J. Cancer Prev. 14, 6109–6114 (2013).

    Article  PubMed  Google Scholar 

  45. Botteri, E. et al. Therapeutic effect of β-blockers in triple-negative breast cancer postmenopausal women. Breast Cancer Res. Treat. 140, 567–575 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. De Giorgi, V. et al. Effect of β-blockers and other antihypertensive drugs on the risk of melanoma recurrence and death. Mayo Clin. Proc. 88, 1196–1203 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Grytli, H. H., Fagerland, M. W., Fossa, S. D., Tasken, K. A. & Haheim, L. L. Use of β-blockers is associated with prostate cancer-specific survival in prostate cancer patients on androgen deprivation therapy. Prostate 73, 250–260 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Grytli, H. H., Fagerland, M. W., Fossa, S. D. & Tasken, K. A. Association between use of β-blockers and prostate cancer-specific survival: a cohort study of 3561 prostate cancer patients with high-risk or metastatic disease. Eur. Urol. 65, 635–641 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Sloan, E. K. et al. The sympathetic nervous system induces a metastatic switch in primary breast cancer. Cancer Res. 70, 7042–7052 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Madden, K. S., Szpunar, M. J. & Brown, E. B. β-adrenergic receptors (β-AR) regulate VEGF and IL-6 production by divergent pathways in high β-AR-expressing breast cancer cell lines. Breast Cancer Res. Treat. 130, 747–758 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Palm, D. et al. The norepinephrine-driven metastasis development of PC-3 human prostate cancer cells in BALB/c nude mice is inhibited by β-blockers. Int. J. Cancer. 118, 2744–2749 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. Hassan, S. et al. Behavioral stress accelerates prostate cancer development in mice. J. Clin. Invest. 123, 874–886 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Pasquier, E. et al. β-blockers increase response to chemotherapy via direct antitumour and anti-angiogenic mechanisms in neuroblastoma. Br. J. Cancer 108, 2485–2494 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wolter, J. K. et al. Anti-tumor activity of the β-adrenergic receptor antagonist propranolol in neuroblastoma. Oncotarget 5, 161–172 (2014).

    Article  PubMed  Google Scholar 

  55. Hasegawa, H. & Saiki, I. Psychosocial stress augments tumor development through β-adrenergic activation in mice. Jpn J. Cancer Res. 93, 729–735 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Goldfarb, Y. et al. Improving postoperative immune status and resistance to cancer metastasis: a combined perioperative approach of immunostimulation and prevention of excessive surgical stress responses. Ann. Surg. 253, 798–810 (2011).

    Article  PubMed  Google Scholar 

  57. Kim-Fuchs, C. et al. Chronic stress accelerates pancreatic cancer growth and invasion: a critical role for β-adrenergic signaling in the pancreatic microenvironment. Brain Behav. Immun. 40, 40–47 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Lamkin, D. M. et al. Chronic stress enhances progression of acute lymphoblastic leukemia via β-adrenergic signaling. Brain Behav. Immun. 26, 635–641 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Inbar, S. et al. Do stress responses promote leukemia progression? An animal study suggesting a role for epinephrine and prostaglandin-E2 through reduced NK activity. PLoS ONE 6, e19246 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hara, M. R. et al. A stress response pathway regulates DNA damage through β2-adrenoreceptors and β-arrestin-1. Nature 477, 349–353 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Hara, M. R., Sachs, B. D., Caron, M. G. & Lefkowitz, R. J. Pharmacological blockade of a β2AR-β-arrestin-1 signaling cascade prevents the accumulation of DNA damage in a behavioral stress model. Cell Cycle 12, 219–224 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Reeder, A. et al. Stress hormones reduce the efficacy of paclitaxel in triple negative breast cancer through induction of DNA damage. Br. J. Cancer 112, 1461–1470 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Armaiz-Pena, G. N. et al. Src activation by β-adrenoreceptors is a key switch for tumour metastasis. Nat. Commun. 4, 1403 (2013).

    Article  CAS  PubMed  Google Scholar 

  64. Shi, M. et al. The β2-adrenergic receptor and Her2 comprise a positive feedback loop in human breast cancer cells. Breast Cancer Res. Treat. 125, 351–362 (2011).

    Article  CAS  PubMed  Google Scholar 

  65. Gu, L., Lau, S. K., Loera, S., Somlo, G. & Kane, S. E. Protein kinase A activation confers resistance to trastuzumab in human breast cancer cell lines. Clin. Cancer Res. 15, 7196–7206 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Chang, M. et al. β-adrenoreceptors reactivate Kaposi's sarcoma-associated herpesvirus lytic replication via PKA-dependent control of viral RTA. J. Virol. 79, 13538–13547 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. zur Hausen, H. Infections Causing Human Cancer, (Wiley-VCH, 2008).

    Google Scholar 

  68. Nilsson, M. B. et al. Stress hormones regulate interleukin-6 expression by human ovarian carcinoma cells through a Src-dependent mechanism. J. Biol. Chem. 282, 29919–29926 (2007).

    Article  CAS  PubMed  Google Scholar 

  69. Shahzad, M. M. et al. Stress effects on FosB- and interleukin-8 (IL8)-driven ovarian cancer growth and metastasis. J. Biol. Chem. 285, 35462–35470 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Yang, R., Lin, Q., Gao, H. B. & Zhang, P. Stress-related hormone norepinephrine induces interleukin-6 expression in GES-1 cells. Braz. J. Med. Biol. Res. 47, 101–109 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Cakir, Y., Plummer, H. K., 3rd, Tithof, P. K. & Schuller, H. M. β-adrenergic and arachidonic acid-mediated growth regulation of human breast cancer cell lines. Int. J. Oncol. 21, 153–157 (2002).

    CAS  PubMed  Google Scholar 

  72. Armaiz-Pena, G. N. et al. Adrenergic regulation of monocyte chemotactic protein 1 leads to enhanced macrophage recruitment and ovarian carcinoma growth. Oncotarget 6, 4266–4273 (2015).

    Article  PubMed  Google Scholar 

  73. Collado-Hidalgo, A., Sung, C. & Cole, S. Adrenergic inhibition of innate anti-viral response: PKA blockade of type I interferon gene transcription mediates catecholamine support for HIV-1 replication. Brain Behav. Immun. 20, 552–563 (2006).

    Article  CAS  PubMed  Google Scholar 

  74. Cole, S. W., Korin, Y. D., Fahey, J. L. & Zack, J. A. Norepinephrine accelerates HIV replication via protein kinase A-dependent effects on cytokine production. J. Immunol. 161, 610–616 (1998).

    CAS  PubMed  Google Scholar 

  75. Glasner, A. et al. Improving survival rates in two models of spontaneous postoperative metastasis in mice by combined administration of a β-adrenergic antagonist and a cyclooxygenase-2 inhibitor. J. Immunol. 184, 2449–2457 (2010).

    Article  CAS  PubMed  Google Scholar 

  76. Lee, J. W. et al. Surgical stress promotes tumor growth in ovarian carcinoma. Clin. Cancer Res. 15, 2695–2702 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Magnon, C. et al. Autonomic nerve development contributes to prostate cancer progression. Science 341, 1236361 (2013).

    Article  PubMed  Google Scholar 

  78. Bruzzone, A. et al. α2-adrenoceptors enhance cell proliferation and mammary tumor growth acting through both the stroma and the tumor cells. Curr. Cancer Drug Targets 11, 763–774 (2011).

    Article  CAS  PubMed  Google Scholar 

  79. Flint, M. S. et al. Chronic exposure to stress hormones promotes transformation and tumorigenicity of 3T3 mouse fibroblasts. Stress 16, 114–121 (2013).

    Article  CAS  PubMed  Google Scholar 

  80. Cao, L. et al. Environmental and genetic activation of a brain–adipocyte BDNF/leptin axis causes cancer remission and inhibition. Cell 142, 52–64 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Cao, L. & During, M. J. What is the brain–cancer connection? Annu. Rev. Neurosci. 35, 331–345 (2012).

    Article  CAS  PubMed  Google Scholar 

  82. Hanoun, M. et al. Acute myelogenous leukemia-induced sympathetic neuropathy promotes malignancy in an altered hematopoietic stem cell niche. Cell Stem Cell 15, 365–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Lang, K. et al. Induction of a metastatogenic tumor cell type by neurotransmitters and its pharmacological inhibition by established drugs. Int. J. Cancer 112, 231–238 (2004).

    Article  CAS  PubMed  Google Scholar 

  84. Drell, T. L. t. et al. Effects of neurotransmitters on the chemokinesis and chemotaxis of MDA-MB-468 human breast carcinoma cells. Breast Cancer Res. Treat. 80, 63–70 (2003).

    Article  CAS  PubMed  Google Scholar 

  85. Landen, C. N. Jr et al. Neuroendocrine modulation of signal transducer and activator of transcription-3 in ovarian cancer. Cancer Res. 67, 10389–10396 (2007).

    Article  CAS  PubMed  Google Scholar 

  86. Sood, A. K. et al. Stress hormone-mediated invasion of ovarian cancer cells. Clin. Cancer Res. 12, 369–375 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Yang, E. V. et al. Norepinephrine up-regulates the expression of vascular endothelial growth factor, matrix metalloproteinase (MMP)-2, and MMP-9 in nasopharyngeal carcinoma tumor cells. Cancer Res. 66, 10357–10364 (2006).

    Article  CAS  PubMed  Google Scholar 

  88. Chakroborty, D., Sarkar, C., Basu, B., Dasgupta, P. S. & Basu, S. Catecholamines regulate tumor angiogenesis. Cancer Res. 69, 3727–3730 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Yang, E. V. et al. Norepinephrine upregulates VEGF, IL-8, and IL-6 expression in human melanoma tumor cell lines: implications for stress-related enhancement of tumor progression. Brain Behav. Immun. 23, 267–275 (2009).

    Article  CAS  PubMed  Google Scholar 

  90. Moretti, S. et al. β-adrenoceptors are upregulated in human melanoma and their activation releases pro-tumorigenic cytokines and metalloproteases in melanoma cell lines. Lab Invest. 93, 279–290 (2013).

    Article  CAS  PubMed  Google Scholar 

  91. Liu, J. et al. The effect of chronic stress on anti-angiogenesis of sunitinib in colorectal cancer models. Psychoneuroendocrinology 52, 130–142 (2015).

    Article  CAS  PubMed  Google Scholar 

  92. Sood, A. K. et al. Adrenergic modulation of focal adhesion kinase protects human ovarian cancer cells from anoikis. J. Clin. Invest. 120, 1515–1523 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Sastry, K. S. et al. Epinephrine protects cancer cells from apoptosis via activation of cAMP-dependent protein kinase and BAD phosphorylation. J. Biol. Chem. 282, 14094–14100 (2007).

    Article  CAS  PubMed  Google Scholar 

  94. Deng, G. H. et al. Exogenous norepinephrine attenuates the efficacy of sunitinib in a mouse cancer model. J. Exp. Clin. Cancer Res. 33, 21 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Dar, A. et al. Rapid mobilization of hematopoietic progenitors by AMD3100 and catecholamines is mediated by CXCR4-dependent SDF-1 release from bone marrow stromal cells. Leukemia 25, 1286–1296 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Lucas, D. et al. Chemotherapy-induced bone marrow nerve injury impairs hematopoietic regeneration. Nat. Med. 19, 695–703 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Lutgendorf, S. K. et al. Social isolation is associated with elevated tumor norepinephrine in ovarian carcinoma patients. Brain Behav. Immun. 25, 250–255 (2011).

    Article  CAS  PubMed  Google Scholar 

  98. Ayala, G. E. et al. Cancer-related axonogenesis and neurogenesis in prostate cancer. Clin. Cancer Res. 14, 7593–7603 (2008).

    Article  CAS  PubMed  Google Scholar 

  99. Voss, M. J. & Entschladen, F. Tumor interactions with soluble factors and the nervous system. Cell Commun. Signal 8, 21 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  100. Guo, K. et al. Interaction of the sympathetic nerve with pancreatic cancer cells promotes perineural invasion through the activation of STAT3 signaling. Mol. Cancer Ther. 12, 264–273 (2013).

    Article  CAS  PubMed  Google Scholar 

  101. Xu, Q. et al. Stromal-derived factor-1α/CXCL12–CXCR4 chemotactic pathway promotes perineural invasion in pancreatic cancer. Oncotarget 6, 4717–4732 (2015).

    PubMed  Google Scholar 

  102. Flierl, M. A. et al. Phagocyte-derived catecholamines enhance acute inflammatory injury. Nature. 449, 721–725 (2007).

    Article  CAS  PubMed  Google Scholar 

  103. Campbell, J. P. et al. Stimulation of host bone marrow stromal cells by sympathetic nerves promotes breast cancer bone metastasis in mice. PLoS Biol. 10, e1001363 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Lu, H. et al. Impact of β-blockers on prostate cancer mortality: a meta-analysis of 16,825 patients. Onco Targets Ther. 8, 985–990 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Wang, H. M. et al. Improved survival outcomes with the incidental use of β-blockers among patients with non-small-cell lung cancer treated with definitive radiation therapy. Ann. Oncol. 24, 1312–1319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Diaz, E. S., Karlan, B. Y. & Li, A. J. Impact of β blockers on epithelial ovarian cancer survival. Gynecol. Oncol. 127, 375–378 (2012).

    Article  CAS  PubMed  Google Scholar 

  107. Watkins, J. L. et al. Clinical impact of selective and non-selective β-blockers on survival in ovarian cancer patients. Cancer (in the press).

  108. Schuller, H. M., Porter, B. & Riechert, A. β-adrenergic modulation of NNK-induced lung carcinogenesis in hamsters. J. Cancer Res. Clin. Oncol. 126, 624–630 (2000).

    Article  CAS  PubMed  Google Scholar 

  109. Pasquier, E. et al. Propranolol potentiates the anti-angiogenic effects and anti-tumor efficacy of chemotherapy agents: implication in breast cancer treatment. Oncotarget 2, 797–809 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  110. Lin, Q. et al. Effect of chronic restraint stress on human colorectal carcinoma growth in mice. PLoS ONE 8, e61435 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Ganz, P. A. & Cole, S. W. Expanding our therapeutic options: β blockers for breast cancer? J. Clin. Oncol. 29, 2612–2616 (2011).

    Article  PubMed  Google Scholar 

  112. Neeman, E., Zmora, O. & Ben-Eliyahu, S. A new approach to reducing postsurgical cancer recurrence: perioperative targeting of catecholamines and prostaglandins. Clin. Cancer Res. 18, 4895–4902 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Horowitz, M., Neeman, E., Sharon, E. & Ben-Eliyahu, S. Exploiting the critical perioperative period to improve long-term cancer outcomes. Nat. Rev. Clin. Oncol. 12, 213–226 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Boucek, R. J. Jr., Kirsh, A. L., Majesky, M. W. & Perkins, J. A. Propranolol responsiveness in vascular tumors is not determined by qualitative differences in adrenergic receptors. Otolaryngol. Head Neck Surg. 149, 772–776 (2013).

    Article  PubMed  Google Scholar 

  115. Mendez-Ferrer, S., Battista, M. & Frenette, P. S. Cooperation of β2- and β3-adrenergic receptors in hematopoietic progenitor cell mobilization. Ann. NY Acad. Sci. 1192, 139–144 (2010).

    Article  CAS  PubMed  Google Scholar 

  116. Magnon, C., Lucas, D. & Frenette, P. S. Trafficking of stem cells. Methods Mol. Biol. 750, 3–24 (2011).

    Article  CAS  PubMed  Google Scholar 

  117. Szpunar, M. J., Burke, K. A., Dawes, R. P., Brown, E. B. & Madden, K. S. The antidepressant desipramine and α2-adrenergic receptor activation promote breast tumor progression in association with altered collagen structure. Cancer Prev. Res. 6, 1262–1272 (2013).

    Article  CAS  Google Scholar 

  118. Lamkin, D. M. et al. α2-adrenergic blockade mimics the enhancing effect of chronic stress on breast cancer progression. Psychoneuroendocrinology 51, 262–270 (2015).

    Article  CAS  PubMed  Google Scholar 

  119. Friedman, G. D., Udaltsova, N. & Habel, L. A. Norepinephrine antagonists and cancer risk. Int. J. Cancer 128, 737–738; author reply 739 (2011).

    Article  CAS  PubMed  Google Scholar 

  120. Quail, D. F. & Joyce, J. A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 19, 1423–1437 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Shan, T. et al. β2-adrenoceptor blocker synergizes with gemcitabine to inhibit the proliferation of pancreatic cancer cells via apoptosis induction. Eur. J. Pharmacol. 665, 1–7 (2011).

    Article  CAS  PubMed  Google Scholar 

  122. Obeid, E. I. & Conzen, S. D. The role of adrenergic signaling in breast cancer biology. Cancer Biomark. 13, 161–169 (2013).

    Article  CAS  PubMed  Google Scholar 

  123. Zhao, C. M. et al. Denervation suppresses gastric tumorigenesis. Sci. Transl Med. 6,250ra115 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Rosas-Ballina, M. et al. Splenic nerve is required for cholinergic antiinflammatory pathway control of TNF in endotoxemia. Proc. Natl Acad. Sci. USA 105, 11008–11013 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  125. Villanueva, M. T. Therapeutics: gastric cancer gets a red carpet treatment. Nat. Rev. Cancer 14, 648–649 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the United States National Institutes of Health (CA083639, CA098258, CA104825, CA109298, CA116778, CA140933, CA151668, CA177909, AG017265, AG033590) and Department of Defense (OC120547, OC093416), the Betty Ann Asche Murray Distinguished Professorship, the Cancer Prevention and Research Institute of Texas (CPRIT RP140106), and the Breast Cancer Research Foundation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Steven W. Cole.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cole, S., Nagaraja, A., Lutgendorf, S. et al. Sympathetic nervous system regulation of the tumour microenvironment. Nat Rev Cancer 15, 563–572 (2015). https://doi.org/10.1038/nrc3978

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc3978

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer