DOI QR코드

DOI QR Code

교감신경계가 암의 발전과 진행에 미치는 영향

The Influence of the Sympathetic Nervous System on the Development and Progression of Cancer

  • 박신형 (동의대학교 한의과대학 병리학교실) ;
  • 지규용 (동의대학교 한의과대학 병리학교실) ;
  • 최영현 (동의대학교 한의과대학 생화학교실)
  • Park, Shin-Hyung (Department of Pathology, College of Korean Medicine, Dongeui University) ;
  • Chi, Gyoo-Yong (Department of Pathology, College of Korean Medicine, Dongeui University) ;
  • Choi, Yung Hyun (Department of Biochemistry, College of Korean Medicine, Dongeui University)
  • 투고 : 2017.09.26
  • 심사 : 2017.12.07
  • 발행 : 2018.01.30

초록

생물체는 스트레스에 대처하기 위한 항상성 유지 기작을 가지고 있으나, 만성적이고 반복적인 스트레스는 카테콜아민과 같은 스트레스 호르몬의 과도한 방출로 인해 인체에 해로운 영향을 미친다. 교감신경계와 암과의 관련성을 분석하는 연구는 스트레스가 암을 악화시킬 수 있다는 오랜 가설을 바탕으로 이루어졌다. 다수의 전임상연구와 역학 연구결과는 교감신경계의 주요 신호전달경로인 ${\beta}$-adrenergic signaling을 조절하는 것이 암의 진행을 억제할 수 있음을 보여주고 있다. 교감신경계의 활성화는 암세포의 oncogene과 DNA repair 유전자 조절, 생존과 사멸 조절, EMT 및 전이 조절, 면역계와 혈관신생 조절, 세포외기질과 간엽세포 조절, 지방세포 조절 등을 통해 암세포와 종양미세환경에 광범위하게 영향을 미칠 수 있다. 오늘날 암의 성장과 관련된 분자적 기전을 차단하는 표적항암치료가 각광받고 있으나, 보상경로의 활성화와 항암제 내성 출현 및 여러 부작용으로 말미암아 임상적 실패를 거듭하면서 암의 생병리를 다방면에서 조절하는 전략이 새로운 치료법으로 대두되고 있다. 본 총설에서는 이러한 암의 전신적 조절인자로서 교감신경계가 암의 형성과 발전에 미치는 영향을 요약하고, 향후 새로운 암 치료전략으로서 ${\beta}$-adrenergic signaling을 조절하는 약물의 임상적 활용가능성에 대해 논의하고자 한다.

Living creatures possess long-conserved mechanisms to maintain homeostasis in response to various stresses. However, chronic and continuous exposure to stress can result in the excessive production of stress hormones, including catecholamines, which have harmful effects on health. Studies on the relationship between the sympathetic nervous system (SNS) and cancer have been conducted based on the traditional hypothesis that stress can promote cancer progression. Many preclinical and epidemiological studies have suggested that the regulation of ${\beta}$-adrenergic signaling, which mediates SNS activity, can suppress the progression of solid tumors. SNS activation has highly pleiotropic effects on tumor biology, as it stimulates oncogenes, survival pathways, the epithelial - mesenchymal transition, and invasion. Moreover, it inhibits DNA repair and programmed cell death and regulates the tumor microenvironment, including immune cells, endothelial cells, the extracellular matrix, mesenchymal cells, and adipocytes. Although targeted therapies on the molecular basis of tumor proliferation are currently receiving increased attention, they have clinical limitations, such as the compensatory activation of other signaling pathways, emergence of drug resistance, and various side effects, which raise the need for pleiotropic cancer regulation. This review summarizes the effects of the SNS on the development and progression of cancer and discusses the clinical perspectives of ${\beta}$-blockade as a novel therapeutic strategy for this disease.

키워드

참고문헌

  1. Adriaenssens, E., Vanhecke, E., Saule, P., Mougel, A., Page, A., Romon, R., Nurcombe, V., Le Bourhis, X. and Hondermarck, H. 2008. Nerve growth factor is a potential therapeutic target in breast cancer. Cancer Res. 68, 346-351. https://doi.org/10.1158/0008-5472.CAN-07-1183
  2. Antoni, M. H., Lutgendorf, S. K., Cole, S. W., Dhabhar, F. S., Sephton, S. E., McDonald, P. G., Stefanek, M. and Sood, A. K. 2006. The influence of bio-behavioural factors on tumour biology: pathways and mechanisms. Nat. Rev. Cancer 6, 240-248. https://doi.org/10.1038/nrc1820
  3. Antoni, M. H., Lutgendorf, S. K., Blomberg, B., Carver, C. S., Lechner, S., Diaz, A., Stagl, J., Arevalo, J. M. and Cole, S. W. 2012. Cognitive-behavioral stress management reverses anxiety-related leukocyte transcriptional dynamics. Biol. Psychiatry 71, 366-372. https://doi.org/10.1016/j.biopsych.2011.10.007
  4. Armaiz-Pena, G. N., Allen, J. K., Cruz, A., Stone, R. L., Nick, A. M., Lin, Y. G., Han, L. Y., Mangala, L. S., Villares, G. J., Vivas-Mejia, P., Rodriguez-Aguayo, C., Nagaraja, A. S., Gharpure, K. M., Wu, Z., English, R. D., Soman, K. V., Shahzad, M. M., Zigler, M., Deavers, M. T., Zien, A., Soldatos, T. G., Jackson, D. B., Wiktorowicz, J. E., Torres- Lugo, M., Young, T., De Geest, K., Gallick, G. E., Bar-Eli, M., Lopez-Berestein, G., Cole, S. W., Lopez, G. E., Lutgendorf, S. K. and Sood, A. K. 2013. Src activation by ${\beta}$-adrenoreceptors is a key switch for tumour metastasis. Nat. Commun. 4, 1403. https://doi.org/10.1038/ncomms2413
  5. Armaiz-Pena, G. N., Cole, S. W., Lutgendorf, S. K. and Sood, A. K. 2013. Neuroendocrine influences on cancer progression. Brain Behav. Immun. 30, S19-S25. https://doi.org/10.1016/j.bbi.2012.06.005
  6. Armaiz-Pena, G. N., Gonzalez-Villasana, V., Nagaraja, A. S., Rodriguez-Aguayo, C., Sadaoui, N. C., Stone, R. L., Matsuo, K., Dalton, H. J., Previs, R. A., Jennings, N. B., Dorniak, P., Hansen, J. M., Arevalo, J. M., Cole, S. W., Lutgendorf, S. K., Sood, A. K. and Lopez-Berestein, G. 2015. Adrenergic regulation of monocyte chemotactic protein 1 leads to enhanced macrophage recruitment and ovarian carcinoma growth. Oncotarget 6, 4266-4273.
  7. Ayala, G. E., Dai, H., Powell, M., Li, R., Ding, Y., Wheeler, T. M., Shine, D., Kadmon, D., Thompson, T., Miles, B. J., Ittmann, M. M. and Rowley, D. 2008. Cancer-related axonogenesis and neurogenesis in prostate cancer. Clin. Cancer Res. 14, 7593-7603. https://doi.org/10.1158/1078-0432.CCR-08-1164
  8. Aydiner, A., Ciftci, R., Karabulut, S. and Kilic, L. 2013. Does ${\beta}$-blocker therapy improve the survival of patients with metastatic non-small cell lung cancer? Asian Pac. J. Cancer Prev. 14, 6109-6114. https://doi.org/10.7314/APJCP.2013.14.10.6109
  9. Barron, T. I., Connolly, R. M., Sharp, L., Bennett, K. and Visvanathan, K. 2011. ${\beta}$ blockers and breast cancer mortality: a population-based study. J. Clin. Oncol. 29, 2635-2644. https://doi.org/10.1200/JCO.2010.33.5422
  10. Boluyt, M. O., Long, X., Eschenhagen, T., Mende, U., Schmitz, W., Crow, M. T. and Lakatta, E. G. 1995. Isoproterenol infusion induces alterations in expression of hypertrophy-associated genes in rat heart. Am. J. Physiol. 2, H638-647.
  11. Bonnans, C., Chou, J. and Werb, Z. 2014. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 12, 786-801.
  12. Botteri, E., Munzone, E., Rotmensz, N., Cipolla, C., De Giorgi, V., Santillo, B., Zanelotti, A., Adamoli, L., Colleoni, M., Viale, G., Goldhirsch, A. and Gandini, S. 2013. Therapeutic effect of ${\beta}$-blockers in triple-negative breast cancer postmenopausal women. Breast Cancer Res. Treat. 140, 567-575. https://doi.org/10.1007/s10549-013-2654-3
  13. Boucek, R. J. Jr., Kirsh, A. L., Majesky, M. W. and Perkins, J. A. 2013. Propranolol responsiveness in vascular tumors is not determined by qualitative differences in adrenergic receptors. Otolaryngol. Head Neck Surg. 149, 772-776. https://doi.org/10.1177/0194599813503445
  14. Boyd, A. L., Salleh, A., Humber, B., Yee, J., Tomes, L. and Kerr, L. R. 2010. Neonatal experiences differentially infl uence mammary gland morphology, estrogen receptor a protein levels, and carcinogenesis in BALB/c mice. Cancer Prev. Res. (Phila) 3, 1398-1408. https://doi.org/10.1158/1940-6207.CAPR-10-0111
  15. Bronzetti, E., Artico, M., Forte, F., Pagliarella, G., Felici, L. M., D'Ambrosio, A., Vespasiani, G. and Bronzetti, B. 2008. A possible role of BDNF in prostate cancer detection. Oncol. Rep. 19, 969-974.
  16. Bruzzone, A. Pinero, C. P., Rojas, P., Romanato, M., Gass, H., Lanari, C. and Lüthy, I. A. 2011. ${\alpha}$2-adrenoceptors enhance cell proliferation and mammary tumor growth acting through both the stroma and the tumor cells. Curr. Cancer Drug Targets 11, 763-774. https://doi.org/10.2174/156800911796191051
  17. Burger, R., Bakker, F., Guenther, A., Baum, W., Schmidt Arras, D., Hideshima, T., Tai, Y. T., Shringarpure, R., Catley, L., Senaldi, G. Gramatzki, M. and Anderson, K. C. 2003. Functional significance of novel neurotrophin-1/B cell-stimulating factor-3 (cardiotrophin-like cytokine) for human myeloma cell growth and survival. Brit. J. Haematol. 123, 869-878. https://doi.org/10.1046/j.1365-2141.2003.04686.x
  18. Calvani, M., Pelon, F., Comito, G., Taddei, M. L., Moretti, S., Innocenti, S., Nassini, R., Gerlini, G., Borgognoni, L., Bambi, F., Giannoni, E., Filippi, L. and Chiarugi, P. 2015. Norepinephrine promotes tumor microenvironment reactivity through ${\beta}$3-adrenoreceptors during melanoma progression. Oncotarget 6, 4615-4632.
  19. Cao, L. Liu, X., Lin, E. J., Wang, C., Choi, E. Y., Riban, V., Lin, B. and During, M. J. 2010. Environmental and genetic activation of a brain-adipocyte BDNF/leptin axis causes cancer remission and inhibition. Cell 142, 52-64. https://doi.org/10.1016/j.cell.2010.05.029
  20. Cao, L. and During, M. J. 2012. What is the brain-cancer connection? Annu. Rev. Neurosci. 35, 331-345. https://doi.org/10.1146/annurev-neuro-062111-150546
  21. Chakroborty, D., Sarkar, C., Basu, B., Dasgupta, P. S. and Basu, S. 2009. Catecholamines regulate tumor angiogenesis. Cancer Res. 69, 3727-3730. https://doi.org/10.1158/0008-5472.CAN-08-4289
  22. Chang, M., Brown, H. J., Collado-Hidalgo, A., Arevalo, J. M., Galic, Z., Symensma, T. L., Tanaka, L., Deng, H., Zack, J. A., Sun, R. and Cole, S. W. 2005. ${\beta}$-adrenoreceptors reactivate Kaposi's sarcoma-associated herpesvirus lytic replication via PKA-dependent control of viral RTA. J. Virol. 79, 13538-13547. https://doi.org/10.1128/JVI.79.21.13538-13547.2005
  23. Chida, Y., Hamer, M., Wardle, J. and Steptoe, A. 2008. Do stress-related psychosocial factors contribute to cancer incidence and survival? Nat. Clin. Pract. Oncol. 5, 466-475. https://doi.org/10.1038/ncponc1134
  24. Cirillo, D., Rachiglio, D. A. M., la Montagna R, Giordano, A. and Normanno, N. 2008. Leptin signaling in breast cancer: an overview. J. Cell. Biochem. 105, 956-964. https://doi.org/10.1002/jcb.21911
  25. Cole, S. W. 2011. Beta-adrenergic regulation of gene expression in cancer. In: Proceeding of the 102nd Annual Meetingofthe American Association for Cancer Research.
  26. Cole, S. W. 2013. Social regulation of human gene expression: mechanisms and implications for public health. Am. J. Publ. Health 103, S84-S92. https://doi.org/10.2105/AJPH.2012.301183
  27. Cole, S. W. 2013. Nervous system regulation of the cancer genome. Brain Behav. Immun. 30, S10-S18. https://doi.org/10.1016/j.bbi.2012.11.008
  28. Cole, S. W. 2014. Human social genomics. PLoS Genet. 10, e1004601. https://doi.org/10.1371/journal.pgen.1004601
  29. Cole, S. W., Korin, Y. D., Fahey, J. L. and Zack, J. A. 1998. Norepinephrine accelerates HIV replication via protein kinase A-dependent effects on cytokine production. J. Immunol. 161, 610-616.
  30. Cole, S. W., Nagaraja, A. S., Lutgendorf, S. K., Green, P. A. and Sood, A. K. 2015. Sympathetic nervous system regulation of the tumour microenvironment. Nat. Rev. Cancer. 15, 563-572. https://doi.org/10.1038/nrc3978
  31. Cole, S. W. and Sood, A. K. 2012. Molecular pathways: beta- adrenergic signaling in cancer. Clin. Cancer Res. 5, 1201- 1206.
  32. Collado-Hidalgo, A., Sung, C. and Cole, S. 2006. 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. https://doi.org/10.1016/j.bbi.2006.01.005
  33. Cook-Mills, J. M., Mokyr, M. B., Cohen, R. L., Perlman, R. L. and Chambers, D. A. 1995. Neurotransmitter suppression of the in vitro generation of a cytotoxic T lymphocyte response against the syngeneic MOPC-315 plasmacytoma. Cancer Immunol. Immunother. 2, 79-87.
  34. Creed, S. J., Le, C. P., Hassan, M., Pon, C. K., Albold, S., Chan, K. T., Berginski, M. E., Huang, Z., Bear, J. E., Lane, J. R., Halls, M. L., Ferrari, D., Nowell, C. J. and Sloan, E. K. 2015. ${\beta}$2-adrenoceptor signaling regulates invadopodia formation to enhance tumor cell invasion. Breast Cancer Res. 17, 145. https://doi.org/10.1186/s13058-015-0655-3
  35. Dal Monte, M., Casini, G., Filippi, L., Nicchia, G. P., Svelto, M. and Bagnoli, P. 2013. Functional involvement of ${\beta}$3-adrenergic receptors in melanoma growth and vascularization. J. Mol. Med. 91, 1407-1419. https://doi.org/10.1007/s00109-013-1073-6
  36. Damon, D. H., Teriele, J. A. and Marko, S. B. 2007. Vascularderived artemin: a determinant of vascular sympathetic innervation? Am. J. Physiol. 293, H266-273
  37. De Giorgi, V., Gandini, S., Grazzini, M., Benemei, S., Marchionni, N. and Geppetti, P. 2013. Effect of ${\beta}$-blockers and other antihypertensive drugs on the risk of melanoma recurrence and death. Mayo Clin. Proc. 88, 1196-1203. https://doi.org/10.1016/j.mayocp.2013.09.001
  38. De Giorgi, V., Grazzini, M., Gandini, S., Benemei, S., Lotti, T., Marchionni, N. and Geppetti, P. 2011. Treatment with b-blockers and reduced disease progression in patients with thick melanoma. Arch. Intern. Med. 171, 779-781.
  39. Deng, G. H., Liu, J., Zhang, J., Wang, Y., Peng, X. C., Wei, Y. Q. and Jiang, Y. 2014. Exogenous norepinephrine attenuates the efficacy of sunitinib in a mouse cancer model. J. Exp. Clin. Cancer Res. 33, 21. https://doi.org/10.1186/1756-9966-33-21
  40. De Rooij, J., Zwartkruis, F. J., Verheijen, M. H., Cool, R. H., Nijman, S. M., Wittinghofer, A. and Bos, J. L. 1998. Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP. Nature 396, 474-477. https://doi.org/10.1038/24884
  41. Diaz, E. S., Karlan, B. Y. and Li, A. J. 2012. Impact of ${\beta}$ blockers on epithelial ovarian cancer survival. Gynecol. Oncol. 127, 375-378. https://doi.org/10.1016/j.ygyno.2012.07.102
  42. Drell, T. L., Joseph, J., Lang, K., Niggemann, B., Zaenker, K. S. and Entschladen, F. 2003. Effects of neurotransmitters on the chemokinesis and chemotaxis of MDA-MB-468 human breast carcinoma cells. Breast Cancer Res. Treat. 80, 63-70. https://doi.org/10.1023/A:1024491219366
  43. Eichmann, A. and Thomas, J. L. 2013. Molecular parallels between neural and vascular development. Cold Spring Harb. Perspect. Med. 3, a006551.
  44. Elenkov, I. J., Wilder, R. L., Chrousos, G. P. and Vizi, E. S. 2000. The sympathetic nerve - an integrative interface between two supersystems: the brain and the immune system. Pharmacol. Rev. 52, 595-638.
  45. Eng, J. W., Reed, C. B., Kokolus, K. M., Pitoniak, R., Utley, A., Bucsek, M. J., Ma, W. W., Repasky, E. A. and Hylander, B. L. 2015. Housing temperature-induced stress drives therapeutic resistance in murine tumour models through ${\beta}2-$ adrenergic receptor activation. Nat. Commun. 6, 6426. https://doi.org/10.1038/ncomms7426
  46. Erler, J. T., Bennewith, K. L., Cox, T. R., Lang, G., Bird, D., Koong, A., Le, Q. T. and Giaccia, A. J. 2009. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell 1, 35-44.
  47. Flierl, M. A., Rittirsch, D., Nadeau, B. A., Chen, A. J., Sarma, J. V., Zetoune, F. S., McGuire, S. R., List, R. P., Day, D. E., Hoesel, L. M., Gao, H., Van Rooijen, N., Huber-Lang, M. S., Neubig, R. R. and Ward, P. A. 2007. Phagocyte-derived catecholamines enhance acute inflammatory injury. Nature 449, 721-725. https://doi.org/10.1038/nature06185
  48. Flint, M. S., Baum, A., Episcopo, B., Knickelbein, K. Z., Liegey Dougall, A. J., Chambers, W. H. and Jenkins, F. J. 2013. Chronic exposure to stress hormones promotes transformation and tumorigenicity of 3T3 mouse fibroblasts. Stress 16, 114-121. https://doi.org/10.3109/10253890.2012.686075
  49. Friedman, G. D., Udaltsova, N. and Habel, L. A. 2011. Norepinephrine antagonists and cancer risk. Int. J. Cancer 128, 737-738.
  50. Fridman, W. H., Pages, F., Sautes-Fridman, C. and Galon, J. 2012. The immune contexture in human tumours: impact on clinical outcome. Nat. Rev. Cancer 4, 298-306.
  51. Garofalo, C. and Surmacz, E. 2006. Leptin and cancer. J. Cell. Physiol. 207, 12-22. https://doi.org/10.1002/jcp.20472
  52. Ganz, P. A. and Cole, S. W. 2011. Expanding our therapeutic options: ${\beta}$ blockers for breast cancer? J. Clin. Oncol. 29, 2612-2616. https://doi.org/10.1200/JCO.2011.35.8820
  53. Gogas, H., Trakatelli, M., Dessypris, N., Terzidis, A., Katsambas, A., Chrousos, G. P. and Petridou, E. T. 2008. Melanoma risk in association with serum leptin levels and lifestyle parameters: a case-control study. Ann. Oncol. 19, 384-389. https://doi.org/10.1093/annonc/mdm464
  54. Goldfarb, Y., Sorski, L., Benish, M., Levi, B., Melamed, R. and Ben-Eliyahu, S. 2011. 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. https://doi.org/10.1097/SLA.0b013e318211d7b5
  55. Glaser, R. and Kiecolt-Glaser J. K. 2005. Stress-induced immune dysfunction: implications for health. Nat. Rev. Immunol. 5, 243-251. https://doi.org/10.1038/nri1571
  56. Grytli, H. H., Fagerland, M. W., Fossa, S. D., Tasken, K. A. and Haheim, L. L. 2013. Use of ${\beta}$-blockers is associated with prostate cancer-specific survival in prostate cancer patients on androgen deprivation therapy. Prostate 73, 250-260. https://doi.org/10.1002/pros.22564
  57. Grytli, H. H., Fagerland, M. W., Fossa, S. D. and Tasken, K. A. 2014. Association between use of ${\beta}$-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. https://doi.org/10.1016/j.eururo.2013.01.007
  58. Guo, K. Ma, Q., Li, J., Wang, Z., Shan, T., Li, W., Xu, Q. And Xie, K. 2013. Interaction of the sympathetic nerve with pancreatic cancer cells promotes perineural invasion through the activation of STAT3 signaling. Mol. Cancer Ther. 12, 264-273. https://doi.org/10.1158/1535-7163.MCT-12-0809
  59. Hanoun, M., Maryanovich, M., Arnal-Estape, A. and Frenette, P. S. 2015. Neural regulation of hematopoiesis, inflammation, and cancer. Neuron 86, 360-373. https://doi.org/10.1016/j.neuron.2015.01.026
  60. Hara, M. R., Kovacs, J. J., Whalen, E. J., Rajagopal, S., Strachan, R. T., Grant, W., Towers, A. J., Williams, B., Lam, C. M., Xiao, K., Shenoy, S. K., Gregory, S. G., Ahn, S., Duckett, D. R. and Lefkowitz, R. J. 2011. A stress response pathway regulates DNA damage through ${\beta}2$-adrenoreceptors and ${\beta}$-arrestin-1. Nature 477, 349-353. https://doi.org/10.1038/nature10368
  61. Hara, M. R., Sachs, B. D., Caron, M. G. and Lefkowitz, R. J. 2013. Pharmacological blockade of a ${\beta}2AR$-${\beta}arrestin$-1 signaling cascade prevents the accumulation of DNA damage in a behavioral stress model. Cell Cycle 12, 219-224. https://doi.org/10.4161/cc.23368
  62. Hasegawa, H. and Saiki, I. 2002. Psychosocial stress augments tumor development through ${\beta}$-adrenergic activation in mice. Jpn. J. Cancer Res. 93, 729-735. https://doi.org/10.1111/j.1349-7006.2002.tb01313.x
  63. Hassan, S., Karpova, Y., Baiz, D., Yancey, D., Pullikuth, A., Flores, A., Register, T., Cline, J. M., D'Agostino, R. Jr., Danial, N. Datta, S. R. and Kulik, G. 2013. Behavioral stress accelerates prostate cancer development in mice. J. Clin. Invest. 123, 874-886.
  64. Heidt, T., Sager, H. B., Courties, G., Dutta, P., Iwamoto, Y., Zaltsman, A., von Zur Muhlen C., Bode, C., Fricchione, G. L., Denninger, J., Lin, C. P., Vinegoni, C., Libby, P., Swirski, F. K., Weissleder, R. and Nahrendorf, M. 2014. Chronic variable stress activates hematopoietic stem cells. Nat. Med. 20, 754-758. https://doi.org/10.1038/nm.3589
  65. Hermes, G. L., Delgado, B., Tretiakova, M., Cavigelli, S. A., Krausz, T., Conzen, S. D. and McClintock, M. K. 2009. Social isolation dysregulates endocrine and behavioral stress while increasing malignant burden of spontaneous mammary tumors. Proc. Natl. Acad. Sci. USA 106, 22393-22398. https://doi.org/10.1073/pnas.0910753106
  66. Hori, Y., Ishii, K., Kanda, H., Iwamoto, Y., Nishikawa, K., Soga, N., Kise, H., Arima, K. and Sugimura, Y. 2011. Naftopidil, a selective ${\alpha}1$-adrenoceptor antagonist, suppresses human prostate tumor growth by altering interactions between tumor cells and stroma. Cancer Prev. Res. 4, 87-96. https://doi.org/10.1158/1940-6207.CAPR-10-0189
  67. Horowitz, M., Neeman, E., Sharon, E. and Ben-Eliyahu, S. 2015. Exploiting the critical perioperative period to improve long-term cancer outcomes. Nat. Rev. Clin. Oncol. 12, 213-226. https://doi.org/10.1038/nrclinonc.2014.224
  68. Huang, S. and Ingber, D. E. 2005. Cell tension, matrix mechanics, and cancer development. Cancer Cell 3, 175-176.
  69. Inbar, S., Neeman, E., Avraham, R., Benish, M., Rosenne, E. and Ben-Eliyahu, S. 2011. 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. https://doi.org/10.1371/journal.pone.0019246
  70. Irwin, M. R. and Cole, S. W. 2011. Reciprocal regulation of the neural and innate immune systems. Nat. Rev. Immunol. 11, 625-632. https://doi.org/10.1038/nri3042
  71. Jimenez-Andrade, J. M., Bloom, A. P., Stake, J. I., Mantyh, W. G., Taylor, R. N., Freeman, K. T., Ghilardi, J. R., Kuskowski, M. A. and Mantyh, P. W. 2010. Pathological sprouting of adult nociceptors in chronic prostate cancer-induced bone pain. J. Neurosci. 30, 14649-14659. https://doi.org/10.1523/JNEUROSCI.3300-10.2010
  72. Kalinichenko, V. V., Mokyr, M. B., Graf, L. H. Jr., Cohen, R. L. and Chambers, D. A. 1999. Norepinephrine-mediated inhibition of antitumor cytotoxic T lymphocyte generation involves a beta-adrenergic receptor mechanism and decreased TNF-alpha gene expression. J. Immunol. 5, 2492- 2499.
  73. Katayama, Y., Battista, M., Kao, W. M., Hidalgo, A., Peired, A. J., Thomas, S. A. and Frenette, P. S. 2006. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell 124, 407-421. https://doi.org/10.1016/j.cell.2005.10.041
  74. Keino-Masu, K., Masu, M., Hinck, L., Leonardo, E. D., Chan, S. S., Culotti, J. G. and Tessier-Lavigne, M. 1996. Deleted in colorectal cancer (DCC) encodes a netrin receptor. Cell 87, 175-185. https://doi.org/10.1016/S0092-8674(00)81336-7
  75. Kim-Fuchs, C., Le, C. P., Pimentel, M. A., Shackleford, D., Ferrari, D., Angst, E., Hollande, F. and Sloan, E. K. 2014. Chronic stress accelerates pancreatic cancer growth and invasion: a critical role for ${\beta}$-adrenergic signaling in the pancreatic microenvironment. Brain Behav. Immun. 40, 40-47. https://doi.org/10.1016/j.bbi.2014.02.019
  76. Kitamura, T., Qian, B. Z. and Pollard, J. W. 2015. Immune cell promotion of metastasis. Nat. Rev. Immunol. 2, 73-86.
  77. Kohm, A. P. and Sanders, V. M. 2001. Norepinephrine and ${\beta}$ 2-adrenergic receptor stimulation regulate $CD4^+$ T and B lymphocyte function in vitro and in vivo. Pharmacol. Rev. 53, 487-525.
  78. Kruttgen, A., Schneider, I. and Weis, J. 2006. The dark side of the NGF family: neurotrophins in neoplasias. Brain Pathol. 16, 304-310. https://doi.org/10.1111/j.1750-3639.2006.00037.x
  79. Kulik, G. A., Hassan, S., Karpova, Y. and Baurin, V. 2011. Behavioral stress protects prostate cancer cells from apoptosis. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research.
  80. Lai, P. C., Chiu, T. H. and Huang, Y. T. 2010. Overexpression of BDNF and TrkB in human bladder cancer specimens. Oncol. Rep. 24, 1265-1270.
  81. Lamkin, D. M., Sloan, E. K., Patel, A. J., Chiang, B. S., Pimentel, M. A., Ma, J. C., Arevalo, J. M., Morizono, K. and Cole, S. W. 2012. Chronic stress enhances progression of acute lymphoblastic leukemia via ${\beta}$-adrenergic signaling. Brain Behav. Immun. 26, 635-641. https://doi.org/10.1016/j.bbi.2012.01.013
  82. Lamkin, D. M., Sung, H. Y., Yang, G. S., David, J. M., Ma, J. C., Cole, S. W. and Sloan, E. K. 2015. ${\alpha}2$-adrenergic blockade mimics the enhancing effect of chronic stress on breast cancer progression. Psychoneuroendocrinology 51, 262-270. https://doi.org/10.1016/j.psyneuen.2014.10.004
  83. Lang, K., Drell, T. L. 4th., Lindecke, A., Niggemann, B., Kaltschmidt, C., Zaenker, K. S. and Entschladen, F. 2004. Induction of a metastatogenic tumor cell type by neurotransmitters and its pharmacological inhibition by established drugs. Int. J. Cancer 112, 231-238. https://doi.org/10.1002/ijc.20410
  84. Landen, C. N. Jr., Lin, Y. G., Armaiz Pena, G. N., Das, P. D., Arevalo, J. M., Kamat, A. A., Han, L. Y., Jennings, N. B., Spannuth, W. A., Thaker, P. H., Lutgendorf, S. K., Savary, C. A., Sanguino, A. M., Lopez-Berestein, G., Cole, S. W. and Sood, A. K. 2007. Neuroendocrine modulation of signal transducer and activator of transcription-3 in ovarian cancer. Cancer Res. 67, 10389-10396. https://doi.org/10.1158/0008-5472.CAN-07-0858
  85. Lemeshow, S. Sorensen, H. T., Phillips, G., Yang, E. V., Antonsen, S., Riis, A. H., Lesinski, G. B., Jackson, R. and Glaser, R. 2011. ${\beta}$-blockers and survival among Danish patients with malignant melanoma: a population-based cohort study. Cancer Epidemiol. Biomarkers Prev. 20, 2273-2279. https://doi.org/10.1158/1055-9965.EPI-11-0249
  86. Leonardo, E. D., Hinck, L., Masu, M., Keino-Masu, K., Ackerman, S. L. and Tessier-Lavigne, M. 1997. Vertebrate homologues of C. elegans UNC-5 are candidate netrin receptors. Nature 386, 833-838. https://doi.org/10.1038/386833a0
  87. Liu, J., Deng, G. H., Zhang, J., Wang, Y., Xia, X. Y., Luo, X. M., Deng, Y. T., He, S. S., Mao, Y. Y., Peng, X. C., Wei, Y. Q. and Jiang, Y. 2015. The effect of chronic stress on antiangiogenesis of sunitinib in Lu colorectal cancer models. Psychoneuroendocrinology 52, 130-142. https://doi.org/10.1016/j.psyneuen.2014.11.008
  88. Lu, P., Weaver, V. M. and Werb, Z. 2012. The extracellular matrix: a dynamic niche in cancer progression. J. Cell Biol. 4, 395-406.
  89. Lutgendorf, S. K., DeGeest, K., Sung, C. Y., Arevalo, J. M., Penedo, F., Lucci, J., Goodheart, M., Lubaroff, D., Farley, D. M., Sood, A. K. and Cole, S. W. 2009. Depression, social support, and ${\beta}$-adrenergic transcription control in human ovarian cancer. Brain Behav. Immun. 23, 176-183. https://doi.org/10.1016/j.bbi.2008.04.155
  90. Lutgendorf, S. K., DeGeest, K., Dahmoush, L., Farley, D., Penedo, F., Bender, D., Goodheart, M., Buekers, T. E., Mendez, L., Krueger, G., Clevenger, L., Lubaroff, D. M., Sood, A. K. and Cole, S. W. 2011. Social isolation is associated with elevated tumor norepinephrine in ovarian carcinoma patients. Brain Behav. Immun. 25, 250-255. https://doi.org/10.1016/j.bbi.2010.10.012
  91. Lutgendorf, S. K., Sood, A. K. and Antoni, M. H. 2010. Host factors and cancer progression: biobehavioral signaling pathways and interventions. J. Clin. Oncol. 28, 4094- 4099. https://doi.org/10.1200/JCO.2009.26.9357
  92. Luttrell, L. M., Ferguson, S. S., Daaka, Y., Miller, W. E., Maudsley, S., Della Rocca, G. J., Lin, F., Kawakatsu, H., Owada, K., Luttrell, D. K., Caron, M. G. and Lefkowitz, R. J. 1999. Beta-arrestin-dependent formation of beta2 adrenergic receptor-Src protein kinase complexes. Science 283, 655-661. https://doi.org/10.1126/science.283.5402.655
  93. Madden, K. S., Szpunar, M. J. and Brown, E. B. 2011. ${\beta}$- adrenergic receptors (${\beta}$-AR) regulate VEGF and IL-6 production by divergent pathways in high ${\beta}$-AR-expressing breast cancer cell lines. Breast Cancer Res. Treat. 130, 747-758. https://doi.org/10.1007/s10549-011-1348-y
  94. Magnon, C., Hall, S. J., Lin, J., Xue, X., Gerber, L., Freedland, S. J. and Frenette, P. S. 2013. Autonomic nerve development contributes to prostate cancer progression. Science 341, 1236361. https://doi.org/10.1126/science.1236361
  95. Mantovani, A., Allavena, P., Sica, A. and Balkwill, F. 2008. Cancer-related inflammation. Nature 7203, 436-444.
  96. Makita, T., Sucov, H. M., Gariepy, C. E., Yanagisawa, M. and Ginty, D. D. 2008. Endothelins are vascular-derived axonal guidance cues for developing sympathetic neurons. Nature 452, 759-763. https://doi.org/10.1038/nature06859
  97. McGregor, L. M., McCune, B. K., Graff, J. R., McDowell, P. R., Romans, K. E., Yancopoulos, G. D., Ball, D. W., Baylin, S. B. and Nelkin, B. D. 1999. Roles of trk family neurotrophin receptors in medullary thyroid carcinoma development and progression. Proc. Nat. Acad. Sci. USA 96, 4540-4545. https://doi.org/10.1073/pnas.96.8.4540
  98. Melhem-Bertrandt, A., Chavez-Macgregor, M., Lei, X., Brown, E. N., Lee, R. T., Meric-Bernstam, F., Sood, A. K., Conzen, S. D., Hortobagyi, G. N. and Gonzalez-Angulo, A. M. 2011. ${\beta}$-blocker use is associated with improved relapse- free survival in patients with triple-negative breast cancer. J. Clin. Oncol. 29, 2645-2652. https://doi.org/10.1200/JCO.2010.33.4441
  99. Minchinton, A. I. and Tannock, I. F. 2006. Drug penetration in solid tumours. Nat. Rev. Cancer 8, 583-592.
  100. Montminy, M. 1997. Transcriptional regulation by cyclic AMP. Annu. Rev. Biochem. 66, 807-822. https://doi.org/10.1146/annurev.biochem.66.1.807
  101. Mantyh, W. G., Jimenez-Andrade, J. M., Stake, J. I., Bloom, A. P., Kaczmarska, M. J., Taylor, R. N., Freeman, K. T., Ghilardi, J. R., Kuskowski, M. A. and Mantyh, P. W. 2010. Blockade of nerve sprouting and neuroma formation markedly attenuates the development of late stage cancer pain. Neuroscience 171, 588-598. https://doi.org/10.1016/j.neuroscience.2010.08.056
  102. Nakai, A., Hayano, Y., Furuta, F., Noda, M. and Suzuki, K. 2014. Control of lymphocyte egress from lymph nodes through beta2-adrenergic receptors. J. Exp. Med. 13, 2583- 2598.
  103. Neeman, E., Zmora, O. and Ben-Eliyahu, S. 2012. A new approach to reducing postsurgical cancer recurrence: perioperative targeting of catecholamines and prostaglandins. Clin. Cancer Res. 18, 4895-4902. https://doi.org/10.1158/1078-0432.CCR-12-1087
  104. Nilsson, M. B., Armaiz-Pena, G., Takahashi, R., Lin, Y. G., Trevino, J., Li, Y., Jennings, N., Arevalo, J., Lutgendorf, S. K., Gallick, G. E., Sanguino, A. M., Lopez-Berestein, G., Cole, S. W. and Sood, A. K. 2007. Stress hormones regulate interleukin-6 expression by human ovarian carcinoma cells through a Src-dependent mechanism. J. Biol. Chem. 282, 29919-29926. https://doi.org/10.1074/jbc.M611539200
  105. Ohta, T., Numata, M., Tsukioka, Y., Futagami, F., Kayahara, M., Kitagawa, H., Nagakawa, T., Yamamoto, M., Wakayama, T., Kitamura, Y., Terada, T. and Nakanuma, Y. 1997. Neurotrophin-3 expression in human pancreatic cancers. J. Pathol. 181, 405-412. https://doi.org/10.1002/(SICI)1096-9896(199704)181:4<405::AID-PATH786>3.0.CO;2-3
  106. Palm, D., Lang, K., Niggemann, B., Drell, T. L., Masur, K., Zaenker, K. S. and Entschladen, F. 2006. The norepinephrine- driven metastasis development of PC-3 human prostate cancer cells in BALB/c nude mice is inhibited by ${\beta}$-blockers. Int. J. Cancer 118, 2744-2749. https://doi.org/10.1002/ijc.21723
  107. Pongratz, G. and Straub, R. H. 2014. The sympathetic nervous response in inflammation. Arthritis Res. Ther. 6, 504.
  108. Pasquier, E., Street, J., Pouchy, C., Carre, M., Gifford, A. J., Murray, J., Norris, M. D., Trahair, T., Andre, N. and Kavallaris, M. 2013. ${\beta}$-blockers increase response to chemotherapy via direct antitumour and antiangiogenic mechanisms in neuroblastoma. Br. J. Cancer 108, 2485-2494. https://doi.org/10.1038/bjc.2013.205
  109. Paszek, M. J., Zahir, N., Johnson, K. R., Lakins, J. N., Rozenberg, G. I., Gefen, A. Reinhart-King, C. A., Margulies, S. S., Dembo, M., Boettiger, D., Hammer, D. A. and Weaver, V. M. 2005. Tensional homeostasis and the malignant phenotype. Cancer Cell 3, 241-254.
  110. Penninx, B. W., Guralnik, J. M., Pahor, M., Ferrucci, L., Cerhan, J. R., Wallace, R. B. and Havlik, R. J. 1998. Chronically depressed mood and cancer risk in older persons. J. Nat. Cancer Inst. 90, 1888-1893. https://doi.org/10.1093/jnci/90.24.1888
  111. Peters, L. J. and Kelly, H. 1977. The influence of stress and stress hormones on the transplantability of a non-immunogenic syngeneic murine tumor. Cancer 39, 14828.
  112. Powe, D. G., Voss, M. J., Zanker, K. S., Habashy, H. O., Green, A. R., Ellis, I. O. and Entschladen, F. 2010. Betablocker drug therapy reduces secondary cancer formation in breast cancer and improves cancer specific survival. Oncotarget 1, 628-638.
  113. Powell, N. D., Sloan, E. K., Bailey, M. T., Arevalo, J. M., Miller, G. E., Chen, E., Kobor, M. S., Reader, B. F., Sheridan, J. F. and Cole, S. W. 2013. Social stress up-regulates inflammatory gene expression in the leukocyte transcriptome via ${\beta}$-adrenergic induction of myelopoiesis. Proc. Natl. Acad. Sci. USA 110, 16574-16579. https://doi.org/10.1073/pnas.1310655110
  114. Reiche, E. M., Nunes, S. O. and Morimoto, H. K. 2004. Stress, depression, the immune system, and cancer. Lancet Oncol. 5, 617-625. https://doi.org/10.1016/S1470-2045(04)01597-9
  115. Salmon, H., Franciszkiewicz, K., Damotte, D., Dieu-Nosjean, M. C., Validire, P., Trautmann, A. Mami-Chouaib, F. and Donnadieu, E. 2012. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J. Clin. Invest. 3, 899-910.
  116. Sastry, K. S., Karpova, Y., Prokopovich, S., Smith, A. J., Essau, B., Gersappe, A., Carson, J. P., Weber, M. J., Register, T. C., Chen, Y. Q., Penn, R. B. and Kulik, G. 2007. Epinephrine protects cancer cells from apoptosis via activation of cAMP-dependent protein kinase and BAD phosphorylation. J. Biol. Chem. 282, 14094-14100. https://doi.org/10.1074/jbc.M611370200
  117. Scheiermann, C., Kunisaki, Y. and Frenette, P. S. 2013. Circadian control of the immune system. Nat. Rev. Immunol. 13, 190-198. https://doi.org/10.1038/nri3386
  118. Schuler, L. A. and Auger, A. P. 2010. Psychosocially infl uenced cancer : diverse early life stress experiences and links to breast cancer. Cancer Prev. Res. (Phila) 3, 1365-1370. https://doi.org/10.1158/1940-6207.CAPR-10-0238
  119. Seo, B. R., Bhardwaj, .P., Choi, S., Gonzalez, J., Andresen Eguiluz, R. C., Wang, K., Mohanan, S., Morris, P. G., Du, B., Zhou, X. K., Vahdat, L. T., Verma, A., Elemento, O., Hudis, C. A., Williams, R. M., Gourdon, D., Dannenberg, A. J. and Fischbach, C. 2015. Obesitydependent changes in interstitial ECM mechanics promote breast tumorigenesis. Sci. Transl. Med. 7, 301ra130. https://doi.org/10.1126/scitranslmed.3010467
  120. Shah, S. M., Carey, I. M., Owen, C. G., Harris, T., Dewilde, S. and Cook, D. G. 2011. Does b-adrenoceptor blocker therapy improve cancer survival? Findings from a populationbased retrospective cohort study. Br. J. Clin. Pharmacol. 72, 157-161. https://doi.org/10.1111/j.1365-2125.2011.03980.x
  121. Shahzad, M. M., Arevalo, J. M., Armaiz-Pena, G. N., Lu, C., Stone, R. L., Moreno-Smith, M., Nishimura, M., Lee, J. W., Jennings, N. B., Bottsford-Miller, J. Vivas-Mejia, P., Lutgendorf, S. K., Lopez-Berestein, G., Bar-Eli, M., Cole, S. W. and Sood, A. K. 2010. Stress effects on FosB- and interleukin-8 (IL8)-driven ovarian cancer growth and metastasis. J. Biol. Chem. 285, 35462-35470. https://doi.org/10.1074/jbc.M110.109579
  122. Shakhar, G. and Ben-Eliyahu, S. 1998. In vivo beta-adrenergic stimulation suppresses natural killer activity and compromises resistance to tumor metastasis in rats. J. Immunol. 7, 3251-3258.
  123. Shan, T., Cui, X., Li, W., Lin, W., Li, Y., Chen, X. and Wu, T. 2014. Novel regulatory program for norepinephrine-induced epithelial-mesenchymal transition in gastric adenocarcinoma cell lines. Cancer Sci. 105, 847-856. https://doi.org/10.1111/cas.12438
  124. Shanley, C. J., Gharaee-Kermani, M., Sarkar, R., Welling, T. H., Kriegel, A., Ford, J. W., Stanley, J. C. and Phan, S. H. 1997. Transforming growth factor-beta 1 increases lysyl oxidase enzyme activity and mRNA in rat aortic smooth muscle cells. J. Vasc. Surg. 3, 446-452.
  125. Shi, M., Liu, D., Duan, H., Qian, L., Wang, L., Niu, L., Zhang, H., Yong, Z., Gong, Z., Song, L., Yu, M., Hu, M., Xia, Q., Shen, B. and Guo, N. 2011. The ${\beta}$2-adrenergic receptor and Her2 comprise a positive feedback loop in human breast cancer cells. Breast Cancer Res. Treat. 125, 351-362. https://doi.org/10.1007/s10549-010-0822-2
  126. Sklar, L. S. and Anisman, H. 1979. Stress and coping factors influence tumor growth. Science 205, 513-515. https://doi.org/10.1126/science.109924
  127. Simon, R. H., Lovett, E. J., Tomaszek, D. and Lundy, J. 1980. Electrical stimulation of the midbrain mediates metastatic tumor growth. Science 209, 1132-1133. https://doi.org/10.1126/science.6250220
  128. Sloan, E. K., Capitanio, J. P., Tarara, R. P., Mendoza, S. P., Mason, W. A. and Cole, S. W. 2007. Social stress enhances sympathetic innervation of primate lymph nodes: mechanisms and implications for viral pathogenesis. J. Neurosci. 27, 8857-8865. https://doi.org/10.1523/JNEUROSCI.1247-07.2007
  129. Sloan, E. K., Capitanio, J. P. and Cole, S. W. 2008. Stress-induced remodeling of lymphoid innervation. Brain Behav. Immun. 22, 15-21.
  130. Sloan, E. K., Capitanio, J. P., Tarara, R. P. and Cole, S. W. 2008. Social temperament and lymph node innervation. Brain Behav. Immun. 22, 717-726. https://doi.org/10.1016/j.bbi.2007.10.010
  131. Sloan, E. K., Priceman, S. J., Cox, B. F., Yu, S., Pimentel, M. A., Tangkanangnukul, V., Arevalo, J. M., Morizono, K., Karanikolas, B. D., Wu, L., Sood, A. K. and Cole, S. W. 2010. The sympathetic nervous system induces a metastatic switch in primary breast cancer. Cancer Res. 18, 7042-7052.
  132. Sood, A. K., Bhatty, R., Kamat, A. A., Landen, C. N., Han, L., Thaker, P. H., Li, Y., Gershenson, D. M., Lutgendorf, S. and Cole, S. W. 2006. Stress hormone-mediated invasion of ovarian cancer cells. Clin. Cancer Res. 12, 369-375. https://doi.org/10.1158/1078-0432.CCR-05-1698
  133. Sood, A. K., Armaiz-Pena, G. N., Halder, J., Nick, A. M., Stone, R. L., Hu, W., Carroll, A. R., Spannuth, W. A., Deavers, M. T., Allen, J. K., Han, L. Y., Kamat, A. A., Shahzad, M. M., McIntyre, B. W., Diaz-Montero, C. M., Jennings, N. B., Lin, Y. G., Merritt, W. M., DeGeest, K., Vivas-Mejia, P. E., Lopez-Berestein, G., Schaller, M. D., Cole, S. W. and Lutgendorf, S. K. 2010. Adrenergic modulation of focal adhesion kinase protects human ovarian cancer cells from anoikis. J. Clin. Invest. 120, 1515-1523. https://doi.org/10.1172/JCI40802
  134. Szpunar, M. J., Burke, K. A., Dawes, R. P., Brown, E. B. and Madden, K. S. 2013. The antidepressant desipramine and ${\alpha}$2-adrenergic receptor activation promote breast tumor progression in association with altered collagen structure. Cancer Prev. Res. 6, 1262-1272. https://doi.org/10.1158/1940-6207.CAPR-13-0079
  135. Thaker, P. H., Han, L. Y., Kamat, A. A., Arevalo, J. M., Takahashi, R., Lu, C., Jennings, N. B., Armaiz-Pena, G., Bankson, J. A., Ravoori, M., Merritt, W. M., Lin, Y. G., Mangala, L. S., Kim, T. J., Coleman, R. L., Landen, C. N., Li, Y., Felix, E., Sanguino, A. M., Newman, R. A., Lloyd, M., Gershenson, D. M., Kundra, V., Lopez-Berestein, G., Lutgendorf, S. K., Cole, S. W. and Sood, A. K. 2006. Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma. Nat. Med. 12, 939-944. https://doi.org/10.1038/nm1447
  136. Voss, M. J. and Entschladen, F. 2010. Tumor interactions with soluble factors and the nervous system. Cell Commun. Signal 8, 21. https://doi.org/10.1186/1478-811X-8-21
  137. Watkins, J. L., Thaker, P. H., Nick, A. M., Ramondetta, L. M., Kumar, S., Urbauer, D. L., Matsuo, K., Squires, K. C., Coleman, R. L., Lutgendorf, S. K., Ramirez, P. T. and Sood, A. K. 2015. Clinical impact of selective and non-selective ${\beta}$-blockers on survival in patients with ovarian cancer. Cancer 121, 3444-3451. https://doi.org/10.1002/cncr.29392
  138. Wauters, M., Considine, R. V. and Van Gool, L. F. 2000. Human leptin: From an adipocyte hormone to an endocrine mediator. Eur. J. Endocrionol. 143, 293-311. https://doi.org/10.1530/eje.0.1430293
  139. Weiner, H. 1992. Perturbing the Organism: The Biology of Stressful Experience, Univ. of Chicago Press, Chicago, USA.
  140. Wingenfeld, K., Whooley, M. A., Neylan, T. C., Otte, C. and Cohen, B. E. 2015. 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. https://doi.org/10.1016/j.psyneuen.2014.10.023
  141. Wolter, J. K., Wolter, N. E., Blanch, A., Partridge, T., Cheng, L., Morgenstern, D. A., Podkowa, M., Kaplan, D. R. and Irwin, M. S. 2014. Anti-tumor activity of the ${\beta}$-adrenergic receptor antagonist propranolol in neuroblastoma. Oncotarget 5, 161-172.
  142. Xu, Q., Wang, Z., Chen, X., Duan, W., Lei, J., Zong, L., Li, X., Sheng, L., Ma, J., Han, L., Li, W., Zhang, L., Guo, K., Ma, Z., Wu, Z., Wu, E. and Ma, Q. 2015. Stromal-derived factor-$1{\alpha}$/CXCL12-CXCR4 chemotactic pathway promotes perineural invasion in pancreatic cancer. Oncotarget 6, 4717-4732.
  143. Yang, E. V., Sood, A. K., Chen, M., Li, Y., Eubank, T. D., Marsh, C. B., Jewell, S., Flavahan, N. A., Morrison, C., Yeh, P. E., Lemeshow, S. and Glaser, R. 2006. 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. https://doi.org/10.1158/0008-5472.CAN-06-2496
  144. Yang, E. V., Kim, S. J., Donovan, E. L., Chen, M., Gross, A. C., Webster Marketon, J. I., Barsky, S. H. and Glaser, R. 2009. 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. https://doi.org/10.1016/j.bbi.2008.10.005
  145. Yang, Z. F., Ho, D. W., Lam, C. T., Luk, J. M., Lum, C. T., Yu, W. C., Poon, R. T. and Fan, S. T. 2005. Identification of brain-derived neurotrophic factor as a novel functional protein in hepatocellular carcinoma. Cancer Res. 65, 21925.
  146. Zage, P. E., Graham, T. C., Zeng, L., Fang, W., Pien, C., Thress, K., Omer, C., Brown, J. L. and Zweidler-McKay, P. A. 2011. The selective Trk inhibitor AZ623 inhibits brain-derived neurotrophic factor-mediated neuroblastoma cell proliferation and signaling and is synergistic with topotecan. Cancer 117, 1321-1391. https://doi.org/10.1002/cncr.25674
  147. Zhang, J., Deng, Y. T., Liu, J., Wang, Y. Q., Yi, T. W., Huang, B. Y., He, S. S., Zheng, B. and Jiang, Y. 2016. Norepinephrine induced epithelial-mesenchymal transition in HT-29 and A549 cells in vitro. J. Cancer Res. Clin. Oncol. 142, 423-435. https://doi.org/10.1007/s00432-015-2044-9
  148. Zhang, X., Odom, D. T., Koo, S. H., Conkright, M. D., Canettieri, G., Best, J., Chen, H., Jenner, R., Herbolsheimer, E., Jacobsen, E., Kadam, S., Ecker, J. R., Emerson, B., Hogenesch, J. B., Unterman, T., Young, R. A. and Montminy, M. 2005. Genome-wide analysis of cAMP-response element binding protein occupancy, phosphorylation, and target gene activation in human tissues. Proc. Natl. Acad. Sci. USA 102, 4459-4464. https://doi.org/10.1073/pnas.0501076102
  149. Zur Hausen, H. 2008. Infections causing human cancer. Wiley-VCH, NJ, USA.