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:

Exercise-dependent regulation of the tumour microenvironment

Abstract

The integrity and composition of the tumour microenvironment (TME) is highly plastic, undergoing constant remodelling in response to instructive signals derived from alterations in the availability and nature of systemic host factors. This 'systemic milieu' is directly modulated by host exposure to modifiable lifestyle factors such as exercise. Host exposure to regular exercise markedly reduces the risk of the primary development of several cancers and might improve clinical outcomes following a diagnosis of a primary disease. However, the molecular mechanisms that underpin the apparent antitumour effects of exercise are poorly understood. In this Opinion article, we explore the putative effects of exercise in reprogramming the interaction between the host and the TME. Specifically, we speculate on the possible effects of exercise on reprogramming 'distant' tissue microenvironments (those not directly involved in the exercise response) by analysing how alterations in the systemic milieu might modulate key TME components to influence cancer hallmarks.

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: Integrated physiological response to acute exercise.
Figure 2: A simplified model of the adaptive response to exercise in skeletal muscle.
Figure 3: Exercise-dependent regulation of the tumour microenvironment.
Figure 4: Timeline of exercise in cancer research.

Similar content being viewed by others

References

  1. Rozhok, A. I. & DeGregori, J. The evolution of lifespan and age-dependent cancer risk. Trends Cancer 2, 552–560 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Wu, S., Powers, S., Zhu, W. & Hannun, Y. A. Substantial contribution of extrinsic risk factors to cancer development. Nature 529, 43–47 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Govindan, R. et al. Genomic landscape of non-small cell lung cancer in smokers and never-smokers. Cell 150, 1121–1134 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Jhappan, C., Noonan, F. P. & Merlino, G. Ultraviolet radiation and cutaneous malignant melanoma. Oncogene 22, 3099–3112 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. [No authors listed.] TOBACCO and carcinoma of the lung. N. Engl. J. Med. 250, 125 (1954).

  6. Richmond, B. G. & Jungers, W. L. Orrorin tugenensis femoral morphology and the evolution of hominin bipedalism. Science 319, 1662–1665 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Bramble, D. M. & Lieberman, D. E. Endurance running and the evolution of Homo. Nature 432, 345–352 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Booth, F. W., Chakravarthy, M. V. & Spangenburg, E. E. Exercise and gene expression: physiological regulation of the human genome through physical activity. J. Physiol. 543, 399–411 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hopkins, B. D., Goncalves, M. D. & Cantley, L. C. Obesity and cancer mechanisms: cancer metabolism. J. Clin. Oncol. 34, 4277–4283 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lee, I. M. et al. Effect of physical inactivity on major non-communicable diseases worldwide: an analysis of burden of disease and life expectancy. Lancet 380, 219–229 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Kohl, H. W. III et al. The pandemic of physical inactivity: global action for public health. Lancet 380, 294–305 (2012).

    Article  PubMed  Google Scholar 

  12. Bauman, A. et al. Tackling obesity: challenges ahead. Lancet 386, 741–742 (2015).

    Article  PubMed  Google Scholar 

  13. Calle, E. E., Rodriguez, C., Walker-Thurmond, K. & Thun, M. J. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U. S. adults. N. Engl. J. Med. 348, 1625–1638 (2003).

    Article  PubMed  Google Scholar 

  14. Pontzer, H. et al. Hunter-gatherer energetics and human obesity. PLoS ONE 7, e40503 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hallal, P. C. et al. Global physical activity levels: surveillance progress, pitfalls, and prospects. Lancet 380, 247–257 (2012).

    Article  PubMed  Google Scholar 

  16. World Health Organization. Global health risks: mortality and burden of disease attributable to selected major risks (WHO, 2009).

  17. Myers, J. et al. Exercise capacity and mortality among men referred for exercise testing. N. Engl. J. Med. 346, 793–801 (2002).

    Article  PubMed  Google Scholar 

  18. Gulati, M. et al. The prognostic value of a nomogram for exercise capacity in women. N. Engl. J. Med. 353, 468–475 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Moore, S. C. et al. Association of leisure-time physical activity with risk of 26 types of cancer in 1.44 million adults. JAMA Intern. Med. 176, 816–825 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Friedenreich, C. M., Neilson, H. K., Farris, M. S. & Courneya, K. S. Physical activity and cancer outcomes: a precision medicine approach. Clin. Cancer Res. 22, 4766–4775 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Betof, A. S., Dewhirst, M. W. & Jones, L. W. Effects and potential mechanisms of exercise training on cancer progression: a translational perspective. Brain Behav. Immun. 30, S75–S87 (2013).

    Article  PubMed  Google Scholar 

  22. Ashcraft, K. A., Peace, R. M., Betof, A. S., Dewhirst, M. W. & Jones, L. W. Efficacy and mechanisms of aerobic exercise on cancer initiation, progression, and metastasis: a critical systematic review of in vivo preclinical data. Cancer Res. 76, 4032–4050 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Neufer, P. D. et al. Understanding the cellular and molecular mechanisms of physical activity-induced health benefits. Cell Metab. 22, 4–11 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Jones, N. L. & Killian, K. J. Exercise limitation in health and disease. N. Engl. J. Med. 343, 632–641 (2000).

    Article  CAS  PubMed  Google Scholar 

  25. Jones, L. W., Eves, N. D., Haykowsky, M., Freedland, S. J. & Mackey, J. R. Exercise intolerance in cancer and the role of exercise therapy to reverse dysfunction. Lancet Oncol. 10, 598–605 (2009).

    Article  PubMed  Google Scholar 

  26. Levine, B. D. VO2, max: what do we know, and what do we still need to know? J. Physiol. 586, 25–34 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Peake, J. M. et al. Modulating exercise-induced hormesis: does less equal more? J. Appl. Physiol. 119, 172–189 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Hawley, J. A., Hargreaves, M., Joyner, M. J. & Zierath, J. R. Integrative biology of exercise. Cell 159, 738–749 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. Fan, W. & Evans, R. M. Exercise Mimetics: Impact on Health and Performance. Cell. Metabolism 25, 242–247 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Egan, B., Hawley, J. A. & Zierath, J. R. SnapShot: exercise metabolism. Cell Metab. 24, 342–342.e1 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Lin, J., Handschin, C. & Spiegelman, B. M. Metabolic control through the PGC-1 family of transcription coactivators. Cell. Metabolism 1, 361–370 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Fan, W. & Evans, R. PPARs and ERRs: molecular mediators of mitochondrial metabolism. Curr. Opin. Cell Biol. 33, 49–54 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Dufour, C. R. et al. Genome-wide orchestration of cardiac functions by the orphan nuclear receptors ERRalpha and gamma. Cell. Metabolism 5, 345–356 (2007).

    Article  CAS  PubMed  Google Scholar 

  34. Kelly, D. P. & Scarpulla, R. C. Transcriptional regulatory circuits controlling mitochondrial biogenesis and function. Genes Dev. 18, 357–368 (2004).

    Article  CAS  PubMed  Google Scholar 

  35. Silveira, L. R., Pilegaard, H., Kusuhara, K., Curi, R. & Hellsten, Y. The contraction induced increase in gene expression of peroxisome proliferator-activated receptor (PPAR)-γ coactivator 1α (PGC-1α), mitochondrial uncoupling protein 3 (UCP3) and hexokinase II (HKII) in primary rat skeletal muscle cells is dependent on reactive oxygen species. Biochim. Biophys. Acta 1763, 969–976 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Irrcher, I., Ljubicic, V. & Hood, D. A. Interactions between ROS and AMP kinase activity in the regulation of PGC-1α transcription in skeletal muscle cells. Am. J. Physiol. Cell Physiol. 296, C116–C123 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. Yan, Z., Okutsu, M., Akhtar, Y. N. & Lira, V. A. Regulation of exercise-induced fiber type transformation, mitochondrial biogenesis, and angiogenesis in skeletal muscle. J. Appl. Physiol. 110, 264–274 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Prior, B. M., Yang, H. T. & Terjung, R. L. What makes vessels grow with exercise training? J. Appl. Physiol. 97, 1119–1128 (2004).

    Article  PubMed  Google Scholar 

  39. Gustafsson, T. & Kraus, W. E. Exercise-induced angiogenesis-related growth and transcription factors in skeletal muscle, and their modification in muscle pathology. Front. Biosci. 6, D75–D89 (2001).

    CAS  PubMed  Google Scholar 

  40. Arany, Z. et al. HIF-independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1α. Nature 451, 1008–1012 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Lindholm, M. E. & Rundqvist, H. Skeletal muscle hypoxia-inducible factor-1 and exercise. Exp. Physiol. 101, 28–32 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Semenza, G. L. Regulation of metabolism by hypoxia-inducible factor 1. Cold Spring Harb. Symp. Quant. Biol. 76, 347–353 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Fraisl, P., Aragones, J. & Carmeliet, P. Inhibition of oxygen sensors as a therapeutic strategy for ischaemic and inflammatory disease. Nat. Rev. Drug Discov. 8, 139–152 (2009).

    Article  CAS  PubMed  Google Scholar 

  44. Wong, B. W., Kuchnio, A., Bruning, U. & Carmeliet, P. Emerging novel functions of the oxygen-sensing prolyl hydroxylase domain enzymes. Trends Biochem. Sci. 38, 3–11 (2013).

    Article  CAS  PubMed  Google Scholar 

  45. Semenza, G. L. Oxygen sensing, homeostasis, and disease. N. Engl. J. Med. 365, 537–547 (2011).

    Article  CAS  PubMed  Google Scholar 

  46. Chazaud, B. Inflammation during skeletal muscle regeneration and tissue remodeling: application to exercise-induced muscle damage management. Immunol. Cell Biol. 94, 140–145 (2016).

    Article  CAS  PubMed  Google Scholar 

  47. Lieber, R. L., Thornell, L. E. & Friden, J. Muscle cytoskeletal disruption occurs within the first 15 min of cyclic eccentric contraction. J. Appl. Physiol. 80, 278–284 (1996).

    Article  CAS  PubMed  Google Scholar 

  48. Nunes-Silva, A. et al. Treadmill exercise induces neutrophil recruitment into muscle tissue in a reactive oxygen species-dependent manner. An intravital microscopy study. PLoS ONE 9, e96464 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Fielding, R. A. et al. Acute phase response in exercise. III. Neutrophil and IL-1ß accumulation in skeletal muscle. Am. J. Physiol. 265, R166–R172 (1993).

    Article  CAS  PubMed  Google Scholar 

  50. Marklund, P. et al. Extensive inflammatory cell infiltration in human skeletal muscle in response to an ultraendurance exercise bout in experienced athletes. J. Appl. Physiol. 114, 66–72 (1985).

    Article  CAS  Google Scholar 

  51. Tidball, J. G. Regulation of muscle growth and regeneration by the immune system. Nat. Rev. Immunol. 17, 165–178 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Neubauer, O. et al. Time course-dependent changes in the transcriptome of human skeletal muscle during recovery from endurance exercise: from inflammation to adaptive remodeling. J. Appl. Physiol. 116, 274–287 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Peake, J. M., Neubauer, O., Walsh, N. P. & Simpson, R. J. Recovery of the immune system after exercise. J. Appl. Physiol. 122, 1077–1087 (1985).

    Article  CAS  Google Scholar 

  54. Arnold, L. et al. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J. Exp. Med. 204, 1057–1069 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Deng, B., Wehling-Henricks, M., Villalta, S. A., Wang, Y. & Tidball, J. G. IL-10 triggers changes in macrophage phenotype that promote muscle growth and regeneration. J. Immunol. 189, 3669–3680 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Tonkin, J. et al. Monocyte/macrophage-derived IGF-1 orchestrates murine skeletal muscle regeneration and modulates autocrine polarization. Mol. Ther. 23, 1189–1200 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ginhoux, F., Schultze, J. L., Murray, P. J., Ochando, J. & Biswas, S. K. New insights into the multidimensional concept of macrophage ontogeny, activation and function. Nat. Immunol. 17, 34–40 (2016).

    Article  CAS  PubMed  Google Scholar 

  58. Goh, J., Goh, K. P. & Abbasi, A. Exercise and adipose tissue macrophages: new frontiers in obesity research? Front. Endocrinol. (Lausanne) 7, 65 (2016).

    Article  Google Scholar 

  59. Furrer, R., Eisele, P. S., Schmidt, A., Beer, M. & Handschin, C. Paracrine cross-talk between skeletal muscle and macrophages in exercise by PGC-1α-controlled BNP. Sci. Rep. 7, 40789 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kotas, M. E. & Medzhitov, R. Homeostasis, inflammation, and disease susceptibility. Cell 160, 816–827 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Karsenty, G. & Olson, E. N. Bone and muscle endocrine functions: unexpected paradigms of inter-organ communication. Cell 164, 1248–1256 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Karsenty, G. & Ferron, M. The contribution of bone to whole-organism physiology. Nature 481, 314–320 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Safdar, A., Saleem, A. & Tarnopolsky, M. A. The potential of endurance exercise-derived exosomes to treat metabolic diseases. Nat. Rev. Endocrinol. 12, 504–517 (2016).

    Article  CAS  PubMed  Google Scholar 

  64. Goodwin, P. J., Meyerhardt, J. A. & Hursting, S. D. Host factors and cancer outcome. J. Clin. Oncol. 28, 4019–4021 (2010).

    Article  PubMed  Google Scholar 

  65. McTiernan, A. Mechanisms linking physical activity with cancer. Nat. Rev. Cancer 8, 205–211 (2008).

    Article  CAS  PubMed  Google Scholar 

  66. Hanahan, D. & Coussens, L. M. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 21, 309–322 (2012).

    Article  CAS  PubMed  Google Scholar 

  67. Aaronson, S. A. Growth factors and cancer. Science 254, 1146–1153 (1991).

    Article  CAS  PubMed  Google Scholar 

  68. Rathmell, J. C., Vander Heiden, M. G., Harris, M. H., Frauwirth, K. A. & Thompson, C. B. In the absence of extrinsic signals, nutrient utilization by lymphocytes is insufficient to maintain either cell size or viability. Mol. Cell 6, 683–692 (2000).

    Article  CAS  PubMed  Google Scholar 

  69. Pavlova, N. N. & Thompson, C. B. The emerging hallmarks of cancer metabolism. Cell Metab. 23, 27–47 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Cairns, R. A., Harris, I. S. & Mak, T. W. Regulation of cancer cell metabolism. Nat. Rev. Cancer 11, 85–95 (2011).

    Article  CAS  PubMed  Google Scholar 

  71. Koppenol, W. H., Bounds, P. L. & Dang, C. V. Otto Warburg's contributions to current concepts of cancer metabolism. Nat. Rev. Cancer 11, 325–337 (2011).

    Article  CAS  PubMed  Google Scholar 

  72. Raitakari, O. T., Porkka, K. V., Rasanen, L. & Viikari, J. S. Relations of life-style with lipids, blood pressure and insulin in adolescents and young adults. The cardiovascular risk young finns study. Atherosclerosis 111, 237–246 (1994).

    Article  CAS  PubMed  Google Scholar 

  73. Barnard, R. J. Effects of life-style modification on serum lipids. Arch. Intern. Med. 151, 1389–1394 (1991).

    Article  CAS  PubMed  Google Scholar 

  74. Stanford, K. I. & Goodyear, L. J. Exercise and type 2 diabetes: molecular mechanisms regulating glucose uptake in skeletal muscle. Adv. Physiol. Educ. 38, 308–314 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Irwin, M. L. et al. Randomized controlled trial of aerobic exercise on insulin and insulin-like growth factors in breast cancer survivors: the Yale Exercise and Survivorship study. Cancer Epidemiol. 18, 306–313 (2009).

    Article  CAS  Google Scholar 

  76. Fairey, A. S. et al. Effects of exercise training on fasting insulin, insulin resistance, insulin-like growth factors, and insulin-like growth factor binding proteins in postmenopausal breast cancer survivors: a randomized controlled trial. Cancer Epidemiol. 12, 721–727 (2003).

    CAS  Google Scholar 

  77. Zhu, Z. et al. Effect of nonmotorized wheel running on mammary carcinogenesis: circulating biomarkers, cellular processes, and molecular mechanisms in rats. Cancer Epidemiol. 17, 1920–1929 (2008).

    Article  CAS  Google Scholar 

  78. Thompson, H. J., Wolfe, P., McTiernan, A., Jiang, W. & Zhu, Z. Wheel running-induced changes in plasma biomarkers and carcinogenic response in the 1-methyl-1-nitrosourea-induced rat model for breast cancer. Cancer Prevention Res. 3, 1484–1492 (2010).

    Article  CAS  Google Scholar 

  79. Xie, L. et al. Effects of dietary calorie restriction or exercise on the PI3K and Ras signaling pathways in the skin of mice. J. Biol. Chem. 282, 28025–28035 (2007).

    Article  CAS  PubMed  Google Scholar 

  80. Glass, O. et al. Differential response to exercise in claudin-low breast cancer. Oncotarget (in the press).

  81. Morikawa, T. et al. Association of CTNNB1 (ß-catenin) alterations, body mass index, and physical activity with survival in patients with colorectal cancer. JAMA 305, 1685–1694 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Klaus, A. & Birchmeier, W. Wnt signalling and its impact on development and cancer. Nat. Rev. Cancer 8, 387–398 (2008).

    Article  CAS  PubMed  Google Scholar 

  83. Hanyuda, A. et al. Survival benefit of exercise differs by tumor IRS1 expression status in colorectal cancer. Ann. Surg. Oncol. 23, 908–917 (2016).

    Article  PubMed  Google Scholar 

  84. Slattery, M. L. et al. Associations among IRS1, IRS2, IGF1, and IGFBP3 genetic polymorphisms and colorectal cancer. Cancer Epidemiol. 13, 1206–1214 (2004).

    CAS  Google Scholar 

  85. Pollak, M. Insulin and insulin-like growth factor signalling in neoplasia. Nat. Rev. Cancer 8, 915–928 (2008).

    Article  CAS  PubMed  Google Scholar 

  86. Kalaany, N. Y. & Sabatini, D. M. Tumours with PI3K activation are resistant to dietary restriction. Nature 458, 725–731 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Jiang, X., Overholtzer, M. & Thompson, C. B. Autophagy in cellular metabolism and cancer. J. Clin. Invest. 125, 47–54 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Amaravadi, R. & Debnath, J. Mouse models address key concerns regarding autophagy inhibition in cancer therapy. Cancer Discov. 4, 873–875 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Yang, A. et al. Autophagy is critical for pancreatic tumor growth and progression in tumors with p53 alterations. Cancer Discov. 4, 905–913 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 12, 401–410 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. He, C. et al. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature 481, 511–515 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. He, C., Sumpter, R. Jr & Levine, B. Exercise induces autophagy in peripheral tissues and in the brain. Autophagy 8, 1548–1551 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Aveseh, M., Nikooie, R. & Aminaie, M. Exercise-induced changes in tumour LDH-B and MCT1 expression are modulated by oestrogen-related receptor alpha in breast cancer-bearing BALB/c mice. J. Physiol. 593, 2635–2648 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Semenza, G. L. Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 3, 721–732 (2003).

    Article  CAS  PubMed  Google Scholar 

  95. Carmeliet, P. & Jain, R. K. Angiogenesis in cancer and other diseases. Nature 407, 249–257 (2000).

    Article  CAS  PubMed  Google Scholar 

  96. Betof, A. S. et al. Modulation of murine breast tumor vascularity, hypoxia and chemotherapeutic response by exercise. J. Natl Cancer Inst. 107, djv040 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Jones, L. W. et al. Effect of aerobic exercise on tumor physiology in an animal model of human breast cancer. J. Appl. Physiol. 108, 343–348 (2010).

    Article  PubMed  Google Scholar 

  98. McCullough, D. J., Stabley, J. N., Siemann, D. W. & Behnke, B. J. Modulation of blood flow, hypoxia, and vascular function in orthotopic prostate tumors during exercise. J. Natl Cancer Inst. 106, dju036 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Schadler, K. L. et al. Tumor vessel normalization after aerobic exercise enhances chemotherapeutic efficacy. Oncotarget 7, 65429–65440 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  100. Kashiwagi, S. et al. Perivascular nitric oxide gradients normalize tumor vasculature. Nat. Med. 14, 255–257 (2008).

    Article  CAS  PubMed  Google Scholar 

  101. Fukumura, D., Kashiwagi, S. & Jain, R. K. The role of nitric oxide in tumour progression. Nat. Rev. Cancer 6, 521–534 (2006).

    Article  CAS  PubMed  Google Scholar 

  102. Psaila, B. & Lyden, D. The metastatic niche: adapting the foreign soil. Nat. Rev. Cancer 9, 285–293 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Erler, J. T. et al. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell 15, 35–44 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Cox, T. R. et al. The hypoxic cancer secretome induces pre-metastatic bone lesions through lysyl oxidase. Nature 522, 106–110 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Sceneay, J. et al. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 72, 3906–3911 (2012).

    Article  CAS  PubMed  Google Scholar 

  106. Chafe, S. C. et al. Carbonic anhydrase IX promotes myeloid-derived suppressor cell mobilization and establishment of a metastatic niche by stimulating G-CSF production. Cancer Res. 75, 996–1008 (2015).

    Article  CAS  PubMed  Google Scholar 

  107. 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 

  108. Gao, D. et al. Endothelial progenitor cells control the angiogenic switch in mouse lung metastasis. Science 319, 195–198 (2008).

    Article  CAS  PubMed  Google Scholar 

  109. Jones, L. W. et al. Modulation of circulating angiogenic factors and tumor biology by aerobic training in breast cancer patients receiving neoadjuvant chemotherapy. Cancer Prevention Res. 6, 925–937 (2013).

    Article  CAS  Google Scholar 

  110. Pearson, M. J. & Smart, N. A. Effect of exercise training on endothelial function in heart failure patients: a systematic review meta-analysis. Int. J. Cardiol. 231, 234–243 (2017).

    Article  CAS  PubMed  Google Scholar 

  111. Jain, R. K. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307, 58–62 (2005).

    Article  CAS  PubMed  Google Scholar 

  112. Hatfield, S. M. et al. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci. Trans. Med. 7, 277ra230 (2015).

    Article  CAS  Google Scholar 

  113. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  114. Kruger, K., Lechtermann, A., Fobker, M., Volker, K. & Mooren, F. C. Exercise-induced redistribution of T lymphocytes is regulated by adrenergic mechanisms. Brain Behav. Immun. 22, 324–338 (2008).

    Article  CAS  PubMed  Google Scholar 

  115. Walsh, N. P. et al. Position statement. Part one: Immune function and exercise. Exerc. Immunol. Rev. 17, 6–63 (2011).

    PubMed  Google Scholar 

  116. Timmerman, K. L., Flynn, M. G., Coen, P. M., Markofski, M. M. & Pence, B. D. Exercise training-induced lowering of inflammatory (CD14+CD16+) monocytes: a role in the anti-inflammatory influence of exercise? J. Leukoc. Biol. 84, 1271–1278 (2008).

    Article  CAS  PubMed  Google Scholar 

  117. Sloan, R. P. et al. Aerobic exercise attenuates inducible TNF production in humans. J. Appl. Physiol. 103, 1007–1011 (1985).

    Article  Google Scholar 

  118. Pyne, D. B. et al. Effects of an intensive 12-wk training program by elite swimmers on neutrophil oxidative activity. Med. Sci. Sports Exerc. 27, 536–542 (1995).

    Article  CAS  PubMed  Google Scholar 

  119. Hack, V., Strobel, G., Weiss, M. & Weicker, H. PMN cell counts and phagocytic activity of highly trained athletes depend on training period. J. Appl. Physiol. 77, 1731–1735 (1985).

    Article  Google Scholar 

  120. Woods, J. A. et al. Effects of 6 months of moderate aerobic exercise training on immune function in the elderly. Mech. Ageing Dev. 109, 1–19 (1999).

    Article  CAS  PubMed  Google Scholar 

  121. Kawanishi, N., Yano, H., Yokogawa, Y. & Suzuki, K. Exercise training inhibits inflammation in adipose tissue via both suppression of macrophage infiltration and acceleration of phenotypic switching from M1 to M2 macrophages in high-fat-diet-induced obese mice. Exerc. Immunol. Rev. 16, 105–118 (2010).

    PubMed  Google Scholar 

  122. Kizaki, T. et al. Adaptation of macrophages to exercise training improves innate immunity. Biochem. Biophys. Res. Commun. 372, 152–156 (2008).

    Article  CAS  PubMed  Google Scholar 

  123. Sugiura, H., Nishida, H., Sugiura, H. & Mirbod, S. M. Immunomodulatory action of chronic exercise on macrophage and lymphocyte cytokine production in mice. Acta Physiol. Scand. 174, 247–256 (2002).

    Article  CAS  PubMed  Google Scholar 

  124. Engblom, C., Pfirschke, C. & Pittet, M. J. The role of myeloid cells in cancer therapies. Nat. Rev. Cancer 16, 447–462 (2016).

    Article  CAS  PubMed  Google Scholar 

  125. Abdalla, D. R., Aleixo, A. A., Murta, E. F. & Michelin, M. A. Innate immune response adaptation in mice subjected to administration of DMBA and physical activity. Oncol. Lett. 7, 886–890 (2014).

    Article  CAS  PubMed  Google Scholar 

  126. Murphy, E. A. et al. Effects of moderate exercise and oat beta-glucan on lung tumor metastases and macrophage antitumor cytotoxicity. J. Appl. Physiol. 97, 955–959 (1985).

    Article  Google Scholar 

  127. Noy, R. & Pollard, J. W. Tumor-associated macrophages: from mechanisms to therapy. Immunity 41, 49–61 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Bai, J. et al. Contact-dependent carcinoma aggregate dispersion by M2a macrophages via ICAM-1 and ß2 integrin interactions. Oncotarget 6, 25295–25307 (2015).

    PubMed  PubMed Central  Google Scholar 

  129. Almeida, P. W. et al. Swim training suppresses tumor growth in mice. J. Appl. Physiol. 107, 261–265 (1985).

    Article  Google Scholar 

  130. Zielinski, M. R., Muenchow, M., Wallig, M. A., Horn, P. L. & Woods, J. A. Exercise delays allogeneic tumor growth and reduces intratumoral inflammation and vascularization. J. Appl. Physiol. 96, 2249–2256 (1985).

    Article  Google Scholar 

  131. McClellan, J. L. et al. Exercise effects on polyp burden and immune markers in the ApcMin/+ mouse model of intestinal tumorigenesis. Int. J. Oncol. 45, 861–868 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Fairey, A. S. et al. Randomized controlled trial of exercise and blood immune function in postmenopausal breast cancer survivors. J. Appl. Physiol. 98, 1534–1540 (1985).

    Article  Google Scholar 

  133. Pedersen, L. et al. Voluntary running suppresses tumor growth through epinephrine- and IL-6-Dependent NK cell mobilization and redistribution. Cell Metab. 23, 554–562 (2016).

    Article  CAS  PubMed  Google Scholar 

  134. Simpson, R. J., Florida-James, G. D., Whyte, G. P. & Guy, K. The effects of intensive, moderate and downhill treadmill running on human blood lymphocytes expressing the adhesion/activation molecules CD54 (ICAM-1), CD18 (ß2 integrin) and CD53. Eur. J. Appl. Physiol. 97, 109–121 (2006).

    Article  CAS  PubMed  Google Scholar 

  135. LaVoy, E. C., Bosch, J. A., Lowder, T. W. & Simpson, R. J. Acute aerobic exercise in humans increases cytokine expression in CD27 but not CD27+ CD8+ T-cells. Brain Behav. Immun. 27, 54–62 (2013).

    Article  CAS  PubMed  Google Scholar 

  136. Silva, L. C. et al. Moderate and intense exercise lifestyles attenuate the effects of aging on telomere length and the survival and composition of T cell subpopulations. Age (Dordr) 38, 24 (2016).

    Article  CAS  Google Scholar 

  137. Wang, J. et al. Effect of exercise training intensity on murine T-regulatory cells and vaccination response. Scand. J. Med. Sci. Sports 22, 643–652 (2012).

    Article  CAS  PubMed  Google Scholar 

  138. Glass, O. K. et al. Effect of aerobic training on the host systemic milieu in patients with solid tumours: an exploratory correlative study. Br. J. Cancer 112, 825–831 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Angelin, A. et al. Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell. Metab. 25, 1282–1293.e7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Facciabene, A. et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and Treg cells. Nature 475, 226–230 (2011).

    Article  CAS  PubMed  Google Scholar 

  141. Carmona-Fontaine, C. et al. Emergence of spatial structure in the tumor microenvironment due to the Warburg effect. Proc. Natl Acad. Sci. USA 110, 19402–19407 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Colegio, O. R. et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 513, 559–563 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Carmona-Fontaine, C. et al. Metabolic origins of spatial organization in the tumor microenvironment. Proc. Natl Acad. Sci. USA 114, 2934–2939 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Roodman, G. D. Mechanisms of bone metastasis. N. Engl. J. Med. 350, 1655–1664 (2004).

    Article  CAS  PubMed  Google Scholar 

  145. Jones, L. W. Precision oncology framework for investigation of exercise as treatment for cancer. J. Clin. Oncol. 33, 4134–4137 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  146. Schmitz, K. H. et al. American College of Sports Medicine roundtable on exercise guidelines for cancer survivors. Med. Sci. Sports Exerc. 42, 1409–1426 (2010).

    Article  PubMed  Google Scholar 

  147. Sasso, J. P. et al. A framework for prescription in exercise-oncology research. J. Cachexia Sarcopenia Muscle 6, 115–124 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  148. van Waart, H. et al. Effect of low-intensity physical activity and moderate- to high-intensity physical exercise during adjuvant chemotherapy on physical fitness, fatigue, and chemotherapy completion rates: results of the PACES randomized clinical trial. J. Clin. Oncol. 33, 1918–1927 (2015).

    Article  CAS  PubMed  Google Scholar 

  149. Courneya, K. S. et al. Effects of exercise dose and type during breast cancer chemotherapy: multicenter randomized trial. J. Natl Cancer Inst. 105, 1821–1832 (2013).

    Article  PubMed  Google Scholar 

  150. Courneya, K. S. et al. Effects of aerobic and resistance exercise in breast cancer patients receiving adjuvant chemotherapy: a multicenter randomized controlled trial. J. Clin. Oncol. 25, 4396–4404 (2007).

    Article  PubMed  Google Scholar 

  151. Courneya, K. S. et al. The colon health and life-long exercise change trial: a randomized trial of the national cancer institute of canada clinical trials group. Curr. Oncol. 15, 279–285 (2008).

    Article  CAS  PubMed  Google Scholar 

  152. Kitano, H. Biological robustness. Nat. Rev. Genet. 5, 826–837 (2004).

    Article  CAS  PubMed  Google Scholar 

  153. Kitano, H. Cancer robustness: tumour tactics. Nature 426, 125 (2003).

    Article  CAS  PubMed  Google Scholar 

  154. Greenhaff, P. L. & Hargreaves, M. 'Systems biology' in human exercise physiology: is it something different from integrative physiology? J. Physiol. 589, 1031–1036 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Baker, J. M., De Lisio, M. & Parise, G. Endurance exercise training promotes medullary hematopoiesis. FASEB J. 25, 4348–4357 (2011).

    Article  CAS  PubMed  Google Scholar 

  156. David, V. et al. Mechanical loading down-regulates peroxisome proliferator-activated receptor gamma in bone marrow stromal cells and favors osteoblastogenesis at the expense of adipogenesis. Endocrinology 148, 2553–2562 (2007).

    Article  CAS  PubMed  Google Scholar 

  157. Wallace, J. M., Ron, M. S. & Kohn, D. H. Short-term exercise in mice increases tibial post-yield mechanical properties while two weeks of latency following exercise increases tissue-level strength. Calcif. Tissue Int. 84, 297–304 (2009).

    Article  CAS  PubMed  Google Scholar 

  158. Pichler, K. et al. RANKL is downregulated in bone cells by physical activity (treadmill and vibration stimulation training) in rat with glucocorticoid-induced osteoporosis. Histol. Histopathol. 28, 1185–1196 (2013).

    CAS  PubMed  Google Scholar 

  159. Croucher, P. I., McDonald, M. M. & Martin, T. J. Bone metastasis: the importance of the neighbourhood. Nat. Rev. Cancer 16, 373–386 (2016).

    Article  CAS  PubMed  Google Scholar 

  160. De Lisio, M. & Parise, G. Characterization of the effects of exercise training on hematopoietic stem cell quantity and function. J. Appl. Physiol. 113, 1576–1584 (1985).

    Article  Google Scholar 

  161. Reagan, M. R. & Rosen, C. J. Navigating the bone marrow niche: translational insights and cancer-driven dysfunction. Nat. Rev. Rheumatol 12, 154–168 (2016).

    Article  CAS  PubMed  Google Scholar 

  162. Roodman, G. D. Genes associate with abnormal bone cell activity in bone metastasis. Cancer Metastasis Rev. 31, 569–578 (2012).

    Article  CAS  PubMed  Google Scholar 

  163. De Lisio, M., Baker, J. M. & Parise, G. Exercise promotes bone marrow cell survival and recipient reconstitution post-bone marrow transplantation, which is associated with increased survival. Exp. Hematol. 41, 143–154 (2013).

    Article  PubMed  Google Scholar 

  164. Shiozawa, Y. et al. Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J. Clin. Invest. 121, 1298–1312 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Pagnotti, G. M. & Styner, M. Exercise regulation of marrow adipose tissue. Front. Endocrinol. (Lausanne) 7, 94 (2016).

    Article  Google Scholar 

  166. Emmons, R., Niemiro, G. M., Owolabi, O. & De Lisio, M. Acute exercise mobilizes hematopoietic stem and progenitor cells and alters the mesenchymal stromal cell secretome. J. Appl. Physiol. 120, 624–632 (2016) (1985).

    Article  CAS  PubMed  Google Scholar 

  167. Liu, F., Poursine-Laurent, J. & Link, D. C. Expression of the G-CSF receptor on hematopoietic progenitor cells is not required for their mobilization by G-CSF. Blood 95, 3025–3031 (2000).

    CAS  PubMed  Google Scholar 

  168. Hardaway, A. L., Herroon, M. K., Rajagurubandara, E. & Podgorski, I. Bone marrow fat: linking adipocyte-induced inflammation with skeletal metastases. Cancer Metastasis Rev. 33, 527–543 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Fazeli, P. K. et al. Marrow fat and bone—new perspectives. J. Clin. Endocrinol. Metab. 98, 935–945 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. American Thoracic Society & American College of Chest Physicians. ATS/ACCP Statement on cardiopulmonary exercise testing. Am. J. Respir. Crit. Care Med. 167, 211–277 (2003).

  171. Davis, J. A. Anaerobic threshold: review of the concept and directions for future research. Med. Sci. Sports Exerc. 17, 6–21 (1985).

    CAS  PubMed  Google Scholar 

  172. Convertino, V. A. Blood volume response to physical activity and inactivity. Am. J. Med. Sci. 334, 72–79 (2007).

    Article  PubMed  Google Scholar 

  173. Bishop, D. J., Granata, C. & Eynon, N. Can we optimise the exercise training prescription to maximise improvements in mitochondria function and content? Biochim. Biophys. Acta 1840, 1266–1275 (2014).

    Article  CAS  PubMed  Google Scholar 

  174. MacDougall, J. D. et al. Muscle performance and enzymatic adaptations to sprint interval training. J. Appl. Physiol. 84, 2138–2142 (1985).

    Article  Google Scholar 

  175. Hickson, R. C., Hagberg, J. M., Ehsani, A. A. & Holloszy, J. O. Time course of the adaptive responses of aerobic power and heart rate to training. Med. Sci. Sports Exerc. 13, 17–20 (1981).

    CAS  PubMed  Google Scholar 

  176. Kreher, J. B. & Schwartz, J. B. Overtraining syndrome: a practical guide. Sports Health 4, 128–138 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  177. Fry, R. W., Morton, A. R. & Keast, D. Periodisation of training stress—a review. Can. J. Sport Sci. 17, 234–240 (1992).

    CAS  PubMed  Google Scholar 

  178. Fry, R. W., Morton, A. R. & Keast, D. Periodisation and the prevention of overtraining. Can. J. Sport Sci. 17, 241–248 (1992).

    CAS  PubMed  Google Scholar 

  179. Rusch, H. P. & Kline, B. E. The effect of exercise on the growth of a mouse tumor. Cancer Res. 4, 116–118 (1944).

    Google Scholar 

  180. Winningham, M. L., MacVicar, M. G., Bondoc, M., Anderson, J. I. & Minton, J. P. Effect of aerobic exercise on body weight and composition in patients with breast cancer on adjuvant chemotherapy. Oncol. Nurs. Forum 16, 683–689 (1989).

    CAS  PubMed  Google Scholar 

  181. Segal, R. et al. Structured exercise improves physical functioning in women with stages I and II breast cancer: results of a randomized controlled trial. J. Clin. Oncol. 19, 657–665 (2001).

    Article  CAS  PubMed  Google Scholar 

  182. Brown, J. K. et al. Nutrition and physical activity during and after cancer treatment: an American Cancer Society guide for informed choices. CA Cancer J. Clin. 53, 268–291 (2003).

    Article  PubMed  Google Scholar 

  183. Holmes, M. D., Chen, W. Y., Feskanich, D., Kroenke, C. H. & Colditz, G. A. Physical activity and survival after breast cancer diagnosis. JAMA 293, 2479–2486 (2005).

    Article  CAS  PubMed  Google Scholar 

  184. Meyerhardt, J. A. et al. Interaction of molecular markers and physical activity on mortality in patients with colon cancer. Clin. Cancer Res. 15, 5931–5936 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

L.W.J. is supported by research grants from the National Cancer Institute, AKTIV Against Cancer and the Memorial Sloan Kettering Cancer Center Support Grant/Core Grant (P30 CA008748). The authors would like to thank N. Eves and anonymous reviewers for insights and feedback on earlier versions of this manuscript as well as W. Underwood for administrative support. The authors apologize to the many authors whose work we were unable to cite owing to space constraints.

Author information

Authors and Affiliations

Authors

Contributions

G.J.K., D.F.Q. and L.W.J. researched data for the article, made substantial contributions to the discussion of content and wrote the manuscript. All authors reviewed and edited the manuscript before submission. G.J.K. and D.F.Q. are joint first authors.

Corresponding author

Correspondence to Lee W. Jones.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Glossary

Caloric restriction

Reduction in calorie consumption.

Cardiorespiratory fitness

The integrative capacity of the pulmonary and cardiovascular systems, the vasculature, blood and skeletal muscle to deliver and/or utilize oxygen from the environment to the skeletal muscle mitochondria.

Chronic exercise

Structured, repeated and purposeful physical activity with the objective of improving health or cardiorespiratory fitness.

Exercise prescription

A specific regimen with details regarding the frequency, duration, modality, intensity and length of exercise training.

Homeostasis

The need to maintain a stable internal environment in the face of pathological and physiological perturbations.

Homeostatic control circuits

Specialized homeostatic systems that operate at the cellular, tissue and systemic levels to regulate physiological processes within predefined ranges ('set points').

Pathological angiogenesis

Dysfunctional or non-productive angiogenesis that results in pathological phenotypes.

Physical activity

Any bodily movement produced by skeletal muscles that requires energy expenditure above resting levels.

Physiological angiogenesis

Functional or productive angiogenesis that results in physiological phenotypes.

Pre-metastatic niche

Secondary organ microenvironments (for example, lung, liver, brain or bone) that undergo molecular and possibly architectural alterations to augment engraftment and survival of subsequent tumour cell arrival.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Koelwyn, G., Quail, D., Zhang, X. et al. Exercise-dependent regulation of the tumour microenvironment. Nat Rev Cancer 17, 620–632 (2017). https://doi.org/10.1038/nrc.2017.78

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc.2017.78

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