Skip to main content

Microenvironment Influences Cancer Cell Mechanics from Tumor Growth to Metastasis

  • Chapter
  • First Online:
Biomechanics in Oncology

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 1092))

Abstract

The microenvironment in a solid tumor includes a multitude of cell types, matrix proteins, and growth factors that profoundly influence cancer cell mechanics by providing both physical and chemical stimulation. This tumor microenvironment, which is both dynamic and heterogeneous in nature, plays a critical role in cancer progression from the growth of the primary tumor to the development of metastatic and drug-resistant tumors. This chapter provides an overview of the biophysical tools used to study cancer cell mechanics and mechanical changes in the tumor microenvironment at different stages of cancer progression, including growth of the primary tumor, local invasion, and metastasis. Quantitative single cell biophysical analysis of intracellular mechanics, cell traction forces, and cell motility can easily be combined with analysis of critical cell fate processes, including adhesion, proliferation, and drug resistance, to determine how changes in mechanics contribute to cancer progression. This biophysical approach can be used to systematically investigate the parameters in the tumor that control cancer cell interactions with the stroma and to identify specific conditions that induce tumor-promoting behavior, along with strategies for inhibiting these conditions to treat cancer. Increased understanding of the underlying biophysical mechanisms that drive cancer progression may provide insight into novel therapeutic approaches in the fight against cancer.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

eBook
USD 16.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 129.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 179.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

References

  1. Mehlen P, Puisieux A (2006) Metastasis: a question of life or death. Nat Rev Cancer 6:449–458

    Article  CAS  PubMed  Google Scholar 

  2. Wirtz D, Konstantopoulos K, Searson PC (2011) The physics of cancer: the role of physical interactions and mechanical forces in metastasis. Nat Rev Cancer 11:522

    Article  CAS  Google Scholar 

  3. Kumar S, Weaver VM (2009) Mechanics, malignancy, and metastasis: the force journey of a tumor cell. Cancer Metastasis Rev 28:113–127

    Article  PubMed  PubMed Central  Google Scholar 

  4. Butcher DT, Alliston T, Weaver VM (2009) A tense situation: forcing tumour progression. Nat Rev Cancer 9:108–122

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Whiteside TL (2008) The tumor microenvironment and its role in promoting tumor growth. Oncogene 27:5904–5912

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Joyce JA (2005) Therapeutic targeting of the tumor microenvironment. Cancer Cell 7:513–520

    Article  CAS  PubMed  Google Scholar 

  7. Joyce JA, Pollard JW (2009) Microenvironmental regulation of metastasis. Nat Rev Cancer 9:239–252

    Article  CAS  PubMed  Google Scholar 

  8. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell 100:57–70

    Article  CAS  PubMed  Google Scholar 

  9. Hanahan D, Weinberg RA, Pan KH, Shay JW, Cohen SN, Taylor MB, Clarke NW, Jayson GC, Eshleman JR, Nowak MA et al (2011) Hallmarks of cancer: the next generation. Cell 144:646–674

    Article  CAS  PubMed  Google Scholar 

  10. Lu P, Weaver VM, Werb Z (2012) The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol 196:395–406

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, Gefen A, Reinhart-King CA, Margulies SS, Dembo M, Boettiger D, Hammer DA, Weaver VM (2005) Tensional homeostasis and the malignant phenotype. Cancer Cell 8:241–254

    Article  CAS  PubMed  Google Scholar 

  12. McGrail DJ, Kieu QMN, Dawson MR (2014) The malignancy of metastatic ovarian cancer cells is increased on soft matrices through a mechanosensitive rho-ROCK pathway. J Cell Sci 127:2621–2626

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. McGrail DJ, Kieu QMN, Iandoli JA, Dawson MR (2015) Actomyosin tension as a determinant of metastatic cancer mechanical tropism. Phys Biol 12:26001

    Article  CAS  Google Scholar 

  14. McGrail DJ, Mezencev R, Kieu QMN, McDonald JF, Dawson MR (2015) SNAIL-induced epithelial-to-mesenchymal transition produces concerted biophysical changes from altered cytoskeletal gene expression. FASEB J 29:1280–1289

    Article  CAS  PubMed  Google Scholar 

  15. Ghosh D, Lili L, McGrail DJ, Matyunina LV, McDonald JF, Dawson MR (2014) Integral role of platelet-derived growth factor in mediating transforming growth factor-β1-dependent mesenchymal stem cell stiffening. Stem Cells Dev 23:245–261

    Article  CAS  PubMed  Google Scholar 

  16. McGrail DJ, Ghosh D, Quach ND, Dawson MR (2012) Differential mechanical response of mesenchymal stem cells and fibroblasts to tumor-secreted soluble factors. PLoS One 7:e33248

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. McAndrews KM, Yi J, McGrail DJ, Dawson MR (2015) Enhanced adhesion of stromal cells to invasive cancer cells regulated by cadherin 11. ACS Chem Biol 10:1932–1938

    Article  CAS  PubMed  Google Scholar 

  18. McGrail DJ, McAndrews KM, Dawson MR (2013) Biomechanical analysis predicts decreased human mesenchymal stem cell function before molecular differences. Exp Cell Res 319:684–696

    Article  CAS  PubMed  Google Scholar 

  19. McAndrews KM, McGrail DJ, Ravikumar N, Dawson MR (2015) Mesenchymal stem cells induce directional migration of invasive breast cancer cells through TGF-β. Sci Rep 5:16941

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Shieh AC (2011) Biomechanical forces shape the tumor microenvironment. Ann Biomed Eng 39:1379–1389

    Article  PubMed  Google Scholar 

  21. Mierke CT (2013) Physical break-down of the classical view on cancer cell invasion and metastasis. Eur J Cell Biol 92:89–104

    Article  CAS  PubMed  Google Scholar 

  22. Jain RK, Martin JD, Stylianopoulos T (2014) The role of mechanical forces in tumor growth and therapy. Annu Rev Biomed Eng 16:321–346

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Stroka KM, Konstantopoulos K (2014) Physical biology in cancer. 4. Physical cues guide tumor cell adhesion and migration. Am J Physiol Cell Physiol 306:C98–C109

    Article  CAS  PubMed  Google Scholar 

  24. Murrell M, Oakes PW, Lenz M, Gardel ML (2015) Forcing cells into shape: the mechanics of actomyosin contractility. Nat Rev Mol Cell Biol 16:486–498

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Karlsson R, Pedersen ED, Wang Z, Brakebusch C (2009) Rho GTPase function in tumorigenesis. Biochim Biophys Acta 1796:91–98

    CAS  PubMed  Google Scholar 

  26. Vega FM, Ridley AJ (2008) Rho GTPases in cancer cell biology. FEBS Lett 582:2093–2101

    Article  CAS  PubMed  Google Scholar 

  27. Sahai E, Marshall CJ (2002) RHO–GTPases and cancer. Nat Rev Cancer 2:133–142

    Article  PubMed  Google Scholar 

  28. Orgaz JL, Herraiz C, Sanz-Moreno V (2014) Rho GTPases modulate malignant transformation of tumor cells. Small GTPases 5:e29019

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Porter AP, Papaioannou A, Malliri A (2016) Deregulation of rho GTPases in cancer. Small GTPases 7:123–138

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Alam SG, Zhang Q, Prasad N, Li Y, Chamala S, Kuchibhotla R, Kc B, Aggarwal V, Shrestha S, Jones AL, Levy SE, Roux KJ, Nickerson JA, Tanmay, Lele P (2016) The mammalian LINC complex regulates genome transcriptional responses to substrate rigidity. Sci Rep 6:38063

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang N, Tytell JD, Ingber DE (2009) Mechanotransduction at a distance: mechanically coupling the extracellular matrix with the nucleus. Nat Rev Mol Cell Biol 10:75–82

    Article  CAS  PubMed  Google Scholar 

  32. Pritchard RH, Huang YY, Terentjev EM (2014) Mechanics of biological networks: from the cell cytoskeleton to connective tissue. Soft matter 10(12):1864–84.

    Google Scholar 

  33. Suresh S (2007) Biomechanics and biophysics of cancer cells. Acta Mater 55:3989–4014

    Article  CAS  Google Scholar 

  34. Fletcher DA, Mullins RD (2010) Cell mechanics and the cytoskeleton. Nature 463:485–492

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Janmey PA, Hvidt S, Lamb J, Stossel TP (1990) Resemblance of actin-binding protein/actin gels to covalently crosslinked networks. Nature 345:89–92

    Article  CAS  PubMed  Google Scholar 

  36. Hall A (1998) Rho GTPases and the actin cytoskeleton. Science 279:80

    Article  Google Scholar 

  37. Winder SJ, Ayscough KR (2005) Actin-binding proteins. J Cell Sci 118:651

    Article  CAS  PubMed  Google Scholar 

  38. Stricker J, Falzone T, Gardel ML (2010) Mechanics of the F-actin cytoskeleton. J Biomech 43:9–14

    Article  PubMed  Google Scholar 

  39. Hall A (2012) Rho family GTPases. Biochem Soc Trans 40:1378–1382

    Article  CAS  PubMed  Google Scholar 

  40. Janmey PA, Euteneuer U, Traub P, Schliwa M (1991) Viscoelastic properties of vimentin compared with other filamentous biopolymer networks. The Journal of cell biology. 113(1):155–60

    Article  CAS  PubMed  Google Scholar 

  41. Liu C-Y, Lin H-H, Tang M-J, Wang Y-K (2015) Vimentin contributes to epithelial-mesenchymal transition cancer cell mechanics by mediating cytoskeletal organization and focal adhesion maturation. Oncotarget 6:15966–15983

    PubMed  PubMed Central  Google Scholar 

  42. Mendez MG, Kojima S-I, Goldman RD (2010) Vimentin induces changes in cell shape, motility, and adhesion during the epithelial to mesenchymal transition. FASEB J 24:1838–1851

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kollmannsberger P, Fabry B (2011) Linear and nonlinear rheology of living cells. Annu Rev Mater Res 41:75–97

    Article  CAS  Google Scholar 

  44. Chen J (2014) Nanobiomechanics of living cells: a review. Interface Focus 4(2):20130055

    Article  PubMed  PubMed Central  Google Scholar 

  45. Hoffman BD, Crocker JC (2009) Cell mechanics: dissecting the physical responses of cells to force. Annu Rev Biomed Eng 11:259–288

    Article  CAS  PubMed  Google Scholar 

  46. Moeendarbary E, Harris AR (2014) Cell mechanics: principles, practices, and prospects. Wiley Interdiscip Rev Syst Biol Med 6:371–388

    Article  PubMed  PubMed Central  Google Scholar 

  47. Wirtz D (2009) Particle-tracking microrheology of living cells: principles and applications. Annu Rev Biophys 38:301–326

    Article  CAS  PubMed  Google Scholar 

  48. Crocker JC, Hoffman BD (2007) Multiple-particle tracking and two-point microrheology in cells. Methods Cell Biol 83:141–178

    Article  CAS  PubMed  Google Scholar 

  49. Hoffman BD, Massiera G, Van Citters KM, Crocker JC (2006) The consensus mechanics of cultured mammalian cells. Proc Natl Acad Sci U S A 103:10259–10264

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Darling EM, Zauscher S, Block JA, Guilak F (2007) A thin-layer model for viscoelastic, stress-relaxation testing of cells using atomic force microscopy: do cell properties reflect metastatic potential? Biophys J 92:1784–1791

    Article  CAS  PubMed  Google Scholar 

  51. Lee JSH, Panorchan P, Hale CM, Khatau SB, Kole TP, Tseng Y, Wirtz D (2006) Ballistic intracellular nanorheology reveals ROCK-hard cytoplasmic stiffening response to fluid flow. J Cell Sci 119:1760–1768

    Article  CAS  PubMed  Google Scholar 

  52. Swaminathan V, Mythreye K, O’Brien ET, Berchuck A, Blobe GC, Superfine R (2011) Mechanical stiffness grades metastatic potential in patient tumor cells and in cancer cell lines. Cancer Res 71:5075–5080

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Icard-Arcizet D, Cardoso O, Richert A, Hénon S (2008) Cell stiffening in response to external stress is correlated to actin recruitment. Biophys J 94:2906–2913

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Pachenari M, Seyedpour SM, Janmaleki M, Shayan SB, Taranejoo S, Hosseinkhani H (2014) Mechanical properties of cancer cytoskeleton depend on actin filaments to microtubules content: investigating different grades of colon cancer cell lines. J Biomech 47:373–379

    Article  CAS  PubMed  Google Scholar 

  55. Guck J, Schinkinger S, Lincoln B, Wottawah F, Ebert S, Romeyke M, Lenz D, Erickson HM, Ananthakrishnan R, Mitchell D, Käs J, Ulvick S, Bilby C (2005) Optical deformability as an inherent cell marker for testing malignant transformation and metastatic competence. Biophys J 88:3689–3698

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Dawson MR, Tseng Y, Lee JSH, McAndrews KM (2014) Handbook of imaging in biological mechanics. CRC Press, Boca Raton, pp 381–388

    Book  Google Scholar 

  57. Lin H-H, Lin H-K, Lin I-H, Chiou Y-W, Chen H-W, Liu C-Y, Harn HI-C, Chiu W-T, Wang Y-K, Shen M-R, Tang M-J, Lin H-H, Lin H-K, Lin I-H, Chiou Y-W, Chen H-W, Liu C-Y, Harn HI-C, Chiu W-T, Wang Y-K, Shen M-R, Tang M-J (2015) Mechanical phenotype of cancer cells: cell softening and loss of stiffness sensing. Oncotarget 6: 20946–20958

    PubMed  PubMed Central  Google Scholar 

  58. Alibert C, Goud B, Manneville JB (2017) Are cancer cells really softer than normal cells? Biol Cell 109:167–189. https://doi.org/10.1111/boc.201600078

    Article  PubMed  Google Scholar 

  59. Petrie RJ, Yamada KM (2013) At the leading edge of three-dimensional cell migration. J Cell Sci 125:5917

    Article  CAS  Google Scholar 

  60. Ridley AJ (2001) Rho GTPases and cell migration. J Cell Sci 114:2713

    CAS  PubMed  Google Scholar 

  61. Whirledge S, Dixon D, Cidlowski JA (2012) Glucocorticoids regulate gene expression and repress cellular proliferation in human uterine leiomyoma cells. Horm Cancer 3:79–92

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Huveneers S, Danen EHJ (2009) Adhesion signaling – crosstalk between integrins, Src and rho. J Cell Sci 122:1059

    Article  CAS  PubMed  Google Scholar 

  63. Munevar S, Wang Y, Dembo M (2001) Traction force microscopy of migrating normal and H-ras transformed 3T3 fibroblasts. Biophys J 80:1744–1757

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Sabass B, Gardel ML, Waterman CM, Schwarz US (2008) High resolution traction force microscopy based on experimental and computational advances. Biophys J 94:207–220

    Article  CAS  PubMed  Google Scholar 

  65. McGrail DJ(2015) Mechanics and malignancy: physical cues and changes that drive tumor progression. Georgia Institute of Technology

    Google Scholar 

  66. Kraning-Rush CM, Califano JP, Reinhart-King CA, Thirumurthi U, Dembo M (2012) Cellular traction stresses increase with increasing metastatic potential. PLoS One 7:e32572

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Koch TM, Münster S, Bonakdar N, Butler JP, Fabry B (2012) 3D traction forces in cancer cell invasion. PLoS One 7:e33476

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Levental I, Georges PC, Janmey PA (2007) Soft biological materials and their impact on cell function. Soft Matter 3:299–306

    Article  CAS  PubMed  Google Scholar 

  69. Swift J, Ivanovska IL, Buxboim A, Harada T, Dingal PCDP, Pinter J, Pajerowski JD, Spinler KR, Shin J-W, Tewari M, Rehfeldt F, Speicher DW, Discher DE (2013) Nuclear Lamin-a scales with tissue stiffness and enhances matrix-directed differentiation. Science 341(6149):1240104

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Acerbi I, Cassereau L, Dean I, Shi Q, Au A, Park C, Chen YY, Liphardt J, Hwang ES, Weaver VM (2015) Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration. Integr Biol 7:1120–1134

    Article  CAS  Google Scholar 

  71. Baker A-M, Bird D, Lang G, Cox TR, Erler JT (2013) Lysyl oxidase enzymatic function increases stiffness to drive colorectal cancer progression through FAK. Oncogene 32:1863–1868

    Article  CAS  PubMed  Google Scholar 

  72. Venkatesh SK, Yin M, Glockner JF, Takahashi N, Araoz PA, Talwalkar JA, Ehman RL (2008) MR elastography of liver tumors: preliminary results. AJR Am J Roentgenol 190:1534–1540

    Article  PubMed  PubMed Central  Google Scholar 

  73. Hoyt K, Castaneda B, Zhang M, Nigwekar P, di Sant’agnese PA, Joseph JV, Strang J, Rubens DJ, Parker KJ (2008) Tissue elasticity properties as biomarkers for prostate cancer. Cancer Biomark 4:213–225

    Article  PubMed  PubMed Central  Google Scholar 

  74. Chaudhuri O, Koshy ST, Branco da Cunha C, Shin J-W, Verbeke CS, Allison KH, Mooney DJ (2014) Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat Mater 13:970–978

    Article  CAS  PubMed  Google Scholar 

  75. Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SFT, Csiszar K, Giaccia A, Weninger W, Yamauchi M, Gasser DL, Weaver VM (2009) Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 139:891–906

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Lee S, Kumar S (2016) Actomyosin stress fiber mechanosensing in 2D and 3D. F1000Research 5:2261

    Article  CAS  Google Scholar 

  77. Kraning-Rush CM, Carey SP, Califano JP, Smith BN, Reinhart-King CA (2011) The role of the cytoskeleton in cellular force generation in 2D and 3D environments. Phys Biol 8:15009

    Article  CAS  Google Scholar 

  78. Doyle AD, Carvajal N, Jin A, Matsumoto K, Yamada KM (2015) Local 3D matrix microenvironment regulates cell migration through spatiotemporal dynamics of contractility-dependent adhesions. Nat Commun 6:8720

    Article  CAS  PubMed  Google Scholar 

  79. Polacheck WJ, Chen CS (2016) Measuring cell-generated forces: a guide to the available tools. Nat Methods 13:415–423

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Discher DE, Janmey P, Wang Y (2005) Tissue cells feel and respond to the stiffness of their substrate. Science 310:1139–1143

    Article  CAS  PubMed  Google Scholar 

  81. Chin L, Xia Y, Discher DE, Janmey PA (2016) Mechanotransduction in cancer. Curr Opin Chem Eng 11:77–84

    Article  PubMed  PubMed Central  Google Scholar 

  82. Tse JR, Engler AJ (2010) Preparation of hydrogel substrates with tunable mechanical properties. Curr Protoc Cell Biol 47:10.16.1–10.16.16

    Article  Google Scholar 

  83. Wolf K, Alexander S, Schacht V, Coussens LM, von Andrian UH, van Rheenen J, Deryugina E, Friedl P (2009) Collagen-based cell migration models in vitro and in vivo. Semin Cell Dev Biol 20:931–941

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Leggett SE, Khoo AS, Wong IY (2017) Multicellular tumor invasion and plasticity in biomimetic materials. Biomater Sci 5(8):1460–1479. https://doi.org/10.1039/C7BM00272F

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Nieman KM, Kenny H a, Penicka CV, Ladanyi A, Buell-Gutbrod R, Zillhardt MR, Romero IL, Carey MS, Mills GB, Hotamisligil GS, Yamada SD, Peter ME, Gwin K, Lengyel E (2011) Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat Med 17:1498–1503

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. McAndrews KM, McGrail DJ, Quach ND, Dawson MR (2014) Spatially coordinated changes in intracellular rheology and extracellular force exertion during mesenchymal stem cell differentiation. Phys Biol 11:56004

    Article  CAS  Google Scholar 

  87. Helmlinger G, Netti PA, Lichtenbeld HC, Melder RJ, Jain RK (1997) Solid stress inhibits the growth of multicellular tumor spheroids. Nat Biotechnol 15:778–783

    Article  CAS  PubMed  Google Scholar 

  88. Stylianopoulos T, Martin JD, Chauhan VP, Jain SR, Diop-Frimpong B, Bardeesy N, Smith BL, Ferrone CR, Hornicek FJ, Boucher Y, Munn LL, Jain RK (2012) Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors. Proc Natl Acad Sci 109:15101–15108

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. McGrail DJ, McAndrews KM, Brandenburg CP, Ravikumar N, Kieu QMN, Dawson MR (2015) Osmotic regulation is required for cancer cell survival under solid stress. Biophys J 109:1334–1337

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Thamilselvan V, Basson MD (2004) Pressure activates colon cancer cell adhesion by inside-out focal adhesion complex and actin cytoskeletal signaling. Gastroenterology 126:8–18

    Article  CAS  PubMed  Google Scholar 

  91. Thamilselvan V, Craig DH, Basson MD (2007) FAK association with multiple signal proteins mediates pressure-induced colon cancer cell adhesion via a Src-dependent PI3K/Akt pathway. FASEB J 21:1730–1741

    Article  CAS  PubMed  Google Scholar 

  92. Tse JM, Cheng G, Tyrrell JA, Wilcox-Adelman SA, Boucher Y, Jain RK, Munn LL (2012) Mechanical compression drives cancer cells toward invasive phenotype. Proc Natl Acad Sci 109:911–916

    Article  CAS  PubMed  Google Scholar 

  93. Adhikari AS, Chai J, Dunn AR (2011) Mechanical load induces a 100-fold increase in the rate of collagen proteolysis by MMP-1. J Am Chem Soc 133:1686–1689

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Roh HD, Boucher Y, Kalnicki S, Buchsbaum R, Bloomer WD, Jain RK (1991) Interstitial hypertension in carcinoma of uterine cervix in patients: possible correlation with tumor oxygenation and radiation response. Cancer Res 51:6695–6698

    CAS  PubMed  Google Scholar 

  95. Milosevic M, Fyles A, Hedley D, Pintilie M, Levin W, Manchul L, Hill R (2001) Interstitial fluid pressure predicts survival in patients with cervix cancer independent of clinical prognostic factors and tumor oxygen measurements. Cancer Res 61:6400–6405

    CAS  PubMed  Google Scholar 

  96. Curti BD, Urba WJ, Alvord WG, Janik JE, Smith JW, Madara K, Longo DL (1993) Interstitial pressure of subcutaneous nodules in melanoma and lymphoma patients: changes during treatment. Cancer Res 53:2204–2207

    CAS  PubMed  Google Scholar 

  97. Hofmann M, Guschel M, Bernd A, Bereiter-Hahn J, Kaufmann R, Tandi C, Wiig H, Kippenberger S (2006) Lowering of tumor interstitial fluid pressure reduces tumor cell proliferation in a xenograft tumor model. Neoplasia 8:89–95

    Article  PubMed  PubMed Central  Google Scholar 

  98. Hofmann M, Schultz M, Bernd A, Bereiter-Hahn J, Kaufmann R, Kippenberger S (2007) Long-term lowering of tumour interstitial fluid pressure reduces Ki-67 expression. J Biomech 40:2324

    Article  PubMed  Google Scholar 

  99. Polacheck WJ, Charest JL, Kamm RD (2011) Interstitial flow influences direction of tumor cell migration through competing mechanisms. Proc Natl Acad Sci U S A 108:11115–11120

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Shields JD, Fleury ME, Yong C, Tomei AA, Randolph GJ, Swartz MA (2007) Autologous chemotaxis as a mechanism of tumor cell homing to lymphatics via interstitial flow and autocrine CCR7 signaling. Cancer Cell 11:526–538

    Article  CAS  PubMed  Google Scholar 

  101. Ng CP, Swartz MA (2003) Fibroblast alignment under interstitial fluid flow using a novel 3-D tissue culture model. Am J Physiol Heart Circ Physiol 284:H1771

    Article  CAS  PubMed  Google Scholar 

  102. Ng CP, Hinz B, Swartz MA (2005) Interstitial fluid flow induces myofibroblast differentiation and collagen alignment in vitro. J Cell Sci 118:4731

    Article  CAS  PubMed  Google Scholar 

  103. Shah AD, Bouchard MJ, Shieh AC, Ohtani O, Saiki I, Baffet G (2015) Interstitial fluid flow increases hepatocellular carcinoma cell invasion through CXCR4/CXCL12 and MEK/ERK signaling. PLoS One 10:e0142337

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Shieh AC, Swartz MA (2011) Regulation of tumor invasion by interstitial fluid flow. Phys Biol 8:15012

    Article  Google Scholar 

  105. Shieh AC, Rozansky HA, Hinz B, Swartz MA (2011) Tumor cell invasion is promoted by interstitial flow-induced matrix priming by stromal fibroblasts. Cancer Res 71:790–800

    Article  CAS  PubMed  Google Scholar 

  106. Wipff P-J, Hinz B (2009) Myofibroblasts work best under stress. J Bodyw Mov Ther 13:121–127

    Article  PubMed  Google Scholar 

  107. Chaffer CL, Weinberg RA (2011) A perspective on cancer cell metastasis. Science 331:1559–1564

    Article  CAS  PubMed  Google Scholar 

  108. Xu W, Mezencev R, Kim B, Wang L, McDonald J, Sulchek T (2012) Cell stiffness is a biomarker of the metastatic potential of ovarian cancer cells. PLoS One 7:e46609–e46609

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Thiery JP, Sleeman JP (2006) Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol 7:131–142

    Article  CAS  PubMed  Google Scholar 

  110. Kalluri R, Weinberg RA (2009) The basics of epithelial-mesenchymal transition. J Clin Invest 119:1420–1428

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Polyak K, Weinberg RA (2009) Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer 9:265–273

    Article  CAS  PubMed  Google Scholar 

  112. Przybylo JA, Radisky DC (2007) Matrix metalloproteinase-induced epithelial-mesenchymal transition: tumor progression at Snail's pace. The international journal of biochemistry & cell biology. 39(6):1082–1088.

    Article  CAS  Google Scholar 

  113. Radisky ES, Radisky DC (2010) Matrix metalloproteinase-induced epithelial-mesenchymal transition in breast cancer. J Mammary Gland Biol Neoplasia 15:201–212

    Article  PubMed  PubMed Central  Google Scholar 

  114. Conklin MW, Eickhoff JC, Riching KM, Pehlke CA, Eliceiri KW, Provenzano PP, Friedl A, Keely PJ (2011) Aligned collagen is a prognostic signature for survival in human breast carcinoma. Am J Pathol 178:1221–1232

    Article  PubMed  PubMed Central  Google Scholar 

  115. Bloom RJ, George JP, Celedon A, Sun SX, Wirtz D, Overall C, Stack MS, Friedl P, Matsudaira P, Krane S, Allen E, Chung D, Weiss SJ (2008) Mapping local matrix remodeling induced by a migrating tumor cell using three-dimensional multiple-particle tracking. Biophys J 95:4077–4088

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Fraley SI, Feng Y, Krishnamurthy R, Kim D-H, Celedon A, Longmore GD, Wirtz D (2010) A distinctive role for focal adhesion proteins in three- dimensional cell motility. Nat Cell Biol 12: 598–604

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Zaman MH, Trapani LM, Sieminski AL, MacKellar D, Gong H, Kamm RD, Wells A, Lauffenburger DA, Matsudaira P, Matsudaira P (2006) Migration of tumor cells in 3D matrices is governed by matrix stiffness along with cell-matrix adhesion and proteolysis. Proc Natl Acad Sci 103: 10889–10894

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Alexander S, Weigelin B, Winkler F, Friedl P (2013) Preclinical intravital microscopy of the tumour-stroma interface: invasion, metastasis, and therapy response. Curr Opin Cell Biol 25:659–671

    Article  CAS  PubMed  Google Scholar 

  119. Clark AG, Vignjevic DM (2015) Modes of cancer cell invasion and the role of the microenvironment. Curr Opin Cell Biol 36:13–22

    Article  CAS  PubMed  Google Scholar 

  120. Paul CD, Mistriotis P, Konstantopoulos K (2016) Cancer cell motility: lessons from migration in confined spaces. Nat Rev Cancer 17:131–140

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Ridley AJ, Schwartz MA, Burridge K, Firtel RA, Ginsberg MH, Borisy G, Parsons JT, Horwitz AR (2003) Cell migration: integrating signals from front to back. Science 302:1704–1709

    Article  CAS  PubMed  Google Scholar 

  122. Friedl P, Wolf K (2003) Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer 3:362–374

    Article  CAS  PubMed  Google Scholar 

  123. Wolf K, Mazo I, Leung H, Engelke K, Von Andrian UH, Deryugina EI, Strongin AY, Bröcker EB, Friedl P (2003) Compensation mechanism in tumor cell migration: Mesenchymal-amoeboid transition after blocking of pericellular proteolysis. J Cell Biol 160:267–277

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Te Boekhorst V, Friedl P (2016) Plasticity of cancer cell invasion - mechanisms and implications for therapy. Adv Cancer Res 132:209

    Article  Google Scholar 

  125. Sabeh F, Shimizu-Hirota R, Weiss SJ (2009) Protease-dependent versus -independent cancer cell invasion programs: three-dimensional amoeboid movement revisited. J Cell Biol 185

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Friedl P (2004) Prespecification and plasticity: shifting mechanisms of cell migration. Curr Opin Cell Biol 16:14–23

    Article  CAS  PubMed  Google Scholar 

  127. Sahai E, Marshall CJ (2003) Differing modes of tumour cell invasion have distinct requirements for rho/ROCK signalling and extracellular proteolysis. Nat Cell Biol 5:711–719

    Article  CAS  PubMed  Google Scholar 

  128. Lee M-H, Wu P-H, Staunton JR, Ros R, Longmore GD, Wirtz D (2012) Mismatch in mechanical and adhesive properties induces pulsating cancer cell migration in epithelial monolayer. Biophys J 102:2731–2741

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Friedl P, Gilmour D (2009) Collective cell migration in morphogenesis, regeneration and cancer. Nat Rev Mol Cell Biol 10:445–457

    Article  CAS  PubMed  Google Scholar 

  130. Friedl P, Wolf K, Zegers MM (2014) Rho-directed forces in collective migration. Nat Cell Biol 16: 208–210

    Article  CAS  PubMed  Google Scholar 

  131. Haeger A, Wolf K, Zegers MM, Friedl P (2015) Collective cell migration: guidance principles and hierarchies. Trends Cell Biol 25:556–566

    Article  PubMed  Google Scholar 

  132. Reymond N, D’Água BB, Ridley AJ (2013) Crossing the endothelial barrier during metastasis. Nat Rev Cancer 13:858–870

    Article  CAS  PubMed  Google Scholar 

  133. Wolf K, Wu YI, Liu Y, Geiger J, Tam E, Overall C, Stack MS, Friedl P (2007) Multi-step pericellular proteolysis controls the transition from individual to collective cancer cell invasion. Nat Cell Biol 9: 893–904

    Article  CAS  PubMed  Google Scholar 

  134. Fu Y, Chin LK, Bourouina T, Liu AQ, VanDongen AMJ (2012) Nuclear deformation during breast cancer cell transmigration. Lab Chip 12:3774

    Article  CAS  PubMed  Google Scholar 

  135. Mitchell MJ, King MR (2013) Computational and experimental models of cancer cell response to fluid shear stress. Front Oncol 3:44

    Article  PubMed  PubMed Central  Google Scholar 

  136. Kienast Y, von Baumgarten L, Fuhrmann M, Klinkert WEF, Goldbrunner R, Herms J, Winkler F (2010) Real-time imaging reveals the single steps of brain metastasis formation. Nat Med 16:116–122

    Article  CAS  PubMed  Google Scholar 

  137. Lengyel E (2010) Ovarian cancer development and metastasis. Am J Pathol 177:1053–1064

    Article  PubMed  PubMed Central  Google Scholar 

  138. Desgrosellier JS, Cheresh DA (2010) Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer 10:9–22

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Seguin L, Desgrosellier JS, Weis SM, Cheresh DA (2015) Integrins and cancer: regulators of cancer stemness, metastasis, and drug resistance. Trends Cell Biol 25:234–240

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Cavallaro U, Christofori G (2004) Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer 4:118–132

    Article  CAS  PubMed  Google Scholar 

  141. Paredes J, Figueiredo J, Albergaria A, Oliveira P, Carvalho J, Ribeiro AS, Caldeira J, Costa ÂM, Simões-Correia J, Oliveira MJ, Pinheiro H, Pinho SS, Mateus R, Reis CA, Leite M, Fernandes MS, Schmitt F, Carneiro F, Figueiredo C, Oliveira C, Seruca R (2012) Epithelial E- and P-cadherins: role and clinical significance in cancer. Biochim Biophys Acta 1826:297–311

    CAS  PubMed  Google Scholar 

  142. Canel M, Serrels A, Frame MC, Brunton VG (2013) E-cadherin–integrin crosstalk in cancer invasion and metastasis. J Cell Sci 126:393

    Article  CAS  PubMed  Google Scholar 

  143. Mitra AK, Sawada K, Tiwari P, Mui K, Gwin K, Lengyel E (2011) Ligand-independent activation of c-met by fibronectin and α5β1-integrin regulates ovarian cancer invasion and metastasis. Oncogene 30:1566–1576

    Article  CAS  PubMed  Google Scholar 

  144. Chu K, Cheng C-J, Ye X, Lee Y-C, Zurita AJ, Chen D-T, Yu-Lee L-Y, Zhang S, Yeh ET, Hu MC-T, Logothetis CJ, Lin S-H (2008) Cadherin-11 promotes the metastasis of prostate cancer cells to bone. Mol Cancer Res 6:1259–1267

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordón-Cardo C, Guise TA, Massagué J (2003) A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3:537–549

    Article  CAS  PubMed  Google Scholar 

  146. Minn AJ, Gupta GP, Siegel PM, Bos PD, Shu W, Giri DD, Viale A, Olshen AB, Gerald WL, Massagué J (2005) Genes that mediate breast cancer metastasis to lung. Nature 436:518–524

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Bos PD, Zhang XH-F, Nadal C, Shu W, Gomis RR, Nguyen DX, Minn AJ, van de Vijver MJ, Gerald WL, Foekens J a, Massagué J (2009) Genes that mediate breast cancer metastasis to the brain. Nature 459:1005–1009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Kostic A, Lynch CD, Sheetz MP (2009) Differential matrix rigidity response in breast cancer cell lines correlates with the tissue tropism. PLoS One 4:e6361–e6361

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Erler JT, Bennewith KL, Cox TR, Lang G, Bird D, Koong A, Le Q-T, Giaccia AJ (2009) Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell 15:35–44

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Elkabets M, Gifford AM, Scheel C, Nilsson B, Reinhardt F, Bray M-A, Carpenter AE, Jirström K, Magnusson K, Ebert BL, Pontén F, Weinberg RA, McAllister SS (2011) Human tumors instigate granulin-expressing hematopoietic cells that promote malignancy by activating stromal fibroblasts in mice. J Clin Invest 121:784–799

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Cox TR, Rumney RMH, Schoof EM, Perryman L, Hye AM, Agrawal A, Bird D, Latif NA, Forrest H, Evans HR, Huggins ID, Lang G, Linding R, Gartland A, Erler JT (2015) The hypoxic cancer secretome induces pre-metastatic bone lesions through lysyl oxidase. Nature 522:106–110

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Hoshino A, Costa-Silva B, Shen T-L, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, Singh S, Williams C, Soplop N, Uryu K, Pharmer L, King T, Bojmar L, Davies AE, Ararso Y, Lyden D et al (2015) Tumour exosome integrins determine organotropic metastasis. Nature 527:329

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Holle AW, Young JL, Spatz JP (2016) In vitro cancer cell–ECM interactions inform in vivo cancer treatment. Adv Drug Deliv Rev 97:270–279

    Article  CAS  PubMed  Google Scholar 

  154. Sherman-Baust CA, Weeraratna AT, Rangel LBA, Pizer ES, Cho KR, Schwartz DR, Shock T, Morin PJ (2003) Remodeling of the extracellular matrix through overexpression of collagen VI contributes to cisplatin resistance in ovarian cancer cells. Cancer Cell 3:377–386

    Article  CAS  PubMed  Google Scholar 

  155. Hatano K, Kikuchi J, Takatoku M, Shimizu R, Wada T, Ueda M, Nobuyoshi M, Oh I, Sato K, Suzuki T, Ozaki K, Mori M, Nagai T, Muroi K, Kano Y, Furukawa Y, Ozawa K (2009) Bortezomib overcomes cell adhesion-mediated drug resistance through downregulation of VLA-4 expression in multiple myeloma. Oncogene 28:231–242

    Article  CAS  Google Scholar 

  156. Schmidmaier R, Baumann P, Simsek M, Dayyani F, Emmerich B, Meinhardt G (2004) The HMG-CoA reductase inhibitor simvastatin overcomes cell adhesion-mediated drug resistance in multiple myeloma by geranylgeranylation of rho protein and activation of rho kinase. Blood 104:1825–1832

    Article  CAS  PubMed  Google Scholar 

  157. Aoudjit F, Vuori K (2012) Integrin signaling in cancer cell survival and chemoresistance. Chemother Res Pract 2012:283181

    PubMed  PubMed Central  Google Scholar 

  158. Wei L, Surma M, Shi S, Lambert-Cheatham N, Shi J (2016) Novel insights into the roles of rho kinase in cancer. Arch Immunol Ther Exp 64: 259–278

    Article  CAS  Google Scholar 

  159. Hong X, Michalski CW, Kong B, Zhang W, Raggi MC, Sauliunaite D, De Oliveira T, Friess H, Kleeff J (2010) ALCAM is associated with chemoresistance and tumor cell adhesion in pancreatic cancer. J Surg Oncol 101:564–569

    Article  CAS  PubMed  Google Scholar 

  160. McGrail DJ, Khambhati NN, Qi MX, Patel KS, Ravikumar N, Brandenburg CP, Dawson MR (2015) Alterations in ovarian cancer cell adhesion drive taxol resistance by increasing microtubule dynamics in a FAK-dependent manner. Sci Rep 5:9529

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michelle R. Dawson .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Ghosh, D., Dawson, M.R. (2018). Microenvironment Influences Cancer Cell Mechanics from Tumor Growth to Metastasis. In: Dong, C., Zahir, N., Konstantopoulos, K. (eds) Biomechanics in Oncology. Advances in Experimental Medicine and Biology, vol 1092. Springer, Cham. https://doi.org/10.1007/978-3-319-95294-9_5

Download citation

Publish with us

Policies and ethics