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.

  • Review Article
  • Published:

Targeting the tumour stroma to improve cancer therapy

Abstract

Cancers are not composed merely of cancer cells alone; instead, they are complex ‘ecosystems’ comprising many different cell types and noncellular factors. The tumour stroma is a critical component of the tumour microenvironment, where it has crucial roles in tumour initiation, progression, and metastasis. Most anticancer therapies target cancer cells specifically, but the tumour stroma can promote the resistance of cancer cells to such therapies, eventually resulting in fatal disease. Therefore, novel treatment strategies should combine anticancer and antistromal agents. Herein, we provide an overview of the advances in understanding the complex cancer cell–tumour stroma interactions and discuss how this knowledge can result in more effective therapeutic strategies, which might ultimately improve patient outcomes.

Key points

  • Tumours comprise cancer cells as well as a stromal compartment with cellular and noncellular components.

  • The tumour stroma has critical roles in cancer development, progression, and metastasis.

  • Typically, anticancer therapies predominantly target cancer cells, and their effect on the tumour stroma is not taken into account.

  • The tumour stroma responds to anticancer therapies by inducing therapeutic resistance, which can ultimately lead to fatal disease.

  • Anticancer therapies should target both cancer cells and the stromal compartment to be effective and result in improved patient outcomes.

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

Fig. 1: Comparison of nonmalignant stroma and tumour stroma.
Fig. 2: Tumour-stroma-mediated chemoresistance.
Fig. 3: Targeting tumour stromal cells in addition to cancer cells.

Similar content being viewed by others

References

  1. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2017. CA Cancer J. Clin. 67, 7–30 (2017).

    Article  PubMed  Google Scholar 

  2. Amend, S. R. & Pienta, K. J. Ecology meets cancer biology: the cancer swamp promotes the lethal cancer phenotype. Oncotarget 6, 9669–9678 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Amend, S. R., Roy, S., Brown, J. S. & Pienta, K. J. Ecological paradigms to understand the dynamics of metastasis. Cancer Lett. 380, 237–242 (2016).

    Article  PubMed  CAS  Google Scholar 

  4. Camacho, D. F. & Pienta, K. J. Disrupting the networks of cancer. Clin. Cancer Res. 18, 2801–2808 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. de Groot, A. E., Roy, S., Brown, J. S., Pienta, K. J. & Amend, S. R. Revisiting seed and soil: examining the primary tumor and cancer cell foraging in metastasis. Mol. Cancer Res. 15, 361–370 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Maley, C. C. et al. Classifying the evolutionary and ecological features of neoplasms. Nat. Rev. Cancer 17, 605–619 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Paget, S. The distribution of secondary growths in cancer of the breast. Lancet 1, 571–573 (1889).

    Article  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  9. Brown, J. M. Vasculogenesis: a crucial player in the resistance of solid tumours to radiotherapy. Br. J. Radiol. 87, 20130686 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Hida, K., Akiyama, K., Ohga, N., Maishi, N. & Hida, Y. Tumour endothelial cells acquire drug resistance in a tumour microenvironment. J. Biochem. 153, 243–249 (2013).

    Article  PubMed  CAS  Google Scholar 

  11. Kibria, G., Hatakeyama, H. & Harashima, H. Cancer multidrug resistance: mechanisms involved and strategies for circumvention using a drug delivery system. Arch. Pharm. Res. 37, 4–15 (2014).

    Article  PubMed  CAS  Google Scholar 

  12. Ruffell, B. & Coussens, L. M. Macrophages and therapeutic resistance in cancer. Cancer Cell 27, 462–472 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. van Beijnum, J. R., Nowak-Sliwinska, P., Huijbers, E. J., Thijssen, V. L. & Griffioen, A. W. The great escape; the hallmarks of resistance to antiangiogenic therapy. Pharmacol. Rev. 67, 441–461 (2015).

    Article  PubMed  CAS  Google Scholar 

  14. Choi, J., Cha, Y. J. & Koo, J. S. Adipocyte biology in breast cancer: from silent bystander to active facilitator. Prog. Lipid Res. 69, 11–20 (2017).

    Article  PubMed  CAS  Google Scholar 

  15. Kozin, S. V. et al. Recruitment of myeloid but not endothelial precursor cells facilitates tumor regrowth after local irradiation. Cancer Res. 70, 5679–5685 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Ribas, A. Adaptive immune resistance: how cancer protects from immune attack. Cancer Discov. 5, 915–919 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Roca, H. et al. Transcription factors OVOL1 and OVOL2 induce the mesenchymal to epithelial transition in human cancer. PLoS ONE 8, e76773 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Mammoto, T. & Ingber, D. E. Mechanical control of tissue and organ development. Development 137, 1407–1420 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Hynes, R. O. & Naba, A. Overview of the matrisome — an inventory of extracellular matrix constituents and functions. Cold Spring Harb. Perspect. Biol. 4, a004903 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Bonnans, C., Chou, J. & Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 15, 786–801 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Hynes, R. O. The extracellular matrix: not just pretty fibrils. Science 326, 1216–1219 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Laurent, G. J., Chambers, R. C., Hill, M. R. & McAnulty, R. J. Regulation of matrix turnover: fibroblasts, forces, factors and fibrosis. Biochem. Soc. Trans. 35, 647–651 (2007).

    Article  PubMed  CAS  Google Scholar 

  23. Alberts, B. et al. Molecular Biology of the Cell. 2nd edn (Garland Publishing, 1989).

  24. Alexander, J. & Cukierman, E. Stromal dynamic reciprocity in cancer: intricacies of fibroblastic-ECM interactions. Curr. Opin. Cell Biol. 42, 80–93 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Buckley, C. D. et al. Fibroblasts regulate the switch from acute resolving to chronic persistent inflammation. Trends Immunol. 22, 199–204 (2001).

    Article  PubMed  CAS  Google Scholar 

  26. Smith, R. S., Smith, T. J., Blieden, T. M. & Phipps, R. P. Fibroblasts as sentinel cells. Synthesis of chemokines and regulation of inflammation. Am. J. Pathol. 151, 317–322 (1997).

    PubMed  PubMed Central  CAS  Google Scholar 

  27. Schneider, E. L., Mitsui, Y., Au, K. S. & Shorr, S. S. Tissue-specific differences in cultured human diploid fibroblasts. Exp. Cell Res. 108, 1–6 (1977).

    Article  PubMed  CAS  Google Scholar 

  28. Zamansky, G. B., Arundel, C., Nagasawa, H. & Little, J. B. Adaptation of human diploid fibroblasts in vitro to serum from different sources. J. Cell Sci. 61, 289–297 (1983).

    PubMed  CAS  Google Scholar 

  29. Porter, K. E. & Turner, N. A. Cardiac fibroblasts: at the heart of myocardial remodeling. Pharmacol. Ther. 123, 255–278 (2009).

    Article  PubMed  CAS  Google Scholar 

  30. Baum, J. & Duffy, H. S. Fibroblasts and myofibroblasts: what are we talking about? J. Cardiovasc. Pharmacol. 57, 376–379 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Bochaton-Piallat, M. L., Gabbiani, G. & Hinz, B. The myofibroblast in wound healing and fibrosis: answered and unanswered questions. F1000Res 5, 752 (2016).

    Article  CAS  Google Scholar 

  32. Hinz, B. et al. The myofibroblast: one function, multiple origins. Am. J. Pathol. 170, 1807–1816 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Dvorak, H. F. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N. Engl. J. Med. 315, 1650–1659 (1986).

    Article  PubMed  CAS  Google Scholar 

  34. Shiga, K. et al. Cancer-associated fibroblasts: their characteristics and their roles in tumor growth. Cancers 7, 2443–2458 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Tuxhorn, J. A. et al. Reactive stroma in human prostate cancer: induction of myofibroblast phenotype and extracellular matrix remodeling. Clin. Cancer Res. 8, 2912–2923 (2002).

    PubMed  CAS  Google Scholar 

  36. Nombela-Arrieta, C., Ritz, J. & Silberstein, L. E. The elusive nature and function of mesenchymal stem cells. Nat. Rev. Mol. Cell Biol. 12, 126–131 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Friedenstein, A. J., Chailakhjan, R. K. & Lalykina, K. S. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 3, 393–403 (1970).

    PubMed  CAS  Google Scholar 

  38. Friedenstein, A. J., Piatetzky-Shapiro, I. I. & Petrakova, K. V. Osteogenesis in transplants of bone marrow cells. J. Embryol. Exp. Morphol. 16, 381–390 (1966).

    PubMed  CAS  Google Scholar 

  39. Friedenstein, A. J., Gorskaja, J. F. & Kulagina, N. N. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp. Hematol. 4, 267–274 (1976).

    PubMed  CAS  Google Scholar 

  40. Piersma, A. H. et al. Characterization of fibroblastic stromal cells from murine bone marrow. Exp. Hematol. 13, 237–243 (1985).

    PubMed  CAS  Google Scholar 

  41. Pittenger, M. F. et al. Multilineage potential of adult human mesenchymal stem cells. Science 284, 143–147 (1999).

    Article  PubMed  CAS  Google Scholar 

  42. Rickard, D. J., Sullivan, T. A., Shenker, B. J., Leboy, P. S. & Kazhdan, I. Induction of rapid osteoblast differentiation in rat bone marrow stromal cell cultures by dexamethasone and BMP-2. Dev. Biol. 161, 218–228 (1994).

    Article  PubMed  Google Scholar 

  43. Wakitani, S., Saito, T. & Caplan, A. I. Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve 18, 1417–1426 (1995).

    Article  PubMed  CAS  Google Scholar 

  44. Horwitz, E. M. et al. Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement. Cytotherapy 7, 393–395 (2005).

    Article  PubMed  CAS  Google Scholar 

  45. Lindner, U., Kramer, J., Rohwedel, J. & Schlenke, P. Mesenchymal stem or stromal cells: toward a better understanding of their biology? Transfus. Med. Hemother 37, 75–83 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Paunescu, V. et al. Tumour-associated fibroblasts and mesenchymal stem cells: more similarities than differences. J. Cell. Mol. Med. 15, 635–646 (2011).

    Article  PubMed  CAS  Google Scholar 

  47. Haniffa, M. A. et al. Adult human fibroblasts are potent immunoregulatory cells and functionally equivalent to mesenchymal stem cells. J. Immunol. 179, 1595–1604 (2007).

    Article  PubMed  CAS  Google Scholar 

  48. Nakagawa, M. et al. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat. Biotechnol. 26, 101–106 (2008).

    Article  PubMed  CAS  Google Scholar 

  49. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    Article  PubMed  CAS  Google Scholar 

  50. Dominici, M. et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8, 315–317 (2006).

    Article  PubMed  CAS  Google Scholar 

  51. Alt, E. et al. Fibroblasts share mesenchymal phenotypes with stem cells, but lack their differentiation and colony-forming potential. Biol. Cell 103, 197–208 (2011).

    Article  PubMed  CAS  Google Scholar 

  52. Battula, V. L. et al. Connective tissue growth factor regulates adipocyte differentiation of mesenchymal stromal cells and facilitates leukemia bone marrow engraftment. Blood 122, 357–366 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Erices, A., Conget, P. & Minguell, J. J. Mesenchymal progenitor cells in human umbilical cord blood. Br. J. Haematol. 109, 235–242 (2000).

    Article  PubMed  CAS  Google Scholar 

  54. Zvaifler, N. J. et al. Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res. 2, 477–488 (2000).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Galotto, M. et al. Stromal damage as consequence of high-dose chemo/radiotherapy in bone marrow transplant recipients. Exp. Hematol. 27, 1460–1466 (1999).

    Article  PubMed  CAS  Google Scholar 

  56. Sanchez-Abarca, L. I. et al. Uptake and delivery of antigens by mesenchymal stromal cells. Cytotherapy 15, 673–678 (2013).

    Article  PubMed  CAS  Google Scholar 

  57. Yagi, H. et al. Mesenchymal stem cells: Mechanisms of immunomodulation and homing. Cell Transplant 19, 667–679 (2010).

    Article  PubMed  Google Scholar 

  58. Moroni, L. & Fornasari, P. M. Human mesenchymal stem cells: a bank perspective on the isolation, characterization and potential of alternative sources for the regeneration of musculoskeletal tissues. J. Cell. Physiol. 228, 680–687 (2013).

    Article  PubMed  CAS  Google Scholar 

  59. Wu, L., Cai, X., Zhang, S., Karperien, M. & Lin, Y. Regeneration of articular cartilage by adipose tissue derived mesenchymal stem cells: perspectives from stem cell biology and molecular medicine. J. Cell. Physiol. 228, 938–944 (2013).

    Article  PubMed  CAS  Google Scholar 

  60. Ducy, P., Schinke, T. & Karsenty, G. The osteoblast: a sophisticated fibroblast under central surveillance. Science 289, 1501–1504 (2000).

    Article  PubMed  CAS  Google Scholar 

  61. Seeman, E. & Delmas, P. D. Bone quality—the material and structural basis of bone strength and fragility. N. Engl. J. Med. 354, 2250–2261 (2006).

    Article  PubMed  CAS  Google Scholar 

  62. Mackie, E. J. Osteoblasts: novel roles in orchestration of skeletal architecture. Int. J. Biochem. Cell Biol. 35, 1301–1305 (2003).

    Article  PubMed  CAS  Google Scholar 

  63. Caetano-Lopes, J., Canhao, H. & Fonseca, J. E. Osteoblasts and bone formation. Acta Reumatol. Port. 32, 103–110 (2007).

    PubMed  Google Scholar 

  64. Gori, F. et al. The expression of osteoprotegerin and RANK ligand and the support of osteoclast formation by stromal-osteoblast lineage cells is developmentally regulated. Endocrinology 141, 4768–4776 (2000).

    Article  PubMed  CAS  Google Scholar 

  65. Harada, S. & Rodan, G. A. Control of osteoblast function and regulation of bone mass. Nature 423, 349–355 (2003).

    Article  PubMed  CAS  Google Scholar 

  66. Muir, H. The chondrocyte, architect of cartilage. Biomechanics, structure, function and molecular biology of cartilage matrix macromolecules. Bioessays 17, 1039–1048 (1995).

    Article  PubMed  CAS  Google Scholar 

  67. Mackie, E. J., Ahmed, Y. A., Tatarczuch, L., Chen, K. S. & Mirams, M. Endochondral ossification: how cartilage is converted into bone in the developing skeleton. Int. J. Biochem. Cell Biol. 40, 46–62 (2008).

    Article  PubMed  CAS  Google Scholar 

  68. Urban, J. P. The chondrocyte: a cell under pressure. Br. J. Rheumatol 33, 901–908 (1994).

    Article  PubMed  CAS  Google Scholar 

  69. Massague, J. & Obenauf, A. C. Metastatic colonization by circulating tumour cells. Nature 529, 298–306 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Pienta, K. J., McGregor, N., Axelrod, R. & Axelrod, D. E. Ecological therapy for cancer: defining tumors using an ecosystem paradigm suggests new opportunities for novel cancer treatments. Transl Oncol. 1, 158–164 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Ostman, A. & Augsten, M. Cancer-associated fibroblasts and tumor growth — bystanders turning into key players. Curr. Opin. Genet. Dev. 19, 67–73 (2009).

    Article  PubMed  CAS  Google Scholar 

  72. Cunha, G. R., Bigsby, R. M., Cooke, P. S. & Sugimura, Y. Stromal-epithelial interactions in adult organs. Cell Differ. 17, 137–148 (1985).

    Article  PubMed  CAS  Google Scholar 

  73. Cunha, G. R., Donjacour, A. A. & Sugimura, Y. Stromal-epithelial interactions and heterogeneity of proliferative activity within the prostate. Biochem. Cell Biol. 64, 608–614 (1986).

    Article  PubMed  CAS  Google Scholar 

  74. Ozdemir, B. C. et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 25, 719–734 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Bhowmick, N. A. et al. TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science 303, 848–851 (2004).

    Article  PubMed  CAS  Google Scholar 

  76. Leight, J. L., Wozniak, M. A., Chen, S., Lynch, M. L. & Chen, C. S. Matrix rigidity regulates a switch between TGF-beta1-induced apoptosis and epithelial-mesenchymal transition. Mol. Biol. Cell 23, 781–791 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Calon, A. et al. Dependency of colorectal cancer on a TGF-beta-driven program in stromal cells for metastasis initiation. Cancer Cell 22, 571–584 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Rhim, A. D. et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 25, 735–747 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Shimoda, M., Jackson, H. W. & Khokha, R. Tumor suppression by stromal TIMPs. Mol. Cell Oncol. 3, e975082 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Cruz-Munoz, W. & Khokha, R. The role of tissue inhibitors of metalloproteinases in tumorigenesis and metastasis. Crit. Rev. Clin. Lab. Sci. 45, 291–338 (2008).

    Article  PubMed  CAS  Google Scholar 

  81. Shimoda, M. et al. Loss of the Timp gene family is sufficient for the acquisition of the CAF-like cell state. Nat. Cell Biol. 16, 889–901 (2014).

    Article  PubMed  CAS  Google Scholar 

  82. Tjomsland, V. et al. Profile of MMP and TIMP expression in human pancreatic stellate cells: regulation by IL-1α and TGFβ and implications for migration of pancreatic cancer cells. Neoplasia 18, 447–456 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Bloomston, M., Shafii, A., Zervos, E. E. & Rosemurgy, A. S. TIMP-1 overexpression in pancreatic cancer attenuates tumor growth, decreases implantation and metastasis, and inhibits angiogenesis. J. Surg. Res. 102, 39–44 (2002).

    Article  PubMed  CAS  Google Scholar 

  84. Langley, R. R. & Fidler, I. J. The seed and soil hypothesis revisited — the role of tumor-stroma interactions in metastasis to different organs. Int. J. Cancer 128, 2527–2535 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Fidler, I. J. et al. Modulation of tumor cell response to chemotherapy by the organ environment. Cancer Metastasis Rev. 13, 209–222 (1994).

    Article  PubMed  CAS  Google Scholar 

  86. Couillard, J., Demers, M., Lavoie, G. & St-Pierre, Y. The role of DNA hypomethylation in the control of stromelysin gene expression. Biochem. Biophys. Res. Commun. 342, 1233–1239 (2006).

    Article  PubMed  CAS  Google Scholar 

  87. Hanson, J. A. et al. Gene promoter methylation in prostate tumor-associated stromal cells. J. Natl Cancer Inst. 98, 255–261 (2006).

    Article  PubMed  CAS  Google Scholar 

  88. Lin, H. J. et al. Breast cancer-associated fibroblasts confer AKT1-mediated epigenetic silencing of Cystatin M in epithelial cells. Cancer Res. 68, 10257–10266 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  90. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 16, 582–598 (2016).

    Article  PubMed  CAS  Google Scholar 

  91. Micallef, L. et al. The myofibroblast, multiple origins for major roles in normal and pathological tissue repair. Fibrogen. Tissue Repair 5, S5 (2012).

    Article  CAS  Google Scholar 

  92. Neuzillet, C. et al. Stromal expression of SPARC in pancreatic adenocarcinoma. Cancer Metastasis Rev. 32, 585–602 (2013).

    Article  PubMed  CAS  Google Scholar 

  93. Ronnov-Jessen, L., Petersen, O. W. & Bissell, M. J. Cellular changes involved in conversion of normal to malignant breast: importance of the stromal reaction. Physiol. Rev. 76, 69–125 (1996).

    Article  PubMed  CAS  Google Scholar 

  94. Chiquet-Ehrismann, R., Mackie, E. J., Pearson, C. A. & Sakakura, T. Tenascin: an extracellular matrix protein involved in tissue interactions during fetal development and oncogenesis. Cell 47, 131–139 (1986).

    Article  PubMed  CAS  Google Scholar 

  95. Kyutoku, M. et al. Role of periostin in cancer progression and metastasis: inhibition of breast cancer progression and metastasis by anti-periostin antibody in a murine model. Int. J. Mol. Med. 28, 181–186 (2011).

    PubMed  CAS  Google Scholar 

  96. Mackie, E. J. et al. Tenascin is a stromal marker for epithelial malignancy in the mammary gland. Proc. Natl Acad. Sci. USA 84, 4621–4625 (1987).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Ouyang, G. et al. Upregulated expression of periostin by hypoxia in non-small-cell lung cancer cells promotes cell survival via the Akt/PKB pathway. Cancer Lett. 281, 213–219 (2009).

    Article  PubMed  CAS  Google Scholar 

  98. Ruan, K., Bao, S. & Ouyang, G. The multifaceted role of periostin in tumorigenesis. Cell. Mol. Life Sci. 66, 2219–2230 (2009).

    Article  PubMed  CAS  Google Scholar 

  99. Mouw, J. K. et al. Tissue mechanics modulate microRNA-dependent PTEN expression to regulate malignant progression. Nat. Med. 20, 360–367 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Kim, W. et al. The integrin-coupled signaling adaptor p130Cas suppresses Smad3 function in transforming growth factor-beta signaling. Mol. Biol. Cell 19, 2135–2146 E07-10-0991 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Liu, J. & Agarwal, S. Mechanical signals activate vascular endothelial growth factor receptor-2 to upregulate endothelial cell proliferation during inflammation. J. Immunol. 185, 1215–1221 (2010).

    Article  PubMed  CAS  Google Scholar 

  102. Pylayeva, Y. et al. Ras- and PI3K-dependent breast tumorigenesis in mice and humans requires focal adhesion kinase signaling. J. Clin. Invest. 119, 252–266 (2009).

    PubMed  PubMed Central  CAS  Google Scholar 

  103. Tomakidi, P. et al. Defects of basement membrane and hemidesmosome structure correlate with malignant phenotype and stromal interactions in HaCaT-Ras xenografts. Differentiation 64, 263–275 (1999).

    Article  PubMed  CAS  Google Scholar 

  104. Kaukonen, R. et al. Normal stroma suppresses cancer cell proliferation via mechanosensitive regulation of JMJD1a-mediated transcription. Nat. Commun. 7, 12237 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Olumi, A. F. et al. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res. 59, 5002–5011 (1999).

    PubMed  CAS  Google Scholar 

  106. Boire, A. et al. PAR1 is a matrix metalloprotease-1 receptor that promotes invasion and tumorigenesis of breast cancer cells. Cell 120, 303–313 (2005).

    Article  PubMed  CAS  Google Scholar 

  107. Sternlicht, M. D. et al. The stromal proteinase MMP3/stromelysin-1 promotes mammary carcinogenesis. Cell 98, 137–146 (1999).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Hotary, K. B. et al. Membrane type I matrix metalloproteinase usurps tumor growth control imposed by the three-dimensional extracellular matrix. Cell 114, 33–45 (2003).

    Article  PubMed  CAS  Google Scholar 

  109. Tang, D. et al. High expression of Galectin-1 in pancreatic stellate cells plays a role in the development and maintenance of an immunosuppressive microenvironment in pancreatic cancer. International journal of cancer. Int. J. Cancer 130, 2337–2348 (2012).

    Article  PubMed  CAS  Google Scholar 

  110. Luttenberger, T. et al. Platelet-derived growth factors stimulate proliferation and extracellular matrix synthesis of pancreatic stellate cells: implications in pathogenesis of pancreas fibrosis. Lab. Invest. 80, 47–55 (2000).

    Article  PubMed  CAS  Google Scholar 

  111. Joesting, M. S. et al. Identification of SFRP1 as a candidate mediator of stromal-to-epithelial signaling in prostate cancer. Cancer Res. 65, 10423–10430 (2005).

    Article  PubMed  CAS  Google Scholar 

  112. Bragado, P. et al. TGF-beta2 dictates disseminated tumour cell fate in target organs through TGF-beta-RIII and p38alpha/beta signalling. Nat. Cell Biol. 15, 1351–1361 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  113. Carstens, J. L. et al. FGFR1-WNT-TGF-beta signaling in prostate cancer mouse models recapitulates human reactive stroma. Cancer Res. 74, 609–620 (2014).

    Article  PubMed  CAS  Google Scholar 

  114. Claffey, K. P. et al. Fibroblast growth factor 2 activation of stromal cell vascular endothelial growth factor expression and angiogenesis. Lab. Invest. 81, 61–75 (2001).

    Article  PubMed  CAS  Google Scholar 

  115. Korc, M. & Friesel, R. E. The role of fibroblast growth factors in tumor growth. Curr. Cancer Drug Targets 9, 639–651 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Crawford, Y. et al. PDGF-C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti-VEGF treatment. Cancer Cell 15, 21–34 (2009).

    Article  PubMed  CAS  Google Scholar 

  117. Pietras, K., Pahler, J., Bergers, G. & Hanahan, D. Functions of paracrine PDGF signaling in the proangiogenic tumor stroma revealed by pharmacological targeting. PLoS Med. 5, e19 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  118. Fukumura, D. et al. Tumor induction of VEGF promoter activity in stromal cells. Cell 94, 715–725 (1998).

    Article  PubMed  CAS  Google Scholar 

  119. Erez, N., Truitt, M., Olson, P., Arron, S. T. & Hanahan, D. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-κB-dependent manner. Cancer Cell 17, 135–147 (2010).

    Article  PubMed  CAS  Google Scholar 

  120. Prabhu, V. V., Warfel, N. A. & El-Deiry, W. S. CTGF-mediated autophagy-senescence transition in tumor stroma promotes anabolic tumor growth and metastasis. Cell Cycle 11, 2592–2593 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Orimo, A. et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 121, 335–348 (2005).

    Article  PubMed  CAS  Google Scholar 

  122. Sun, Y. X. et al. Expression and activation of alpha v beta 3 integrins by SDF-1/CXC12 increases the aggressiveness of prostate cancer cells. Prostate 67, 61–73 (2007).

    Article  PubMed  CAS  Google Scholar 

  123. Sun, Y. X. et al. Expression of CXCR4 and CXCL12 (SDF-1) in human prostate cancers (PCa) in vivo. J. Cell. Biochem. 89, 462–473 (2003).

    Article  PubMed  CAS  Google Scholar 

  124. LeBedis, C., Chen, K., Fallavollita, L., Boutros, T. & Brodt, P. Peripheral lymph node stromal cells can promote growth and tumorigenicity of breast carcinoma cells through the release of IGF-I and EGF. International journal of cancer. J. Int. Cancer 100, 2–8 (2002).

    Article  CAS  Google Scholar 

  125. Sainaghi, P. P. et al. Gas6 induces proliferation in prostate carcinoma cell lines expressing the Axl receptor. J. Cell. Physiol. 204, 36–44 (2005).

    Article  PubMed  CAS  Google Scholar 

  126. Shiozawa, Y. et al. GAS6/AXL axis regulates prostate cancer invasion, proliferation, and survival in the bone marrow niche. Neoplasia 12, 116–127 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Taichman, R. S. et al. GAS6 receptor status is associated with dormancy and bone metastatic tumor formation. PLoS ONE 8, e61873 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Quante, M. et al. Bone marrow-derived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth. Cancer Cell 19, 257–272 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  129. Hugo, H. J. et al. Contribution of fibroblast and mast cell (afferent) and tumor (efferent) IL-6 effects within the tumor microenvironment. Cancer Microenviron. 5, 83–93 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Cheng, N., Chytil, A., Shyr, Y., Joly, A. & Moses, H. L. Transforming growth factor-beta signaling-deficient fibroblasts enhance hepatocyte growth factor signaling in mammary carcinoma cells to promote scattering and invasion. Mol. Cancer Res. 6, 1521–1533 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Tait, L. R. et al. Dynamic stromal-epithelial interactions during progression of MCF10DCIS.com xenografts. International journal of cancer. J. Int. Cancer 120, 2127–2134 (2007).

    Article  CAS  Google Scholar 

  132. Rajaram, M., Li, J., Egeblad, M. & Powers, R. S. System-wide analysis reveals a complex network of tumor-fibroblast interactions involved in tumorigenicity. PLoS Genet. 9, e1003789 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Elkhattouti, A., Hassan, M. & Gomez, C. R. Stromal fibroblast in age-related cancer: role in tumorigenesis and potential as novel therapeutic target. Front. Oncol. 5, 158 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  134. Pazolli, E. et al. Chromatin remodeling underlies the senescence-associated secretory phenotype of tumor stromal fibroblasts that supports cancer progression. Cancer Res. 72, 2251–2261 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Luo, X. et al. Stromal-initiated changes in the bone promote metastatic niche development. Cell Rep. 14, 82–92 (2016).

    Article  PubMed  CAS  Google Scholar 

  136. Pazolli, E. et al. Senescent stromal-derived osteopontin promotes preneoplastic cell growth. Cancer Res. 69, 1230–1239 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  137. Ao, Z. et al. Identification of cancer-associated fibroblasts in circulating blood from patients with metastatic breast cancer. Cancer Res. 75, 4681–4687 (2015).

    Article  PubMed  CAS  Google Scholar 

  138. Jones, M. L., Siddiqui, J., Pienta, K. J. & Getzenberg, R. H. Circulating fibroblast-like cells in men with metastatic prostate cancer. Prostate 73, 176–181 (2013).

    Article  PubMed  CAS  Google Scholar 

  139. Bystricky, B. et al. Relationship between circulating tumor cells and annexin A2 in early breast cancer patients. Anticancer Res. 37, 2727–2734 (2017).

    Article  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Bergfeld, S. A., Blavier, L. & DeClerck, Y. A. Bone marrow-derived mesenchymal stromal cells promote survival and drug resistance in tumor cells. Mol. Cancer Ther. 13, 962–975 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Worthley, D. L. et al. Bone marrow cells as precursors of the tumor stroma. Exp. Cell Res. 319, 1650–1656 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Brennen, W. N., Chen, S., Denmeade, S. R. & Isaacs, J. T. Quantification of mesenchymal stem cells (MSCs) at sites of human prostate cancer. Oncotarget 4, 106–117 (2013).

    Article  PubMed  Google Scholar 

  144. Arina, A. et al. Tumor-associated fibroblasts predominantly come from local and not circulating precursors. Proc. Natl Acad. Sci. USA 113, 7551–7556 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. Jung, Y. et al. Annexin 2-CXCL12 interactions regulate metastatic cell targeting and growth in the bone marrow. Mol. Cancer Res. 13, 197–207 (2015).

    Article  PubMed  CAS  Google Scholar 

  146. Mishra, A., Shiozawa, Y., Pienta, K. J. & Taichman, R. S. Homing of cancer cells to the bone. Cancer Microenviron. 4, 221–235 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  147. 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  PubMed  PubMed Central  CAS  Google Scholar 

  148. Wobus, M. et al. Breast carcinoma cells modulate the chemoattractive activity of human bone marrow-derived mesenchymal stromal cells by interfering with CXCL12. International journal of cancer. Int. J. Cancer 136, 44–54 (2015).

    Article  PubMed  CAS  Google Scholar 

  149. Wang, N. et al. Prostate cancer cells preferentially home to osteoblast-rich areas in the early stages of bone metastasis — evidence from in vivo models. J. Bone Miner. Res. 29, 2688–2696 (2014).

    Article  PubMed  CAS  Google Scholar 

  150. Shimo, T. et al. The role of sonic hedgehog signaling in osteoclastogenesis and jaw bone destruction. PLoS ONE 11, e0151731 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Heller, E. et al. Hedgehog signaling inhibition blocks growth of resistant tumors through effects on tumor microenvironment. Cancer Res. 72, 897–907 (2012).

    Article  PubMed  CAS  Google Scholar 

  152. Johnson, R. W. et al. TGF-beta promotion of Gli2-induced expression of parathyroid hormone-related protein, an important osteolytic factor in bone metastasis, is independent of canonical Hedgehog signaling. Cancer Res. 71, 822–831 (2011).

    Article  PubMed  CAS  Google Scholar 

  153. Mundy, G. R. Metastasis to bone: causes, consequences and therapeutic opportunities. Nature reviews. Cancer 2, 584–593 (2002).

    Article  PubMed  CAS  Google Scholar 

  154. Guise, T. A. The vicious cycle of bone metastases. J. Musculoskelet. Neuronal Interact. 2, 570–572 (2002).

    PubMed  CAS  Google Scholar 

  155. Jeong, H. M., Cho, S. W. & Park, S. I. Osteoblasts are the centerpiece of the metastatic bone microenvironment. Endocrinol. Metab. 31, 485–492 (2016).

    Article  CAS  Google Scholar 

  156. Li, X. Q. et al. ITGBL1 Is a Runx2 transcriptional target and promotes breast cancer bone metastasis by activating the TGFβ signaling pathway. Cancer Res. 75, 3302–3313 (2015).

    Article  PubMed  CAS  Google Scholar 

  157. Sottnik, J. L., Dai, J., Zhang, H., Campbell, B. & Keller, E. T. Tumor-induced pressure in the bone microenvironment causes osteocytes to promote the growth of prostate cancer bone metastases. Cancer Res. 75, 2151–2158 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  158. Cunha, G. R. Epithelio-mesenchymal interactions in primordial gland structures which become responsive to androgenic stimulation. Anat. Rec. 172, 179–195 (1972).

    Article  PubMed  CAS  Google Scholar 

  159. Cunha, G. R. Tissue interactions between epithelium and mesenchyme of urogenital and integumental origin. Anat. Rec. 172, 529–541 (1972).

    Article  PubMed  CAS  Google Scholar 

  160. Cunha, G. R. The role of androgens in the epithelio-mesenchymal interactions involved in prostatic morphogenesis in embryonic mice. Anat. Rec. 175, 87–96 (1973).

    Article  PubMed  CAS  Google Scholar 

  161. Aboseif, S., El-Sakka, A., Young, P. & Cunha, G. Mesenchymal reprogramming of adult human epithelial differentiation. Differentiation 65, 113–118 (1999).

    Article  PubMed  CAS  Google Scholar 

  162. Hayward, S. W. et al. Malignant transformation in a nontumorigenic human prostatic epithelial cell line. Cancer Res. 61, 8135–8142 (2001).

    PubMed  CAS  Google Scholar 

  163. Barcellos-Hoff, M. H. & Ravani, S. A. Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Res. 60, 1254–1260 (2000).

    PubMed  CAS  Google Scholar 

  164. Yasuda, K. et al. Fibroblasts induce expression of FGF4 in ovarian cancer stem-like cells/cancer-initiating cells and upregulate their tumor initiation capacity. Lab. Invest. 94, 1355–1369 (2014).

    Article  PubMed  CAS  Google Scholar 

  165. Zhao, X. L. et al. High-mobility group box 1 released by autophagic cancer-associated fibroblasts maintains the stemness of luminal breast cancer cells. J. Pathol. 243, 376–389 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  166. Vermeulen, L. et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat. Cell Biol. 12, 468–476 (2010).

    Article  PubMed  CAS  Google Scholar 

  167. Del Pozo Martin, Y. et al. Mesenchymal cancer cell-stroma crosstalk promotes niche activation, epithelial reversion, and metastatic colonization. Cell Rep. 13, 2456–2469 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  168. Du, H. & Che, G. Genetic alterations and epigenetic alterations of cancer-associated fibroblasts. Oncol. Lett. 13, 3–12 (2017).

    Article  PubMed  CAS  Google Scholar 

  169. Kurose, K. et al. Frequent somatic mutations in PTEN and TP53 are mutually exclusive in the stroma of breast carcinomas. Nat. Genet. 32, 355–357 (2002).

    Article  PubMed  CAS  Google Scholar 

  170. Kurose, K. et al. Genetic model of multi-step breast carcinogenesis involving the epithelium and stroma: clues to tumour-microenvironment interactions. Hum. Mol. Genet. 10, 1907–1913 (2001).

    Article  PubMed  CAS  Google Scholar 

  171. Tanwar, P. S., Zhang, L., Roberts, D. J. & Teixeira, J. M. Stromal deletion of the APC tumor suppressor in mice triggers development of endometrial cancer. Cancer Res. 71, 1584–1596 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  172. Kode, A. et al. Leukaemogenesis induced by an activating beta-catenin mutation in osteoblasts. Nature 506, 240–244 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  173. Calvo, F. et al. Mechanotransduction and YAP-dependent matrix remodelling is required for the generation and maintenance of cancer-associated fibroblasts. Nat. Cell Biol. 15, 637–646 (2013).

    Article  PubMed  CAS  Google Scholar 

  174. Procopio, M. G. et al. Combined CSL and p53 downregulation promotes cancer-associated fibroblast activation. Nat. Cell Biol. 17, 1193–1204 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  175. Scherz-Shouval, R. et al. The reprogramming of tumor stroma by HSF1 is a potent enabler of malignancy. Cell 158, 564–578 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Tyekucheva, S. et al. Stromal and epithelial transcriptional map of initiation progression and metastatic potential of human prostate cancer. Nat. Commun. 8, 420 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  177. Bauer, M. et al. Heterogeneity of gene expression in stromal fibroblasts of human breast carcinomas and normal breast. Oncogene 29, 1732–1740 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Nakagawa, H. et al. Role of cancer-associated stromal fibroblasts in metastatic colon cancer to the liver and their expression profiles. Oncogene 23, 7366–7377 (2004).

    Article  PubMed  CAS  Google Scholar 

  179. Sato, N., Maehara, N. & Goggins, M. Gene expression profiling of tumor-stromal interactions between pancreatic cancer cells and stromal fibroblasts. Cancer Res. 64, 6950–6956 (2004).

    Article  PubMed  CAS  Google Scholar 

  180. Singer, C. F. et al. Differential gene expression profile in breast cancer-derived stromal fibroblasts. Breast Cancer Res. Treatment 110, 273–281 (2008).

    Article  CAS  Google Scholar 

  181. Horie, M. et al. TBX4 is involved in the super-enhancer-driven transcriptional programs underlying features specific to lung fibroblasts. Am. J. Physiol. Lung Cell. Mol. Physiol. 314, L177–L191 (2018).

    Article  PubMed  CAS  Google Scholar 

  182. Marks, D. L., Olson, R. L. & Fernandez-Zapico, M. E. Epigenetic control of the tumor microenvironment. Epigenomics 8, 1671–1687 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. Mathot, P. et al. DNA methylation signal has a major role in the response of human breast cancer cells to the microenvironment. Oncogenesis 6, e390 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  184. Rodriguez-Canales, J. et al. Identification of a unique epigenetic sub-microenvironment in prostate cancer. J. Pathol. 211, 410–419 (2007).

    Article  PubMed  CAS  Google Scholar 

  185. Albrengues, J. et al. Epigenetic switch drives the conversion of fibroblasts into proinvasive cancer-associated fibroblasts. Nat. Commun. 6, 10204 (2015).

    Article  PubMed  CAS  Google Scholar 

  186. Albrengues, J. et al. LIF mediates proinvasive activation of stromal fibroblasts in cancer. Cell Rep. 7, 1664–1678 (2014).

    Article  PubMed  CAS  Google Scholar 

  187. Jiang, L. et al. Global hypomethylation of genomic DNA in cancer-associated myofibroblasts. Cancer Res. 68, 9900–9908 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  188. Velaei, K., Samadi, N., Barazvan, B. & Soleimani Rad, J. Tumor microenvironment-mediated chemoresistance in breast cancer. Breast 30, 92–100 (2016).

    Article  PubMed  Google Scholar 

  189. Ayala, G. et al. Reactive stroma as a predictor of biochemical-free recurrence in prostate cancer. Clin. Cancer Res. 9, 4792–4801 (2003).

    PubMed  CAS  Google Scholar 

  190. Mhawech-Fauceglia, P. et al. Stromal expression of fibroblast activation protein alpha (FAP) predicts platinum resistance and shorter recurrence in patients with epithelial ovarian cancer. Cancer Microenviron. 8, 23–31 (2015).

    Article  PubMed  CAS  Google Scholar 

  191. Olive, K. P. et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324, 1457–1461 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  192. Damiano, J. S., Hazlehurst, L. A. & Dalton, W. S. Cell adhesion-mediated drug resistance (CAM-DR) protects the K562 chronic myelogenous leukemia cell line from apoptosis induced by BCR/ABL inhibition, cytotoxic drugs, and gamma-irradiation. Leukemia 15, 1232–1239 (2001).

    Article  PubMed  CAS  Google Scholar 

  193. Hazlehurst, L. A. & Dalton, W. S. Mechanisms associated with cell adhesion mediated drug resistance (CAM-DR) in hematopoietic malignancies. Cancer Metastasis Rev. 20, 43–50 (2001).

    Article  PubMed  CAS  Google Scholar 

  194. Hazlehurst, L. A., Damiano, J. S., Buyuksal, I., Pledger, W. J. & Dalton, W. S. Adhesion to fibronectin via beta1 integrins regulates p27kip1 levels and contributes to cell adhesion mediated drug resistance (CAM-DR). Oncogene 19, 4319–4327 (2000).

    Article  PubMed  CAS  Google Scholar 

  195. Landowski, T. H., Olashaw, N. E., Agrawal, D. & Dalton, W. S. Cell adhesion-mediated drug resistance (CAM-DR) is associated with activation of NF-kappa B (RelB/p50) in myeloma cells. Oncogene 22, 2417–2421 (2003).

    Article  PubMed  CAS  Google Scholar 

  196. Hazlehurst, L. A., Argilagos, R. F. & Dalton, W. S. Beta1 integrin mediated adhesion increases Bim protein degradation and contributes to drug resistance in leukaemia cells. Br. J. Haematol. 136, 269–275 (2007).

    Article  PubMed  CAS  Google Scholar 

  197. Lwin, T. et al. Cell adhesion induces p27Kip1-associated cell-cycle arrest through down-regulation of the SCFSkp2 ubiquitin ligase pathway in mantle-cell and other non-Hodgkin B-cell lymphomas. Blood 110, 1631–1638 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  198. Jain, R. K. Transport of molecules in the tumor interstitium: a review. Cancer Res. 47, 3039–3051 (1987).

    PubMed  CAS  Google Scholar 

  199. Young, J. S., Lumsden, C. E. & Stalker, A. L. The significance of the tissue pressure of normal testicular and of neoplastic (Brown-Pearce carcinoma) tissue in the rabbit. J. Pathol. Bacteriol. 62, 313–333 (1950).

    Article  PubMed  CAS  Google Scholar 

  200. DuFort, C. C., DelGiorno, K. E. & Hingorani, S. R. Mounting pressure in the microenvironment: fluids, solids, and cells in pancreatic ductal adenocarcinoma. Gastroenterology 150, 1545–1557.e2 (2016).

    Article  PubMed  Google Scholar 

  201. Provenzano, P. P. & Hingorani, S. R. Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer. Br. J. Cancer 108, 1–8 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  202. Wegner, C. S. et al. Dynamic contrast-enhanced MRI of the microenvironment of pancreatic adenocarcinoma xenografts. Acta Oncol. 56, 1754–1762 (2017).

    Article  PubMed  Google Scholar 

  203. Provenzano, P. P. et al. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 21, 418–429 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Ozerdem, U. & Hargens, A. R. A simple method for measuring interstitial fluid pressure in cancer tissues. Microvasc. Res. 70, 116–120 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  205. Munson, J. M., Bellamkonda, R. V. & Swartz, M. A. Interstitial flow in a 3D microenvironment increases glioma invasion by a CXCR4-dependent mechanism. Cancer Res. 73, 1536–1546 (2013).

    Article  PubMed  CAS  Google Scholar 

  206. Hirth, J. et al. The effect of an individual’s cytochrome CYP3A4 activity on docetaxel clearance. Clin. Cancer Res. 6, 1255–1258 (2000).

    PubMed  CAS  Google Scholar 

  207. Alonso, S. et al. Human bone marrow niche chemoprotection mediated by cytochrome P450 enzymes. Oncotarget 6, 14905–14912 (2015).

    PubMed  PubMed Central  Google Scholar 

  208. Alonso, S. et al. Hedgehog and retinoid signaling alters multiple myeloma microenvironment and generates bortezomib resistance. J. Clin. Invest. 126, 4460–4468 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  209. Xu, K. et al. Autophagy induction contributes to the resistance to methotrexate treatment in rheumatoid arthritis fibroblast-like synovial cells through high mobility group box chromosomal protein 1. Arthritis Res. Ther. 17, 374 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  210. Huber, R. M. et al. DNA damage induces GDNF secretion in the tumor microenvironment with paracrine effects promoting prostate cancer treatment resistance. Oncotarget 6, 2134–2147 (2015).

    Article  PubMed  Google Scholar 

  211. Sun, Y. et al. Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat. Med. 18, 1359–1368 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  212. Gilbert, L. A. & Hemann, M. T. DNA damage-mediated induction of a chemoresistant niche. Cell 143, 355–366 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  213. Steinbichler, T. B., Metzler, V., Pritz, C., Riechelmann, H. & Dudas, J. Tumor-associated fibroblast-conditioned medium induces CDDP resistance in HNSCC cells. Oncotarget 7, 2508–2518 (2016).

    Article  PubMed  Google Scholar 

  214. Peiris-Pages, M., Sotgia, F. & Lisanti, M. P. Chemotherapy induces the cancer-associated fibroblast phenotype, activating paracrine Hedgehog-GLI signalling in breast cancer cells. Oncotarget 6, 10728–10745 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  215. Hu, Y. et al. Fibroblast-derived exosomes contribute to chemoresistance through priming cancer stem cells in colorectal cancer. PLoS ONE 10, e0125625 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  216. Wang, W. et al. Effector T cells abrogate stroma-mediated chemoresistance in ovarian cancer. Cell 165, 1092–1105 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  217. Baskar, R., Lee, K. A., Yeo, R. & Yeoh, K. W. Cancer and radiation therapy: current advances and future directions. Int. J. Med. Sci. 9, 193–199 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  218. Cordes, N. Integrin-mediated cell-matrix interactions for prosurvival and antiapoptotic signaling after genotoxic injury. Cancer Lett. 242, 11–19 (2006).

    Article  PubMed  CAS  Google Scholar 

  219. Cordes, N., Seidler, J., Durzok, R., Geinitz, H. & Brakebusch, C. β1-integrin-mediated signaling essentially contributes to cell survival after radiation-induced genotoxic injury. Oncogene 25, 1378–1390 (2006).

    Article  PubMed  CAS  Google Scholar 

  220. Hellevik, T. et al. Cancer-associated fibroblasts from human NSCLC survive ablative doses of radiation but their invasive capacity is reduced. Radiat. Oncol. 7, 59 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  221. Mantoni, T. S., Lunardi, S., Al-Assar, O., Masamune, A. & Brunner, T. B. Pancreatic stellate cells radioprotect pancreatic cancer cells through beta1-integrin signaling. Cancer Res. 71, 3453–3458 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  222. Park, C. C., Zhang, H. J., Yao, E. S., Park, C. J. & Bissell, M. J. Beta1 integrin inhibition dramatically enhances radiotherapy efficacy in human breast cancer xenografts. Cancer Res. 68, 4398–4405 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  223. Puthawala, K. et al. Inhibition of integrin alpha(v)beta6, an activator of latent transforming growth factor-beta, prevents radiation-induced lung fibrosis. Am. J. Respir. Crit. Care Med. 177, 82–90 (2008).

    Article  PubMed  CAS  Google Scholar 

  224. Chargari, C., Clemenson, C., Martins, I., Perfettini, J. L. & Deutsch, E. Understanding the functions of tumor stroma in resistance to ionizing radiation: emerging targets for pharmacological modulation. Drug Resist. Updat. 16, 10–21 (2013).

    Article  PubMed  CAS  Google Scholar 

  225. Kamochi, N. et al. Irradiated fibroblast-induced bystander effects on invasive growth of squamous cell carcinoma under cancer-stromal cell interaction. Cancer Sci. 99, 2417–2427 (2008).

    Article  PubMed  CAS  Google Scholar 

  226. Ohuchida, K. et al. Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res. 64, 3215–3222 (2004).

    Article  PubMed  CAS  Google Scholar 

  227. Mitsuhashi, A. et al. Fibrocyte-like cells mediate acquired resistance to anti-angiogenic therapy with bevacizumab. Nat. Commun. 6, 8792 (2015).

    Article  PubMed  CAS  Google Scholar 

  228. Yoshida, T. et al. Podoplanin-positive cancer-associated fibroblasts in the tumor microenvironment induce primary resistance to EGFR-TKIs in lung adenocarcinoma with EGFR mutation. Clin. Cancer Res. 21, 642–651 (2015).

    Article  PubMed  CAS  Google Scholar 

  229. Mueller, K. L. et al. Fibroblast-secreted hepatocyte growth factor mediates epidermal growth factor receptor tyrosine kinase inhibitor resistance in triple-negative breast cancers through paracrine activation of Met. Breast Cancer Res. 14, R104 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  230. Straussman, R. et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 487, 500–504 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  231. Hirata, E. et al. Intravital imaging reveals how BRAF inhibition generates drug-tolerant microenvironments with high integrin beta1/FAK signaling. Cancer Cell 27, 574–588 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  232. Singh, M. et al. Stromal androgen receptor in prostate development and cancer. Am. J. Pathol. 184, 2598–2607 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  233. Schweizer, M. T. et al. Effect of bipolar androgen therapy for asymptomatic men with castration-resistant prostate cancer: results from a pilot clinical study. Sci. Transl Med. 7, 269ra2 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  234. Wikstrom, P., Marusic, J., Stattin, P. & Bergh, A. Low stroma androgen receptor level in normal and tumor prostate tissue is related to poor outcome in prostate cancer patients. Prostate 69, 799–809 (2009).

    Article  PubMed  CAS  Google Scholar 

  235. Li, Y. et al. Decrease in stromal androgen receptor associates with androgen-independent disease and promotes prostate cancer cell proliferation and invasion. J. Cell. Mol. Med. 12, 2790–2798 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  236. Holton, S. E., Bergamaschi, A., Katzenellenbogen, B. S. & Bhargava, R. Integration of molecular profiling and chemical imaging to elucidate fibroblast-microenvironment impact on cancer cell phenotype and endocrine resistance in breast cancer. PLOS ONE 9, e96878 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  237. Witkiewicz, A. K. et al. An absence of stromal caveolin-1 expression predicts early tumor recurrence and poor clinical outcome in human breast cancers. Am. J. Pathol. 174, 2023–2034 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  238. Mercier, I. et al. Human breast cancer-associated fibroblasts (CAFs) show caveolin-1 downregulation and RB tumor suppressor functional inactivation: Implications for the response to hormonal therapy. Cancer Biol. Ther. 7, 1212–1225 (2008).

    Article  PubMed  CAS  Google Scholar 

  239. Pardoll, D. M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12, 252–264 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  240. Gajewski, T. F., Schreiber, H. & Fu, Y. X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 14, 1014–1022 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  241. Silzle, T., Randolph, G. J., Kreutz, M. & Kunz-Schughart, L. A. The fibroblast: sentinel cell and local immune modulator in tumor tissue. International journal of cancer. J. Int. Cancer 108, 173–180 (2004).

    Article  CAS  Google Scholar 

  242. Talts, J. F., Wirl, G., Dictor, M., Muller, W. J. & Fassler, R. Tenascin-C modulates tumor stroma and monocyte/macrophage recruitment but not tumor growth or metastasis in a mouse strain with spontaneous mammary cancer. J. Cell Sci. 112, 1855–1864 (1999).

    PubMed  CAS  Google Scholar 

  243. Salmon, H. et al. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J. Clin. Invest. 122, 899–910 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  244. Martinet, L. et al. Human solid tumors contain high endothelial venules: association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res. 71, 5678–5687 (2011).

    Article  PubMed  CAS  Google Scholar 

  245. Singh, S., Ross, S. R., Acena, M., Rowley, D. A. & Schreiber, H. Stroma is critical for preventing or permitting immunological destruction of antigenic cancer cells. J. Exp. Med. 175, 139–146 (1992).

    Article  PubMed  CAS  Google Scholar 

  246. Kraman, M. et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science 330, 827–830 (2010).

    Article  PubMed  CAS  Google Scholar 

  247. Li, X. et al. Stromal PD-L1 expression is associated with better disease-free survival in triple-negative breast cancer. Am. J. Clin. Pathol. 146, 496–502 (2016).

    Article  PubMed  CAS  Google Scholar 

  248. Miyoshi, H. et al. PD-L1 expression on neoplastic or stromal cells is respectively a poor or good prognostic factor for adult T-cell leukemia/lymphoma. Blood 128, 1374–1381 (2016).

    Article  PubMed  CAS  Google Scholar 

  249. Pines, M., Knopov, V., Genina, O., Lavelin, I. & Nagler, A. Halofuginone, a specific inhibitor of collagen type I synthesis, prevents dimethylnitrosamine-induced liver cirrhosis. J. Hepatol. 27, 391–398 (1997).

    Article  PubMed  CAS  Google Scholar 

  250. Zion, O. et al. Inhibition of transforming growth factor beta signaling by halofuginone as a modality for pancreas fibrosis prevention. Pancreas 38, 427–435 (2009).

    Article  PubMed  CAS  Google Scholar 

  251. Juarez, P. et al. Halofuginone inhibits the establishment and progression of melanoma bone metastases. Cancer Res. 72, 6247–6256 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  252. Kultti, A. et al. 4-Methylumbelliferone inhibits hyaluronan synthesis by depletion of cellular UDP-glucuronic acid and downregulation of hyaluronan synthase 2 and 3. Exp. Cell Res. 315, 1914–1923 (2009).

    Article  PubMed  CAS  Google Scholar 

  253. Hajime, M. et al. Inhibitory effect of 4-methylesculetin on hyaluronan synthesis slows the development of human pancreatic cancer in vitro and in nude mice. International journal of cancer. J. Int. Cancer 120, 2704–2709 (2007).

    Article  CAS  Google Scholar 

  254. Wong, K. M., Horton, K. J., Coveler, A. L., Hingorani, S. R. & Harris, W. P. Targeting the tumor stroma: the biology and clinical development of pegylated recombinant human hyaluronidase (PEGPH20). Curr. Oncol. Rep. 19, 47 (2017).

    Article  PubMed  CAS  Google Scholar 

  255. 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  PubMed  PubMed Central  CAS  Google Scholar 

  256. Erler, J. T. et al. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature 440, 1222–1226 (2006).

    Article  PubMed  CAS  Google Scholar 

  257. Cox, T. R. et al. LOX-mediated collagen crosslinking is responsible for fibrosis-enhanced metastasis. Cancer Res. 73, 1721–1732 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  258. Gilkes, D. M. et al. Collagen prolyl hydroxylases are essential for breast cancer metastasis. Cancer Res. 73, 3285–3296 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  259. Levental, K. R. et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 139, 891–906 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  260. Miller, B. W. et al. Targeting the LOX/hypoxia axis reverses many of the features that make pancreatic cancer deadly: inhibition of LOX abrogates metastasis and enhances drug efficacy. EMBO Mol. Med. 7, 1063–1076 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  261. Sherman, M. H. et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell 159, 80–93 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  262. Chronopoulos, A. et al. ATRA mechanically reprograms pancreatic stellate cells to suppress matrix remodelling and inhibit cancer cell invasion. Nat. Commun. 7, 12630 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  263. Alvarez, R. et al. Stromal disrupting effects of nab-paclitaxel in pancreatic cancer. Br. J. Cancer 109, 926–933 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  264. Von Hoff, D. D. et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J. Clin. Oncol. 29, 4548–4554 (2011).

    Article  CAS  Google Scholar 

  265. Bonomi, A. et al. Human amniotic mesenchymal stromal cells (hAMSCs) as potential vehicles for drug delivery in cancer therapy: an in vitro study. Stem Cell Res. Ther. 6, 155 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  266. Levy, O. et al. A prodrug-doped cellular Trojan Horse for the potential treatment of prostate cancer. Biomaterials 91, 140–150 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  267. Kidd, S. et al. Direct evidence of mesenchymal stem cell tropism for tumor and wounding microenvironments using in vivo bioluminescent imaging. Stem Cells 27, 2614–2623 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  268. Brennen, W. N., Denmeade, S. R. & Isaacs, J. T. Mesenchymal stem cells as a vector for the inflammatory prostate microenvironment. Endocr. Relat. Cancer 20, R269–R290 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  269. Clezardin, P. Mechanisms of action of bisphosphonates in oncology: a scientific concept evolving from antiresorptive to anticancer activities. Bonekey Rep. 2, 267 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  270. Weilbaecher, K. N., Guise, T. A. & McCauley, L. K. Cancer to bone: a fatal attraction. Nat. Rev. Cancer 11, 411–425 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  271. Terpos, E. et al. Bone antiresorptive agents in the treatment of bone metastases associated with solid tumours or multiple myeloma. Bonekey Rep. 4, 744 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  272. Dhesy-Thind, S. et al. Use of adjuvant bisphosphonates and other bone-modifying agents in breast cancer: a cancer care Ontario and American Society of Clinical Oncology Practice Guideline. J. Clin. Oncol. 35, 2062–2081 (2017).

    Article  PubMed  Google Scholar 

  273. Shore, N. D. Radium-223 dichloride for metastatic castration-resistant prostate cancer: the urologist’s perspective. Urology 85, 717–724 (2015).

    Article  PubMed  Google Scholar 

  274. Nilsson, S. et al. Two-year survival follow-up of the randomized, double-blind, placebo-controlled phase II study of radium-223 chloride in patients with castration-resistant prostate cancer and bone metastases. Clin. Genitourin. Cancer 11, 20–26 (2013).

    Article  PubMed  Google Scholar 

  275. Liu, R., Li, H., Liu, L., Yu, J. & Ren, X. Fibroblast activation protein: a potential therapeutic target in cancer. Cancer Biol. Ther. 13, 123–129 (2012).

    Article  PubMed  CAS  Google Scholar 

  276. Hofheinz, R. D. et al. Stromal antigen targeting by a humanised monoclonal antibody: an early phase II trial of sibrotuzumab in patients with metastatic colorectal cancer. Onkologie 26, 44–48 (2003).

    PubMed  CAS  Google Scholar 

  277. Scott, A. M. et al. A Phase I dose-escalation study of sibrotuzumab in patients with advanced or metastatic fibroblast activation protein-positive cancer. Clin. Cancer Res. 9, 1639–1647 (2003).

    PubMed  CAS  Google Scholar 

  278. Mersmann, M. et al. Human antibody derivatives against the fibroblast activation protein for tumor stroma targeting of carcinomas. Int. J. Cancer 92, 240–248 (2001).

    Article  PubMed  CAS  Google Scholar 

  279. Welt, S. et al. Antibody targeting in metastatic colon cancer: a phase I study of monoclonal antibody F19 against a cell-surface protein of reactive tumor stromal fibroblasts. J. Clin. Oncol. 12, 1193–1203 (1994).

    Article  PubMed  CAS  Google Scholar 

  280. Erickson, H. K. et al. Antibody-maytansinoid conjugates are activated in targeted cancer cells by lysosomal degradation and linker-dependent intracellular processing. Cancer Res. 66, 4426–4433 (2006).

    Article  PubMed  CAS  Google Scholar 

  281. Ostermann, E. et al. Effective immunoconjugate therapy in cancer models targeting a serine protease of tumor fibroblasts. Clin. Cancer Res. 14, 4584–4592 (2008).

    Article  PubMed  CAS  Google Scholar 

  282. Fischer, E. et al. Radioimmunotherapy of fibroblast activation protein positive tumors by rapidly internalizing antibodies. Clin. Cancer Res. 18, 6208–6218 (2012).

    Article  PubMed  CAS  Google Scholar 

  283. LeBeau, A. M., Brennen, W. N., Aggarwal, S. & Denmeade, S. R. Targeting the cancer stroma with a fibroblast activation protein-activated promelittin protoxin. Mol. Cancer Ther. 8, 1378–1386 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  284. Froeling, F. E. et al. Retinoic acid-induced pancreatic stellate cell quiescence reduces paracrine Wnt-beta-catenin signaling to slow tumor progression. Gastroenterology 141, 1486–1497 (2011).

    Article  PubMed  CAS  Google Scholar 

  285. Guan, J. et al. Retinoic acid inhibits pancreatic cancer cell migration and EMT through the downregulation of IL-6 in cancer associated fibroblast cells. Cancer Lett. 345, 132–139 (2014).

    Article  PubMed  CAS  Google Scholar 

  286. Ghiaur, G. et al. Regulation of human hematopoietic stem cell self-renewal by the microenvironment’s control of retinoic acid signaling. Proc. Natl Acad. Sci. USA 110, 16121–16126 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  287. Mehra, R. et al. Characterization of bone metastases from rapid autopsies of prostate cancer patients. Clin. Cancer Res. 17, 3924–3932 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  288. Bai, A. et al. GP369, an FGFR2-IIIb-specific antibody, exhibits potent antitumor activity against human cancers driven by activated FGFR2 signaling. Cancer Res. 70, 7630–7639 (2010).

    Article  PubMed  CAS  Google Scholar 

  289. Chae, Y. K. et al. Inhibition of the fibroblast growth factor receptor (FGFR) pathway: the current landscape and barriers to clinical application. Oncotarget 8, 16052–16074 (2017).

    PubMed  Google Scholar 

  290. Katoh, M. & Nakagama, H. FGF receptors: cancer biology and therapeutics. Med. Res. Rev. 34, 280–300 (2014).

    Article  PubMed  CAS  Google Scholar 

  291. Bello, E. et al. E-3810 is a potent dual inhibitor of VEGFR and FGFR that exerts antitumor activity in multiple preclinical models. Cancer Res. 71, 1396–1405 (2011).

    Article  PubMed  CAS  Google Scholar 

  292. Gozgit, J. M. et al. Ponatinib (AP24534), a multitargeted pan-FGFR inhibitor with activity in multiple FGFR-amplified or mutated cancer models. Mol. Cancer Ther. 11, 690–699 (2012).

    Article  PubMed  CAS  Google Scholar 

  293. Biswas, S. et al. Inhibition of TGF-beta with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J. Clin. Invest. 117, 1305–1313 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  294. Domanska, U. M. et al. CXCR4 inhibition enhances radiosensitivity, while inducing cancer cell mobilization in a prostate cancer mouse model. Clin. Exp. Metastasis 31, 829–839 (2014).

    Article  PubMed  CAS  Google Scholar 

  295. Domanska, U. M. et al. CXCR4 inhibition with AMD3100 sensitizes prostate cancer to docetaxel chemotherapy. Neoplasia 14, 709–718 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  296. Feig, C. et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl Acad. Sci. USA 110, 20212–20217 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  297. Jackson, H. J., Rafiq, S. & Brentjens, R. J. Driving CAR T-cells forward. Nat. Rev. Clin. Oncol. 13, 370–383 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  298. Stromnes, I. M. et al. T cells engineered against a native antigen can surmount immunologic and physical barriers to treat pancreatic ductal adenocarcinoma. Cancer Cell 28, 638–652 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  299. Wang, L. C. et al. Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol. Res. 2, 154–166 (2014).

    Article  PubMed  CAS  Google Scholar 

  300. Kakarla, S. et al. Antitumor effects of chimeric receptor engineered human T cells directed to tumor stroma. Mol. Ther. 21, 1611–1620 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  301. Chen, M. et al. A whole-cell tumor vaccine modified to express fibroblast activation protein induces antitumor immunity against both tumor cells and cancer-associated fibroblasts. Sci. Rep. 5, 14421 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  302. Gottschalk, S., Yu, F., Ji, M., Kakarla, S. & Song, X. T. A vaccine that co-targets tumor cells and cancer associated fibroblasts results in enhanced antitumor activity by inducing antigen spreading. PLOS ONE 8, e82658 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  303. Loeffler, M., Kruger, J. A., Niethammer, A. G. & Reisfeld, R. A. Targeting tumor-associated fibroblasts improves cancer chemotherapy by increasing intratumoral drug uptake. J. Clin. Invest. 116, 1955–1962 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  304. Wen, Y. et al. Immunotherapy targeting fibroblast activation protein inhibits tumor growth and increases survival in a murine colon cancer model. Cancer Sci. 101, 2325–2332 (2010).

    Article  PubMed  CAS  Google Scholar 

  305. Mantovani, A. et al. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 14, 399–416 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  306. Martinez, F. O. & Gordon, S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 6, 13 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  307. Long, K. B. et al. IFNγ and CCL2 cooperate to redirect tumor-infiltrating monocytes to degrade fibrosis and enhance chemotherapy efficacy in pancreatic carcinoma. Cancer Discov. 6, 400–413 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  308. Winograd, R. et al. Induction of T-cell immunity overcomes complete resistance to PD-1 and CTLA-4 blockade and improves survival in pancreatic carcinoma. Cancer Immunol. Res. 3, 399–411 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  309. Zippelius, A., Schreiner, J., Herzig, P. & Muller, P. Induced PD-L1 expression mediates acquired resistance to agonistic anti-CD40 treatment. Cancer Immunol. Res. 3, 236–244 (2015).

    Article  PubMed  CAS  Google Scholar 

  310. Beatty, G. L. et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 331, 1612–1616 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  311. Tada, H. et al. Reprogrammed chondrocytes engineered to produce IL-12 provide novel ex vivo immune-gene therapy for cancer. Immunotherapy 9, 239–248 (2017).

    Article  PubMed  CAS  Google Scholar 

  312. Danaei, G. et al. Causes of cancer in the world: comparative risk assessment of nine behavioural and environmental risk factors. Lancet 366, 1784–1793 (2005).

    Article  PubMed  Google Scholar 

  313. Mandelker, D. et al. Mutation detection in patients with advanced cancer by universal sequencing of cancer-related genes in tumor and normal DNA versus guideline-based germline testing. JAMA 318, 825–835 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  314. Tomasetti, C. & Vogelstein, B. Cancer etiology. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science 347, 78–81 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  315. Dry, J. R., Yang, M. & Saez-Rodriguez, J. Looking beyond the cancer cell for effective drug combinations. Genome Med. 8, 125 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  316. Micke, P. & Ostman, A. Tumour-stroma interaction: cancer-associated fibroblasts as novel targets in anti-cancer therapy? Lung Cancer 45 (Suppl. 2), S163–S175 (2004).

    Article  PubMed  Google Scholar 

  317. Zhang, P., Lehmann, B. D., Shyr, Y. & Guo, Y. The utilization of formalin fixed-paraffin-embedded specimens in high throughput genomic studies. Int. J. Genom. 2017, 1926304 (2017).

    Google Scholar 

  318. Dietrich, D. et al. Analysis of DNA methylation of multiple genes in microdissected cells from formalin-fixed and paraffin-embedded tissues. J. Histochem. Cytochem. 57, 477–489 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  319. Paulsson, J. & Micke, P. Prognostic relevance of cancer-associated fibroblasts in human cancer. Semin. Cancer Biol. 25, 61–68 (2014).

    Article  PubMed  CAS  Google Scholar 

  320. Liao, Y., Ni, Y., He, R., Liu, W. & Du, J. Clinical implications of fibroblast activation protein-alpha in non-small cell lung cancer after curative resection: a new predictor for prognosis. J. Cancer Res. Clin. Oncol. 139, 1523–1528 (2013).

    Article  PubMed  CAS  Google Scholar 

  321. Edlund, K. et al. CD99 is a novel prognostic stromal marker in non-small cell lung cancer Int. J. Cancer 131, 2264–2273 (2012).

    Article  PubMed  CAS  Google Scholar 

  322. Ishikawa, S. et al. Matrix metalloproteinase-2 status in stromal fibroblasts, not in tumor cells, is a significant prognostic factor in non-small-cell lung cancer. Clin. Cancer Res. 10, 6579–6585 (2004).

    Article  PubMed  CAS  Google Scholar 

  323. Ono, S. et al. Podoplanin-positive cancer-associated fibroblasts could have prognostic value independent of cancer cell phenotype in stage I lung squamous cell carcinoma: usefulness of combining analysis of both cancer cell phenotype and cancer-associated fibroblast phenotype. Chest 143, 963–970 (2013).

    Article  PubMed  Google Scholar 

  324. Saito, R. A. et al. Forkhead box F1 regulates tumor-promoting properties of cancer-associated fibroblasts in lung cancer. Cancer Res. 70, 2644–2654 (2010).

    Article  PubMed  CAS  Google Scholar 

  325. Monti, D. et al. Pilot study demonstrating metabolic and anti-proliferative effects of in vivo anti-oxidant supplementation with N-acetylcysteine in breast cancer. Semin. Oncol. 44, 226–232 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  326. Chang, H. Y. et al. Gene expression signature of fibroblast serum response predicts human cancer progression: similarities between tumors and wounds. PLoS Biol. 2, E7 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  327. Chen, J. L. et al. Stromal responses among common carcinomas correlated with clinicopathologic features. Clin. Cancer Res. 19, 5127–5135 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  328. Finak, G. et al. Stromal gene expression predicts clinical outcome in breast cancer. Nat. Med. 14, 518–527 (2008).

    Article  PubMed  CAS  Google Scholar 

  329. Navab, R. et al. Prognostic gene-expression signature of carcinoma-associated fibroblasts in non-small cell lung cancer. Proc. Natl Acad. Sci. USA 108, 7160–7165 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  330. Planche, A. et al. Identification of prognostic molecular features in the reactive stroma of human breast and prostate cancer. PLoS ONE 6, e18640 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

The work of K.C.P., K.C.V., and A.E.d.G. is supported by National Cancer Institute (NCI) grants U54CA143803, CA163124, CA093900, and CA143055, as well as by the Prostate Cancer Foundation and the Patrick C. Walsh Fund. The work of K.C.V. is supported by NCI grant F32CA206394. The authors are grateful to S. Amend for editing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

K.C.V. and A.E.d.G. wrote the manuscript and prepared the display items. K.C.P. contributed to discussions about the contents of the manuscript. All authors reviewed and edited the manuscript before submission.

Corresponding author

Correspondence to Kenneth C. Valkenburg.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related Links

ClinicalTrials.gov: https://clinicaltrials.gov/ct2/home

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Valkenburg, K.C., de Groot, A.E. & Pienta, K.J. Targeting the tumour stroma to improve cancer therapy. Nat Rev Clin Oncol 15, 366–381 (2018). https://doi.org/10.1038/s41571-018-0007-1

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41571-018-0007-1

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