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:

Fibroblasts in cancer

Key Points

  • Fibroblasts are a key cellular component of tumours.

  • Activated fibroblasts (which are sometimes referred to as myofibroblasts) that are found in association with cancer cells are known as carcinoma-associated fibroblasts (CAFs).

  • The role of fibroblasts in the origin and initiation of cancer invasion is poorly understood, but recent evidence indicates that they can provide oncogenic signals to the transformed epithelia in a paracrine fashion.

  • Some studies indicate that myofibroblasts might facilitate angiogenesis and cancer progression.

  • Epithelial-to-mesenchymal transition is a source for converting epithelial cells into fibroblast-like cells in various tissues.

  • Fibroblasts might also have a role in metastasis.

  • Therapies against CAFs are being considered as a way to control cancer.

Abstract

Tumours are known as wounds that do not heal — this implies that cells that are involved in angiogenesis and the response to injury, such as endothelial cells and fibroblasts, have a prominent role in the progression, growth and spread of cancers. Fibroblasts are associated with cancer cells at all stages of cancer progression, and their structural and functional contributions to this process are beginning to emerge. Their production of growth factors, chemokines and extracellular matrix facilitates the angiogenic recruitment of endothelial cells and pericytes. Fibroblasts are therefore a key determinant in the malignant progression of cancer and represent an important target for cancer therapies.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Activated fibroblasts.
Figure 2: Tumour–stroma interactions during different stages of mammary ductal carcinoma progression.
Figure 3: Functions of activated fibroblasts in the tumour stroma.
Figure 4: Regulation of epithelial-to-mesenchymal transition by the microenvironment.

Similar content being viewed by others

References

  1. Kalluri, R. Basement membranes: structure, assembly and role in tumour angiogenesis. Nature Rev. Cancer 3, 422–433 (2003). A comprehensive review on ECM and tumour angiogenesis.

    Article  CAS  Google Scholar 

  2. Folkman, J. Tumor angiogenesis: therapeutic implications. N. Engl. J. Med. 285, 1182–1186 (1971). The first publication to indicate that angiogenesis is a target for controlling cancer.

    Article  CAS  PubMed  Google Scholar 

  3. O'Reilly, M. S. et al. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell 88, 277–285 (1997).

    Article  CAS  PubMed  Google Scholar 

  4. Ishii, G. et al. Bone-marrow-derived myofibroblasts contribute to the cancer-induced stromal reaction. Biochem. Biophys. Res. Commun. 309, 232–240 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Kim, K. J. et al. Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 362, 841–844 (1993).

    Article  CAS  PubMed  Google Scholar 

  6. Coussens, L. M. & Werb, Z. Inflammation and cancer. Nature 420, 860–867 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. de Visser, K. E., Korets, L. V. & Coussens, L. M. De novo carcinogenesis promoted by chronic inflammation is B lymphocyte dependent. Cancer Cell 7, 411–423 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Tlsty, T. D. & Hein, P. W. Know thy neighbor: stromal cells can contribute oncogenic signals. Curr. Opin. Genet. Dev. 11, 54–59 (2001). An interesting review of tumour stromal cells and their role in oncogenic manipulation.

    Article  CAS  PubMed  Google Scholar 

  9. Elenbaas, B. & Weinberg, R. A. Heterotypic signaling between epithelial tumor cells and fibroblasts in carcinoma formation. Exp. Cell Res. 264, 169–184 (2001).

    Article  CAS  PubMed  Google Scholar 

  10. Mueller, M. M. & Fusenig, N. E. Friends or foes — bipolar effects of the tumour stroma in cancer. Nature Rev. Cancer 4, 839–849 (2004). An in-depth review about tumour stroma.

    Article  CAS  Google Scholar 

  11. Virchow, R. Die Cellularpathologie in Ihrer Begruendung auf Physiologische und Pathologische Gewebelehre (Hirschwald, A., Berlin, Germany, 1858).

    Google Scholar 

  12. Duvall, M. Atlas d'Embryologie (Masson, Paris, 1879).

    Google Scholar 

  13. Tarin, D. & Croft, C. B. Ultrastructural features of wound healing in mouse skin. J. Anat. 105, 189–190 (1969).

    CAS  PubMed  Google Scholar 

  14. Tomasek, J. J., Gabbiani, G., Hinz, B., Chaponnier, C. & Brown, R. A. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nature Rev. Mol. Cell Biol. 3, 349–363 (2002).

    Article  CAS  Google Scholar 

  15. Parsonage, G. et al. A stromal address code defined by fibroblasts. Trends Immunol. 26, 150–156 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Rodemann, H. P. & Muller, G. A. Characterization of human renal fibroblasts in health and disease: II. In vitro growth, differentiation, and collagen synthesis of fibroblasts from kidneys with interstitial fibrosis. Am. J. Kidney Dis. 17, 684–686 (1991).

    Article  CAS  PubMed  Google Scholar 

  17. Chang, H. Y. et al. Diversity, topographic differentiation, and positional memory in human fibroblasts. Proc. Natl Acad. Sci. USA 99, 12877–12882 (2002). This study compares primary fibroblasts that were isolated from various sites and sheds new light on the vast diversity of fibroblasts.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Simian, M. et al. The interplay of matrix metalloproteinases, morphogens and growth factors is necessary for branching of mammary epithelial cells. Development 128, 3117–3131 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. Wiseman, B. S. & Werb, Z. Stromal effects on mammary gland development and breast cancer. Science 296, 1046–1049 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Castor, C. W., Wilson, S. M., Heiss, P. R. & Seidman, J. C. Activation of lung connective tissue cells in vitro. Am. Rev. Respir. Dis. 120, 101–106 (1979).

    CAS  PubMed  Google Scholar 

  21. Muller, G. A. & Rodemann, H. P. Characterization of human renal fibroblasts in health and disease: I. Immunophenotyping of cultured tubular epithelial cells and fibroblasts derived from kidneys with histologically proven interstitial fibrosis. Am. J. Kidney Dis. 17, 680–683. (1991).

    Article  CAS  PubMed  Google Scholar 

  22. Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57–70 (2000). A comprehensive review on cancer.

    Article  CAS  PubMed  Google Scholar 

  23. 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  CAS  PubMed  Google Scholar 

  24. Dvorak, H. F., Form, D. M., Manseau, E. J. & Smith, B. D. Pathogenesis of desmoplasia. I. Immunofluorescence identification and localization of some structural proteins of line 1 and line 10 guinea pig tumors and of healing wounds. J. Natl Cancer Inst. 73, 1195–1205 (1984).

    CAS  PubMed  Google Scholar 

  25. Dolberg, D. S., Hollingsworth, R., Hertle, M. & Bissell, M. J. Wounding and its role in RSV-mediated tumor formation. Science 230, 676–678 (1985).

    Article  CAS  PubMed  Google Scholar 

  26. Sieweke, M. H., Thompson, N. L., Sporn, M. B. & Bissell, M. J. Mediation of wound-related Rous sarcoma virus tumorigenesis by TGF-β. Science 248, 1656–1660 (1990).

    Article  CAS  PubMed  Google Scholar 

  27. Brown, L. F. et al. Vascular stroma formation in carcinoma in situ, invasive carcinoma, and metastatic carcinoma of the breast. Clin. Cancer Res. 5, 1041–1056 (1999).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  29. Senger, D. R. et al. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science 219, 983–985 (1983).

    Article  CAS  PubMed  Google Scholar 

  30. Dvorak, H. F. et al. Distribution of vascular permeability factor (vascular endothelial growth factor) in tumors: concentration in tumor blood vessels. J. Exp. Med. 174, 1275–1278 (1991).

    Article  CAS  PubMed  Google Scholar 

  31. Feng, D. et al. Ultrastructural localization of the vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) receptor-2 (FLK-1, KDR) in normal mouse kidney and in the hyperpermeable vessels induced by VPF/VEGF-expressing tumors and adenoviral vectors. J. Histochem. Cytochem. 48, 545–556 (2000).

    Article  CAS  PubMed  Google Scholar 

  32. Leung, D. W., Cachianes, G., Kuang, W. J., Goeddel, D. V. & Ferrara, N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 246, 1306–1309 (1989).

    Article  CAS  PubMed  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  CAS  PubMed  Google Scholar 

  34. Schedin, P. & Elias, A. Multistep tumorigenesis and the microenvironment. Breast Cancer Res. 6, 93–101 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Shekhar, M. P., Pauley, R. & Heppner, G. Host microenvironment in breast cancer development: extracellular matrix–stromal cell contribution to neoplastic phenotype of epithelial cells in the breast. Breast Cancer Res. 5, 130–135 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. van Kempen, L. C., Ruiter, D. J., van Muijen, G. N. & Coussens, L. M. The tumor microenvironment: a critical determinant of neoplastic evolution. Eur. J. Cell Biol. 82, 539–548 (2003).

    Article  PubMed  Google Scholar 

  37. Brown, L. F., Dvorak, A. M. & Dvorak, H. F. Leaky vessels, fibrin deposition, and fibrosis: a sequence of events common to solid tumors and to many other types of disease. Am. Rev. Respir. Dis. 140, 1104–1107 (1989).

    Article  CAS  PubMed  Google Scholar 

  38. Folkman, J., Watson, K., Ingber, D. & Hanahan, D. Induction of angiogenesis during the transition from hyperplasia to neoplasia. Nature 339, 58–61 (1989).

    Article  CAS  PubMed  Google Scholar 

  39. 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  CAS  PubMed  Google Scholar 

  40. 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  CAS  PubMed  PubMed Central  Google Scholar 

  41. Ishihara, A., Yoshida, T., Tamaki, H. & Sakakura, T. Tenascin expression in cancer cells and stroma of human breast cancer and its prognostic significance. Clin. Cancer Res. 1, 1035–1041 (1995).

    CAS  PubMed  Google Scholar 

  42. Brunner, A. et al. Prognostic significance of tenascin-C-expression in superficial and invasive bladder cancer. J. Clin. Pathol. 57, 927–931 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. De Wever, O. & Mareel, M. Role of myofibroblasts at the invasion front. Biol. Chem. 383, 55–67 (2002).

    Article  CAS  PubMed  Google Scholar 

  44. Bertrand, P. et al. Hyaluronan (hyaluronic acid) and hyaluronectin in the extracellular matrix of human breast carcinomas: comparison between invasive and non-invasive areas. Int. J. Cancer 52, 1–6 (1992).

    Article  CAS  PubMed  Google Scholar 

  45. Barsky, S. H. & Gopalakrishna, R. Increased invasion and spontaneous metastasis of BL6 melanoma with inhibition of the desmoplastic response in C57 BL/6 mice. Cancer Res. 47, 1663–1667 (1987).

    CAS  PubMed  Google Scholar 

  46. Netti, P. A., Berk, D. A., Swartz, M. A., Grodzinsky, A. J. & Jain, R. K. Role of extracellular matrix assembly in interstitial transport in solid tumors. Cancer Res. 60, 2497–2503 (2000).

    CAS  PubMed  Google Scholar 

  47. Brown, E. B., Boucher, Y., Nasser, S. & Jain, R. K. Measurement of macromolecular diffusion coefficients in human tumors. Microvasc. Res. 67, 231–236 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Hynes, R. O. Integrins: versatility, modulation, and signaling in cell adhesion. Cell 69, 11–25 (1992).

    Article  CAS  PubMed  Google Scholar 

  49. Ryan, G. B. et al. Myofibroblasts in an avascular fibrous tissue. Lab. Invest. 29, 197–206 (1973).

    CAS  PubMed  Google Scholar 

  50. Ryan, G. B. et al. Myofibroblasts in human granulation tissue. Hum. Pathol. 5, 55–67 (1974).

    Article  CAS  PubMed  Google Scholar 

  51. Tsukada, T., McNutt, M. A., Ross, R. & Gown, A. M. HHF35, a muscle actin-specific monoclonal antibody. II. Reactivity in normal, reactive, and neoplastic human tissues. Am. J. Pathol. 127, 389–402 (1987).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Schor, S. L., Schor, A. M., Grey, A. M. & Rushton, G. Foetal and cancer patient fibroblasts produce an autocrine migration-stimulating factor not made by normal adult cells. J. Cell Sci. 90, 391–399 (1988).

    Article  CAS  PubMed  Google Scholar 

  53. Durning, P., Schor, S. L. & Sellwood, R. A. Fibroblasts from patients with breast cancer show abnormal migratory behaviour in vitro. Lancet 2, 890–892 (1984).

    Article  CAS  PubMed  Google Scholar 

  54. Barsky, S. H., Green, W. R., Grotendorst, G. R. & Liotta, L. A. Desmoplastic breast carcinoma as a source of human myofibroblasts. Am. J. Pathol. 115, 329–333 (1984).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Sappino, A. P., Skalli, O., Jackson, B., Schurch, W. & Gabbiani, G. Smooth-muscle differentiation in stromal cells of malignant and non-malignant breast tissues. Int. J. Cancer 41, 707–712 (1988).

    Article  CAS  PubMed  Google Scholar 

  56. Gabbiani, G. The myofibroblast in wound healing and fibrocontractive diseases. J. Pathol. 200, 500–503 (2003).

    Article  CAS  PubMed  Google Scholar 

  57. Ronnov-Jessen, L. & Petersen, O. W. Induction of α-smooth muscle actin by transforming growth factor-β1 in quiescent human breast gland fibroblasts. Implications for myofibroblast generation in breast neoplasia. Lab. Invest. 68, 696–707 (1993).

    CAS  PubMed  Google Scholar 

  58. Lohr, M. et al. Transforming growth factor-β1 induces desmoplasia in an experimental model of human pancreatic carcinoma. Cancer Res. 61, 550–555 (2001).

    CAS  PubMed  Google Scholar 

  59. Aoyagi, Y. et al. Overexpression of TGF-β by infiltrated granulocytes correlates with the expression of collagen mRNA in pancreatic cancer. Br. J. Cancer 91, 1316–1326 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Akhurst, R. J. TGF-β antagonists: why suppress a tumor suppressor? J. Clin. Invest. 109, 1533–1536 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Bronzert, D. A. et al. Synthesis and secretion of platelet-derived growth factor by human breast cancer cell lines. Proc. Natl Acad. Sci. USA 84, 5763–5767 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Forsberg, K., Valyi-Nagy, I., Heldin, C. H., Herlyn, M. & Westermark, B. Platelet-derived growth factor (PDGF) in oncogenesis: development of a vascular connective tissue stroma in xenotransplanted human melanoma producing PDGF-BB. Proc. Natl Acad. Sci. USA 90, 393–397 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Shao, Z. M., Nguyen, M. & Barsky, S. H. Human breast carcinoma desmoplasia is PDGF initiated. Oncogene 19, 4337–4345 (2000).

    Article  CAS  PubMed  Google Scholar 

  64. Armelin, H. A. Pituitary extracts and steroid hormones in the control of 3T3 cell growth. Proc. Natl Acad. Sci. USA 70, 2702–2706 (1973).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Folkman, J. et al. A heparin-binding angiogenic protein — basic fibroblast growth factor — is stored within basement membrane. Am. J. Pathol. 130, 393–400 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Strutz, F. et al. Basic fibroblast growth factor expression is increased in human renal fibrogenesis and may mediate autocrine fibroblast proliferation. Kidney Int. 57, 1521–1538. (2000).

    Article  CAS  PubMed  Google Scholar 

  67. Hay, E. D. An overview of epithelio–mesenchymal transformation. Acta Anat. 154, 8–20 (1995).

    Article  CAS  PubMed  Google Scholar 

  68. Thiery, J. P. Epithelial–mesenchymal transitions in tumor progression. Nature Rev. Cancer 2, 442–454 (2002). A good review on the role of EMT in cancer.

    Article  CAS  Google Scholar 

  69. Kalluri, R. & Neilson, E. G. Epithelial–mesenchymal transition and its implications for fibrosis. J. Clin. Invest. 112, 1776–1784 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Fukino, K. et al. Combined total genome loss of heterozygosity scan of breast cancer stroma and epithelium reveals multiplicity of stromal targets. Cancer Res. 64, 7231–7236 (2004).

    Article  CAS  PubMed  Google Scholar 

  71. Tuhkanen, H. et al. Genetic alterations in the peritumoral stromal cells of malignant and borderline epithelial ovarian tumors as indicated by allelic imbalance on chromosome 3p. Int. J. Cancer 109, 247–252 (2004).

    Article  CAS  PubMed  Google Scholar 

  72. Ellsworth, D. L. et al. Genomic patterns of allelic imbalance in disease free tissue adjacent to primary breast carcinomas. Breast Cancer Res. Treat. 88, 131–139 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  74. Petersen, O. W. et al. Epithelial to mesenchymal transition in human breast cancer can provide a nonmalignant stroma. Am. J. Pathol. 162, 391–402 (2003). An interesting study indicating that cancer cells can contribute to the accumulation of stromal fibroblasts by undergoing a complete EMT.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Zeisberg, M. & Kalluri, R. The role of epithelial-to-mesenchymal transition in renal fibrosis. J. Mol. Med. 82, 175–181 (2004).

    Article  PubMed  Google Scholar 

  76. Zeisberg, M. et al. BMP-7 counteracts TGF-β1-induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nature Med. 9, 964–968 (2003). The first study to indicate that targeting EMT can reduce the numbers of tissue fibroblasts and reverse fibrosis.

    Article  CAS  PubMed  Google Scholar 

  77. Iwano, M. et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J. Clin. Invest. 110, 341–350. (2002). Provides evidence that FSP1 protein is a fibroblast-specific protein, and provides further evidence relating to the role of EMT in organ fibrosis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Kopelovich, L. Genetic predisposition to cancer in man: in vitro studies. Int. Rev. Cytol. 77, 63–88 (1982).

    Article  CAS  PubMed  Google Scholar 

  79. Schor, S. L. et al. Occurrence of a fetal fibroblast phenotype in familial breast cancer. Int. J. Cancer 37, 831–836 (1986).

    Article  CAS  PubMed  Google Scholar 

  80. Kuperwasser, C. et al. Reconstruction of functionally normal and malignant human breast tissues in mice. Proc. Natl Acad. Sci. USA 101, 4966–4971 (2004). Demonstrates that fibroblasts aid mammary epithelia in forming functional breast tissue, and carcinogenesis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Bhowmick, N. A. et al. TGF-β signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science 303, 848–851 (2004). Indicates that deletion of TGFβ type II receptor in FSP1-positive cells leads to the emergence of epithelial tumours.

    Article  CAS  PubMed  Google Scholar 

  82. 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). Indicates a prominent role for fibroblasts in tumour angiogenesis facilitated by SDF1.

    Article  CAS  PubMed  Google Scholar 

  83. Olumi, A. F. et al. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res. 59, 5002–5011 (1999). A study that used co-culture methods to elucidate the role of CAFs in cancer progression.

    CAS  PubMed  Google Scholar 

  84. Dimanche-Boitrel, M. T. et al. In vivo and in vitro invasiveness of a rat colon-cancer cell line maintaining E-cadherin expression: an enhancing role of tumor-associated myofibroblasts. Int. J. Cancer 56, 512–521 (1994).

    Article  CAS  PubMed  Google Scholar 

  85. 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  CAS  PubMed  Google Scholar 

  86. Stetler-Stevenson, W. G., Aznavoorian, S. & Liotta, L. A. Tumor cell interactions with the extracellular matrix during invasion and metastasis. Annu. Rev. Cell Biol. 9, 541–573 (1993).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Lochter, A. et al. Matrix metalloproteinase stromelysin-1 triggers a cascade of molecular alterations that leads to stable epithelial-to-mesenchymal conversion and a premalignant phenotype in mammary epithelial cells. J. Cell Biol. 139, 1861–1872 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Olaso, E. et al. Tumor-dependent activation of rodent hepatic stellate cells during experimental melanoma metastasis. Hepatology 26, 634–642 (1997).

    Article  CAS  PubMed  Google Scholar 

  90. Olaso, E. et al. Proangiogenic role of tumor-activated hepatic stellate cells in experimental melanoma metastasis. Hepatology 37, 674–685 (2003).

    Article  CAS  PubMed  Google Scholar 

  91. Cornil, I. et al. Fibroblast cell interactions with human melanoma cells affect tumor cell growth as a function of tumor progression. Proc. Natl Acad. Sci. USA 88, 6028–6032 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Grum-Schwensen, B. et al. Suppression of tumor development and metastasis formation in mice lacking the S100A4mts1 gene. Cancer Res. 65, 3772–3780 (2005).

    Article  CAS  PubMed  Google Scholar 

  93. Bhowmick, N. A., Neilson, E. G. & Moses, H. L. Stromal fibroblasts in cancer initiation and progression. Nature 432, 332–337 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Siegel, P. M. & Massague, J. Cytostatic and apoptotic actions of TGF-β in homeostasis and cancer. Nature Rev. Cancer 3, 807–821 (2003).

    Article  CAS  Google Scholar 

  95. Bierie, B. & Moses, H. L. TGF-β and cancer. Cytokine Growth Factor Rev. 17, 29–40 (2006).

    Article  CAS  PubMed  Google Scholar 

  96. Derynck, R., Akhurst, R. J. & Balmain, A. TGF-β signaling in tumor suppression and cancer progression. Nature Genet. 29, 117–129 (2001).

    Article  CAS  PubMed  Google Scholar 

  97. Cheng, N. et al. Loss of TGF-β type II receptor in fibroblasts promotes mammary carcinoma growth and invasion through upregulation of TGF-α-, MSP- and HGF-mediated signaling networks. Oncogene 24, 5053–5068 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  99. Rettig, W. J. et al. Regulation and heteromeric structure of the fibroblast activation protein in normal and transformed cells of mesenchymal and neuroectodermal origin. Cancer Res. 53, 3327–3335 (1993).

    CAS  PubMed  Google Scholar 

  100. Wang, X. M., Yu, D. M., McCaughan, G. W. & Gorrell, M. D. Fibroblast activation protein increases apoptosis, cell adhesion, and migration by the LX-2 human stellate cell line. Hepatology 42, 935–945 (2005).

    Article  CAS  PubMed  Google Scholar 

  101. 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).

    CAS  PubMed  Google Scholar 

  102. Zeisberg, M. et al. Bone morphogenic protein-7 inhibits progression of chronic renal fibrosis associated with two genetic mouse models. Am. J. Physiol. Renal Physiol. 285, F1060–F1067 (2003).

    Article  CAS  PubMed  Google Scholar 

  103. Zeisberg, M., Strutz, F. & Muller, G. A. Role of fibroblast activation in inducing interstitial fibrosis. J. Nephrol. 13 (Suppl. 3), S111–S120 (2000).

    PubMed  Google Scholar 

  104. Nakamura, T. et al. Hepatocyte growth factor prevents tissue fibrosis, remodeling, and dysfunction in cardiomyopathic hamster hearts. Am. J. Physiol. Heart Circ. Physiol. 288, H2131–H2139 (2005).

    Article  CAS  PubMed  Google Scholar 

  105. Kim, W. H., Matsumoto, K., Bessho, K. & Nakamura, T. Growth inhibition and apoptosis in liver myofibroblasts promoted by hepatocyte growth factor leads to resolution from liver cirrhosis. Am. J. Pathol. 166, 1017–1028 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Dai, C. et al. Intravenous administration of hepatocyte growth factor gene ameliorates diabetic nephropathy in mice. J. Am. Soc. Nephrol. 15, 2637–2647 (2004).

    Article  CAS  PubMed  Google Scholar 

  107. Isaka, Y. et al. Electroporation-mediated HGF gene transfection protected the kidney against graft injury. Gene Ther. 12, 815–820 (2005).

    Article  CAS  PubMed  Google Scholar 

  108. Yang, J., Dai, C. & Liu, Y. Hepatocyte growth factor gene therapy and angiotensin II blockade synergistically attenuate renal interstitial fibrosis in mice. J. Am. Soc. Nephrol. 13, 2464–2477 (2002).

    Article  CAS  PubMed  Google Scholar 

  109. McDonald, G. A. et al. Relaxin increases ubiquitin-dependent degradation of fibronectin in vitro and ameliorates renal fibrosis in vivo. Am. J. Physiol. Renal Physiol. 285, F59–F67 (2003).

    Article  CAS  PubMed  Google Scholar 

  110. Heeg, M. H. et al. The antifibrotic effects of relaxin in human renal fibroblasts are mediated in part by inhibition of the Smad2 pathway. Kidney Int. 68, 96–109 (2005).

    Article  CAS  PubMed  Google Scholar 

  111. Samuel, C. S. et al. Relaxin modulates cardiac fibroblast proliferation, differentiation, and collagen production and reverses cardiac fibrosis in vivo. Endocrinology 145, 4125–4133 (2004).

    Article  CAS  PubMed  Google Scholar 

  112. Seibold, J. R. et al. Recombinant human relaxin in the treatment of scleroderma. A randomized, double-blind, placebo-controlled trial. Ann. Intern. Med. 132, 871–879 (2000).

    Article  CAS  PubMed  Google Scholar 

  113. Binder, C. et al. Elevated concentrations of serum relaxin are associated with metastatic disease in breast cancer patients. Breast Cancer Res. Treat. 87, 157–166 (2004).

    Article  CAS  PubMed  Google Scholar 

  114. Binder, C., Hagemann, T., Husen, B., Schulz, M. & Einspanier, A. Relaxin enhances in-vitro invasiveness of breast cancer cell lines by up-regulation of matrix metalloproteases. Mol. Hum. Reprod. 8, 789–796 (2002).

    Article  CAS  PubMed  Google Scholar 

  115. Lazarides, E. & Balzer, D. R. Jr. Specificity of desmin to avian and mammalian muscle cells. Cell 14, 429–438 (1978).

    Article  CAS  PubMed  Google Scholar 

  116. Franke, W. W., Schmid, E., Osborn, M. & Weber, K. Different intermediate-sized filaments distinguished by immunofluorescence microscopy. Proc. Natl Acad. Sci. USA 75, 5034–5038 (1978).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Franke, W. W., Schmid, E., Osborn, M. & Weber, K. Intermediate-sized filaments of human endothelial cells. J. Cell Biol. 81, 570–580 (1979).

    Article  CAS  PubMed  Google Scholar 

  118. Mork, C., van Deurs, B. & Petersen, O. W. Regulation of vimentin expression in cultured human mammary epithelial cells. Differentiation 43, 146–156 (1990).

    Article  CAS  PubMed  Google Scholar 

  119. Schmid, E. et al. Distribution of vimentin and desmin filaments in smooth muscle tissue of mammalian and avian aorta. Exp. Cell Res. 137, 329–340 (1982).

    Article  CAS  PubMed  Google Scholar 

  120. Strutz, F. et al. Identification and characterization of a fibroblast marker: FSP1. J. Cell Biol. 130, 393–405 (1995).

    Article  CAS  PubMed  Google Scholar 

  121. Vogel, W., Gish, G. D., Alves, F. & Pawson, T. The discoidin domain receptor tyrosine kinases are activated by collagen. Mol. Cell 1, 13–23 (1997).

    Article  CAS  PubMed  Google Scholar 

  122. Goldsmith, E. C. et al. Organization of fibroblasts in the heart. Dev. Dyn. 230, 787–794 (2004).

    Article  CAS  PubMed  Google Scholar 

  123. Ramirez-Montagut, T. et al. FAPα, a surface peptidase expressed during wound healing, is a tumor suppressor. Oncogene 23, 5435–5446 (2004).

    Article  CAS  PubMed  Google Scholar 

  124. Gardner, H., Kreidberg, J., Koteliansky, V. & Jaenisch, R. Deletion of integrin α1 by homologous recombination permits normal murine development but gives rise to a specific deficit in cell adhesion. Dev. Biol. 175, 301–313 (1996).

    Article  CAS  PubMed  Google Scholar 

  125. Sudhakar, A. et al. Human α1 type IV collagen NC1 domain exhibits distinct antiangiogenic activity mediated by α1β1 integrin. J. Clin. Invest. 115, 2801–2810 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Mussini, E., Hutton, J. J. Jr & Udenfriend, S. Collagen proline hydroxylase in wound healing, granuloma formation, scurvy, and growth. Science 157, 927–929 (1967).

    Article  CAS  PubMed  Google Scholar 

  127. Langness, U. & Udenfriend, S. Collagen biosynthesis in nonfibroblastic cell lines. Proc. Natl Acad. Sci. USA 71, 50–51 (1974).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Florin, L. et al. Cre recombinase-mediated gene targeting of mesenchymal cells. Genesis 38, 139–144 (2004).

    Article  CAS  PubMed  Google Scholar 

  129. Hogan, B. L. & Yingling, J. M. Epithelial–mesenchymal interactions and branching morphogenesis of the lung. Curr. Opin. Genet. Dev. 8, 481–486 (1998).

    Article  CAS  PubMed  Google Scholar 

  130. Clayton, A. et al. Cellular activation through the ligation of intercellular adhesion molecule-1. J. Cell Sci. 111, 443–453 (1998).

    Article  CAS  PubMed  Google Scholar 

  131. Strieter, R. M. et al. Monocyte chemotactic protein gene expression by cytokine-treated human fibroblasts and endothelial cells. Biochem. Biophys. Res. Commun. 162, 694–700 (1989).

    Article  CAS  PubMed  Google Scholar 

  132. Rollins, B. J., Stier, P., Ernst, T. & Wong, G. G. The human homolog of the JE gene encodes a monocyte secretory protein. Mol. Cell. Biol. 9, 4687–4695 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Duda, D. G. et al. Differential transplantability of tumor-associated stromal cells. Cancer Res. 64, 5920–5924 (2004).

    Article  CAS  PubMed  Google Scholar 

  134. Grupp, C. et al. A novel model to study renal myofibroblast formation in vitro. Kidney Int. 59, 543–553 (2001).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The work from the author's laboratories was supported by the National Institutes of Health (NIH) and the Center for Matrix Biology (R.K.). M.Z. is funded by a NIH training grant to BIDMC. We would like to extend our special thanks to H. Dvorak and M. A. Grant for critical reading of this manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Raghu Kalluri.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

National Cancer Institute

bladder cancer

breast cancer

melanoma

FURTHER INFORMATION

Raghu Kalluri's web page

Glossary

Stroma

Extracellular matrix, mesenchymal cells and a scaffold consisting of blood and lymphoid vessels, nerves and inflammatory cells.

Cancer-associated fibroblast

Activated fibroblasts within desmoplastic lesions that are associated with malignant tumours and often express α-smooth-muscle actin.

Extracellular matrix

A network of fibrous proteins such as collagens and fibronectins, which are embedded in a visco-elastic gel of glycosaminoglycans, proteoglycans and glycoproteins. These filamentous proteins provide tensile strength and channels for communication by and movement of cells.

Fibrillar ECM

Unorganized, loosely assembled form of ECM. The most important constituents of fibrillar ECM are fibronectin and type I collagen, which are thought to serve as a scaffold for other ECM components.

Basement membrane

A specialized extracellular matrix present at the baso-lateral side of cells. It appears as an amorphous, dense and proteinaceous structure. This term is used interchangeably with basal lamina.

Matrix metalloproteinases

Endopeptidases that belong to a family of zinc-dependent proteases with more than 21 human forms. Their main substrates are matrix molecules such as collagen, but many non-matrix substrates have also been identified recently.

Fibrosis

An excessive deposition of extracellular matrix, which leads to the destruction of organ structure and the impairment of organ function. The main mediators of tissue fibrosis are activated fibroblasts.

Co-culture experiment

In vitro experiment in which cancer cells are either exposed to conditioned media from CAFs or directly cultured in the presence of CAFs.

Reconstitution experiment

Study in which a suspension of CAFs and tumour cells (often in a ratio that favours the CAFs) are injected into mice. Such studies demonstrated that the presence of CAFs enhanced tumour progression.

Hox genes

The Hox genes encode a family of homeodomain transcription factors that determine positional identity along the anterior–posterior and secondary axis in animals.

Myofibroblasts

The myofibroblast was initially identified by means of electron microscopy as a fibroblast with features that are more typical of smooth muscle cells, such as possessing bundles of microfilaments and maintaining gap junctions. Myofibroblasts are commonly detected by their expression of α-smooth-muscle actin.

NOD/SCID mice

Mice with multiple defects in innate immunity, including a largely reduced natural-killer-cell activity and a twofold-reduced bone-marrow cellularity.

Cleared-fat-pad transplantation system

A cleared fat pad, devoid of any epithelium, into which mouse mammary epithelial cells are engrafted before puberty, causing an anatomically normal mammary gland to develop. However, human mammary epithelial cells injected into the cleared mouse fat pad do not form a mammary gland.

Senescence

A programme that is activated by normal cells in various situations of stress to prevent further proliferation. Senescence occurs in response to telomere capping, DNA damage, oxidative stress or activation of oncogenes.

Epithelial-to-mesenchymal transition

A process that is necessary for embryonic development, tumour progression and organ fibrosis. During EMT epithelial characteristics are lost and the mesenchymal phenotype is acquired.

Pericyte

Mesenchymal cells that wrap around endothelial cells in microvessels. They share the common basement membrane with the vascular endothelial cells.

Stellate cells

Fibroblast-like cells in the liver that express glial fibrillary acidic protein and nestin, and are thought to be neural-crest-derived.

Scleroderma

Fibrosis involving the skin.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kalluri, R., Zeisberg, M. Fibroblasts in cancer. Nat Rev Cancer 6, 392–401 (2006). https://doi.org/10.1038/nrc1877

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc1877

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing