Thromb Haemost 2005; 93(04): 676-681
DOI: 10.1160/TH05-01-0054
Theme Issue Article
Schattauer GmbH

Plasminogen activation and cancer

Keld Danø
1   Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
,
Niels Behrendt
1   Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
,
Gunilla Høyer-Hansen
1   Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
,
Morten Johnsen
2   Institute of Molecular Biology, University of Copenhagen, Copenhagen, Denmark
,
Leif R. Lund
1   Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
,
Michael Ploug
1   Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
,
John Rømer
1   Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
› Author Affiliations
Further Information

Publication History

Received 24 January 2005

Accepted after revision 14 February 2005

Publication Date:
14 December 2017 (online)

Summary

Breakdown of the extracellular matrix is crucial for cancer invasion and metastasis. It is accomplished by the concerted action of several proteases, including the serine protease plasmin and a number of matrix metalloproteases. The activity of each of these proteases is regulated by an array of activators, inhibitors and cellular receptors. Thus, the generation of plasmin involves the pro-enzyme plasminogen, the urokinase type plasminogen activator uPA and its pro-enzyme pro-uPA, the uPA inhibitor PAI-1, the cell surface uPA receptor uPAR, and the plasmin inhibitor α2-antiplasmin. Furthermore, the regulation of extracellular proteolysis in cancer involves a complex interplay between cancer cells and non-malignant stromal cells in the expression of the molecular components involved. For some types of cancer, this cellular interplay mimics that observed in the tissue of origin during non-neoplastic tissue remodelling processes. We propose that cancer invasion can be considered as uncontrolled tissue remodelling. Inhibition of extracellular proteases is an attractive approach to cancer therapy. Because proteases have many different functions in the normal organism, efficient inhibition will have toxic side effects. In cancer invasion, like in normal tissue remodelling processes, there appears to be a functional overlap between different extracellular proteases. This redundancy means that combinations of protease inhibitors must be used. Such combination therapy, however, is also likely to increase toxicity. Therefore for each type of cancer, a combination of protease inhibitors that is optimised with respect to both maximal therapeutic effect and minimal toxic side effects need to be identified.

 
  • References

  • 1 Rømer J. Skin cancer and wound healing. Tissuespecific similarities in extracellular proteolysis. APMIS 2003; (Suppl. 107) 111: 1-36.
  • 2 Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2002; 2: 161-73.
  • 3 Andreasen PA, Kjøller L, Christensen L. et al. The Urokinase-type plasminogen activator system in cancer metastasis. A review. Int J Cancer 1997; 72: 1-22.
  • 4 Ploug M. Structure-function relationships in the interaction between the urokinase- type plasminogen activator and its receptor. Curr Pharmaceut Design 2003; 9: 1499-528.
  • 5 Behrendt N. The urokinase receptor (uPAR) and the uPAR-associated protein (uPARAP/Endo 180): membrane proteins engaged in matrix turnover during tissue remodeling. Biol Chem 2004; 385: 103-36.
  • 6 Johnsen M, Lund LR, Rømer J. et al. Cancer invasion and tissue remodeling: Common themes in proteolytic matrix degradation. Curr Opin Cell Biol 1998; 10: 667-71.
  • 7 Almholt K, Lund LR, Rygaard J. et al. Reduced metastasis of transgenic mammary cancer in urokinase-deficient mice. Int J Cancer 2005; 113: 525-32.
  • 8 Danø K, Andreasen PA, Grøndahl-Hansen J. et al. Plasminogen activators, tissue degradation, and cancer. Adv Cancer Res 1985; 44: 139-266.
  • 9 Danø K, Rømer J, Nielsen BS. et al. Cancer invasion and tissue remodeling – cooperation of protease systems and cell types. APMIS 1999; 107: 120-7.
  • 10 Collen D, Lijnen HR. Thrombolytic agents. Thromb Haemost 2005; 93: 627-30.
  • 11 Melchor JP, Strickland S. Tissue plasminogen activator in central nervous system physiology and pathology. Thromb Haemost 2005; 93: 655-60.
  • 12 Carmeliet P, Schoonjans L, Kieckens L. et al. Physiological consequences of loss of plasminogen activator gene function in mice. Nature 1994; 368: 419-24.
  • 13 Bugge TH, Flick MJ, Danton MJ. et al. Urokinasetype plasminogen activator is effective in fibrin clearance in the absence of its receptor or tissue-type plasminogen activator. Proc Natl Acad Sci USA 1996; 93: 5899-904.
  • 14 Selvarajan S, Lund LR, Takeuchi T. et al. A plasma kallikrein-dependent cascade required for adipocyte differentiation. Nature Cell Biol 2001; 3: 267-75.
  • 15 Loskutoff DJ, Curriden SA, Hu G, Deng G. Regulation of cell adhesion by PAI-1. APMIS 1999; 107: 54-61.
  • 16 Skriver L, Nielsen LS, Stephens R. et al. Plasminogen activator released as inactive proenzyme from murine cells transformed by sarcoma virus. Eur J Biochem 1982; 124: 409-14.
  • 17 Petersen LC, Lund LR, Nielsen LS. et al. One-chain urokinase-type plasminogen activator from human sarcoma cells is a proenzyme with little or no intrinsic activity. J Biol Chem 1988; 263: 11189-95.
  • 18 Vassalli JD, Baccino D, Belin D. A cellular binding site for the Mr 55,000 form of the human plasminogen activator, urokinase. J Cell Biol 1985; 100: 86-92.
  • 19 Roldan AL, Cubellis MV, Masucci MT. et al. Cloning and expression of the receptor for human urokinase plasminogen activator, a central molecule in cell surface plasmin dependent proteolysis. EMBO J 1990; 9: 467-74.
  • 20 Llinas P, Le Du MH, Gårdsvoll H. et al. Crystal structure of the human urokinase plasminogen activator receptor. Thromb Haemost 2005; 93: A8 [abstract #26].
  • 21 Ellis V, Behrendt N, Danø K. Plasminogen activation by receptor-bound urokinase: a kinetic study with both cell-associated and isolated receptor. J Biol Chem 1991; 266: 12752-8.
  • 22 Bugge TH, Suh TT, Flick MJ. et al. The receptor for urokinase-type plasminogen activator is not essential for mouse development or fertility. J Biol Chem 1995; 270: 16886-94.
  • 23 Zhou HM, Nichols A, Meda P. et al. Urokinase-type plasminogen activator and its receptor synergize to promote pathogenic proteolysis. EMBO J 2000; 19: 4817-26.
  • 24 Bolon I, Zhou HM, Charron Y. et al. Plasminogen mediates the pathological effects of urokinase-type plasminogen activator overexpression. Am J Pathol 2004; 164: 2299-304.
  • 25 Liu S, Aaronson H, Mitola DJ. et al. Potent antitumor activity of a urokinase-activated engineered anthrax toxin. Proc Natl Acad Sci U S A 2003; 100: 657-62.
  • 26 Kjøller L. The urokinase plasminogen activator receptor in the regulation of the actin cytoskeleton and cell mobility. Biol Chem 2002; 383: 5-19.
  • 27 Mazzieri R, Blasi F. The urokinase receptor and the regulation of cell proliferation. Thromb Haemost 2005; 93: 641-6.
  • 28 May AE, Kanse SM, Lund LR. et al. Urokinase receptor (CD87) regulates leukocyte recruitment via beta 2 integrins in vivo. J Exp Med 1998; 188: 1029-37.
  • 29 Gyetko MR, Sud S, Kendall T. et al. Urokinase receptor- deficient mice have impaired neutrophil recruitment in response to pulmonary Pseudomonas aeruginosa infection. J Immunol 2000; 165: 1513-9.
  • 30 Rijneveld AW, Levi M, Florquin S. et al. Urokinase receptor is necessary for adequate host defense against pneumococcal pneumonia. J Immunol 2002; 168: 3507-11.
  • 31 Unkeless JC, Danø K, Kellerman GM. et al. Fibrinolysis associated with oncogenic transformation: Partial purification and characterization of the cell factor – a plasminogen activator. J Biol Chem 1974; 249: 4295-305.
  • 32 Danø K, Reich E. Serine enzymes released by cultured neoplastic cells. J Exp Med 1978; 147: 745-57.
  • 33 Skriver L, Larsson I L, Kielberg V. et al. Immunocytochemical localization of urokinase-type plasminogen activator in Lewis lung carcinoma. J Cell Biol 1984; 99: 753-8.
  • 34 Grøndahl JH, Ralfkiær E, Kirkeby LT. et al. Localization of urokinase-type plasminogen activator in stromal cells in adenocarcinomas of the colon in humans. Am J Pathol 1991; 138: 111-7.
  • 35 Pyke C, Kristensen P, Ralfkiær E. et al. Urokinasetype plasminogen activator is expressed in stromal cells and its receptor in cancer cells at invasive foci in human colon adenocarcinomas. Am J Pathol 1991; 138: 1059-67.
  • 36 Danø K, Grrndahl JH, Eriksen J. et al. The receptor for urokinase plasminogen activator: Stromal cell involvement in extracellular proteolysis during cancer invasion. In. Proteolysis and Protein Turnover A.J.. Barrett & J. Bond (eds.); Portland Press, London: 1993: 239-45.
  • 37 Nielsen BS, Sehested M, Timshel S. et al. Messenger RNA for urokinase plasminogen activator is expressed in myofibroblasts adjacent to cancer cells in human breast cancer. Lab Invest 1996; 74: 168-77.
  • 38 Nielsen BS, Sehested M, Dunn S. et al. Urokinase plasminogen activator is localized in stromal cells in ductal breast cancer. Lab Invest 2001; 81: 1485-501.
  • 39 Usher PA, Thomsen OF, Iversen P. et al. Expression of urokinase plasminogen activator, its receptor and type-1 inhibitor in malignant and benign prostate tissue. Int J Cancer 2005; 113: 870-80.
  • 40 Rømer J, Lund LR, Eriksen J. et al. Differential expression of urokinase-type plasminogen activator and its type-1 inhibitor during healing of mouse skin wounds. J Invest Dermatol 1991; 97: 803-11.
  • 41 Pyke C, Græm N, Ralfkiær E. et al. Receptor for urokinase is present in tumor-associated macrophages in ductal breast carcinoma. Cancer Res 1993; 53: 1911-5.
  • 42 Rømer J, Pyke C, Lund LR. et al. Cancer cell expression of urokinase-type plasminogen activator receptor mRNA in squamous cell carcinomas of the skin. J Invest Dermatol 2001; 116: 353-8.
  • 43 Grøndahl-Hansen J, Christensen IJ, Rosenquist C. et al. High levels of urokinase-type plasminogen activator and its inhibitor PAI-1 in cytosolic extracts of breast carcinomas are associated with poor prognosis. Cancer Res 1993; 53: 2513-21.
  • 44 Grøndahl-Hansen J, Peters HA, van Putten WLJ. et al. Prognostic significance of the receptor for urokinase plasminogen activator in breast cancer. Clin Cancer Res 1995; 1: 1079-87.
  • 45 Stephens RW, Nielsen HJ, Christensen IJ. et al. Plasma urokinase receptor in colorectal cancer patients: Relation to prognosis. J. Natl Cancer Inst 1999; 91: 869-74.
  • 46 Harbeck N, Kates RE, Look MP. et al. Enhanced benefit from adjuvant chemotherapy in breast cancer patients classified high-risk according to urokinasetype plasminogen activator (uPA) and plasminogen activator inhibitor type 1 (n = 3424). Cancer Res 2002; 62: 4617-22.
  • 47 Almasi CE, Høyer GH, Christensen IJ. et al. Prognostic impact of liberated domain I of the urokinase plasminogen activator receptor in squamous cell lung cancer tissue. Lung Cancer; in press.:
  • 48 Almholt K, Johnsen M. Stromal cell involvement in cancer. Recent Results Cancer Res 2003; 162: 31-42.
  • 49 Guy C, Cardiff RD, Muller WJ. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol Cell Biol 1992; 12: 954-61.
  • 50 Hewitt R, Danø K. Stromal cell expression of components of matrix-degrading protease systems in human cancer. Enzyme Protein 1996; 49: 163-73.
  • 51 Wolf C, Rouyer N, Lutz Y. et al. Stromelysin 3 belongs to a subgroup of proteinases expressed by breast carcinoma fibroblastic cells and possible implicated in tumor progression. Proc Natl Acad Sci U S A 1993; 90: 1843-7.
  • 52 Heppner KJ, Matrisian LM, Jensen RA. et al. Expression of most matrix metalloproteinases family members in breast cancer represents a tumor-induced host response. Am J Pathol 1996; 149: 273-82.
  • 53 Lefebvre O, Wolf C, Limacker JM. et al. The breast cancer-associated stromelysin-3 gene is expressed during mouse mammary gland apoptosis. J Cell Biol 1992; 119: 997-1002.
  • 54 Lund LR, Rømer J, Thomasset N. et al. Two distinct phases of apoptosis in mammary gland involution:proteinase- independent and -dependent pathways. Development 1996; 122: 181-93.
  • 55 Grøndahl-Hansen J, Lund LR, Ralfkiær E. et al. Urokinase and tssue-type plasminogen activators in keratinocytes during wound reepithelialization in vivo. J Invest Dermatol 1988; 90: 790-5.
  • 56 Rømer J, Lund LR, Eriksen J. et al. The receptor for urokinase-type plasminogen activator is expressed by keratinocytes at the leading edge during re-epithelialization of mouse skin wounds. J Invest Dermatol 1994; 102: 519-22.
  • 57 Lund LR, Rømer J, Bugge TH. et al. Functional overlap between two classes of matrix degrading proteases in wound healing. EMBO J 1999; 18: 4645-56.
  • 58 Madlener M, Parks WC, Werner S. Matrix metalloproteinases (MMPs) and their physiological inhibitors (TIMPs) are differentially expressed during excisional skin wound repair. Exp Cell Res 1998; 242: 201-10.
  • 59 Pyke C, Ralfkiær E, Huhtala P. et al. Localization of messenger RNA for Mr 72,000 and 92,000 Type IV collagenases in human skin cancers by in situ hybridizations. Cancer Res 1992; 52: 1336-41.
  • 60 Ariola K, Johansson N, Kariniemi AL. et al. Human collagenase-3 is expressed in malignant squamous epithelium of the skin. J Invest Dermatol 1997; 109: 225-31.
  • 61 Larsson I L, Skriver L, Nielsen LS. et al. Distribution of urokinase-type plasminogen activator immunoreactivity in the mouse. J Cell Biol 1984; 98: 894-03.
  • 62 Kristensen P, Eriksen J, Danø K. Localization of urokinase-type plasminogen activator messenger RNA in the normal mouse by in situ hybridization. J Histochem Cytochem 1991; 39: 341-9.
  • 63 Rømer J, Bugge TH, Pyke C. et al. Impaired wound healing in mice with a disrupted plasminogen gene. Nature Med 1996; 2: 287-92.
  • 64 Bugge TH, Kombrinck KW, Flick MJ. et al. Loss of fibrinogen rescues mice from the pleiotropic effects of plasminogen deficiency. Cell 1996; 87: 709-19.
  • 65 Solberg H, Rinkenberger J, Danø K, et al. A functional overlap of plasminogen and MMPs regulates vascularization during placental development. Development 2003; 130: 4439-50.
  • 66 Lund LR, Bjørn SF, Sternlicht MD. et al. Lactational competence and involution of the mouse mammary gland require plasminogen. Development 2000; 127: 4481-92.
  • 67 Oh J, Takahashi R, Adachi E. et al. Mutations in two matrix metalloproteinase genes, MMP-2 and MT1-MMP, are synthetic lethal in mice. Oncogene 2004; 23: 5041-8.
  • 68 Bugge TH, Lund LR, Kombrinck KW. et al. Reduced metastasis of polyoma virus middle T antigeninduced mammary cancer in plasminogen-deficient mice. Oncogene 1998; 16: 3097-04.
  • 69 Coussens LM, Fingleton B, Matrisian LM. Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science 2002; 295: 2387-92.
  • 70 Freije JM, Balbin M, Pendas AM. et al. Matrix metalloproteinases and tumor progression. Adv Exp Med Biol 2003; 532: 91-107.
  • 71 Heissig B, Hattori K, Dias S. et al. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell 2002; 109: 625-37.
  • 72 Arbeit J, Munger K, Howley P. et al. Progressive squamous epithelial neoplasia in K14-human papillomavirus type 16 transgenic mice. J Virol 1994; 68: 358-68.
  • 73 Coussens LM, Tinkle CL, Hanahan D et al. MMP-9 supplied by bone marrow-derived cells contributes to skin carcinogenesis. Cell 2000; 103: 481-90.
  • 74 Yu HR, Schultz RM. Relationship between secreted urokinase plasminogen activator activity and metastatic potential in murine B16 cells transfected with human urokinase sense and antisense genes. Cancer Res 1990; 50: 7623-33.
  • 75 Ossowski L, Reich E. Antibodies to plasminogen activator inhibit human tumor metastasis. Cell 1983; 35: 611-9.
  • 76 Gutierrez LS, Schulman A, Brito-Robinson T et al. Tumor development is retarded in mice lacking the gene for urokinase-type plasminogen activator or its inhibitor, plasminogen activator inhibitor-1. Cancer Res 2000; 60: 5839-47.
  • 77 Frandsen TL, Holst-Hansen C, Nielsen BS. et al. Direct evidence of the importance of stromal urokinase plasminogen activator (uPA) in the growth of an experimental human breast cancer using a combined uPA gene-disrupted and immunodeficient xenograft model [erratum appears in Cancer Res 2002;62:330]. Cancer Res 2001; 61: 532-7.
  • 78 Min HY, Doyle LV, Vitt CR. et al. Urokinase receptor antagonists inhibit angiogenesis and primary tumor growth in syngeneic mice. Cancer Res 1996; 56: 2428-33.
  • 79 Ploug M, Østergaard S, Gårdsvoll H. et al. Peptidederived antagonists of the urokinase receptor. Affinity maturation by combinatorial chemistry, identification of functional epitopes, and inhibitory effect on cancer cell intravasation. Biochemistry 2001; 40: 12157-68.
  • 80 Schuster V, Mingers A, Seidenspinner S. et al. Homozygous mutations in the plasminogen gene of two unrelated girls with ligneous conjunctivitis. Blood 1997; 90: 958-66.
  • 81 Bugge TH, Flick MJ, Daugherty CC. et al. Plasminogen deficiency causes severe thrombosis but is compatible with development and reproduction. Genes Dev 1995; 9: 794-807.
  • 82 Liu S, Schubert RL, Bugge TH. et al. Anthrax toxin: structures, functions and tumour targeting. Expert Opin Biol Ther 2003; 3: 843-53.