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.

  • Perspectives
  • Published:

Validating matrix metalloproteinases as drug targets and anti-targets for cancer therapy

Abstract

The matrix metalloproteinases (MMPs) mediate homeostasis of the extracellular environment. They have multiple signalling activities that are commonly altered during tumorigenesis and that might serve as intervention points for anticancer drugs. However, there are many criteria to consider in validating MMPs as drug targets and for the development of MMP inhibitors. The inhibition of some MMPs could have pro-tumorigenic effects (making them anti-targets), counterbalancing the benefits of target inhibition. These effects might partially account for the failure of MMP inhibitors in clinical trials. What are the major challenges in MMP target validation and MMP-inhibitor-drug development?

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

Access options

Buy this article

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

Figure 1: Systems biology of the protease web.
Figure 2: Matrix metalloproteinase signal dynamic changes in the extracellular environment.

Similar content being viewed by others

References

  1. Drews, J. Drug discovery: a historical perspective. Science 287, 1960–1964 (2000).

    CAS  PubMed  Google Scholar 

  2. Lindsay, M. A. Target discovery. Nature Rev. Drug Discov. 2, 831–838 (2003).

    CAS  Google Scholar 

  3. Williams, M. Target validation. Curr. Opin. Pharmacol. 3, 571–577 (2003).

    CAS  PubMed  Google Scholar 

  4. López-Otín, C. & Overall, C. M. Protease degradomics: a new challenge for proteomics. Nature Rev. Mol. Cell Biol. 3, 509–519 (2002).

    Google Scholar 

  5. Puente, X. S., Sanchez, L. M., Overall, C. M. & López-Otín, C. Human and mouse proteases: a comparative genomic approach. Nature Rev. Genet. 4, 544–558 (2003).

    CAS  PubMed  Google Scholar 

  6. Koopec, K. K., Bozyczko-Coyne, D. & Williams, M. Target identification and validation in drug discovery: the role of proteomics. Biochem. Pharmacol. 69, 1133–1139 (2005).

    Google Scholar 

  7. Liotta, L. A. et al. Metastatic potential correlates with enzymatic degradation of basement membrane collagen. Nature 284, 67–68 (1980).

    CAS  PubMed  Google Scholar 

  8. Stetler-Stevenson, W. G. Progelatinase A activation during tumor cell invasion. Invasion Metastasis 14, 259–268 (1994).

    CAS  PubMed  Google Scholar 

  9. Nagase, H. & Woessner, J. F. Matrix metalloproteinases. J. Biol. Chem. 274, 21491–21494 (1999).

    CAS  PubMed  Google Scholar 

  10. Egeblad, M. & Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nature Rev. Cancer 2, 161–174 (2002).

    CAS  Google Scholar 

  11. Minn, A. J. et al. Genes that mediate breast cancer metastasis to lung. Nature 436, 518–524 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Minn, A. J. et al. Distinct organ-specific metastatic potential of individual breast cancer cells and primary tumors. J. Clin. Invest. 115, 44–55 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Radisky, D. C. et al. Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability. Nature 436, 123–127 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Weigelt, B., Peterse, J. L. & van't Veer, L. J. Breast cancer metastasis: markers and models. Nature Rev. Cancer 5, 591–602 (2005).

    CAS  Google Scholar 

  15. McCullagh, K., Wadsworth, H. & Hann, M. Carboxyalkyl peptide derivatives. European Patent EP126,974, 1–111 (1984).

  16. Zucker, S., Cao, J. & Chen, W. T. Critical appraisal of the use of matrix metalloproteinase inhibitors in cancer treatment. Oncogene 19, 6642–6650 (2000).

    CAS  PubMed  Google Scholar 

  17. Overall, C. M. & López-Otín, C. Strategies for MMP inhibition in cancer: innovations for the post-trial era. Nature Rev. Cancer 2, 657–672 (2002).

    CAS  Google Scholar 

  18. Coussens, L. M., Fingleton, B. & Matrisian, L. M. Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science 295, 2387–2392 (2002).

    CAS  PubMed  Google Scholar 

  19. Fingleton, B. Matrix metalloproteinase inhibitors for cancer therapy: the current situation and future prospects. Expert Opin. Ther. Targets 7, 385–397 (2003).

    CAS  PubMed  Google Scholar 

  20. Kumagai, K. et al. Inhibition of matrix metalloproteinases prevents allergen-induced airway inflammation in a murine model of asthma. J. Immunol. 162, 4212–4219 (1999).

    CAS  PubMed  Google Scholar 

  21. McCawley, L. J. & Matrisian, L. M. Matrix metalloproteinases: they're not just for matrix anymore! Curr. Opin. Cell Biol. 13, 534–540 (2001).

    CAS  PubMed  Google Scholar 

  22. Aoki, T., Sumii, T., Mori, T., Wang, X. & Lo, E. H. Blood–brain barrier disruption and matrix metalloproteinase-9 expression during reperfusion injury: mechanical versus embolic focal ischemia in spontaneously hypertensive rats. Stroke 33, 2711–2717 (2002).

    PubMed  Google Scholar 

  23. Overall, C. M. Dilating the degradome: matrix metalloproteinase-2 cuts to the heart of the matter. Biochem. J. 383, E5–E7 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Parks, W. C., Wilson, C. L. & López-Boado, Y. S. Matrix metalloproteinases as modulators of inflammation and innate immunity. Nature Rev. Immunol 4, 617–629 (2004).

    CAS  Google Scholar 

  25. Bergers, G., Javaherian, K., Lo, K. M., Folkman, J. & Hanahan, D. Effects of angiogenesis inhibitors on multistage carcinogenesis in mice. Science 284, 808–812 (1999).

    CAS  PubMed  Google Scholar 

  26. Boehm, T., Folkman, J., Browder, T. & O'Reilly, M. S. Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance. Nature 390, 404–407 (1997).

    CAS  Google Scholar 

  27. McQuibban, G. A. et al. Matrix metalloproteinase activity inactivates the chemokine stromal-cell derived factor-1. J. Biol. Chem. 276, 43503–43508 (2001).

    CAS  PubMed  Google Scholar 

  28. McQuibban, G. A. et al. Matrix metalloproteinase processing of monocyte chemoattractant proteins generates CC chemokine receptor antagonists with anti-inflammatory properties in vivo. Blood 100, 1160–1167 (2002).

    CAS  PubMed  Google Scholar 

  29. Epstein, R. J. The CXCL12–CXCR4 chemotactic pathway as a target of adjuvant breast cancer therapies. Nature Rev. Cancer 4, 901–909 (2004).

    CAS  Google Scholar 

  30. Balkwill, F. Cancer and the chemokine network. Nature Rev. Cancer 4, 540–550 (2004).

    CAS  Google Scholar 

  31. Richardson, P. G. et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. N. Engl. J. Med. 348, 2609–2617 (2003).

    CAS  PubMed  Google Scholar 

  32. Overall, C. M. Molecular determinants of metalloproteinase substrate specificity: matrix metalloproteinase substrate binding domains, modules, and exosites. Mol. Biotechnol. 22, 51–86 (2002).

    CAS  PubMed  Google Scholar 

  33. Topol, E. J. Failing the public health — Rofecoxib, Merck, and the FDA. N. Engl. J. Med. 351, 1707–1709 (2004).

    CAS  PubMed  Google Scholar 

  34. Psaty, B. M. & Furburg, C. D. COX-2 inhibitors — lessons in drug safety. N. Engl. J. Med. 352, 1133–1135 (2005).

    CAS  PubMed  Google Scholar 

  35. Tanne, J. H. Merck faces ongoing claims after Texan ruling on Rofecoxib. Br. Med. J. 331, 471 (2005).

    Google Scholar 

  36. Fidler, I. J. The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nature Rev. Cancer 3, 453–458 (2003).

    CAS  Google Scholar 

  37. Sodek, J. & Overall, C. M. in The Biological Mechanisms of Tooth Eruption and Root Resorption (ed. Davidovitch, Z.) 303–311 (EBSCO Media, Birmingham, 1988).

  38. Engelholm, L. H. et al. uPARAP/Endo180 is essential for cellular uptake of collagen and promotes fibroblast collagen adhesion. J. Cell Biol. 160, 1009–1015 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Curino, A. C. et al. Intracellular collagen degradation mediated by uPARAP/Endo180 is a major pathway of extracellular matrix turnover during malignancy. J. Cell Biol. 169, 977–985 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Schenk, S. & Quaranta, V. Tales from the crypt[ic] sites of the extracellular matrix. Trends Cell Biol. 13, 366–375 (2003).

    CAS  PubMed  Google Scholar 

  41. Pirila, E. et al. Matrix metalloproteinases process the laminin-5 γ2-chain and regulate epithelial cell migration. Biochem. Biophys. Res. Commun. 303, 1012–1017 (2003).

    CAS  PubMed  Google Scholar 

  42. Handsley, M. M. & Edwards, D. R. Metalloproteinases and their inhibitors in tumor angiogenesis. Int. J. Cancer 115, 849–860 (2005).

    CAS  PubMed  Google Scholar 

  43. O'Reilly, M. S., Holmgren, L., Chen, C. & Folkman, J. Angiostatin induces and sustains dormancy of human primary tumors in mice. Nature Med. 2, 689–692 (1996).

    CAS  Google Scholar 

  44. Tam, E. M., Morrison, C. J., Wu, Y. I., Stack, M. S. & Overall, C. M. Membrane protease proteomics: isotope-coded affinity tag MS identification of undescribed MT1-matrix metalloproteinase substrates. Proc. Natl Acad. Sci. USA 101, 6917–6922 (2004).

    CAS  PubMed  Google Scholar 

  45. Fingleton, B., Vargo-Gogola, T., Crawford, H. C. & Matrisian, L. M. Matrilysin (MMP-7) expression selects for cells with reduced sensitivity to apoptosis. Neoplasia 3, 459–468 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Balbin, M. et al. Loss of collagenase-2 confers increased skin tumor susceptibility to male mice. Nature Genet. 35, 252–257 (2003).

    CAS  PubMed  Google Scholar 

  47. McQuibban, G. A. et al. Inflammation dampened by gelatinase A cleavage of monocyte chemoattractant protein-3. Science 289, 1202–1206 (2000).

    CAS  PubMed  Google Scholar 

  48. Fowlkes, J. L., Thrailkill, K. M., Serra, D. M., Suzuki, K. & Nagase, H. Matrix metalloproteinases as insulin-like growth factor binding protein-degrading proteinases. Prog. Growth Factor Res. 6, 255–263 (1995).

    CAS  PubMed  Google Scholar 

  49. Hemers, E. et al. Insulin-like growth factor binding protein-5 is a target of matrix metalloproteinase-7: implications for epithelial-mesenchymal signaling. Cancer Res. 65, 7363–7369 (2005).

    CAS  PubMed  Google Scholar 

  50. Davies, N. M. & Jamali, F. COX-2 selective inhibitors cardiac toxicity: getting to the heart of the matter. J. Pharm. Pharm. Sci. 7, 332–336 (2004).

    CAS  PubMed  Google Scholar 

  51. Khokha, R. et al. Antisense RNA-induced reduction in murine TIMP levels confers oncogenicity on Swiss 3T3 cells. Science 243, 947–950 (1989).

    CAS  PubMed  Google Scholar 

  52. Hanahan, D. & Folkman, J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86, 353–364 (1996).

    CAS  PubMed  Google Scholar 

  53. Arbeit, J. M., Munger, K., Howley, P. M. & Hanahan, D. Progressive squamous epithelial neoplasia in K14-human papillomavirus type 16 transgenic mice. J. Virol. 68, 4358–4368 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Ben-Baruch, A. Host microenvironment in breast cancer development: inflammatory cells, cytokines and chemokines in breast cancer progression: reciprocal tumor–microenvironment interactions. Breast Cancer Res. 5, 31–36 (2003).

    CAS  PubMed  Google Scholar 

  56. Ben-Baruch, A. Inflammation-associated immune suppression in cancer: the roles played by cytokines, chemokines and additional mediators. Semin. Cancer Biol. 16, 38–52 (2006).

    CAS  PubMed  Google Scholar 

  57. Brummelkamp, T. R., Bernards, R. & Agami, R. A system for stable expression of short interfering RNAs in mammalian cell. Science 296, 550–553 (2002).

    CAS  PubMed  Google Scholar 

  58. Bramhall, S. R. et al. Marimastat as maintenance therapy for patients with advanced gastric cancer: a randomised trial. Br. J. Cancer 86, 1864–1870 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Bramhall, S. R. et al. A double-blind placebo-controlled, randomised study comparing gemcitabine and marimastat with gemcitabine and placebo as first line therapy in patients with advanced pancreatic cancer. Br. J. Cancer 87, 161–167 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Groves, M. D. et al. Phase II trial of temozolomide plus the matrix metalloproteinase inhibitor, marimastat, in recurrent and progressive glioblastoma multiforme. J. Clin. Oncol. 20, 1383–1388 (2002).

    CAS  PubMed  Google Scholar 

  61. King, J., Zhao, J., Clingan, P. & Morris, D. Randomised double blind placebo control study of adjuvant treatment with the metalloproteinase inhibitor, Marimastat in patients with inoperable colorectal hepatic metastases: significant survival advantage in patients with musculoskeletal side-effects. Anticancer Res. 23, 639–645 (2003).

    CAS  PubMed  Google Scholar 

  62. van't Veer, L. J. et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 415, 530–536 (2002).

    CAS  Google Scholar 

  63. Murray, G. I., Duncan, M. E., O'Neil, P., Melvin, W. T. & Fothergill, J. E. Matrix metalloproteinase-1 is associated with poor prognosis in colorectal cancer. Nature Med. 2, 461–462 (1996).

    CAS  PubMed  Google Scholar 

  64. Itoh, T. et al. Reduced angiogenesis and tumor progression in gelatinase A-deficient mice. Cancer Res. 58, 1048–1051 (1998).

    CAS  PubMed  Google Scholar 

  65. Kruger, A. et al. Antimetastatic activity of a novel mechanism-based gelatinase inhibitor. Cancer Res. 65, 3523–356 (2005).

    PubMed  Google Scholar 

  66. Tester, A. M. et al. Pro-matrix metalloproteinase-2 transfection increases orthotopic primary growth and experimental metastasis of MDA-MB-231 human breast cancer cells in nude mice. Cancer Res. 64, 652–658 (2004).

    CAS  PubMed  Google Scholar 

  67. Bergers, G. et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nature Cell Biol. 2, 737–744 (2000).

    CAS  PubMed  Google Scholar 

  68. Davidson, B. et al. Altered expression of metastasis-associated and regulatory molecules in effusions from breast cancer patients: a novel model for tumor progression. Clin. Cancer Res. 10, 7335–7346 (2004).

    CAS  PubMed  Google Scholar 

  69. Wilson, C. L., Heppner, K. J., Labosky, P. A., Hogan, B. L. & Matrisian, L. M. Intestinal tumorigenesis is suppressed in mice lacking the metalloproteinase matrilysin. Proc. Natl Acad. Sci. USA 94, 1402–1407 (1997).

    CAS  PubMed  Google Scholar 

  70. Crawford, H. C., Scoggins, C. R., Washington, M. K. & Matrisian, L. M. Matrix metalloproteinase-7 is expressed by pancreatic cancer precursors and regulates acinar-to-ductal metaplasia in exocrine pancrease. J. Clin. Invest. 109, 1437–1344 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Wagenaar-Miller, R. A. et al. Cooperative effects of matrix metalloproteinase and cyclooxygenase-2 inhibition on intestinal adenoma reduction. Br. J. Cancer 88, 1445–1452 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Montel, V. et al. Altered metastatic behavior of human breast cancer cells after experimental manipulation of matrix metalloproteinase 8 gene expression. Cancer Res. 64, 1687–1694 (2004).

    CAS  PubMed  Google Scholar 

  73. Coussens, L. M., Tinkle, C. L., Hanahan, D. & Werb, Z. MMP-9 supplied by bone marrow-derived cells contributes to skin carcinogenesis. Cell 103, 481–490 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Witty, J. P., Lempka, T., Coffey, R. J. Jr & Matrisian, L. M. Decreased tumor formation in 7,12-dimethylbenzanthracene-treated stromelysin-1 transgenic mice is associated with alterations in mammary epithelial cell apoptosis. Cancer Res. 55, 1401–1406 (1995).

    CAS  PubMed  Google Scholar 

  75. McCawley, L. J., Crawford, H. C., King, L. E. Jr, Mudgett, J. & Matrisian, L. M. A protective role for matrix metalloproteinase-3 in squamous cell carcinoma. Cancer Res. 64, 6965–6972 (2004).

    CAS  PubMed  Google Scholar 

  76. Gorrin Rivas, M. J. et al. Expression of human macrophage metalloelastase gene in hepatocellular carcinoma: correlation with angiostatin generation and its clinical significance. Hepatology 28, 986–993 (1998).

    CAS  PubMed  Google Scholar 

  77. Yang, W. et al. Human macrophage metalloelastase gene expression in colorectal carcinoma and its clinicopathologic significance. Cancer 91, 1277–1283 (2001).

    CAS  PubMed  Google Scholar 

  78. Overall, C. M. & Sodek, J. Concanavalin A produces a matrix-degradative phenotype in human fibroblasts. Induction and endogenous activation of collagenase, 72-kDa gelatinase, and Pump-1 is accompanied by the suppression of the tissue inhibitor of matrix metalloproteinases. J. Biol. Chem. 265, 21141–21151 (1990).

    CAS  PubMed  Google Scholar 

  79. Holmbeck, K. et al. MT1-MMP-deficient mice develop dwarfism, osteopenia, arthritis, and connective tissue disease due to inadequate collagen turnover. Cell 99, 81–92 (1999).

    CAS  PubMed  Google Scholar 

  80. Tam, E. M., Moore, T. R., Butler, G. S. & Overall, C. M. Characterization of the distinct collagen binding, helicase and cleavage mechanisms of matrix metalloproteinase 2 and 14 (gelatinase A and MT1-MMP): the differential roles of the MMP hemopexin C domains and the MMP-2 fibronectin type II modules in collagen triple helicase activities. J. Biol. Chem. 279, 43336–4344 (2004).

    CAS  PubMed  Google Scholar 

  81. Overall, C. M. et al. Protease degradomics: mass spectrometry discovery of protease substrates and the CLIP-CHIP, a dedicated DNA microarray of all human proteases and inhibitors. Biol. Chem. 385, 493–504 (2004).

    CAS  PubMed  Google Scholar 

  82. Kang, Y. et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3, 537–549 (2003).

    CAS  PubMed  Google Scholar 

  83. Muller, A. et al. Involvement of chemokine receptors in breast cancer metastasis. Nature 410, 50–56 (2001).

    CAS  Google Scholar 

  84. Nevo, I. et al. The tumor microenvironment: CXCR4 is associated with distinct protein expression patterns in neuroblastoma cells. Immunol. Lett. 92, 163–169 (2004).

    CAS  PubMed  Google Scholar 

  85. Zhang, K. et al. Metalloproteinase cleavage of the chemokine SDF-1α induces neuronal apoptosis in HIV encephalitis. Nature Neurosci. 6, 1064–1071 (2003).

    CAS  PubMed  Google Scholar 

  86. Neumark, E., Sagi-Assif, O., Shalmon, B., Ben-Baruch, A. & Witz, I. P. Progression of mouse mammary tumors: MCP-1-TNFα cross-regulatory pathway and clonal expression of promalignancy and antimalignancy factors. Int. J. Cancer 106, 879–886 (2003).

    CAS  PubMed  Google Scholar 

  87. Basset, P. et al. A novel metalloproteinase gene specifically expressed in stromal cells of breast carcinomas. Nature 348, 699–704 (1990).

    CAS  PubMed  Google Scholar 

  88. Boulay, A. et al. High cancer cell death in syngeneic tumors developed in host mice deficient for the stromelysin-3 matrix metalloproteinase. Cancer Res. 61, 2189–2193 (2001).

    CAS  PubMed  Google Scholar 

  89. Andarawewa, K. L. et al. Stromelysin-3 is a potent negative regulator of adipogenesis participating to cancer cell–adipocyte interaction/crosstalk at the tumor invasive front. Cancer Res. 65, 10862–10871 (2005).

    CAS  PubMed  Google Scholar 

  90. Deng, H., Guo, R. F., Li, W. M., Zhao, M. & Lu, Y. Y. Matrix metalloproteinase 11 depletion inhibits cell proliferation in gastric cancer cells. Biochem. Biophys. Res. Commun. 326, 274–281 (2005).

    CAS  PubMed  Google Scholar 

  91. Pendas, A. M. et al. Diet-induced obesity and reduced skin cancer susceptibility in matrix metalloproteinase 19-deficient mice. Mol. Cell Biol. 24, 5304–5313 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Kang, T. et al. Subcellular distribution and cytokine- and chemokine-regulated secretion of leukolysin/MT6-MMP/MMP-25 in neutrophils. J. Biol. Chem. 276, 21960–21968 (2001).

    CAS  PubMed  Google Scholar 

  93. Gururajan, R., Grenet, J., Lahti, J. M. & Kidd, V. J. Isolation and characterization of two novel metalloproteinase genes linked to the Cdc2L locus on human chromosome 1p36.3. Genomics 52, 101–106 (1998).

    CAS  PubMed  Google Scholar 

  94. Porter, S. et al. Dysregulated expression of adamalysin-thrombospondin genes in human breast carcinoma. Clin. Cancer Res. 10, 2429–2440 (2004).

    CAS  PubMed  Google Scholar 

  95. Riddick, A. C. et al. Identification of degradome components associated with prostate cancer progression by expression analysis of human prostatic tissues. Br. J. Cancer 92, 2171–2180 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Greenbaum, D., Medzihradszky, K. F., Burlingame, A. & Bogyo, M. Epoxide electrophiles as activity-dependent cysteine protease profiling and discovery tools. Chem. Biol. 7, 569–581 (2000).

    CAS  PubMed  Google Scholar 

  97. Abersold, R. & Mann, M. Mass spectrometry-based proteomics. Nature 422, 198–207 (2003).

    Google Scholar 

  98. De Leenheer, A. P. & Thienpont, L. M. Application of isotope dilution-mass spectrometry in clinical chemistry, pharmacokinetics, and toxicology. Mass Spectrom. Rev. 11, 249–307 (1992).

    CAS  Google Scholar 

  99. Gygi, S. P. et al. Quantitative analysis of complex protein mixtures using isotope-coded affinity tags. Nature Biotechnol. 17, 994–999 (1999).

    CAS  Google Scholar 

  100. Saghatelian, A., Jessani, N., Joseph, A., Humphrey, M. & Cravatt, B. F. Activity-based probes for the proteomic profiling of metalloproteases. Proc. Natl Acad. Sci. USA 101, 10000–10005 (2004).

    CAS  PubMed  Google Scholar 

  101. Puerta, D. T. & Cohen, S. M. Examination of novel zinc-binding groups for use in matrix metalloproteinase inhibitors. Inorg. Chem. 42, 3423–3430 (2003).

    CAS  PubMed  Google Scholar 

  102. Matziari, M., Beau, F., Cuniasse, P., Dive, V. & Yiotakis, A. Evaluation of P1′-diversified phosphinic peptides leads to the development of highly selective inhibitors of MMP-11. J. Med. Chem. 47, 325–336 (2004).

    CAS  PubMed  Google Scholar 

  103. Cuniasse, P. et al. Future challenges facing the development of specific active-site-directed synthetic inhibitors of MMPs. Biochimie 87, 393–402 (2005).

    CAS  PubMed  Google Scholar 

  104. Bertini, I. et al. Conformational variability of matrix metalloproteinases: beyond a single 3D structure. Proc. Natl Acad. Sci. USA 102, 5334–5339 (2005).

    CAS  PubMed  Google Scholar 

  105. Moy, F. J. et al. Impact of mobility on structure-based drug design for the MMPs. J. Am. Chem. Soc. 124, 12658–12659 (2002).

    CAS  PubMed  Google Scholar 

  106. Morales, R. et al. Crystal structures of novel non-peptidic, non-zinc chelating inhibitors bound to MMP-12. J. Mol. Biol. 341, 1063–1076 (2004).

    CAS  PubMed  Google Scholar 

  107. Dublanchet, A. C. et al. Structure-based design and synthesis of novel non-zinc chelating MMP-12 inhibitors. Bioorg. Med. Chem. Lett. 15, 3787–3790 (2005).

    CAS  PubMed  Google Scholar 

  108. Lukacova, V. et al. Similarity of binding sites of human matrix metalloproteinases. J. Biol. Chem. 279, 14194–14200 (2004).

    CAS  PubMed  Google Scholar 

  109. Butler, G. S. & Overall, C. M. Proteomic validation of protease drug targets. Pharmacoproteomics of matrix metalloproteinase inhibitor drugs using isotope-coded affinity tag labelling and tandem mass spectrometry. Curr. Pharm. Des. (in the press).

  110. Overall, C. M. & Kleifeld, O. Towards third generation matrix metalloproteinase inhibitors for cancer therapy. Br. J. Cancer (in the press).

Download references

Acknowledgements

This work was supported by grants from the National Cancer Institute of Canada with funds raised by the Canadian Cancer Association, the Canadian Breast Cancer Research Alliance special programme grant on metastasis, and with a Centre Grant from the Michael Smith Research Foundation. We gratefully acknowledgement the useful discussions of these concepts with Joerg Eder, Novartis Institutes of Biomedical Research, Basel, Switzerland.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Christopher M. Overall.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

National Cancer Institute

breast cancer

FURTHER INFORMATION

Cancer degradome

Christopher Overall's laboratory, web page

Human, mouse and rat degradomes

MEROPS peptidase database

The CLIP-CHIP

The International Proteolysis Society

Peptidase nomenclature web site of the NC-IUBMB

Rights and permissions

Reprints and permissions

About this article

Cite this article

Overall, C., Kleifeld, O. Validating matrix metalloproteinases as drug targets and anti-targets for cancer therapy. Nat Rev Cancer 6, 227–239 (2006). https://doi.org/10.1038/nrc1821

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc1821

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