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 proteases: successes, failures and future prospects

Key Points

  • Proteases catalyse the hydrolyis of peptide bonds in proteins, often in a very precise way, and are thereby involved in the control of a number of important physiological processes including cell-cycle progression, DNA replication, cell proliferation and cell death, as well as the immune response

  • Protease signalling varies from a simple direct cleavage of a substrate to a complex cascade organization or a protease network, and requires tight regulation

  • Excessive proteolytic activity often leads to disease but can be prevented by blocking the appropriate proteases, which has been explored therapeutically since the 1950s.

  • Angiotensin-converting enzyme (ACE) inhibitors, which were introduced in 1981 for the treatment of various cardiovascular diseases (hypertension, heart failure, heart attack and so on), are still the major blockbusters among protease inhibitors on the market. On the other hand, various broad-spectrum matrix metalloprotease (MMP) inhibitors failed in advanced clinical trials for cancer and rheumatoid arthritis treatment in the 1990s because of various drawbacks.

  • Identification of endogenous protease substrates and other physiological protease ligands is a key issue in understanding protease signalling pathways and an essential part of the identification and validation of protease targets

  • Therapeutic inhibition of validated protease targets can be achieved either by large or small molecules. Large-molecule approaches include protein-type inhibitors that mimick physiological inhibitors and neutralizing antibodies. The development of small-molecule inhibitors, however, is by far the most popular approach. An ideal inhibitor would be a non-covalent, reversible inhibitor with excellent selectivity, good bioavailability and no side effects. The major issues in inhibitor design are still bioavailability and toxicity.

  • The most advanced inhibitors in clinical trials are the renin inhibitors aliskiren (SPP100) for the treatment of hypertension and end-organ damage for which an NDA was filed in 2006, and the dipeptidyl peptidase IV (DPP IV) inhibitors sitagliptin (MK-0431) and vildagliptin (LAF 327) for the treatment of type 2 diabetes, for which NDAs were also filed in 2006. Balicatib (AAE581), the most advanced among the cathepsin K inhibitors for osteoporosis treatment, successfully passed Phase II trials in 2005. Diabetes type 2 and osteoporosis are completely new therapeutic areas, which is encouraging for the future.

  • Proteases, such as kallikrein 3 (prostate-specific antigen) and plasminogen activator, are important diagnostic and prognostic disease markers.

  • The future of protease-based drug discovery efforts probably lies in the cardiovascular, inflammatory, infectious disease, cancer and neurodegeneration areas.

Abstract

Until fairly recently, proteases were considered primarily to be protein-degrading enzymes. However, this view has dramatically changed and proteases are now seen as extremely important signalling molecules that are involved in numerous vital processes. Protease signalling pathways are strictly regulated, and the dysregulation of protease activity can lead to pathologies such as cardiovascular and inflammatory diseases, cancer, osteoporosis and neurological disorders. Several small-molecule drugs targeting proteases are already on the market and many more are in development. The status of human protease research and prospects for future protease-targeted drugs are reviewed here, with reference to some key examples where protease drugs have succeeded or failed.

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: Protease basics.
Figure 2: The renin–angiotensin system and drug discovery.
Figure 3: Regulation of protease activity.
Figure 4: New protease inhibitors in development.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Rawlings, N. D., Tolle, D. P. & Barrett, A. J. MEROPS: the peptidase database. Nucleic Acids Res. 32 (Database issue), D160–D164 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Barrett, A. J., Rawlings, N. D. & Woessner, J. F. Jr (eds) Handbook of Proteolytic Enzymes 2nd edn (Academic, Amsterdam, 2004). An essential book for every protease lab. Contains a lot of information about the majority of known proteases from various organisms.

    Google Scholar 

  4. Davie, E. W. & Neurath, H. Identification of a peptide released during autocatalytic activation of trypsinogen. J. Biol. Chem. 212, 515–529 (1955).

    Article  CAS  PubMed  Google Scholar 

  5. Davie, E. W. & Ratnoff, O. D. Waterfall sequence for intrinsic blood clotting. Science 145, 1310–1312 (1964).

    Article  CAS  PubMed  Google Scholar 

  6. MacFarland, R. G. An enzyme cascade in the blood clotting mechanism, and its function as a biochemical amplifier. Nature 202, 498–499 (1964). References 4–6 are classical works describing protease activation by limited proteolysis.

    Article  Google Scholar 

  7. Lopez-Otin, C. & Overall, C. M. Protease degradomics: a new challenge for proteomics. Nature Rev. Mol. Cell Biol. 3, 509–519 (2002). A valuable collection of different approaches for system-wide analysis of proteases.

    Article  CAS  Google Scholar 

  8. Thornberry, N. A. et al. A novel heterodimeric cysteine protease is required for interleukin-1b processing in monocytes. Nature 356, 768–774 (1992).

    Article  CAS  PubMed  Google Scholar 

  9. Blobel, C. P. ADAMs: key components in EGFR signalling and development. Nature Rev. Mol. Cell Biol. 6, 32–43 (2005).

    Article  CAS  Google Scholar 

  10. Thomas, G. Furin at the cutting edge: from protein traffic to embryiogenesis and disease. Nature Rev. Mol. Cell Biol. 3, 756–766 (2002).

    Article  CAS  Google Scholar 

  11. Weihofen, A. & Martoglio, B. Intramembrane-cleaving proteases:controlled liberation of proteins and bioactive peptides. Trends Cell Biol. 13, 71–78 (2003).

    Article  CAS  PubMed  Google Scholar 

  12. Glickman, M. J. & Ciechanover, A. The ubiquitin-proteasome proteolytic pathway: destruction for the sake of construction. Physiol. Rev. 82, 373–428 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. Davie, E. W., Fujikawa, K. & Kisiel, W. The coagulation cascade: initiation, maintenance, and regulation. Biochemistry 30, 10363–10370 (1991).

    Article  CAS  PubMed  Google Scholar 

  14. Koblinski, J. E., Ahram, M. & Sloane, B. F. Unraveling the role of proteases in cancer. Clin. Chim. Acta 291, 113–135 (2000).

    Article  CAS  PubMed  Google Scholar 

  15. Stennicke, H. S., Ryan, C. A. & Salvesen, G. S. Reprieval from execution: the molecular basis of caspase inhibition. Trends Biochem. Sci. 27, 94–101 (2002).

    Article  CAS  PubMed  Google Scholar 

  16. Thome, M. et al. Viral FLICE-inhibitory proteins (FLIPs) prevent apoptosis induced by death receptors. Nature 386, 517–521 (1997).

    Article  CAS  PubMed  Google Scholar 

  17. Cuconati, A. & White, E. Viral homologs of BCL-2: role of apoptosis in the regulation of virus infection. Genes Dev. 16, 2465–2478 (2002).

    Article  CAS  PubMed  Google Scholar 

  18. Gustafsson, D. et al. A new oral anticoagulant: the 50-year challenge. Nature Rev. Drug Discov. 3, 649–659 (2004).

    Article  CAS  Google Scholar 

  19. Abbenante, G. & Fairlie, D. P. Protease inhibitors in the clinic. Med. Chem. 1, 71–104 (2005). An excellent collection of the current data on protease inhibitors in clinical and preclinical research.

    Article  CAS  PubMed  Google Scholar 

  20. Rueff, J. & Katus, H. A. New antithrombotic drugs on the horizon. Expert Opin. Investig. Drugs 12, 781–797 (2003).

    Article  Google Scholar 

  21. Hirsch, J. Current anticoagulant therapy — unmet clinical needs. Thromb. Res. 109, S1–S8 (2003).

    Article  CAS  Google Scholar 

  22. Zaman, M. A., Oparil, S. & Calhoun, D. A. Drugs targeting the renin–angiotensin–aldosterone system. Nature Rev. Drug Discov. 1, 621–636 (2002).

    Article  CAS  Google Scholar 

  23. Acharya, K. R., Sturrock, E. D., Riordan, J. F. & Ehlers, M. R. ACE revisited: a new target for structure-based drug design. Nature Rev. Drug Discov. 2, 891–902 (2003).

    Article  CAS  Google Scholar 

  24. Natesh, R., Schwager, S. L., Sturrock, E. D. & Acharya, K. R. Crystal structure of the human angiotensin-converting enzyme–lisinopril complex. Nature 421, 551–554 (2003).

    Article  CAS  PubMed  Google Scholar 

  25. van Esch, J. H. et al. Selective angiotensin-converting enzyme C-domain inhibition is sufficient to prevent angiotensin I-induced vasoconstriction. Hypertension 45, 120–125 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Georgiadis, D. et al. Roles of the two active sites of somatic angiotensin-converting enzyme in the cleavage of angiotensin I and bradykinin. Circ. Res. 93, 148–154 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Robl, J. A. et al. Dual metalloprotease inhibitors: mercaptoacetyl-based fused heterocyclic dipeptide mimetics as inhibitors of angiotensin-converting enzyme and neutral endopeptidase. J. Med. Chem. 40, 1570–1577 (1997).

    Article  CAS  PubMed  Google Scholar 

  28. Kostis, J. B. et al. Omapatrilat and enalapril in patients with hypertension: the Omapatrilat Cardiovascular Treatment vs. Enalapril (OCTAVE) trial. Am. J. Hypertens. 17, 103–111 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  30. 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).

    Article  CAS  PubMed  Google Scholar 

  31. Coussens, L. M., Fingleton, B. & Matrisian, L. M. Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science 295, 2387–2392 (2002). An excellent critical analysis of failure of MMP inhibitors in clinical trials.

    Article  CAS  PubMed  Google Scholar 

  32. Neefjes, J. & Dantuma, N. P. Fluorescent probes for proteolysis: tools for drug discovery. Nature Rev. Drug Discov. 3, 58–69 (2004).

    Article  CAS  Google Scholar 

  33. Baruch, A., Jeffery, D. A. & Bogyo, M. Enzyme activity — it's all about image. Trends Cell Biol. 14, 29–35 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Blum, G. et al. Dynamic imaging of protease activity with fluorescently quenched activity based probes. Nature Chem. Biol. 1, 203–209 (2005). A demonstration of in vivo monitoring protease activity in a living mouse, an extremely valuable technique in evaluating the physiological role of a protease in health and disease.

    Article  CAS  Google Scholar 

  35. Page, I. H. & Helmer, O. M. A crystalline pressor substance (angiotonin) resulting from the interaction between renin and renin activator. J. Exp. Med. 71, 29 (1940).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Skeggs, L. T., Kahn, J. R. & Shumway, N. P. The preparation and function of the hypertensin-converting enzyme. J. Exp. Med. 103, 295–299 (1956).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Matthews, D. J. & Wells, J. A. Substrate phage: selection of protease substrates by monovalent phage display. Science 260, 1113–1117 (1993). The first report on the use of phage display for determining substrate specificity of a protease.

    Article  CAS  PubMed  Google Scholar 

  38. Harris, J. L., Peterson, E. P., Hudig, D., Thornberry, N. A. & Craik, C. S. Definition and redesign of the extended substrate specificity of granzyme B. J. Biol. Chem. 273, 27364–27373 (1998).

    Article  CAS  PubMed  Google Scholar 

  39. Harris, J. L. et al. Rapid and general profiling of protease specificity by using combinatorial fluorogenic substrate libraries. Proc. Natl Acad. Sci. USA 97, 7754–7759 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Berger, A. B., Vitorino, P. M. & Bogyo, M. Activity-based protein profiling: applications to biomarker discovery, in vivo imaging and drug discovery. Am. J. Pharmacogenomics 4, 371–381 (2004).

    Article  CAS  PubMed  Google Scholar 

  41. Thornberry, N. A. et al. A combinatorial approach defines specificities of members of the caspase family and granzyme B. J. Biol. Chem. 272, 17907–17911 (1997). The first paper to demonstrate the power of combinatorial chemistry in defining the substrate specificity of a protease.

    Article  CAS  PubMed  Google Scholar 

  42. Turk, B. E., Huang, L. L., Piro, E. T. & Cantley, L. C. Determination of protease cleavage site motifs using mixture-based oriented peptide libraries. Nature Biotechnol. 19, 661–667 (2001).

    Article  CAS  Google Scholar 

  43. Roberds, S. L. et al. BACE knockout mice are healthy despite lacking the primary b-secretase activity in brain: implications for Alzheimer's disease therapeutics. Hum. Mol. Genet. 10, 1317–1324 (2001). An excellent example of target validation using a mouse knockout model.

    Article  CAS  PubMed  Google Scholar 

  44. Gocheva, V. et al. Distinct roles for cysteine cathepsin genes in multistage tumorigenesis. Genes Dev. 20, 543–556 (2006). A good example how knockout mice models can be combined with a disease model in order to identify the appropriate targets.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Fingleton, B. Matrix metalloproteinases as valid clinical targets. Curr. Pharm. Des. (in the press).

  46. Yuan, J., Shaham, S., Ledoux, S., Ellis, H. M. & Horvitz, H. M. The C. elegans cell death gene ced-3 encodes a protein similar to mammalian interleukin-1b-converting enzyme. Cell 75, 641–652 (1993).

    Article  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 (1999).

    Article  Google Scholar 

  48. Tam, E. M., Morrison, C. J., Wu, Y. I., Stack, S. M. & 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bredemeyer, A. J. et al. A proteomic approach for the discovery of protease substrates. Proc. Natl Acad. Sci. USA 101, 11785–11790 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Van Damme, P. et al. Caspase-specific and nonspecific in vivo protein processing during Fas-induced apoptosis. Nature Meth. 2, 771–777 (2005). This paper demonstrates the power of proteomics in determining in vivo protease substrates.

    Article  CAS  Google Scholar 

  51. Medema, J. P. et al. FLICE is activated by association with the CD95 death-inducing signaling complex (DISC). EMBO J. 16, 2794–2804 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Reed, J. C. Apoptosis based therapies. Nature Rev. Drug Discov. 1, 111–121 (2002).

    Article  CAS  Google Scholar 

  53. Eigenbrot, C. & Kirchhofer, D. New insight into how tissue factor allosterically regulates Factor VIIa. Trends Cardiovasc. Med. 12, 19–26 (2002).

    Article  CAS  PubMed  Google Scholar 

  54. Olson, S. T., Björk, I. & Bock, S. C. Identification of critical molecular interactions mediating heparin activation of antithrombin. Trends Cardiovasc. Med. 12, 198–205 (2002).

    Article  CAS  PubMed  Google Scholar 

  55. Bode, W. & Huber, R. Natural protein proteinase inhibitors and their interaction with proteinases. Eur. J. Biochem. 204, 433–451 (1992).

    Article  CAS  PubMed  Google Scholar 

  56. Bode, W. & Huber, R. Structural basis of the endoproteinase–protein inhibitor interaction. Biochim. Biophys. Acta 1477, 241–252 (2000).

    Article  CAS  PubMed  Google Scholar 

  57. Turk, B., Turk, D. & Salvesen, G. S. Regulating cysteine protease activity: essential role of protease inhibitors as guardians and regulators. Curr. Pharm. Des. 8, 1623–1637 (2002).

    Article  CAS  PubMed  Google Scholar 

  58. Giugliano, R. P. et al. Addition of a tissue-factor/factor VIIa inhibitor to standard treatments in NSTE-ACS managed with an early invasive strategy: results of the phase 2 ANTHEM-TIMI 32 double-blind randomized clinical trial. Am. Heart Assoc. Sci. Sessions Dallas, Texas, USA 13–16 Nov (2005).

  59. Vlasuk, G. P. Structural and functional characterization of tick anticoagulant peptide (TAP): a potent and selective inhibitor of blood coagulation Factor Xa. Thromb. Haemost. 70, 212–216 (1993).

    Article  CAS  PubMed  Google Scholar 

  60. Tuszynski, G. P., Gasic, T. B. & Gasic, G. J. Isolation and characterization of antistasin. J. Biol. Chem. 262, 9718–9723 (1987).

    Article  CAS  PubMed  Google Scholar 

  61. Armstrong, W. B. et al. Clinical modulation of oral leukoplakia and protease activity by Bowman–Birk inhibitor concentrate in a phase IIa chemoprevention trial. Clin. Cancer Res. 6, 4684–4691 (2000).

    CAS  PubMed  Google Scholar 

  62. Chen, Y. W., Huang, S. C., Lin-Shiau, S. Y. & Lin, J. K. Bowman–Birk inhibitor abates proteasome function and suppresses the proliferation of MCF7 breast cancer cells through accumulation of MAP kinase phosphatase-1. Carcinogenesis 26, 1296–1306 (2005).

    Article  CAS  PubMed  Google Scholar 

  63. Weimann, L. M. Fully human therapeutic monoclonal antibodies. J. Immunother. 29, 1–9 (2005).

    Google Scholar 

  64. Reuning, U. et al. Urokinase-type plasminogen activator (uPA) and its receptor (uPAR): development of antagonists of uPA/uPAR interaction and their effects in vitro and in vivo. Curr. Pharm. Des. 9, 1529–1543 (2003).

    Article  CAS  PubMed  Google Scholar 

  65. Premzl, A., Zavašnik-Bergant, V., Turk, V. & Kos, J. Intracellular and extracellular cathepsin B facilitate invasion of MCF-10A neoT cells through reconstituted extracellular matrix in vitro. Exp. Cell Res. 283, 206–214 (2003).

    Article  CAS  PubMed  Google Scholar 

  66. Novartis Institute for Tropical Diseases. New technologies for high-throughput screening and lead discovery of anti-viral compounds. Dengue Digest [online], (2005).

  67. McGovern, S. L., Helfand, B. T., Feng, B. & Shoichet, B. K. A specific mechanism of nonspecific inhibition. J. Med. Chem. 46, 4265–4272 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. De Clercq, E. Antiviral drugs in current clinical use. J. Clin. Virol. 30, 115–133 (2004).

    Article  CAS  PubMed  Google Scholar 

  69. Rozman-Pungercˇar, J. et al. Inhibition of papain-like cysteine proteases and legumain by 'caspase-specific' inhibitors: when reaction mechanism is more important than specificity. Cell Death Diff. 10, 881–888 (2003). A good example of the selectivity problems of irreversible protease inhibitors.

    Article  CAS  Google Scholar 

  70. Smyth, T. P. Substrate variants versus transition state analogues as noncovalent, reversible enzyme inhibitors. Bioorg. Med. Chem. 12, 4081–4088 (2004).

    Article  CAS  PubMed  Google Scholar 

  71. Thurmond, R. L. et al. Identification of a potent and selective noncovalent cathepsin S inhibitor. J. Pharmacol. Exp. Ther. 308, 268–276 (2004).

    Article  CAS  PubMed  Google Scholar 

  72. Altmann, E., Green, J. & Tintelnot-Blomley, M. Arylaminoethyl amides as inhibitors of the cysteine protease cathepsin K-investigating P1′ substituents. Bioorg. Med. Chem. Lett. 13, 1997–2001 (2003).

    Article  CAS  PubMed  Google Scholar 

  73. Hardy, J. A. & Wells, J. A. Searching for new allosteric sites in enzymes. Curr. Opin. Struct. Biol. 14, 706–715 (2004).

    Article  CAS  PubMed  Google Scholar 

  74. Hardy, J. A., Lam, J., Nguyen, J. T., O'Brien, T. & Wells, J. A. Discovery of an allosteric site in the caspases. Proc. Natl Acad. Sci. USA 101, 12461–12466 (2004). Excellent work identifying the first small-molecule allosteric protease inhibitor.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Schimmer, A. D. et al. Small-molecule antagonists of apoptosis suppressor XIAP exhibit broad antitumor activity. Cancer Cell 5, 25–35 (2004). The first report demonstrating that potentiating protease activity by inhibiting endogenous protease inhibitors by small molecules can be a useful therapeutic approach.

    Article  CAS  PubMed  Google Scholar 

  76. Bijnens, A. P., Gils, A., Knockaert, I., Stassen, J. M. & Declerck, P. J. Importance of the hinge region between a-helix F and the main part of serpins, based upon identification of the epitope of plasminogen activator inhibitor type 1 neutralizing antibodies. J. Biol. Chem. 275, 6375–6380 (2000).

    Article  CAS  PubMed  Google Scholar 

  77. Rupin, A., Martin, F., Vallez, M. O., Bonhomme, E. & Verbeuren, T. J. Inactivation of plasminogen activator inhibitor-1 accelerates thrombolysis of platelet-rich thrombus in rat mesenteric arterioles. Thromb. Haemost. 86, 1528–1531 (2001).

    Article  CAS  PubMed  Google Scholar 

  78. Stanton, A. Therapeutic potential of renin inhibitors in the management of cardiovascular disorders. Am. J. Cardiovasc. Drugs 3, 389–394 (2003).

    Article  CAS  PubMed  Google Scholar 

  79. Menard, J. et al. Dose-dependent effects of the renin inhibitor zankiren HCl after a single oral dose in mildly sodium-depleted normotensive subjects. Circulation 91, 330–338 (1995).

    Article  CAS  PubMed  Google Scholar 

  80. Nussberger, J., Wuerzner, G., Jensen, C. & Brunner, H. R. Angiotensin II suppression in humans by the orally active renin inhibitor Aliskiren (SPP100): comparison with enalapril. Hypertension 39, E1–E8 (2002).

    Article  CAS  PubMed  Google Scholar 

  81. Wood, J. M., Schnell, C. R., Cumin, F., Menard, J. & Webb, R. L. Aliskiren, a novel, orally effective renin inhibitor, lowers blood pressure in marmosets and sponataneously hypertensive rats. J. Hypertens. 23, 417–426 (2005).

    Article  CAS  PubMed  Google Scholar 

  82. Mervaala, E. et al. Blood pressure-independent effects in rats with human renin and angiotensinogen genes. Hypertension 35, 587–594 (2000).

    Article  CAS  PubMed  Google Scholar 

  83. Mentlein, R., Gallwitz, B. & Schmidt, W. E. Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide glucagon-like peptide-1 (7–36) amide, peptide histidine methionine and is responsible for their degradation in human serum. Eur. J. Biochem. 214, 829–835 (1993).

    Article  CAS  PubMed  Google Scholar 

  84. Kieffer, T. J., McIntosh, C. H. S. & Pederson, R. A. Degradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IV. Endocrinology 136, 3585–3596 (1995).

    Article  CAS  PubMed  Google Scholar 

  85. Marguet, D. et al. Enhanced insulin secretion and improved glucose tolerance in mice lacking CD26. Proc. Natl Acad. Sci. USA 97, 6874–6879 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Nagakura, T. et al. Improved glucose tolerance via enhanced glucose-dependent insulin secretion in dipeptidyl peptidase IV-deficient Fischer rats. Biochem. Biophys. Res. Commun. 284, 501–506 (2001).

    Article  CAS  PubMed  Google Scholar 

  87. Deacon, C. F., Ahren, B. & Holst, J. J. Inhibitors of dipeptidyl peptidase IV: a novel approach for the prevention and treatment of type 2 diabetes? Exp. Opin. Investig. Drugs 13, 1091–1102 (2004).

    Article  CAS  Google Scholar 

  88. Nielsen, L. L. Incretin mimetics and DPP-IV inhibitors for the treatment of type 2 diabetes. Drug Discov. Today 10, 703–710 (2005).

    Article  CAS  PubMed  Google Scholar 

  89. Demuth, H. U., McIntosh, C. H. & Pederson, R. A. Type 2 diabetes--therapy with dipeptidyl peptidase IV inhibitors. Biochim. Biophys. Acta 1751, 33–44 (2005).

    Article  CAS  PubMed  Google Scholar 

  90. Augustyns, K., Van der Veken, P., Senten, K. & Haemers, A. The therapeutic potential of inhibitors of dipeptidyl peptidase IV (DPP IV) and related proline-specific dipeptidyl aminopeptidases. Curr. Med. Chem. 12, 971–998 (2005).

    Article  CAS  PubMed  Google Scholar 

  91. Kim, D. et al. (2R)-4-oxo-4-[3-(trifluoromethyl)-5,6-dihydro[1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]-1-(2,4,5-trifluorophenyl)butan-2-amine: a potent, orally active dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes. J. Med. Chem. 48, 141–151 (2005).

    Article  CAS  PubMed  Google Scholar 

  92. Augeri, D. J. et al. Discovery and preclinical profile of Saxagliptin (BMS-477118): a highly potent, long-acting, orally active dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes. J. Med. Chem. 48, 5025–5037 (2005).

    Article  CAS  PubMed  Google Scholar 

  93. Moser, B., Wolf, M., Walz, A. & Loetscher, P. Chemokines: multiple levels of leukocyte migration control. Trends Immunol. 25, 75–84 (2004).

    Article  CAS  PubMed  Google Scholar 

  94. Yan, S., Marguet, D., Dobers, J., Reutter, W. & Fan, H. Deficiency of CD26 results in a change of cytokine and immunoglobulin secretion after stimulation by pokeweed mitogen. Eur. J. Immunol. 33, 1519–1527 (2003).

    Article  CAS  PubMed  Google Scholar 

  95. Gelb, B. D., Shi, G.-P., Chapman, H. A. & Desnick, R. J. Pycnodysostosis is caused by a deficiency of cathepsin K. Science 273, 1236–1238 (1996).

    Article  CAS  PubMed  Google Scholar 

  96. Saftig, P. et al. Impaired osteoclastic bone resorption leads to osteopetrosis in cathepsin K-deficient mice. Proc. Natl Acad. Sci. USA 95, 13453–13458 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Gowen, M. et al. Cathepsin K knockout mice develop osteopetrosis due to a deficit in matrix degradation but not demineralization. J. Bone Miner. Res. 14, 1654–1663 (1999).

    Article  CAS  PubMed  Google Scholar 

  98. Kiviranta, R. et al. Accelerated turnover of metaphyseal trabecular bone in mice overexpressing cathepsin K. J. Bone Miner. Res. 16, 1444–1452 (2001).

    Article  CAS  PubMed  Google Scholar 

  99. Inaoka, T. et al. Molecular cloning of human cDNA for cathepsin K: novel cysteine proteinase predominantly expressed in bone. Biochem. Biophys. Res. Commun. 206, 89–96 (1995).

    Article  CAS  PubMed  Google Scholar 

  100. Grabowska, U., Chambers, T. J. & Shiroo, M. Recent developments in cathepsin K inhibitor design. Curr. Opin. Drug Discov. Devel. 8, 619–630 (2005).

    CAS  Google Scholar 

  101. Stroup, G. B. et al. Potent and selective inhibition of human cathepsin K leads to inhibition of bone resorption in vivo in a nonhuman primate. J. Bone Miner. Res. 16, 1739–1746 (2001).

    Article  CAS  PubMed  Google Scholar 

  102. Palmer, J. T. et al. Design and synthesis of tri-ring P3 benzamide-containing aminonitriles as potent, selective, orally effective inhibitors of cathepsin K. J. Med. Chem. 48, 7520–7534 (2005).

    Article  CAS  PubMed  Google Scholar 

  103. Turk, D., Guncˇar, G., Podobnik, M. & Turk, B. Revised definition of substrate binding sites of papain-like cysteine proteases. Biol. Chem. 379, 137–147 (1998).

    Article  CAS  PubMed  Google Scholar 

  104. Misbach, M. et al. AAE581, a potent and highly specific cathepsin K inhibitor, prevents bone resorption after oral treatment in rat and monkey. Ann. Mtg Am. Soc. Bone Miner. Res. Nashville, Tennessee, USA 23–27 Sept (2005).

  105. Falgueyret, J. P. et al. Lysosomotropism of basic cathepsin K inhibitors contributes to increased cellular potencies against off-target cathepsins and reduced functional selectivity. J. Med. Chem. 48, 7535–7543 (2005).

    Article  CAS  PubMed  Google Scholar 

  106. Deaton, D. N. & Kumar, S. Cathepsin K inhibitors: their potential as anti-osteoporosis agents. Progr. Med. Chem. 42, 245–375 (2004). Excellent review paper focused on the development of cathepsin K inhibitors, which contains useful general information about medicinal chemistry.

    Article  CAS  Google Scholar 

  107. Tavares, F. X., Deaton, D. N., Miller, A. B., Miller, L. R. & Wright, L. L. Ketoheterocycle-based inhibitors of cathepsin K: a novel entry into the synthesis of peptidic ketoheterocycles. Bioorg. Med. Chem. Lett. 15, 3891–3895 (2005).

    Article  CAS  PubMed  Google Scholar 

  108. Brown, S. et al. Potent and selective mechanism-based inhibition of gelatinases. J. Am. Chem. Soc. 122, 6799–6800 (2000).

    Article  CAS  Google Scholar 

  109. Krüger, A. et al. Antimetastatic activity of a novel mechanism-based gelatinase inhibitor. Cancer Res. 65, 3523–3526 (2005).

    Article  PubMed  Google Scholar 

  110. Gu, Z. et al. A highly specific inhibitor of matrix metalloproteinase-9 rescues laminin from proteolysis and neurons from apoptosis in transient focal cerebral ischemia. J. Neurosci. 25, 6401–6408 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Ikejiri, M. et al. Potent mechanism-based inhibitors for matrix metalloproteinases. J. Biol. Chem. 280, 33992–34002 (2005).

    Article  CAS  PubMed  Google Scholar 

  112. Look, M. P. & Foekens, J. A. Clinical relevance of the urokinase plasminogen activator system in breast cancer. Acta Pathol. Microbiol. Immunol. Scand. 107, 150–159 (1999).

    Article  CAS  Google Scholar 

  113. Dixon, S. C., Knopf, K. B. & Figg, W. D. The control of prostate-specific antigen expression and gene regulation by pharmacological agents. Pharmacol. Rev. 53, 73–91 (2001).

    CAS  PubMed  Google Scholar 

  114. Kos, J. & Lah, T. T. Cysteine proteinases and their endogenous inhibitors: target proteins for prognosis, diagnosis and therapy in cancer. Oncol. Rep. 5, 1349–1361 (1998).

    CAS  PubMed  Google Scholar 

  115. Lai, W. T., Chang, C. H., Tang, Y., Bronson, R. & Tung, C. H. Early diagnosis of osteoarthritis using cathepsin B sensitive near-infrared fluorescent probes. Osteoar. Cartil. 12, 239–244 (2004).

    Article  Google Scholar 

  116. Cazzulo, J. J., Stoka, V. & Turk, V. The major cysteine proteinase of Trypanosoma cruzi: a valid target for chemotherapy of Chagas disease. Curr. Pharm. Des. 7, 1143–1156 (2001).

    Article  CAS  Google Scholar 

  117. Schechter, I. & Berger, A. On the size of the active site of proteinases. I. Papain. Biochem. Biophys. Res. Commun. 27, 157–162 (1967).

    Article  CAS  PubMed  Google Scholar 

  118. Enari, M. et al. A caspase-activated DNase that degrades DNA during apoptosis, and its inhibitor ICAD. Nature 391, 43–50 (1998).

    Article  CAS  PubMed  Google Scholar 

  119. Rawson, R. B. et al. Complementation cloning of S2P, a gene encoding a putative metalloprotease required for intramembrane cleavage of SREBPs. Mol. Cell 1, 47–57 (1997).

    Article  CAS  PubMed  Google Scholar 

  120. Wilkinson, K. D. Ubiquitination and deubiquitination: targeting of proteins for degradation by the proteasome. Semin. Cell. Dev. Biol. 11, 141–148 (2000).

    Article  CAS  PubMed  Google Scholar 

  121. Turk, D. et al. Structure of human dipeptidyl peptidase I (cathepsin C): exclusion domain added to an endopeptidase framework creates the machine for activation of granular serine proteases. EMBO J. 20, 6570–6582 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Silverman, G. A. et al. The serpins are an expanding superfamily of structurally similar but funtionally diverse proteins: evolution, mechanism of inhibition, novel functions, and a revised nomenclature. J. Biol. Chem. 276, 33293–33296 (2001).

    Article  CAS  PubMed  Google Scholar 

  123. Stubbs, M. T. et al. The refined 2.4Å X-ray crystal structure of recombinant human stefin B in complex with the cysteine proteinase papain: a novel type of proteinase inhibitor interaction. EMBO J. 9, 1939–1947 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Gomis-Ruth, F. X. et al. Mechanism of inhibition of the human matrix metalloproteinase stromelysin-1 by TIMP-1. Nature 389, 77–81 (1997).

    Article  CAS  PubMed  Google Scholar 

  125. Shiozaki, E. N. et al. Mechanism of XIAP-mediated inhibition of caspase-9. Mol. Cell 11, 519–527 (2003).

    Article  CAS  PubMed  Google Scholar 

  126. Deveraux, Q., Takahashi, R., Salvesen, G. S. & Reed, J. C. X-linked IAP is a direct inhibitor of cell death proteases. Nature 388, 300–304 (1997).

    Article  CAS  PubMed  Google Scholar 

  127. Turk, B., Turk, D. & Turk, V. Lysosomal cysteine proteases: more than scavengers. Biochim. Biophys. Acta 1477, 98–111 (2000).

    Article  CAS  PubMed  Google Scholar 

  128. Bevec, T., Stoka, V., Pungercic, G., Dolenc, I. & Turk, V. Major histocompatibility complex class II-associated p41 invariant chain fragment is a strong inhibitor of lysosomal cathepsin L. J. Exp. Med. 183, 1331–1338 (1996).

    Article  CAS  PubMed  Google Scholar 

  129. Nature Publishing Group. Signalling scissors: new perspectives on proteases [online], (2004).

Download references

Acknowledgements

Research in the Turk laboratory is supported by grants from Slovene Research Agency. I wish to thank primarily G. Salvesen for numerous discussions, similar way of thinking and permission to use information and figures from his summary of the Horizon Symposium 'Signalling scissors: new perspectives on proteases' (see Further information). Also thanks to H. P. Nestler for providing most of the data for Table 1, G. Guncˇar for help with figure preparation, D. Turk and V. Turk for valuable discussions and critical reading of the manuscript, and D. Deaton, D. Percival, E. Altmann, R. Thurmond, N. Borkakoti, U. Grabowska, B. Gerhartz, V. Dive, and many others for sharing their data with me. I would also like to acknowledge the authors of the other 300,000 papers found in Medline using protease/proteolysis/proteinase as keywords, whose work contributed to our current understanding of proteolysis but could not have been cited here because of space limitations.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The author declares no competing financial interests.

Related links

Related links

DATABASES

OMIM

Alzheimer's disease

Paget's disease

Pycnodysostosis

Rheumatoid arthritis

Type 2 diabetes

FURTHER INFORMATION

MEROPS database

Signalling scissors: new perspectives on proteases

Glossary

Zymogen

A zymogen or pro-enzyme is an inactive enzyme precursor. A zymogen requires a biochemical change (such as a hydrolysis reaction revealing the active site, or changing the conformation to reveal the active site) for it to become an active enzyme.

Autocatalytic

The proteolytic conversion of a zymogen protease molecule into its mature form by molecules of the same kind.

Extracellular matrix degradation

The extracellular matrix are non-celullar components of tissues, and are primarily composed of various glycoproteins, proteoglycans and hyaluronic acid. In various disease states such as cancer and osteoarthritis, ECM is degraded by various proteases, including matrix metalloproteases and the cathepsins.

Positional scanning library

A combinatorial chemistry approach in which individual positions in the molecule are kept defined with the other positions being degenerate to yield 'positional libraries'. These compound mixtures are then tested. The most active moieties of the defined (and therefore known) positions are subsequently combined to yield the screening result, the active molecule.

Mature protease

The catalytically active form of a protease that results from proteolytic processing of its zymogen (inactive pro-enzyme).

Warhead

A reactive functional group, which covalently binds to amino-acid residues of the target enzyme.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Turk, B. Targeting proteases: successes, failures and future prospects. Nat Rev Drug Discov 5, 785–799 (2006). https://doi.org/10.1038/nrd2092

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd2092

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