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:

A decade of fragment-based drug design: strategic advances and lessons learned

Key Points

  • Fragment-based drug design is based on screening smaller numbers of compounds (typically several thousand) in the hopes of finding low-affinity fragments (Kd values in the high micromolar to millimolar range), in contrast to conventional high-throughput screening (HTS), which attempts to evaluate as many compounds as technologically possible (typically a million or more) in the hopes of finding relatively potent drug leads (Kd values ideally less than 1 μM).

  • The combination of broader sampling of the potential chemical universe than HTS and increased hit rates for molecules of low complexity makes fragment-based screening a powerful tool for lead generation. Fragment-based screening is also less prone to artefacts as the low-molecular-mass compounds tend to be more soluble and the methods of detection are simpler and more robust.

  • Two-dimensional, isotope-edited nuclear magnetic resonance (NMR) spectroscopy was the first approach used in fragment-based drug design. It is well suited to this purpose as NMR chemical shifts are exquisitely sensitive to ligand binding, and problems with compound interference can be solved by spectral editing.

  • During the past decade, the popularity of fragment-based screening has grown at a remarkable rate in both industry and academia. A range of different strategies have been developed, including alternative NMR-based approaches that obviate the need for isotope labelling, approaches based on X-ray-crystallography and fragment tethering, which are discussed here.

  • The ability to obtain NMR or X-ray crystal structures on fragment leads has a dramatic influence on the success of fragment-based drug design.

  • The successful applications of fragment-based drug design have provided ample support that the use of fragments could, in many cases, be the most direct route to the best achievable balance between potency and pharmacokinetics.

Abstract

Since the early 1990s, several technological and scientific advances — such as combinatorial chemistry, high-throughput screening and the sequencing of the human genome — have been heralded as remedies to the problems facing the pharmaceutical industry. The use of these technologies in some form is now well established at most pharmaceutical companies; however, the return on investment in terms of marketed products has not met expectations. Fragment-based drug design is another tool for drug discovery that has emerged in the past decade. Here, we describe the development and evolution of fragment-based drug design, analyse the role that this approach can have in combination with other discovery technologies and highlight the impact that fragment-based methods have made in progressing new medicines into the clinic.

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: Applications of the 'SAR by NMR' method for fragment-based design.
Figure 2: Trends in the application of fragment-based screening.
Figure 3: Percentage of protein-binding sites that can be targeted with small, drug-like molecules as a function of hit rate from NMR-based fragment screening.
Figure 4: Comparing high-throughput screening and fragment-based screening.
Figure 5: Importance of structural information to the success of fragment-based drug design.
Figure 6: Relationship between potency and molecular mass during fragment optimization.

Similar content being viewed by others

References

  1. Butcher, E. C. Can cell systems biology rescue drug discovery? Nature Rev. Drug Discov. 4, 461–467 (2005).

    Article  CAS  Google Scholar 

  2. van der Greef, J. & McBurney, R. N. Innovation: Rescuing drug discovery: in vivo systems pathology and systems pharmacology. Nature Rev. Drug Discov. 4, 961–967 (2005).

    Article  CAS  Google Scholar 

  3. Hardy, L. W. & Peet, N. P. The multiple orthogonal tools approach to define molecular causation in the validation of druggable targets. Drug Discov. Today 9, 117–126 (2004).

    Article  CAS  Google Scholar 

  4. Betz, U. A., Farquhar, R. & Ziegelbauer, K. Genomics: success or failure to deliver drug targets? Curr. Opin. Chem. Biol. 9, 387–391 (2005).

    Article  CAS  Google Scholar 

  5. Zambrowicz, B. P. & Sands, A. T. Knockouts model the 100 best-selling drugs — will they model the next 100? Nature Rev. Drug Discov. 2, 38–51 (2003).

    Article  CAS  Google Scholar 

  6. Macarron, R. Critical review of the role of HTS in drug discovery. Drug Discov. Today 11, 277–279 (2006). An important analysis of the current and future impact of high-throughput screening on the drug discovery process.

    Article  Google Scholar 

  7. Silverman, L., Campbell, R. & Broach, J. R. New assay technologies for high-throughput screening. Curr. Op. Chem. Biol. 2, 397–403 (1998).

    Article  CAS  Google Scholar 

  8. Oprea, T. I. & Matter, H. Integrating virtual screening in lead discovery. Curr. Opin. Chem. Biol. 8, 349–358 (2004).

    Article  CAS  Google Scholar 

  9. Shoichet, B. K. Virtual screening of chemical libraries. Nature 432, 862–865 (2004).

    Article  CAS  Google Scholar 

  10. Scapin, G. Structural biology and drug discovery. Curr. Pharm. Des. 12, 2087–2097 (2006).

    Article  CAS  Google Scholar 

  11. Brown, D. & Superti-Furga, G. Rediscovering the sweet spot in drug discovery. Drug Discov. Today 8, 1067–1077 (2003).

    Article  Google Scholar 

  12. Bohm, H. J. Site-directed structure generation by fragment-joining. Persp. Drug. Disc. Design 3, 21–33 (1995).

    Article  Google Scholar 

  13. Miranker, A. & Karplus, M. Functionality maps of binding sites: a multiple copy simultaneous search method. Proteins 11, 29–34 (1991).

    Article  CAS  Google Scholar 

  14. Hajduk, P. J., Meadows, R. P. & Fesik, S. W. NMR-based screening in drug discovery. Q. Rev. Biophys. 32, 211–240 (1999).

    Article  CAS  Google Scholar 

  15. Shuker, S. B., Hajduk, P. J., Meadows, R. P. & Fesik, S. W. Discovering high-affinity ligands for proteins: SAR by NMR. Science 274, 1531–1534 (1996). A seminal paper describing the SAR by NMR method.

    Article  CAS  Google Scholar 

  16. Hajduk, P. J. et al. Discovery of potent nonpeptide inhibitors of stromelysin using SAR by NMR. J. Am. Chem. Soc. 119, 5818–5827 (1997). Application of the SAR by NMR concept to matrix metalloproteinases, ultimately resulting in the first compound designed using a fragment-based approach to reach the clinic (see reference 18 below).

    Article  CAS  Google Scholar 

  17. Beckett, R. P., Davidson, A. H., Drummond, A. H., Huxley, P. & Whittaker, M. Recent advances in matrix metalloproteinase inhibitor research. Drug. Disc. Today 1, 16–26 (1996).

    Article  CAS  Google Scholar 

  18. Wada, C. K. et al. Phenoxyphenyl sulfone N-formylhydroxylamines (Retorhydroxamates) as potent, selective, orally bioavailable matrix metalloproteinase inhibitors. J. Med. Chem. 45, 219–232 (2002).

    Article  CAS  Google Scholar 

  19. Oltersdorf, T. et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 435, 677–681 (2005). The most recent example of the SAR by NMR approach applied to a protein–protein interaction target, ultimately resulting in a compound that is currently in Phase I clinical trials for the treatment of cancer.

    Article  CAS  Google Scholar 

  20. Kirkin, V., Joos, S. & Zornig, M. The role of Bcl-2 family members in tumorigenesis. Biochim. Biophys. Acta 1644, 229–249 (2004).

    Article  CAS  Google Scholar 

  21. Petros, A. M. et al. Discovery of a potent inhibitor of the antiapoptotic protein Bcl-xL from NMR and parallel synthesis. J. Med. Chem. 49, 656–663 (2006).

    Article  CAS  Google Scholar 

  22. Lepre, C. A., Moore, J. M. & Peng, J. W. Theory and applications of NMR-based screening in pharmaceutical research. Chem. Rev. 104, 3641–3676 (2004).

    Article  CAS  Google Scholar 

  23. Meyer, B. & Peters, T. NMR spectroscopy techniques for screening and identifying ligand binding to protein receptors. Angew. Chem. Int. Ed. Engl. 42, 864–890 (2003). An excellent overview of the numerous applications of NMR in fragment-based screening.

    Article  CAS  Google Scholar 

  24. Dalvit, C. et al. A general NMR method for rapid, efficient, and reliable biochemical screening. J. Am. Chem. Soc. 125, 14620–14625 (2003).

    Article  CAS  Google Scholar 

  25. London, R. E. Theoretical analysis of the inter-ligand overhauser effect: a new approach for mapping structural relationships of macromolecular ligands. J. Magn. Reson. 141, 301–311 (1999).

    Article  CAS  Google Scholar 

  26. Becattini, B. et al. Structure-activity relationships by interligand NOE-based design and synthesis of antiapoptotic compounds targeting Bid. Proc. Natl Acad. Sci. USA 103, 12602–12606 (2006).

    Article  CAS  Google Scholar 

  27. Lin, M., Shapiro, M. J. & Wareing, J. R. Diffusion-edited NMR-affinity NMR for direct observation of molecular interactions. J. Am. Chem. Soc. 119, 5249–5250 (1997).

    Article  CAS  Google Scholar 

  28. Fejzo, J. et al. The SHAPES strategy: An NMR-based approach for lead generation in drug discovery. Chem. Biol. 6, 755–769 (1999).

    Article  CAS  Google Scholar 

  29. Stockman, B. J. NMR spectroscopy as a tool for structure-based drug design. Prog. Nucl. Magn. Reson. Spectrosc. 33, 109–151 (1998).

    Article  CAS  Google Scholar 

  30. Dalvit, C. et al. NMR-based screening with competition water-ligand observed via gradient spectroscopy experiments: detection of high-affinity ligands. J. Med. Chem. 45, 2610–2614 (2002).

    Article  CAS  Google Scholar 

  31. Nienaber, V. L. et al. Discovering novel ligands for macromolecules using X-ray crystallographic screening. Nature Biotechnol. 18, 1105–1108 (2000).

    Article  CAS  Google Scholar 

  32. Carr, R. A., Congreve, M., Murray, C. W. & Rees, D. C. Fragment-based lead discovery: leads by design. Drug Discov. Today 10, 987–992 (2005). A concise description of the theory, advantages and applications of fragment-based drug design.

    Article  CAS  Google Scholar 

  33. Erlanson, D. A., Wells, J. A. & Braisted, A. C. Tethering: fragment-based drug discovery. Annu. Rev. Biophys. Biomol. Struct. 33, 199–223 (2004).

    Article  CAS  Google Scholar 

  34. Martin, Y. C. Challenges and prospects for computational aids to molecular diversity. Perspect. Drug Discov. Des. 7–8, 159–172 (1997).

    Google Scholar 

  35. Jacoby, E. et al. Key aspects of the Novartis compound collection enhancement project for the compilation of a comprehensive chemogenomics drug discovery screening collection. Curr. Top. Med. Chem. 5, 397–411 (2005).

    Article  CAS  Google Scholar 

  36. Davis, A. M., Keeling, D. J., Steele, J., Tomkinson, N. P. & Tinker, A. C. Components of successful lead generation. Curr. Top. Med. Chem. 5, 421–439 (2005).

    Article  CAS  Google Scholar 

  37. Fink, T., Bruggesser, H. & Reymond, J. L. Virtual exploration of the small-molecule chemical universe below 160 Daltons. Angew. Chem. Int. Ed. Engl. 44, 1504–1508 (2005).

    Article  CAS  Google Scholar 

  38. Hann, M. M., Leach, A. R. & Harper, G. Molecular complexity and its impact on the probability of finding leads for drug discovery. J. Chem. Inf. Comput. Sci. 41, 856–864 (2001). The authors provide a sound theoretical basis for the success of fragment-based screening based on molecular complexity and basic principles of molecular recognition.

    Article  CAS  Google Scholar 

  39. Schuffenhauer, A. et al. Library design for fragment based screening. Curr. Top. Med. Chem. 5, 751–762 (2005).

    Article  CAS  Google Scholar 

  40. Hajduk, P. J., Huth, J. R. & Fesik, S. W. Druggability indices for protein targets derived from NMR-based screening data. J. Med. Chem. 48, 2518–2525 (2005). An analysis that establishes the utility of using fragment-based screening not only for lead identification, but also for characterizing the druggability of protein targets with small-molecule ligands. A computational analysis of protein druggability is also described.

    Article  CAS  Google Scholar 

  41. McGovern, S. L., Caselli, E., Grigorieff, N. & Shoichet, B. K. A common mechanism underlying promiscuous inhibitors from virtual and high-throughput screening. J. Med. Chem. 45, 1712–1722 (2002).

    Article  CAS  Google Scholar 

  42. Huth, J. R. et al. ALARM NMR: a rapid and robust experimental method to detect reactive false positives in biochemical screens. J. Am. Chem. Soc. 127, 217–224 (2005).

    Article  CAS  Google Scholar 

  43. Hajduk, P. J. & Burns, D. J. Integration of NMR and high-throughput screening. Comb. Chem. High Throughput Screen. 5, 613–621 (2002).

    Article  CAS  Google Scholar 

  44. Qian, J. et al. Discovery of novel inhibitors of Bcl-xL using multiple high-throughput screening platforms. Anal. Biochem. 328, 131–138 (2004).

    Article  CAS  Google Scholar 

  45. Abad-Zapatero, C. & Metz, J. T. Ligand efficiency indices as guideposts for drug discovery. Drug Discov. Today 10, 464–469 (2005).

    Article  Google Scholar 

  46. Hopkins, A. L., Groom, C. R. & Alex, A. Ligand efficiency: a useful metric for lead selection. Drug Discov. Today 9, 430–431 (2004). This work (together with reference 45) provides some benchmarks for evaluating the usefulness of drug leads based on relationships between potency and mass.

    Article  Google Scholar 

  47. Oprea, T. I., Davis, A. M., Teague, S. J. & Leeson, P. D. Is there a difference between leads and drugs? A historical perspective. J. Chem. Inf. Comput. Sci. 41, 1308–1315 (2001). An important analysis of the process of lead optimization, in which quantitative measures of the changes in mass, hydrophobicity and other physicochemical properties that occur during synthetic optimization are given.

    Article  CAS  Google Scholar 

  48. Lipinski, C. A. Drug-like properties and the causes of poor solubility and poor permeability. J. Pharmacol. Toxicol. Methods 44, 235–249 (2000). A landmark paper correlating oral bioavailability with various physicochemical properties such as mass and hydrophobicity. The guidelines described in this paper are now used throughout the pharmaceutical industry.

    Article  CAS  Google Scholar 

  49. Hajduk, P. J. Fragment-based drug design: how big is too big? J. Med. Chem. 49, 6972–6976 (2006). A retrospective analysis of the process of fragment optimization, in which it is demonstrated that the ultimate ligand efficiency of an optimized inhibitor is dictated by the fragment core, thereby enabling a quantitative evaluation of lead selection and optimization.

    Article  CAS  Google Scholar 

  50. Lipinski, C., Lombardo, F., Dominy, B. & Feeney, P. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Delivery Rev. 23, 3–25 (1997).

    Article  CAS  Google Scholar 

  51. Wunberg, T. et al. Improving the hit-to-lead process: data-driven assessment of drug-like and lead-like screening hits. Drug Discov. Today 11, 175–180 (2006).

    Article  CAS  Google Scholar 

  52. Wolfson, W. Fragmentary solutions Astex Therapeutics puts the pieces together. Chem. Biol. 13, 799–801 (2006).

    Article  CAS  Google Scholar 

  53. Barker, J., Courtney, S., Hesterkamp, T., Ullmann, D. & Whittaker, M. Fragment screening by biochemical assay. Expert Opin. Drug Discov. 1, 225–236 (2006).

    Article  CAS  Google Scholar 

  54. Vanwetswinkel, S. et al. TINS, target immobilized NMR screening: an efficient and sensitive method for ligand discovery. Chem. Biol. 12, 207–216 (2005).

    Article  CAS  Google Scholar 

  55. Lehn, J. M. & Eliseev, A. V. Dynamic combinatorial chemistry. Science 291, 2331–2332 (2001).

    Article  CAS  Google Scholar 

  56. Otto, S., Furlan, R. L. & Sanders, J. K. Recent developments in dynamic combinatorial chemistry. Curr. Opin. Chem. Biol. 6, 321–327 (2002).

    Article  CAS  Google Scholar 

  57. Vajda, S. & Guarnieri, F. Characterization of protein–ligand interaction sites using experimental and computational methods. Curr. Opin. Drug Discov. Devel. 9, 354–362 (2006).

    CAS  PubMed  Google Scholar 

  58. Hajduk, P. J. et al. Design of adenosine kinase inhibitors from the NMR-based screening of fragments. J. Med. Chem. 43, 4781–4786 (2000).

    Article  CAS  Google Scholar 

  59. Liu, G. et al. Novel p-arylthio cinnamides as antagonists of leukocyte function-associated antigen-1/ intracellular adhesion molecule-1 interaction. 2. Mechanism of inhibition and structure-based improvement of pharmaceutical properties. J. Med. Chem. 44, 1202–1210 (2001).

    Article  CAS  Google Scholar 

  60. Szczepankiewicz, B. G. et al. Discovery of a potent, selective protein tyrosine phosphatase 1B inhibitor using a linked-fragment strategy. J. Am. Chem. Soc. 125, 4087–4096 (2003).

    Article  CAS  Google Scholar 

  61. Sanders, W. J. et al. Discovery of potent inhibitors of dihydroneopterin aldolase using CrystaLEAD high-throughput X-ray crystallographic screening and structure-directed lead optimization. J. Med. Chem. 47, 1709–1718 (2004).

    Article  CAS  Google Scholar 

  62. Astex Therapeutics. Clincal candidates: AT13387. Astex Therapeutics web site[online].

  63. Gill, A. L. et al. Identification of novel p38a MAP kinase inhibitors using fragment-based lead generation. J. Med. Chem. 48, 414–426 (2005).

    Article  CAS  Google Scholar 

  64. Lange, G. et al. Requirements for specific binding of low affinity inhibitor fragments to the SH2 domain of (pp60)Src are identical to those for high affinity binding of full length inhibitors. J. Med. Chem. 46, 5184–5195 (2003).

    Article  CAS  Google Scholar 

  65. Lesuisse, D. et al. SAR and X-ray. A new approach combining fragment-based screening and rational drug design: application to the discovery of nanomolar inhibitors of Src SH2. J. Med. Chem. 45, 2379–2387 (2002).

    Article  CAS  Google Scholar 

  66. Forino, M. et al. Efficient synthetic inhibitors of anthrax lethal factor. Proc. Natl Acad. Sci. USA 102, 9499–9504 (2005).

    Article  CAS  Google Scholar 

  67. Card, G. L. et al. A family of phosphodiesterase inhibitors discovered by cocrystallography and scaffold-based drug design. Nature Biotech. 23, 201–207 (2005).

    Article  CAS  Google Scholar 

  68. Oblak, M. et al. In silico fragment-based discovery of indolin-2-one analogues as potent DNA gyrase inhibitors. Bioorg. Med. Chem. Lett. 15, 5207–5210 (2005).

    Article  CAS  Google Scholar 

  69. Blaney, J., Nienaber, V. L. & Burley, S. in Fragment-Based Approaches in Drug Discovery (eds Jahnke, W. & Erlanson, D. A.) 215–248 (Wiley–VCH, Weinheim, Germany, 2006).

    Book  Google Scholar 

  70. Warner, S. L. et al. Identification of a lead small-molecule inhibitor of the Aurora kinases using a structure-assisted, fragment-based approach. Mol. Cancer Ther. 5, 1764–1773 (2006).

    Article  CAS  Google Scholar 

  71. Zhu, Z. et al. Heterocyclic aspartyl protease inhibitors. US Patent 20060111370 (2006).

  72. Chan, T-Y. et al. Kinase inhibitors. European Patent WO2006081230 (2006).

  73. Sunesis. Pipeline programs: SNS-314. Sunesis web site[online].

  74. Braisted, A. C. et al. Discovery of a potent small molecule IL-2 inhibitor through fragment assembly. J. Am. Chem. Soc. 125, 3714–3715 (2003).

    Article  CAS  Google Scholar 

  75. Choong, I. C. et al. Identification of potent and selective small-molecule inhibitors of caspase-3 through the use of extended tethering and structure-based drug design. J. Med. Chem. 45, 5005–5022 (2002).

    Article  CAS  Google Scholar 

  76. O'Brien, T. et al. Structural analysis of caspase-1 inhib-itors derived from Tethering. Acta Crystallograph. Sect. F Struct. Biol. Cryst. Commun. 61, 451–458 (2005).

    Article  CAS  Google Scholar 

  77. Sem, D. in Fragment-Based Approaches in Drug Discovery (eds Jahnke, W. & Erlanson, D. A.) 149–177 (Wiley–VCH, Weinheim, Germany, 2006). One of a series of chapters in a book dedicated to the theory, implementation and application of fragment-based drug design.

    Google Scholar 

  78. Moore, J. et al. Leveraging structural approaches: applications of NMR-based screening and X-ray crystallography for inhibitor design. J. Synchrotron Radiat. 11, 97–100 (2004).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to thank R. Artis (Plexxikon), M. Congreve (Astex), D. Erlanson (Sunesis), R. Hubbard (Vernalis), W. Jahnke (Novartis), C. Lepre (Vertex), and D. Wyss (Schering–Plough) for help in gathering the data for Table 1.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Philip J. Hajduk.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Glossary

Forward and reverse genetics

Forward genetics approaches involve proceeding from phenotype to genotype by positional cloning or candidate-gene analysis. Reverse genetics approaches involve proceeding from genotype to phenotype through gene-manipulation techniques.

New chemical entity

A medication containing an active ingredient that has not been previously approved for marketing in any form.

Kd values

The equilibrium dissociation constant of a compound that reflects the concentration needed to reach half-maximal saturation of binding sites. Kd reflects the strength of binding of a compound to its specific binding site.

Pharmacophore

The ensemble of steric and electronic features that is necessary to ensure optimal interactions with a specific biological target structure and to trigger (or to block) its biological response.

Two-dimensional, isotope-edited nuclear magnetic resonance (NMR) spectroscopy

NMR experiments that exploit nuclear coupling to correlate the chemical shifts of protons with other NMR-active nuclei, most often carbon-13 or nitrogen-15.

Structure–activity relationships

Correlations that are constructed between the features of chemical structure in a set of candidate compounds and parameters of biological activity, such as potency, selectivity and toxicity.

IC50 value

The half maximal inhibitory concentration. Represents the concentration of an inhibitor that is required for 50% inhibition of a biological or molecular process.

Druggability

The ability of a target to be modulated by a lead candidate that has the requisite physicochemical and absorption, distribution, metabolism and excretion properties for development as a drug candidate.

Rule-of-five

The 'rule of five' identifies several key properties that should be considered for compounds with oral delivery in mind. These properties are molecular mass <500 Da, cLogP <5, number of hydrogen-bond donors <5 and number of hydrogen-bond acceptors <10.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hajduk, P., Greer, J. A decade of fragment-based drug design: strategic advances and lessons learned. Nat Rev Drug Discov 6, 211–219 (2007). https://doi.org/10.1038/nrd2220

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd2220

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