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:

Small molecules, big targets: drug discovery faces the protein–protein interaction challenge

Key Points

  • Protein–protein interactions (PPIs) are increasingly being targeted by drug discovery groups, and there exists great scope for therapeutic modulation of this target class in disease.

  • The array of structurally interacting elements through which proteins interact with one another is wide and resists clear-cut classification. However, broad divisions can be made by grouping interactions based upon the globular or peptidic nature of the proteins.

  • Some strategies for developing inhibitors against a given PPI may have more traction against certain classes of PPIs than others; for example, fragment-based drug discovery has shown particular promise in targeting bromodomains, as have peptide mimetics in mimicking β-strands.

  • We examine case studies representative of the various structural types of PPI and discuss the lessons learnt from each.

  • A summary of current status of inhibitors in clinical trials against different targets is presented.

Abstract

Protein–protein interactions (PPIs) are of pivotal importance in the regulation of biological systems and are consequently implicated in the development of disease states. Recent work has begun to show that, with the right tools, certain classes of PPI can yield to the efforts of medicinal chemists to develop inhibitors, and the first PPI inhibitors have reached clinical development. In this Review, we describe the research leading to these breakthroughs and highlight the existence of groups of structurally related PPIs within the PPI target class. For each of these groups, we use examples of successful discovery efforts to illustrate the research strategies that have proved most useful.

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: Structural classification of protein–protein interactions.
Figure 2: Strategies for developing peptidomimetics.
Figure 3: Interaction between a globular protein and a helical peptide with a discontinuous epitope: BCL-2–BH3 domain.
Figure 4: Interaction between a globular protein and a peptide with a continuous epitope: XIAP–caspase 9.
Figure 5: Interaction between a globular protein and a peptide with a continuous epitope: KEAP1–NRF2.
Figure 6: Interaction between a globular protein and globular protein through a discontinuous epitope: IL-2–IL-2R.
Figure 7: Interaction between a globular protein and a peptide with an anchoring residue: BRD4.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

  1. 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).

    CAS  PubMed  Google Scholar 

  2. Huggins, D. J., Marsh, M. & Payne, M. C. Thermodynamic properties of water molecules at a protein–protein interaction surface. J. Chem. Theory Comput. 7, 3514–3522 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Clackson, T. & Wells, J. A. A hot spot of binding energy in a hormone–receptor interface. Science 267, 383–386 (1995). A seminal paper presenting the crystal structure of human growth hormone and a domain of its receptor and systematically mutating interface residues to Ala.

    CAS  PubMed  Google Scholar 

  4. Bogan, A. A. & Thorn, K. S. Anatomy of hot spots in protein interfaces. J. Mol. Biol. 280, 1–9 (1998). This paper examines the residue composition of hot spots at PPI interfaces, demonstrating an enrichment for certain amino acids. The importance of solvent occlusion around hot spot residues is also discussed.

    CAS  PubMed  Google Scholar 

  5. Smith, M. C. & Gestwicki, J. E. Features of protein–protein interactions that translate into potent inhibitors: topology, surface area and affinity. Expert Rev. Mol. Med. 14, e16 (2012).

    PubMed  PubMed Central  Google Scholar 

  6. Chakrabarti, P. & Janin, J. Dissecting protein–protein recognition sites. Proteins 47, 334–343 (2002).

    CAS  PubMed  Google Scholar 

  7. Keskin, O., Ma, B. & Nussinov, R. Hot regions in protein–protein interactions: the organization and contribution of structurally conserved hot spot residues. J. Mol. Biol. 345, 1281–1294 (2005).

    CAS  PubMed  Google Scholar 

  8. Ma, B., Elkayam, T., Wolfson, H. & Nussinov, R. Protein–protein interactions: structurally conserved residues distinguish between binding sites and exposed protein surfaces. Proc. Natl Acad. Sci. USA 100, 5772–5777 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Davis, F. P. & Sali, A. The overlap of small molecule and protein binding sites within families of protein structures. PLoS Comput. Biol. 6, e1000668 (2010).

    PubMed  PubMed Central  Google Scholar 

  10. Hu, Z., Ma, B., Wolfson, H. & Nussinov, R. Conservation of polar residues as hot spots at protein interfaces. Proteins 39, 331–342 (2000).

    CAS  PubMed  Google Scholar 

  11. Sheinerman, F. B., Norel, R. & Honig, B. Electrostatic aspects of protein–protein interactions. Curr. Opin. Struct. Biol. 10, 153–159 (2000).

    CAS  PubMed  Google Scholar 

  12. Rao, V. S., Srinivas, K., Sujini, G. N. & Kumar, G. N. Protein–protein interaction detection: methods and analysis. Int. J. Proteom. 2014, 147648 (2014).

    Google Scholar 

  13. Lehner, B. & Fraser, A. G. A first-draft human protein-interaction map. Genome Biol. 5, R63 (2004).

    PubMed  PubMed Central  Google Scholar 

  14. Zhang, Q. C. et al. Structure-based prediction of protein–protein interactions on a genome-wide scale. Nature 490, 556–560 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Winter, A. et al. Biophysical and computational fragment-based approaches to targeting protein–protein interactions: applications in structure-guided drug discovery. Q. Rev. Biophys. 45, 383–426 (2012).

    CAS  PubMed  Google Scholar 

  16. Szklarczyk, D. et al. The STRING database in 2011: functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res. 39, D561–D568 (2011).

    CAS  PubMed  Google Scholar 

  17. Higueruelo, A. P. et al. Atomic interactions and profile of small molecules disrupting protein–protein interfaces: the TIMBAL database. Chem. Biol. Drug Des. 74, 457–467 (2009).

    CAS  PubMed  Google Scholar 

  18. Basse, M. J. et al. 2P2Idb: a structural database dedicated to orthosteric modulation of protein–protein interactions. Nucleic Acids Res. 41, D824–D827 (2013).

    CAS  PubMed  Google Scholar 

  19. Morelli, X., Bourgeas, R. & Roche, P. Chemical and structural lessons from recent successes in protein–protein interaction inhibition (2P2I). Curr. Opin. Chem. Biol. 15, 475–481 (2011).

    CAS  PubMed  Google Scholar 

  20. Bickerton, G. R., Higueruelo, A. P. & Blundell, T. L. Comprehensive, atomic-level characterization of structurally characterized protein–protein interactions: the PICCOLO database. BMC Bioinformat. 12, 313 (2011).

    Google Scholar 

  21. Jones, S. & Thornton, J. M. Principles of protein–protein interactions. Proc. Natl Acad. Sci. USA 93, 13–20 (1996). A classic paper reviewing PPIs, discussing concepts such as homo- and hetero-dimers as well as obligate and non-obligate complexes.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Miller, S. The structure of interfaces between subunits of dimeric and tetrameric proteins. Protein Eng. 3, 77–83 (1989).

    CAS  PubMed  Google Scholar 

  23. Larsen, T. A., Olson, A. J. & Goodsell, D. S. Morphology of protein–protein interfaces. Structure 6, 421–427 (1998).

    CAS  PubMed  Google Scholar 

  24. Keskin, O., Gursoy, A., Ma, B. & Nussinov, R. Principles of protein–protein interactions: what are the preferred ways for proteins to interact? Chem. Rev. 108, 1225–1244 (2008).

    CAS  PubMed  Google Scholar 

  25. Arkin, M. R., Tang, Y. & Wells, J. A. Small-molecule inhibitors of protein–protein interactions: progressing toward the reality. Chem. Biol. 21, 1102–1114 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Blundell, T. L. et al. Structural biology and bioinformatics in drug design: opportunities and challenges for target identification and lead discovery. Phil. Trans. R. Soc. B 361, 413–423 (2006). This paper introduces the idea that protein structure can aid target selection, in particular the benefits of targeting a globular protein that orders a flexible peptide upon binding are proposed.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Wendt, M. D. Protein–protein interactions as drug targets. Top. Med. Chem. 8, 1–56 (2012).

    CAS  Google Scholar 

  28. Rickert, M., Wang, X., Boulanger, M. J., Goriatcheva, N. & Garcia, K. C. The structure of interleukin-2 complexed with its alpha receptor. Science 308, 1477–1480 (2005). This paper shows the crystal structure of IL-2 in complex with IL-2Rα, revealing the complex nature of the globular protein–globular protein interaction.

    CAS  PubMed  Google Scholar 

  29. Jubb, H., Blundell, T. L. & Ascher, D. B. Flexibility and small pockets at protein–protein interfaces: new insights into druggability. Prog. Biophys. Mol. Biol. 119, 2–9 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Fletcher, S. & Prochownik, E. V. Small-molecule inhibitors of the Myc oncoprotein. Biochim. Biophys. Acta 1849, 525–543 (2015).

    CAS  PubMed  Google Scholar 

  31. Baell, J. B. & Holloway, G. A. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J. Med. Chem. 53, 2719–2740 (2010).

    CAS  PubMed  Google Scholar 

  32. Follis, A. V., Hammoudeh, D. I., Wang, H., Prochownik, E. V. & Metallo, S. J. Structural rationale for the coupled binding and unfolding of the c-Myc oncoprotein by small molecules. Chem. Biol. 15, 1149–1155 (2008).

    CAS  PubMed  Google Scholar 

  33. Hammoudeh, D. I., Follis, A. V., Prochownik, E. V. & Metallo, S. J. Multiple independent binding sites for small-molecule inhibitors on the oncoprotein c-Myc. J. Am. Chem. Soc. 131, 7390–7401 (2009).

    CAS  PubMed  Google Scholar 

  34. Pellegrini, L. et al. Insights into DNA recombination from the structure of a RAD51–BRCA2 complex. Nature 420, 287–293 (2002). The only published structure of human RAD51 published to date, revealing an interaction with a peptide derived from BRCA2.

    CAS  PubMed  Google Scholar 

  35. Scott, D. E. et al. Small-molecule inhibitors that target protein–protein interactions in the RAD51 family of recombinases. ChemMedChem 10, 296–303 (2014).

    PubMed  PubMed Central  Google Scholar 

  36. Christ, F. et al. Rational design of small-molecule inhibitors of the LEDGF/p75-integrase interaction and HIV replication. Nat. Chem. Biol. 6, 442–448 (2010). A successful rational design of inhibitors binding to LEDGF (also known as p75) at a site bound by an inter-helix loop of viral HIV-1 integrase.

    CAS  PubMed  Google Scholar 

  37. Fuller, J. C., Burgoyne, N. J. & Jackson, R. M. Predicting druggable binding sites at the protein–protein interface. Drug Discov. Today 14, 155–161 (2009).

    CAS  PubMed  Google Scholar 

  38. Jochim, A. L. & Arora, P. S. Systematic analysis of helical protein interfaces reveals targets for synthetic inhibitors. ACS Chem. Biol. 5, 919–923 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Wells, J. A. & McClendon, C. L. Reaching for high-hanging fruit in drug discovery at protein–protein interfaces. Nature 450, 1001–1009 (2007).

    CAS  PubMed  Google Scholar 

  40. Vassilev, L. T. et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 303, 844–848 (2004).

    CAS  PubMed  Google Scholar 

  41. Grasberger, B. L. et al. Discovery and cocrystal structure of benzodiazepinedione HDM2 antagonists that activate p53 in cells. J. Med. Chem. 48, 909–912 (2005).

    CAS  PubMed  Google Scholar 

  42. Allen, J. G. et al. Discovery and optimization of chromenotriazolopyrimidines as potent inhibitors of the mouse double minute 2-tumor protein 53 protein–protein interaction. J. Med. Chem. 52, 7044–7053 (2009).

    CAS  PubMed  Google Scholar 

  43. Blackburn, T. J. et al. Diaryl- and triaryl-pyrrole derivatives: inhibitors of the MDM2-p53 and MDMX-p53 protein–protein interactions. MedChemComm 4, 1297–1304 (2013).

    CAS  PubMed  Google Scholar 

  44. Kenny, C. H. et al. Development of a fluorescence polarization assay to screen for inhibitors of the FtsZ/ZipA interaction. Anal. Biochem. 323, 224–233 (2003).

    CAS  PubMed  Google Scholar 

  45. White, P. W. et al. Inhibition of human papillomavirus DNA replication by small molecule antagonists of the E1–E2 protein interaction. J. Biol. Chem. 278, 26765–26772 (2003).

    CAS  PubMed  Google Scholar 

  46. Ferrari, S., Pellati, F. & Costi, M. P. in Disruption of Protein–Protein Interfaces (ed. Mangani, S.) 31–60 (Springer Berlin Heidelberg, 2013).

    Google Scholar 

  47. Hajduk, P. J. & Greer, J. A decade of fragment-based drug design: strategic advances and lessons learned. Nat. Rev. Drug. Discov. 6, 211–219 (2007). An excellent review of fragment-based drug discovery with valuable historical insight.

    CAS  PubMed  Google Scholar 

  48. Whittaker, M. Picking up the pieces with FBDD or FADD: invest early for future success. Drug Discov. Today 14, 623–624 (2009).

    PubMed  Google Scholar 

  49. Blundell, T. L., Jhoti, H. & Abell, C. High-throughput crystallography for lead discovery in drug design. Nat.Rev. Drug. Discov. 1, 45–54 (2002).

    CAS  PubMed  Google Scholar 

  50. Coyne, A. G., Scott, D. E. & Abell, C. Drugging challenging targets using fragment-based approaches. Curr. Opin. Chem. Biol. 14, 299–307 (2010).

    CAS  PubMed  Google Scholar 

  51. Turnbull, A., Boyd, S. & Walse, B. Fragment-based drug discovery and protein–protein interactions. Res. Rep. Biochem. 4, 13–26 (2014).

    CAS  Google Scholar 

  52. Ciulli, A., Williams, G., Smith, A. G., Blundell, T. L. & Abell, C. Probing hot spots at protein-ligand binding sites: a fragment-based approach using biophysical methods. J. Med. Chem. 49, 4992–5000 (2006).

    CAS  PubMed  Google Scholar 

  53. Scott, D. E. et al. Using a fragment-based approach to target protein–protein interactions. ChemBioChem 14, 332–342 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Lo, M. C. et al. Evaluation of fluorescence-based thermal shift assays for hit identification in drug discovery. Anal. Biochem. 332, 153–159 (2004).

    CAS  PubMed  Google Scholar 

  55. Navratilova, I. & Hopkins, A. L. Emerging role of surface plasmon resonance in fragment-based drug discovery. Future. Med. Chem. 3, 1809–1820 (2011).

    CAS  PubMed  Google Scholar 

  56. Lepre, C. A., Connolly, P. J. & Moore, J. M. in Drug Design (eds Merz, K. M. Jr., Ringe, D. & Reynolds, C. H.) 41–58 (Cambridge Univ. Press, 2010).

    Google Scholar 

  57. Davies, T. G. & Tickle, I. J. Fragment screening using X-ray crystallography. Top. Curr. Chem. 317, 33–59 (2012).

    CAS  PubMed  Google Scholar 

  58. Turnbull, W. B. & Daranas, A. H. On the value of c: can low affinity systems be studied by isothermal titration calorimetry? J. Am. Chem. Soc. 125, 14859–14866 (2003).

    CAS  PubMed  Google Scholar 

  59. Erlanson, D. A. et al. Site-directed ligand discovery. Proc. Natl Acad. Sci. USA 97, 9367–9372 (2000). The first report of covalent 'tethering' as a technique for hit discovery and lead development against challenging drug discovery targets.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Morley, A. D. et al. Fragment-based hit identification: thinking in 3D. Drug Discov. Today 18, 1221–1227 (2013).

    PubMed  Google Scholar 

  61. Van Molle, I. et al. Dissecting fragment-based lead discovery at the von Hippel-Lindau protein:hypoxia inducible factor 1α protein–protein interface. Chem. Biol. 19, 1300–1312 (2012).

    CAS  PubMed  Google Scholar 

  62. Jhoti, H., Williams, G., Rees, D. C. & Murray, C. W. The 'rule of three' for fragment-based drug discovery: where are we now? Nat. Rev. Drug. Discov. 12, 644–645 (2013).

    CAS  PubMed  Google Scholar 

  63. Fry, D. C. et al. Deconstruction of a nutlin: dissecting the binding determinants of a potent protein–protein interaction inhibitor. ACS Med. Chem. Lett. 4, 660–665 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Barelier, S., Pons, J., Marcillat, O., Lancelin, J.-M. & Krimm, I. Fragment-based deconstruction of Bcl-xL inhibitors. J. Med. Chem. 53, 2577–2588 (2010).

    CAS  PubMed  Google Scholar 

  65. Peat, T. S. et al. Small molecule inhibitors of the LEDGF site of human immunodeficiency virus integrase identified by fragment screening and structure based design. PLoS ONE 7, e40147 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Ichihara, O., Barker, J., Law, R. J. & Whittaker, M. Compound design by fragment-linking. Mol. Inform. 30, 298–306 (2011).

    CAS  PubMed  Google Scholar 

  67. de Vega, M. J., Martin-Martinez, M. & Gonzalez-Muniz, R. Modulation of protein–protein interactions by stabilizing/mimicking protein secondary structure elements. Curr. Top. Med. Chem. 7, 33–62 (2007).

    PubMed  Google Scholar 

  68. Craik, D. J., Fairlie, D. P., Liras, S. & Price, D. The future of peptide-based drugs. Chem. Biol. Drug Des. 81, 136–147 (2013).

    CAS  PubMed  Google Scholar 

  69. Henchey, L. K., Jochim, A. L. & Arora, P. S. Contemporary strategies for the stabilization of peptides in the α-helical conformation. Curr. Opin. Chem. Biol. 12, 692–697 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Walensky, L. D. & Bird, G. H. Hydrocarbon-stapled peptides: principles, practice, and progress. J. Med. Chem. 57, 6275–6288 (2014). A review of the concept of 'stapled peptides' — hydrocarbon chains holding peptides into a fixed conformation.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Chu, Q. et al. Towards understanding cell penetration by stapled peptides. Med. Chem. Commun. 6, 111–119 (2014).

    Google Scholar 

  72. Kritzer, J. A. Stapled peptides: magic bullets in nature's arsenal. Nat. Chem. Biol. 6, 566–567 (2010).

    CAS  PubMed  Google Scholar 

  73. Liu, J., Wang, D., Zheng, Q., Lu, M. & Arora, P. S. Atomic structure of a short α-helix stabilized by a main chain hydrogen-bond surrogate. J. Am. Chem. Soc. 130, 4334–4337 (2008).

    CAS  PubMed  Google Scholar 

  74. Seebach, D. & Gardiner, J. β-peptidic peptidomimetics. Acc. Chem. Res. 41, 1366–1375 (2008).

    CAS  PubMed  Google Scholar 

  75. Sadowsky, J. D. et al. Chimeric (α/β + α)-peptide ligands for the BH3-recognition cleft of Bcl-XL: critical role of the molecular scaffold in protein surface recognition. J. Am. Chem. Soc. 127, 11966–11968 (2005). An investigation of both α- and β-amino acid peptides, and hybrids of both, as inhibitors of the BH3–BCL-X L interaction.

    CAS  PubMed  Google Scholar 

  76. Fletcher, S. & Hamilton, A. D. Protein surface recognition and proteomimetics: mimics of protein surface structure and function. Curr. Opin. Chem. Biol. 9, 632–638 (2005).

    CAS  PubMed  Google Scholar 

  77. Chen, L. et al. p53 α-helix mimetics antagonize p53/MDM2 interaction and activate p53. Mol. Cancer Ther. 4, 1019–1025 (2005).

    CAS  PubMed  Google Scholar 

  78. Davis, J. M., Truong, A. & Hamilton, A. D. Synthesis of a 2,3';6',3''-terpyridine scaffold as an α-helix mimetic. Org. Lett. 7, 5405–5408 (2005).

    CAS  PubMed  Google Scholar 

  79. Shaginian, A. et al. Design, synthesis, and evaluation of an α-helix mimetic library targeting protein–protein interactions. J. Am. Chem. Soc. 131, 5564–5572 (2009). In this study a 400-member triaryl amide library, designed as α-helix mimetics, was screened against the MDM2–p53 PPI.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Bayly, A. R., White, A. J. P. & Spivey, A. C. Design and synthesis of a prototype scaffold for five-residue α-helix mimetics. Eur. J. Org. Chem. 25, 5566–5569 (2013).

    Google Scholar 

  81. Wei, C. Q., Li, B., Guo, R., Yang, D. & Burke, T. R. Jr. Development of a phosphatase-stable phosphotyrosyl mimetic suitably protected for the synthesis of high-affinity Grb2 SH2 domain-binding ligands. Bioorg. Med. Chem. Lett. 12, 2781–2784 (2002).

    CAS  PubMed  Google Scholar 

  82. Wei, C. Q. et al. Macrocyclization in the design of Grb2 SH2 domain-binding ligands exhibiting high potency in whole-cell systems. J. Med. Chem. 46, 244–254 (2003).

    CAS  PubMed  Google Scholar 

  83. Loughlin, W. A., Tyndall, J. D. A., Glenn, M. P., Hill, T. A. & Fairlie, D. P. Beta-strand mimetics. Chem. Rev. 110, 32–69 (2010).

    Google Scholar 

  84. Rognan, D. Rational design of protein–protein interaction inhibitors. Med. Chem. Commun. 6, 51–60 (2015).

    CAS  Google Scholar 

  85. Sugaya, N. & Furuya, T. Dr. PIAS: an integrative system for assessing the druggability of protein–protein interactions. BMC Bioinformatics 12, 50 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Walter, P., Metzger, J., Thiel, C. & Helms, V. Predicting where small molecules bind at protein–protein interfaces. PLoS ONE 8, e58583 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Meireles, L. M., Domling, A. S. & Camacho, C. J. ANCHOR: a web server and database for analysis of protein–protein interaction binding pockets for drug discovery. Nucleic Acids Res. 38, W407–W411 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Bienstock, R. J. Computational drug design targeting protein–protein interactions. Curr. Pharm. Des. 18, 1240–1254 (2012).

    CAS  PubMed  Google Scholar 

  89. Grimme, D., González-ruiz, D. & Gohlke, H. in Physico-Chemical and Computational Approaches to Drug Discovery (eds Luque, J. & Barril, X.) 319–359 (2012).

    Google Scholar 

  90. Falchi, F., Caporuscio, F. & Recanatini, M. Structure-based design of small-molecule protein–protein interaction modulators: the story so far. Future Med. Chem. 6, 343–357 (2014).

    CAS  PubMed  Google Scholar 

  91. Johnson, D. K. & Karanicolas, J. Druggable protein interaction sites are more predisposed to surface pocket formation than the rest of the protein surface. PLoS Comput. Biol. 9, e1002951 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. 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).

    CAS  PubMed  Google Scholar 

  93. Brown, S. P. & Hajduk, P. J. Effects of conformational dynamics on predicted protein druggability. ChemMedChem 1, 70–72 (2006).

    CAS  PubMed  Google Scholar 

  94. Tan, Y. S., Spring, D. R., Abell, C. & Verma, C. The use of chlorobenzene as a probe molecule in molecular dynamics simulations. J. Chem. Inf. Model. 54, 1821–1827 (2014).

    CAS  PubMed  Google Scholar 

  95. Sledz, P. et al. From crystal packing to molecular recognition: prediction and discovery of a binding site on the surface of polo-like kinase 1. Angew. Chem. Int. Ed. Engl. 50, 4003–4006 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Vogler, M., Dinsdale, D., Dyer, M. J. & Cohen, G. M. Bcl-2 inhibitors: small molecules with a big impact on cancer therapy. Cell Death Differ. 16, 360–367 (2009).

    CAS  PubMed  Google Scholar 

  97. Souers, A. J. et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat. Med. 19, 202–208 (2013).

    CAS  PubMed  Google Scholar 

  98. Liu, X. Q., Dai, S. D., Zhu, Y. N., Marrack, P. & Kappler, J. W. The structure of a Bcl-xL/Bim fragment complex: implications for bim function. Immunity 19, 341–352 (2003).

    CAS  PubMed  Google Scholar 

  99. Oberstein, A., Jeffrey, P. D. & Shi, Y. G. Crystal structure of the Bcl-X-L–beclin 1 peptide complex — beclin 1 is a novel BH3-only protein. J. Biol. Chem. 282, 13123–13132 (2007).

    CAS  PubMed  Google Scholar 

  100. Lee, E. F. et al. Conformational changes in Bcl-2 pro-survival proteins determine their capacity to bind ligands. J. Biol. Chem. 284, 30508–30517 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Petros, A. M. et al. Rationale for Bcl-xL/Bad peptide complex formation from structure, mutagenesis, and biophysical studies. Protein Sci. 9, 2528–2534 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Lee, E. F. et al. Crystal structure of ABT-737 complexed with Bcl-xL: implications for selectivity of antagonists of the Bcl-2 family. Cell Death Differ. 14, 1711–1713 (2007).

    CAS  PubMed  Google Scholar 

  103. Lee, E. F. et al. High-resolution structural characterization of a helical α/β-peptide foldamer bound to the anti-apoptotic protein Bcl-xL . Angew. Chem. Int. Ed. Engl. 48, 4318–4322 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Sattler, M. et al. Structure of Bcl-xL–Bak peptide complex: recognition between regulators of apoptosis. Science 275, 983–986 (1997).

    CAS  PubMed  Google Scholar 

  105. Wang, J. L. et al. Structure-based discovery of an organic compound that binds Bcl-2 protein and induces apoptosis of tumor cells. Proc. Natl Acad. Sci. USA 97, 7124–7129 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Enyedy, I. J. et al. Discovery of small-molecule inhibitors of Bcl-2 through structure-based computer screening. J. Med. Chem. 44, 4313–4324 (2001).

    CAS  PubMed  Google Scholar 

  107. Lugovskoy, A. A. et al. A novel approach for characterizing protein ligand complexes: molecular basis for specificity of small-molecule Bcl-2 inhibitors. J. Am. Chem. Soc. 124, 1234–1240 (2002).

    CAS  PubMed  Google Scholar 

  108. Mukherjee, P., Desai, P., Zhou, Y. D. & Avery, M. Targeting the BH3 domain mediated protein–protein interaction of Bcl-xL through virtual screening. J. Chem. Inf. Model. 50, 906–923 (2010).

    CAS  PubMed  Google Scholar 

  109. Real, P. J. et al. Breast cancer cells can evade apoptosis-mediated selective killing by a novel small molecule inhibitor of Bcl-2. Cancer Res. 64, 7947–7953 (2004).

    CAS  PubMed  Google Scholar 

  110. Wang, G. et al. Structure-based design of potent small-molecule inhibitors of anti-apoptotic Bcl-2 proteins. J. Med. Chem. 49, 6139–6142 (2006).

    CAS  PubMed  Google Scholar 

  111. Zhou, H. et al. Design of Bcl-2 and Bcl-xL inhibitors with subnanomolar binding affinities based upon a new scaffold. J. Med. Chem. 55, 4664–4682 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Biros, S. M. et al. Heterocyclic α-helix mimetics for targeting protein–protein interactions. Bioorg. Med. Chem. Lett. 17, 4641–4645 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Yin, H. & Hamilton, A. D. Terephthalamide derivatives as mimetics of the helical region of Bak peptide target Bcl-xL protein. Bioorg. Med. Chem. Lett. 14, 1375–1379 (2004).

    CAS  PubMed  Google Scholar 

  114. Antuch, W. et al. Design and modular parallel synthesis of a MCR derived α-helix mimetic protein–protein interaction inhibitor scaffold. Bioorg. Med. Chem. Lett. 16, 1740–1743 (2006).

    CAS  PubMed  Google Scholar 

  115. Walensky, L. D. et al. Activation of apoptosis in vivo by a hydrocarbon-stapled BH3 helix. Science 305, 1466–1470 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Stewart, M. L., Fire, E., Keating, A. E. & Walensky, L. D. The MCL-1 BH3 helix is an exclusive MCL-1 inhibitor and apoptosis sensitizer. Nat. Chem. Biol. 6, 595–601 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Perez, H. L. et al. Identification of a phenylacylsulfonamide series of dual Bcl-2/Bcl-xL antagonists. Bioorg. Med. Chem. Lett. 22, 3946–3950 (2012).

    CAS  PubMed  Google Scholar 

  118. Degterev, A. et al. Identification of small-molecule inhibitors of interaction between the BH3 domain and Bcl-xL. Nat. Cell Biol. 3, 173–182 (2001).

    CAS  PubMed  Google Scholar 

  119. Tse, C. et al. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 68, 3421–3428 (2008).

    CAS  PubMed  Google Scholar 

  120. Sleebs, B. E. et al. Quinazoline sulfonamides as dual binders of the proteins B-cell lymphoma 2 and B-cell lymphoma extra long with potent proapoptotic cell-based activity. J. Med. Chem. 54, 1914–1926 (2011).

    CAS  PubMed  Google Scholar 

  121. Tanaka, Y. et al. Discovery of potent Mcl-1/Bcl-xL dual inhibitors by using a hybridization strategy based on structural analysis of target proteins. J. Med. Chem. 56, 9635–9645 (2013).

    CAS  PubMed  Google Scholar 

  122. Mahon, A. B., Miller, S. E., Joy, S. T. & Arora, P. S. Rational design strategies for developing synthetic inhibitors of helical protein interfaces. Top. Med. Chem. 8, 197–230 (2012).

    CAS  Google Scholar 

  123. Zhao, Y., Aguilar, A., Bernard, D. & Wang, S. Small-molecule inhibitors of the MDM2-p53 protein–protein interaction (MDM2 Inhibitors) in clinical trials for cancer treatment. J. Med. Chem. 58, 1038–1052 (2015).

    CAS  PubMed  Google Scholar 

  124. Srinivasula, S. M. et al. A conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and apoptosis. Nature 410, 112–116 (2001).

    CAS  PubMed  Google Scholar 

  125. Huang, Y., Rich, R. L., Myszka, D. G. & Wu, H. Requirement of both the second and third BIR domains for the relief of X-linked inhibitor of apoptosis protein (XIAP)-mediated caspase inhibition by Smac. J. Biol. Chem. 278, 49517–49522 (2003).

    CAS  PubMed  Google Scholar 

  126. Sun, H. Y. et al. Structure-based design of potent, conformationally constrained Smac mimetics. J. Am. Chem. Soc. 126, 16686–16687 (2004).

    CAS  PubMed  Google Scholar 

  127. Oost, T. K. et al. Discovery of potent antagonists of the antiapoptotic protein XIAP for the treatment of cancer. J. Med. Chem. 47, 4417–4426 (2004).

    CAS  PubMed  Google Scholar 

  128. Cai, Q. et al. A potent and orally active antagonist (SM-406/AT-406) of multiple inhibitor of apoptosis proteins (IAPs) in clinical development for cancer treatment. J. Med. Chem. 54, 2714–2726 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Cossu, F. et al. Designing Smac-mimetics as antagonists of XIAP, cIAP1, and cIAP2. Biochem. Biophys. Res. Commun. 378, 162–167 (2009).

    CAS  PubMed  Google Scholar 

  130. Monfardini, I. et al. Screening multicomponent reactions for X-linked inhibitor of apoptosis-baculoviral inhibitor of apoptosis protein repeats domain binder. J. Med. Chem. 54, 890–900 (2011).

    CAS  PubMed  Google Scholar 

  131. Huang, J. W. et al. Fragment-based design of small molecule X-linked inhibitor of apoptosis protein inhibitors. J. Med. Chem. 51, 7111–7118 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Sun, H. et al. Design, synthesis, and characterization of a potent, nonpeptide, cell-permeable, bivalent Smac mimetic that concurrently targets both the BIR2 and BIR3 domains in XIAP. J. Am. Chem. Soc. 129, 15279–15294 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Wu, H., Tschopp, J. & Lin, S. C. Smac mimetics and TNFα: a dangerous liaison? Cell 131, 655–658 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Kester, R. F. et al. Optimization of benzodiazepinones as selective inhibitors of the X-linked inhibitor of apoptosis protein (XIAP) second baculovirus IAP repeat (BIR2) domain. J. Med. Chem. 56, 7788–7803 (2013).

    CAS  PubMed  Google Scholar 

  135. Donnell, A. F. et al. Benzazepinones and benzoxazepinones as antagonists of inhibitor of apoptosis proteins (IAPs) selective for the second baculovirus IAP repeat (BIR2) domain. J. Med. Chem. 56, 7772–7787 (2013).

    CAS  PubMed  Google Scholar 

  136. Padmanabhan, B. et al. Structural basis for defects of Keap1 activity provoked by its point mutations in lung cancer. Mol. Cell 21, 689–700 (2006).

    CAS  PubMed  Google Scholar 

  137. Tong, K. I. et al. Different electrostatic potentials define ETGE and DLG motifs as hinge and latch in oxidative stress response. Mol. Cell. Biol. 27, 7511–7521 (2007). This paper proposes a hinge and latch model to explain how the KEAP1–NRF2 system senses and responds to oxidative and electrophilic stress.

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Lo, S. C., Li, X., Henzl, M. T., Beamer, L. J. & Hannink, M. Structure of the Keap1:Nrf2 interface provides mechanistic insight into Nrf2 signaling. EMBO J. 25, 3605–3617 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Chen, Y., Inoyama, D., Kong, A. N., Beamer, L. J. & Hu, L. Kinetic analyses of Keap1–Nrf2 interaction and determination of the minimal Nrf2 peptide sequence required for Keap1 binding using surface plasmon resonance. Chem. Biol. Drug Des. 78, 1014–1021 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Hancock, R. et al. Peptide inhibitors of the Keap1-Nrf2 protein–protein interaction. Free Radic. Biol. Med. 52, 444–451 (2012).

    CAS  PubMed  Google Scholar 

  141. Hancock, R., Schaap, M., Pfister, H. & Wells, G. Peptide inhibitors of the Keap1–Nrf2 protein–protein interaction with improved binding and cellular activity. Org. Biomol. Chem. 11, 3553–3557 (2013).

    CAS  PubMed  Google Scholar 

  142. Hu, L. et al. Discovery of a small-molecule inhibitor and cellular probe of Keap1–Nrf2 protein–protein interaction. Bioorg. Med. Chem. Lett. 23, 3039–3043 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Marcotte, D. et al. Small molecules inhibit the interaction of Nrf2 and the Keap1 Kelch domain through a non-covalent mechanism. Bioorg. Med.Chem. 21, 4011–4019 (2013).

    CAS  PubMed  Google Scholar 

  144. Jiang, Z. Y. et al. Discovery of potent Keap1–Nrf2 protein–protein interaction inhibitor based on molecular binding determinants analysis. J. Med. Chem. 57, 2736–2745 (2014). In this study a potent inhibitor of the KEAP1–NRF2 interaction was designed based upon a reported ligand of KEAP1 and a consideration of hot spot residues from the NRF2 peptide ligand.

    CAS  PubMed  Google Scholar 

  145. Sun, H. P. et al. Novel protein–protein interaction inhibitor of Nrf2–Keap1 discovered by structure-based virtual screening. MedChemComm 5, 93–98 (2014).

    CAS  Google Scholar 

  146. Zhuang, C., Narayanapillai, S., Zhang, W., Sham, Y. Y. & Xing, C. Rapid identification of Keap1–Nrf2 small-molecule inhibitors through structure-based virtual screening and hit-based substructure search. J. Med.Chem. 57, 1121–1126 (2014).

    CAS  PubMed  Google Scholar 

  147. Ducki, S. & Bennett, E. Protein–protein interactions: recent progress in the development of selective PDZ inhibitors. Curr. Chem. Biol. 3, 1–13 (2009).

    Google Scholar 

  148. Cox, D., Brennan, M. & Moran, N. Integrins as therapeutic targets: lessons and opportunities. Nat.Rev. Drug. Discov. 9, 804–820 (2010).

    CAS  PubMed  Google Scholar 

  149. Patel, A., Dharmarajan, V. & Cosgrove, M. S. Structure of WDR5 bound to mixed lineage leukemia protein-1 peptide. J. Biol. Chem. 283, 32158–32161 (2008).

    CAS  PubMed  Google Scholar 

  150. Patel, A., Vought, V. E., Dharmarajan, V. & Cosgrove, M. S. A conserved arginine-containing motif crucial for the assembly and enzymatic activity of the mixed lineage leukemia protein-1 core complex. J. Biol. Chem. 283, 32162–32175 (2008).

    CAS  PubMed  Google Scholar 

  151. Karatas, H. et al. High-affinity, small-molecule peptidomimetic inhibitors of MLL1/WDR5 protein–protein interaction. J. Am. Chem. Soc. 135, 669–682 (2013).

    CAS  PubMed  Google Scholar 

  152. Wilson, C. G. & Arkin, M. R. Small-molecule inhibitors of IL-2/IL-2R: lessons learned and applied. Curr. Top. Microbiol. Immunol. 348, 25–59 (2011).

    CAS  PubMed  Google Scholar 

  153. Tilley, J. W. et al. Identification of a small molecule inhibitor of the IL-2/IL-2Rα receptor interaction which binds to IL-2. J. Am. Chem. Soc. 119, 7589–7590 (1997).

    CAS  Google Scholar 

  154. Arkin, M. R. et al. Binding of small molecules to an adaptive protein–protein interface. Proc. Natl Acad. Sci. USA 100, 1603–1608 (2003). In this paper the flexibility and adaptivity of certain protein–protein interfaces is highlighted, with a small molecule that binds a pocket in IL-2 that is not seen in the unbound IL-2 structure. The technique of fragment 'tethering' was then applied to find other hits for this cryptic pocket.

    CAS  PubMed  PubMed Central  Google Scholar 

  155. 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). The first description of 'tethering' to discover a high potency inhibitor against a PPI interface.

    CAS  PubMed  Google Scholar 

  156. Raimundo, B. C. et al. Integrating fragment assembly and biophysical methods in the chemical advancement of small-molecule antagonists of IL-2: an approach for inhibiting protein–protein interactions. J. Med. Chem. 47, 3111–3130 (2004).

    CAS  PubMed  Google Scholar 

  157. Hyde, J., Braisted, A. C., Randal, M. & Arkin, M. R. Discovery and characterization of cooperative ligand binding in the adaptive region of interleukin-2. Biochemistry 42, 6475–6483 (2003).

    CAS  PubMed  Google Scholar 

  158. Arrowsmith, C. H., Bountra, C., Fish, P. V., Lee, K. & Schapira, M. Epigenetic protein families: a new frontier for drug discovery. Nat. Rev. Drug. Discov. 11, 384–400 (2012).

    CAS  PubMed  Google Scholar 

  159. Furdas, S. D., Carlino, L., Sippl, W. & Jung, M. Inhibition of bromodomain-mediated protein–protein interactions as a novel therapeutic strategy. MedChemComm 3, 123–134 (2012).

    CAS  Google Scholar 

  160. Filippakopoulos, P. et al. Histone recognition and large-scale structural analysis of the human bromodomain family. Cell 149, 214–231 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Filippakopoulos, P. et al. Selective inhibition of BET bromodomains. Nature 468, 1067–1073 (2010). In this paper the authors report a small molecule with high potency and a remarkable selectivity for a small subset of human bromodomains.

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Mirguet, O. et al. Discovery of epigenetic regulator I-BET762: lead optimization to afford a clinical candidate inhibitor of the BET bromodomains. J. Med. Chem. 56, 7501–7515 (2013).

    CAS  PubMed  Google Scholar 

  163. Zimmermann, G. et al. Small molecule inhibition of the KRAS-PDEδ interaction impairs oncogenic KRAS signalling. Nature 497, 638–642 (2013). The first report of small molecules that disrupt KRAS localization to PDEδ by binding in the prenyl binding pocket of PDEδ.

    CAS  PubMed  Google Scholar 

  164. Ku, B., Liang, C., Jung, J. U. & Oh, B. H. Evidence that inhibition of BAX activation by BCL-2 involves its tight and preferential interaction with the BH3 domain of BAX. Cell Res. 21, 627–641 (2011).

    CAS  PubMed  Google Scholar 

  165. Bruncko, M. et al. Studies leading to potent, dual inhibitors of Bcl-2 and Bcl-xL. J. Med. Chem. 50, 641–662 (2007).

    CAS  PubMed  Google Scholar 

  166. Robb, R. J., Rusk, C. M. & Neeper, M. P. Structure-function relationships for the interleukin 2 receptor: location of ligand and antibody binding sites on the Tac receptor chain by mutational analysis. Proc. Natl Acad. Sci. USA 85, 5654–5658 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Sauve, K. et al. Localization in human interleukin 2 of the binding site to the alpha chain (p55) of the interleukin 2 receptor. Proc. Natl Acad. Sci. USA 88, 4636–4640 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Thanos, C. D., DeLano, W. L. & Wells, J. A. Hot-spot mimicry of a cytokine receptor by a small molecule. Proc. Natl Acad. Sci. USA 103, 15422–15427 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank T. Blundell and H. Jubb for helpful discussions. J.S., D.E.S. and A.R.B. thank the Wellcome Trust for funding.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Chris Abell or John Skidmore.

Ethics declarations

Competing interests

J.S. owns shares in GlaxoSmithKline. C.A. is co-founder and advisor for Astex Pharmaceuticals. A.B. is an employee of Vertex.

Related links

PowerPoint slides

Glossary

Globular protein

A protein whose peptide chains are folded to form a broadly spherical shape. This term is often extended to mean a non-membrane-bound protein in which multiple regions of the peptide chain combine to give a defined tertiary structure with one or more binding or active sites.

Hot spots

Regions of a binding surface that contribute a disproportionately large amount to the interaction energy of a pair of proteins or a protein and a ligand.

Alanine scanning

A set of experiments in which amino acids in a protein are sequentially mutated to Ala in order to estimate the contribution of the individual side chains to the binding energy of a protein–protein interaction.

Discontinuous epitope

A binding site at which amino acids from different regions of the protein sequence combine to interact with the partner protein. For example, periodic side chains on one face of a helix or residues from a group of neighbouring chains in a globular protein.

Intrinsically disordered peptide

A peptide lacking a fixed 3D structure when in a monomeric state. Such species may adopt a more ordered structure when interacting with other proteins or ligands.

Druggability

An assessment of the ease with which a drug can be developed to interact with a given protein target. In this case, it has a relatively narrow meaning — namely the ease of generating a small molecule with reasonable affinity.

Pharmacophore

The features of a ligand responsible for its binding to a protein, which are reduced to an abstract representation lacking an underlying framework of bonds or rings

Apoptosis

Programmed cell death controlled by a complex network of interactions including protein–protein interactions.

Ubiquitylation

The attachment of a small protein, ubiquitin, onto another protein — often as a prelude to controlled degradation of the labelled protein.

ADMET

Absorption, distribution, metabolism, excretion and toxicity — key properties of a potential drug that depend, in part, on the physicochemical properties of a molecule.

PDZ domain

A protein domain containing a series of Gly-Leu-Gly-Phe repeating units. Named after the postsynaptic density protein 95 (PSD95), the Drosophilia melanogaster tumour suppressor discs large 1 (Dlg1; also known as Dlg-A) and the tight-junction associated protein zonula occludens 1 (ZO1).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Scott, D., Bayly, A., Abell, C. et al. Small molecules, big targets: drug discovery faces the protein–protein interaction challenge. Nat Rev Drug Discov 15, 533–550 (2016). https://doi.org/10.1038/nrd.2016.29

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd.2016.29

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research