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.

  • Opinion
  • Published:

ESKAPEing the labyrinth of antibacterial discovery

An Erratum to this article was published on 14 August 2015

This article has been updated

Abstract

Antimicrobial drug resistance is a growing threat to global public health. Multidrug resistance among the 'ESKAPE' organisms — encompassing Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp. — is of particular concern because they are responsible for many serious infections in hospitals. Although some promising agents are in the pipeline, there is an urgent need for new antibiotic scaffolds. However, antibacterial researchers have struggled to identify new small molecules with meaningful cellular activity, especially those effective against multidrug-resistant Gram-negative pathogens. This difficulty ultimately stems from an incomplete understanding of efflux systems and compound permeation through bacterial membranes. This Opinion article describes findings from target-based and phenotypic screening efforts carried out at AstraZeneca over the past decade, discusses some of the subsequent chemistry challenges and concludes with a description of new approaches comprising a combination of computational modelling and advanced biological tools which may pave the way towards the discovery of new antibacterial agents.

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: Mean LogD values for internal AstraZeneca antibacterial project compounds and for exemplar hits from other disease areas.
Figure 2: The relationship over time between the biochemical potency against Pseudomonas aeruginosa MurC and the cLogD of newly synthesized programme compounds.
Figure 3: Gram-negative and Gram-positive cell walls.
Figure 4: Effect of porin point mutations on antibiotic transport.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

Change history

  • 14 July 2015

    In reference 67 the journal name was incorrect. It has now been corrected online and in print.

  • 14 August 2015

    In the legend of Figure 3, Gram-negative and Gram-positive bacteria were incorrectly labelled. This has been corrected in the online version of the article.

References

  1. World Health Organization. Antimicrobial Resistance: Global Report on Surveillance http://apps.who.int/iris/bitstream/10665/112642/1/9789241564748_eng.pdf?ua=1 (WHO 2014).

  2. Boucher, H. W. et al. 10 × '20 progress — development of new drugs active against Gram-negative Bacilli: an update from the Infectious Diseases Society of America. Clin. Infect. Dis. 56, 1685–1694 (2013).

    Article  Google Scholar 

  3. Rex, J. H., Goldberger, M., Eisenstein, B. I. & Harney, C. The evolution of the regulatory framework for antibacterial agents. Ann. NY Acad. Sci. 1323, 11–21 (2014).

    Article  Google Scholar 

  4. Payne, D. J., Gwynn, M. N., Holmes, D. J. & Pompliano, D. L. Drugs for bad bugs: confronting the challenges of antibacterial discovery. Nat. Rev. Drug Discov. 6, 29–40 (2007). This paper provides an overview of the HTS efforts directed at the identification of novel antibacterials at GSK, which comprised the study of more than 300 genes and 70 high-throughput screens over a period of 7 years. Furthermore, a thorough analysis of potential reasons for failure is presented along with perspectives on how to improve the likelihood of success through focusing on broader chemical diversity.

    Article  CAS  Google Scholar 

  5. O'Shea, R. & Moser, H. E. Physicochemical properties of antibacterial compounds: implications for drug discovery. J. Med. Chem. 51, 2871–2878 (2008).

    Article  CAS  Google Scholar 

  6. Eren, E. et al. Substrate specificity within a family of outer membrane carboxylate channels. PLoS Biol. 10, e1001242 (2012).

    Article  CAS  Google Scholar 

  7. de Jonge, B. L. et al. Discovery of inhibitors of 4′-phosphopantetheine adenylyltransferase (PPAT) to validate PPAT as a target for antibacterial therapy. Antimicrob. Agents Chemother. 57, 6005–6015 (2013).

    Article  CAS  Google Scholar 

  8. Copeland, R. A. Evaluation of Enzyme Inhibitors in Drug Discovery: A Guide for Medicinal Chemists and Pharmacologists. 2nd edn (John Wiley & Sons, 2013).

    Book  Google Scholar 

  9. Roemer, T., Davies, J., Giaever, G. & Nislow, C. Bugs, drugs and chemical genomics. Nat. Chem. Biol. 8, 46–56 (2012).

    Article  CAS  Google Scholar 

  10. Buurman, E. T. et al. In vitro validation of acetyltransferase activity of GlmU as an antibacterial target in Haemophilus influenzae. J. Biol. Chem. 286, 40734–40742 (2011).

    Article  CAS  Google Scholar 

  11. Mills, S. D. et al. Novel bacterial NAD+-dependent DNA ligase inhibitors with broad-spectrum activity and antibacterial efficacy in vivo. Antimicrob. Agents Chemother. 55, 1088–1096 (2011).

    Article  CAS  Google Scholar 

  12. Lajiness, M. S., Maggiora, G. M. & Shanmugasundaram, V. Assessment of the consistency of medicinal chemists in reviewing sets of compounds. J. Med. Chem. 47, 4891–4896 (2004).

    Article  CAS  Google Scholar 

  13. Sanguinetti, M. C. & Tristani-Firouzi, M. hERG potassium channels and cardiac arrhythmia. Nature 440, 463–469 (2006).

    Article  CAS  Google Scholar 

  14. Seidler, J., McGovern, S. L., Doman, T. N. & Shoichet, B. K. Identification and prediction of promiscuous aggregating inhibitors among known drugs. J. Med. Chem. 46, 4477–4486 (2003).

    Article  CAS  Google Scholar 

  15. Clinical and Laboratory Standards Institute. CLSI document M07-A9: Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard 9th edn (CLSI, 2012).

  16. Uria-Nickelsen, M. et al. Novel topoisomerase inhibitors: microbiological characterisation and in vivo efficacy of pyrimidines. Int. J. Antimicrob. Agents 41, 363–371 (2013).

    Article  CAS  Google Scholar 

  17. Keating, T. A. et al. In vivo validation of thymidylate kinase (TMK) with a rationally designed, selective antibacterial compound. ACS Chem. Biol. 7, 1866–1872 (2012).

    Article  CAS  Google Scholar 

  18. Brown, D. G., May-Dracka, T. L., Gagnon, M. M. & Tommasi, R. Trends and exceptions of physical properties on antibacterial activity for Gram-positive and Gram-negative pathogens. J. Med. Chem. 57, 10144–10161 (2014).

    Article  CAS  Google Scholar 

  19. Hann, M. M. Molecular obesity, potency and other addictions in drug discovery. Med. Chem. Commun. 2, 349–355 (2011).

    Article  CAS  Google Scholar 

  20. Leeson, P. D. & Springthorpe, B. The influence of drug-like concepts on decision-making in medicinal chemistry. Nat. Rev. Drug Discov. 6, 881–890 (2007).

    Article  CAS  Google Scholar 

  21. Wenlock, M. C., Austin, R. P., Barton, P., Davis, A. M. & Leeson, P. D. A comparison of physiochemical property profiles of development and marketed oral drugs. J. Med. Chem. 46, 1250–1256 (2003).

    Article  CAS  Google Scholar 

  22. Hernandez, V. et al. Discovery of a novel class of boron-based antibacterials with activity against Gram-negative bacteria. Antimicrob. Agents Chemother. 57, 1394–1403 (2013).

    Article  CAS  Google Scholar 

  23. Rees, D. C., Congreve, M., Murray, C. W. & Carr, R. Fragment-based lead discovery. Nat. Rev. Drug Discov. 3, 660–672 (2004).

    Article  CAS  Google Scholar 

  24. Ling, L. L. et al. A new antibiotic kills pathogens without detectable resistance. Nature 517, 455–459 (2015).

    Article  CAS  Google Scholar 

  25. Brown, D. G., Lister, T. & May-Dracka, T. L. New natural products as new leads for antibacterial drug discovery. Bioorg. Med. Chem. Lett. 24, 413–418 (2014).

    Article  CAS  Google Scholar 

  26. Cabeen, M. T. & Jacobs-Wagner, C. Bacterial cell shape. Nat. Rev. Micro 3, 601–610 (2005).

    Article  CAS  Google Scholar 

  27. Silhavy, T. J., Kahne, D. & Walker, S. The bacterial cell envelope. Cold Spring Harb. Perspect. Biol. 2, a000414 (2010).

    Article  Google Scholar 

  28. Denyer, S. P. & Maillard, J. Y. Cellular impermeability and uptake of biocides and antibiotics in Gram-negative bacteria. J. Appl. Microbiol. 92, 35S–45S (2002).

    Article  Google Scholar 

  29. Hancock, R. E. & Brinkman, F. S. Function of pseudomonas porins in uptake and efflux. Annu. Rev. Microbiol. 56, 17–38 (2002).

    Article  CAS  Google Scholar 

  30. Biswas, S., Mohammad, M. M., Patel, D. R., Movileanu, L. & van den Berg, B. Structural insight into OprD substrate specificity. Nature Struct. Mol. Biol. 14, 1108–1109 (2007). This paper provides an encompassing structural and biophysical rationale for the adoption of the OccD and OccK nomenclature standard for P. aeruginosa outer-membrane carboxylate channels (porins). Additionally, it presents and summarizes the main methods that are presently at the forefront of studying porin dynamics and elucidating substrate selectivity: single-channel electrophysiology and radioactive-tracer direct measurement (or competition). The authors also demonstrate the selective binding and transport of antibiotics by these channels.

    Article  CAS  Google Scholar 

  31. Ceccarelli, M., Vargiu, A. & Ruggerone, P. A kinetic Monte Carlo approach to investigate antibiotic translocation through bacterial porins. J. Phys. Condens. Matter 24, 104012 (2012).

    Article  Google Scholar 

  32. Eren, E. et al. Toward understanding the outer membrane uptake of small molecules by Pseudomonas aeruginosa. J. Biol. Chem. 288, 12042–12053 (2013).

    Article  CAS  Google Scholar 

  33. Kattner, C., Zaucha, J., Jaenecke, F., Zachariae, U. & Tanabe, M. Identification of a cation transport pathway in Neisseria meningitidis PorB. Proteins 81, 830–840 (2013).

    Article  CAS  Google Scholar 

  34. Shaw, D. E. et al. Atomic-level characterization of the structural dynamics of proteins. Science 330, 341–346 (2010). This paper describes a landmark 1-millisecond molecular-dynamics simulation of bovine pancreatic trypsin inhibitor (BPTI) as well as simulations of FiP35 and villin, which demonstrate that not only are long timescale simulations stable — a point which was previously in doubt — but they are also capable of accurately reproducing the experimentally observed behaviour of these systems, including the kinetics of protein folding to the native state, in addition to the detail these methods provide at atomic resolution.

    Article  CAS  Google Scholar 

  35. Laio, A. Parrinello, M. Escaping free-energy minima. Proc. Natl Acad. Sci. USA. 99, 12562–12566 (2002).

    Article  CAS  Google Scholar 

  36. Isabella, V. M. et al. Towards the rational design of carbapenem uptake in Pseudomonas aeruginosa. Chem.Biol. 22, 535–547 (2015). Using a multidisciplinary approach, including genetics, molecular dynamic simulations and medicinal chemistry, these authors discovered a novel mechanism of carbapenem uptake in P. aeruginosa , which led to the generation of novel carbapenems with altered uptake properties.

    Article  CAS  Google Scholar 

  37. van Opijnen, T., Lazinski, D. W. & Camilli, A. Genome-wide fitness and genetic interactions determined by Tn-seq, a high-throughput massively parallel sequencing method for microorganisms. Curr. Protoc. Microbiol. 36, 1E.3.1–1E.3.24 (2015). This study describes Tn-seq, a newly developed genomic platform that deploys high-throughput next-generation sequencing on saturated transposon-insertion libraries of microorganisms to decipher complex fitness phenotypes on a global scale.

    Article  Google Scholar 

  38. Tamber, S. & Hancock, R. E. W. Involvement of two related porins, OprD and OpdP, in the uptake of arginine by Pseudomonas aeruginosa. FEMS Microbiol. Lett. 260, 23–29 (2006).

    Article  CAS  Google Scholar 

  39. Möllmann, U., Heinisch, L., Bauernfeind, A., Köhler, T. & Ankel-Fuchs, D. Siderophores as drug delivery agents: application of the “Trojan Horse” strategy. Biometals 22, 615–624 (2009).

    Article  Google Scholar 

  40. deCarvalho, C. C. & Fernandes, P. Siderophores as “Trojan Horses”: tackling multidrug resistance? Frontiers Microbiol. 5, 290 (2014).

    Google Scholar 

  41. Rivault, F. et al. Synthesis of pyochelin–norfloxacin conjugates. Bioorg. Med. Chem. Lett. 17, 640–644 (2007).

    Article  CAS  Google Scholar 

  42. Page, M. G. P. Siderophore conjugates. Ann. NY Acad. Sci. 1277, 115–126 (2013).

    Article  CAS  Google Scholar 

  43. Page, M. G. P. et al. In vitro and in vivo properties of BAL30376, a β-lactam and dual β-lactamase inhibitor combination with enhanced activity against Gram-negative Bacilli that express multiple β-lactamases. Antimicrob. Agents Chemother. 55, 1510–1519 (2011).

    Article  CAS  Google Scholar 

  44. Tomaras, A. P. et al. Adaptation-based resistance to siderophore-conjugated antibacterial agents by Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 57, 4197–4207 (2013).

    Article  CAS  Google Scholar 

  45. Ricci, V. & Piddock, L. Accumulation of garenoxacin by Bacteroides fragilis compared with that of five fluoroquinolones. J. Antimicrob. Chemother. 52, 605–609 (2003).

    Article  CAS  Google Scholar 

  46. Williams, K. J. & Piddock, L. J. Accumulation of rifampicin by Escherichia coli and Staphylococcus aureus. J. Antimicrob. Chemother. 42, 597–603 (1998).

    Article  CAS  Google Scholar 

  47. Bhat, J., Narayan, A., Venkatraman, J. & Chatterji, M. LC–MS based assay to measure intracellular compound levels in Mycobacterium smegmatis: linking compound levels to cellular potency. J. Microbiol. Methods 94, 152–158 (2013).

    Article  CAS  Google Scholar 

  48. Davis, T. D., Gerry, C. J. & Tan, D. S. General platform for systematic quantitative evaluation of small-molecule permeability in bacteria. ACS Chem. Biol. 9, 2535–2544 (2014).

    Article  CAS  Google Scholar 

  49. Rubakhin, S. S., Lanni, E. J. & Sweedler, J. V. Progress toward single cell metabolomics. Curr. Opin. Biotechnol. 24, 95–104 (2013).

    Article  CAS  Google Scholar 

  50. IMI. Translocation: molecular basis of the bacterial cell wall permeability. Innovative Medicines Initiative http://www.imi.europa.eu/content/translocation (2008).

  51. May, M. Drug development: time for teamwork. Nature 509, S4–S5 (2014).

    Article  CAS  Google Scholar 

  52. Valentino, M. D. et al. Genes contributing to Staphylococcus aureus fitness in abscess- and infection-related ecologies. mBio 5, e01729-14 (2014).

    Article  Google Scholar 

  53. Subashchandrabose, S., Smith, S. N., Spurbeck, R. R., Kole, M. M. & Mobley, H. L. T. Genome-wide detection of fitness genes in uropathogenic Escherichia coli during systemic infection. PLoS Pathog. 9, e1003788 (2013).

    Article  Google Scholar 

  54. Palace, S. G., Proulx, M. K., Lu, S., Baker, R. E. & Goguen, J. D. Genome-wide mutant fitness profiling identifies nutritional requirements for optimal growth of Yersinia pestis in deep tissue. mBio 5, e01385-14 (2014).

    Article  Google Scholar 

  55. Wang, N., Ozer, E. A., Mandel, M. J. & Hauser, A. R. Genome-wide identification of Acinetobacter baumannii genes necessary for persistence in the lung. mBio 5, e01163-14 (2014).

    Article  Google Scholar 

  56. Skurnik, D. et al. Enhanced in vivo fitness of carbapenem-resistant oprD mutants of Pseudomonas aeruginosa revealed through high-throughput sequencing. Proc. Natl Acad. Sci. USA 110, 20747–20752 (2013).

    Article  CAS  Google Scholar 

  57. Turner, K. H., Everett, J., Trivedi, U., Rumbaugh, K. P. & Whiteley, M. Requirements for Pseudomonas aeruginosa acute burn and chronic surgical wound infection. PLoS Genet. 10, e1004518 (2014).

    Article  Google Scholar 

  58. Moule, M. G. et al. Genome-wide saturation mutagenesis of Burkholderia pseudomallei K96243 predicts essential genes and novel targets for antimicrobial development. mBio 5, e00926-13 (2014).

    Article  Google Scholar 

  59. Gallagher, L. A., Shendure, J. & Manoil, C. Genome-scale identification of resistance functions in Pseudomonas aeruginosa using Tn-seq. mBio 2, e00315-10 (2011).

    Article  Google Scholar 

  60. Cabot, G. et al. Pseudomonas aeruginosa ceftolozane-tazobactam resistance development requires multiple mutations leading to overexpression and structural modification of AmpC. Antimicrob. Agents Chemother. 58, 3091–3099 (2014).

    Article  Google Scholar 

  61. Jones, C. J. et al. ChIP-Seq and RNA-Seq reveal an AmrZ-mediated mechanism for cyclic di-GMP synthesis and biofilm development by Pseudomonas aeruginosa. PLoS Pathog. 10, e1003984 (2014).

    Article  Google Scholar 

  62. Hua, X., Chen, Q., Li, X. & Yu, Y. Global transcriptional response of Acinetobacter baumannii to a subinhibitory concentration of tigecycline. Int. J. Antimicrob. Agents 44, 337–344 (2014).

    Article  CAS  Google Scholar 

  63. Tan, S. Y.-Y. et al. Comparative systems biology analysis and mode of action of the isothiocyanate compound iberin on Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 58, 6648–6659 (2014).

    Article  Google Scholar 

  64. Armengaud, J. Microbiology and proteomics, getting the best of both worlds! Environ. Microbiol. 15, 12–23 (2013).

    Article  CAS  Google Scholar 

  65. Chavali, A. K., D'Auria, K. M., Hewlett, E. L., Pearson, R. D. & Papin, J. A. A metabolic network approach for the identification and prioritization of antimicrobial drug targets. Trends Microbiol. 20, 113–123 (2012).

    Article  CAS  Google Scholar 

  66. Lebeis, S. L. & Kalman, D. Aligning antimicrobial drug discovery with complex and redundant host-pathogen interactions. Cell Host Microbe 5, 114–122 (2009).

    Article  CAS  Google Scholar 

  67. Nayar, A.S. et al. Novel antibacterial targets and compounds revealed by a high-throughput cell wall reporter assay. J. Bacteriol. 197, 1726–1734 (2015)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This review represents many years of hard work by former members of the AstraZeneca Infection Innovative Medicine and Early Development department, whom the authors would like to gratefully acknowledge for their intelligence, creativity and dedication to the advancement of this important field of research.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ruben Tommasi.

Ethics declarations

Competing interests

All authors are current or former employees of AstraZeneca and, as such, may hold stock in the company.

Related links

FURTHER INFORMATION

Discuva website

DATABASES

RCSB Protein Data Bank

PowerPoint slides

Glossary

cLogD

The 'computed' LogD, using a predictive model. The values in this paper were calculated using AstraZeneca's proprietary model, AZlogD7.4.

Hill slope

A measure of binding or linkage cooperativity that can reflect a deviation from simple 1:1 stoichiometry (for which the Hill slope is equal to 1). The term is named in honour of A. V. Hill, the recipient of the 1922 Nobel Prize in Physiology or Medicine.

IC50

Inhibitor concentration at 50% effect. This is a generalized measure of inhibitory potency for a dose response, and dependent upon the conditions of the specific assay. Ideally, the relationship between this result and the intrinsic binding inhibition constant (Ki) may be understood from mechanism of inhibition studies.

LogD

The distribution coefficient, measured as the relative partitioning of all ionizable forms of a small molecule between a hydrophobic (octanol) and aqueous phase buffered to a particular pH, usually 7.4. This term describes the relative hydrophobicity of a chemical compound and is different to the related partition coefficient, logP, which describes the partitioning of only the neutral (non-ionized) form of the compound between phases.

Minimum inhibitory concentration

This measurement reflects the lowest concentration of compound that visibly inhibits the growth of an organism after overnight incubation.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tommasi, R., Brown, D., Walkup, G. et al. ESKAPEing the labyrinth of antibacterial discovery. Nat Rev Drug Discov 14, 529–542 (2015). https://doi.org/10.1038/nrd4572

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd4572

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