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:

Antibiotic resistance breakers: can repurposed drugs fill the antibiotic discovery void?

An Erratum to this article was published on 29 January 2016

This article has been updated

Key Points

  • Concern over antibiotic resistance is growing. Resistance of up to 50% has been reported in some regions, including resistance to carbapenems, our current last line of defence.

  • New classes of antibiotics are needed, particularly against Gram-negative bacteria. However, even if the scientific hurdles can be overcome, it could take decades before sufficient numbers of such antibiotics become available.

  • As an interim solution, antibiotic resistance could be 'broken' by co-administering appropriate non-antibiotic drugs with failing antibiotics.

  • Several marketed drugs that do not currently have antibacterial indications can directly kill bacteria, reduce the antibiotic minimum inhibitory concentration when used in combination with existing antibiotics, modulate host defence through effects on host innate immunity, particularly inflammation and autophagy, or a combination of these three.

  • This article discusses how such 'antibiotic resistance breakers' (ARBs) could contribute to reducing the antibiotic resistance problem, and analyses a priority list of candidates for further investigation.

Abstract

Concern over antibiotic resistance is growing, and new classes of antibiotics, particularly against Gram-negative bacteria, are needed. However, even if the scientific hurdles can be overcome, it could take decades for sufficient numbers of such antibiotics to become available. As an interim solution, antibiotic resistance could be 'broken' by co-administering appropriate non-antibiotic drugs with failing antibiotics. Several marketed drugs that do not currently have antibacterial indications can either directly kill bacteria, reduce the antibiotic minimum inhibitory concentration when used in combination with existing antibiotics and/or modulate host defence through effects on host innate immunity, in particular by altering inflammation and autophagy. This article discusses how such 'antibiotic resistance breakers' could contribute to reducing the antibiotic resistance problem, and analyses a priority list of candidates for further investigation.

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: Sites of antibacterial action and mechanisms of resistance.

Similar content being viewed by others

Change history

  • 29 January 2016

    The article contained incorrect information regarding the susceptible bacteria and clinical trial phase of the aztreonam–avibactam combination, and incorrect nomenclature for Salmonella enterica subsp. enterica serovar Typhimurium. These errors have been corrected in the online version of the article.

References

  1. Rice, L. B. Federal funding for the study of antimicrobial resistance in nosocomial pathogens: no ESKAPE. J. Infect. Dis. 197, 1079–1081 (2008).

    PubMed  Google Scholar 

  2. Woodford, N., Wareham, D. W., Guerra, B. & Teale, C. Carbapenemase-producing Enterobacteriaceae and non-Enterobacteriaceae from animals and the environment: an emerging public health risk of our own making? J. Antimicrob. Chemother. 69, 287–291 (2014).

    CAS  PubMed  Google Scholar 

  3. Davis, S. C. Infections and the rise of antimicrobial resistance. UK Government [online], (2015)

  4. Centers for Disease Contol and Prevention. Antibiotic resistance threats in the United States, 2013. CDC [online], (2013).

  5. Bassetti, M. & Righi, E. Eravacycline for the treatment of intra-abdominal infections. Expert Opin. Investigat. Drugs 23, 1575–1584 (2014).

    CAS  Google Scholar 

  6. Walkty, A. et al. In vitro activity of plazomicin against 5015 Gram-negative and Gram-positive clinical isolates obtained from patients in Canadian hospitals as part of the CANWARD study, 2011–2012. Antimicrob. Agents Chemother. 58, 2554–2563 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Zhanel, G. G. et al. Ceftazidime–avibactam: a novel cephalosporin/β-lactamase inhibitor combination. Drugs. 73, 159–177 (2013).

    CAS  PubMed  Google Scholar 

  8. Zhanel, G. G. et al. Ceftolozane/tazobactam: a novel cephalosporin/β-lactamase inhibitor combination with activity against multidrug-resistant gram-negative bacilli. Drugs. 74, 31–51 (2014).

    CAS  PubMed  Google Scholar 

  9. White, A. R. et al. Augmentin (amoxicillin/clavulanate) in the treatment of community-acquired respiratory tract infection: a review of the continuing development of an innovative antimicrobial agent. J. Antimicrob. Chemother. 53 (Suppl. 1), i3–i20 (2004).

    CAS  PubMed  Google Scholar 

  10. Prabhudesai, P. P. et al. The efficacy and safety of amoxicillin-clavulanic acid 1000/125mg twice daily extended release (XR) tablet for the treatment of bacterial community-acquired pneumonia in adults. J. Indian Med. Assoc. 109, 124–127 (2011).

    CAS  PubMed  Google Scholar 

  11. Coates, A. & Hu, Y. in Novel Antimicrobial Agents and Strategies Ch. 2 (eds Phoenix, D. A., Harris, F. & Dennison, S. R.) (Wiley, 2014).

    Google Scholar 

  12. Blair, J. M., Richmond, G. E. & Piddock, L. J. Multidrug efflux pumps in Gram-negative bacteria and their role in antibiotic resistance. Future Microbiol. 9, 1165–1177 (2014).

    CAS  PubMed  Google Scholar 

  13. Amsden, G. W. Anti-inflammatory effects of macrolides — an under-appreciated benefit in the treatment of community-acquired respiratory tract infections and chronic inflammatory pulmonary conditions? J. Antimicrob. Chemother. 55, 10–21 (2005).

    CAS  PubMed  Google Scholar 

  14. Kudoh, S. et al. Improvement of survival in patients with diffuse panbronchiolitis treated with low dose erythromycin. Amer. J. Resp. Crit. Care Med. 157, 1829–1832 (1998).

    CAS  Google Scholar 

  15. Kudoh, S. et al. Clinical effects of low-dose long-term erythromycin chemotherapy on diffuse panbronchiolitis. Nihon Kyobu Shikkan Gakkai Zasshi 25, 632–642 (in Japanese) (1987).

    CAS  PubMed  Google Scholar 

  16. Tateda, K. et al. Azithromycin inhibits quorum sensing in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 45, 1930–1933 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Molinari, G. et al. Inhibition of Pseudomonas aeruginosa virulence factors by subinhibitory concentrations of azithromycin and other macrolide antibiotics. J. Antimicrob. Chemother. 31, 681–688 (1993).

    CAS  PubMed  Google Scholar 

  18. Nguyen, T. et al. Potential role of macrolide antibiotics in the management of cystic fibrosis lung disease. Curr. Opin. Pulmonary Med. 8, 521–528 (2002).

    Google Scholar 

  19. Karabay, O. et al. A new effect of acetylsalicylic acid? Significantly lower prevalence of nasal carriage of Staphylococcus aureus among patients receiving orally administered acetylsalicylic acid. Infect. Control Hosp. Epidemiol. 27, 318–319 (2006).

    PubMed  Google Scholar 

  20. Sedlacek, M. et al. Aspirin treatment is associated with a significantly decreased risk of Staphylococcus aureus bacteremia in hemodialysis patients with tunneled catheters. Am. J. Kidney Dis. 49, 401–408 (2007).

    CAS  PubMed  Google Scholar 

  21. Mazumdar, K. et al. Diclofenac in the management of E. coli urinary tract infections. In Vivo 20, 613–619 (2006).

    CAS  PubMed  Google Scholar 

  22. Mazumdar, K. et al. The anti-inflammatory non-antibiotic helper compound diclofenac: an antibacterial drug target. Eur. J. Clin. Microbiol. Infect. Dis. 28, 881–891 (2009).

    CAS  PubMed  Google Scholar 

  23. Pongkorpsakol, P. et al. Inhibition of cAMP-activated intestinal chloride secretion by diclofenac: cellular mechanism and potential application in cholera. PLoS Negl. Trop. Dis. 8, e3119 (2014).

    PubMed  PubMed Central  Google Scholar 

  24. Vilaplana, C. et al. Ibuprofen therapy resulted in significantly decreased tissue bacillary loads and increased survival in a new murine experimental model of active tuberculosis. J. Infect. Dis. 208, 199–202 (2013).

    CAS  PubMed  Google Scholar 

  25. Eisen, D. P. et al. Low-dose aspirin and ibuprofen sterilizing effects on Mycobacterium tuberculosis suggest safe new adjuvant therapies for tuberculosis. J. Infect. Diseases 208, 1925–1927 (2013).

    CAS  Google Scholar 

  26. Cicerale, S., Lucas, L. J. & Keast, R. S. Antimicrobial, antioxidant and anti-inflammatory phenolic activities in extra virgin olive oil. Curr. Opin. Biotechnol. 23, 129–135 (2012).

    CAS  PubMed  Google Scholar 

  27. Pettengill, M. et al. Ivermectin inhibits growth of Chlamydia trachomatis in epithelial cells. PLoS ONE 7, e48456 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhang, X. et al. Ivermectin inhibits LPS-induced production of inflammatory cytokines and improves LPS-induced survival in mice. Inflamm. Res. 57, 524–529 (2008).

    CAS  PubMed  Google Scholar 

  29. Schlievert, P. M. et al. Effect of glycerol monolaurate on bacterial growth and toxin production. Antimicrob. Agents Chemother. 36, 626–631 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Projan, S. J. et al. Glycerol monolaurate inhibits the production of β-lactamase, toxic shock syndrome toxin-1, and other staphylococcal exoproteins by interfering with signal transduction. J. Bacteriol. 176, 4204–4209 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhao, X. et al. Activation of AMPK attenuates neutrophil proinflammatory activity and decreases the severity of acute lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 295, 497–504 (2008).

    Google Scholar 

  32. Tsoyi, K. et al. Metformin inhibits HMGB1 release in LPS-treated RAW 264.7 cells and increases survival rate of endotoxaemic mice. Br. J. Pharmacol. 162, 1498–1508 (2010).

    Google Scholar 

  33. Rogers, A. C. et al. Activation of AMPK inhibits cholera toxin stimulated chloride secretion in human and murine intestine. PLoS ONE 8, e69050 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Yuk, J. M. et al. Vitamin D3 induces autophagy in human monocytes/ macrophages via cathelicidin. Cell Host Microbe 6, 231–234 (2009).

    CAS  PubMed  Google Scholar 

  35. Montoya, D. et al. IL-32 is a molecular marker of a host defense network in human tuberculosis. Sci. Transl. Med. 20, 250 (2014).

    Google Scholar 

  36. Dittmar, W. et al. Microbiological laboratory studies with ciclopiroxolamine. Drug Res. 31, 1317–1322 (1981).

    CAS  Google Scholar 

  37. Carlson-Banning, K. M. et al. Toward repurposing Ciclopirox as an antibiotic against drug-resistant Acinetobacter baumannii, Escherichia coli, and Klebsiella pneumoniae. PLoS ONE 8, e69646 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Niewerth, M. et al. Ciclopirox olamine treatment affects the expression pattern of Candida albicans genes encoding virulence factors, iron metabolism proteins, and drug resistance factors. Antimicrob. Agents Chemother. 47, 1805–1817 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Dihazi, G. H. et al. Impact of the antiproliferative agent ciclopirox olamine treatment on stem cells proteome. World J. Stem Cells 5, 9–25 (2013).

    PubMed  PubMed Central  Google Scholar 

  40. Zhou, H. et al. Ciclopirox induces autophagy through reactive oxygen species-mediated activation of JNK signaling pathway. Oncotarget 5, 10140–10150 (2014).

    PubMed  PubMed Central  Google Scholar 

  41. Weir, S. J. et al. The repositioning of the anti-fungal agent ciclopirox olamine as a novel therapeutic agent for the treatment of haematologic malignancy. J. Clin. Pharm. Ther. 36, 128–134 (2011).

    CAS  PubMed  Google Scholar 

  42. Eberhard, Y. et al. Chelation of intracellular iron with the antifungal agent ciclopirox olamine induces cell death in leukemia and myeloma cells. Blood 114, 3064–3073 (2009).

    CAS  PubMed  Google Scholar 

  43. Kellner, H. M. et al. Pharmacokinetics and biotransformation of the antimycotic drug ciclopiroxolamine in animals and man after topical and systemic administration. Arzneimittelforschung 31, 1337–1353 (in German) (1981).

    CAS  PubMed  Google Scholar 

  44. Minden, M. D. et al. Oral ciclopirox olamine displays biological activity in a phase I study in patients with advanced hematologic malignancies. Am. J. Hematol. 89, 363–368 (2014).

    CAS  PubMed  Google Scholar 

  45. Ejim, L. et al. Combinations of antibiotics and nonantibiotic drugs enhance antimicrobial efficacy. Nat. Chem. Biol. 7, 348–350 (2011).

    CAS  PubMed  Google Scholar 

  46. Taylor. P. L. et al. A forward chemical screen identifies antibiotic adjuvants in Escherichia coli. ACS Chem. Biol. 7, 1547–1555 (2012).

    CAS  PubMed  Google Scholar 

  47. Tascini, C. et al. Synergistic activity of colistin plus rifampin against colistin-resistant KPC-producing Klebsiella pneumoniae. Antimicrob. Agents Chemother. 57, 3990–3993 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Yu, H.-H. et al. Antimicrobial activity of berberine alone and in combination with ampicillin or oxacillin against methicillin-resistant Staphylococcus aureus. J. Med. Food 8, 454–461 (2005).

    CAS  PubMed  Google Scholar 

  49. Kim, S.-H. et al. Inhibition of the bacterial surface protein anchoring transpeptidase sortase by isoquinoline alkaloids. Biosci. Biotechnol. Biochem. 68, 421–424 (2004).

    CAS  PubMed  Google Scholar 

  50. Domadia, P. N. Berberine targets assembly of Escherichia coli cell division protein FtsZ. Biochemistry 47, 3225–3234 (2008).

    CAS  PubMed  Google Scholar 

  51. Chu, M. et al. Role of berberine in anti-bacterial as a high-affinity LPS antagonist binding to TLR4/MD-2 receptor. BMC Complement. Altern. Med. 14, 89 (2014).

    PubMed  PubMed Central  Google Scholar 

  52. Jin, J. L. et al. Antibacterial mechanisms of berberine and reasons for little resistance of bacteria. Chinese Herbal Med. 3, 27–35 (2010).

    Google Scholar 

  53. Li, H.-M. et al. Berberine protects against lipopolysaccharide-induced intestinal injury in mice via α 2 adrenoceptor-independent mechanisms. Acta Pharmacol. Sin. 32, 1364–1372 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Jeong, H. W. et al. Berberine suppresses proinflammatory responses through AMPK activation in macrophages. Am. J. Physiol. Endocrinol. Metab. 296, 955–964 (2009).

    Google Scholar 

  55. Zhang, M. & Chen, L. Berberine in type 2 diabetes therapy: a new perspective for an old antidiarrheal drug? Acta Pharmaceutica Sinica B 2, 379–386 (2012).

    CAS  Google Scholar 

  56. Zhang, H. et al. Berberine lowers blood glucose in type 2 diabetes mellitus patients through increasing insulin receptor expression. Metabolism 59, 285–292 (2009).

    PubMed  Google Scholar 

  57. Yin, J., Xing, H. & Ye, J. Efficacy of berberine in patients with type 2 diabetes mellitus. Metabolism 57, 712–717 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Fürst, R. & Zündorf, I. Plant-derived anti-inflammatory compounds: hopes and disappointments regarding the translation of preclinical knowledge into clinical progress. Mediators Inflamm. 2014, 146832 (2014).

    PubMed  PubMed Central  Google Scholar 

  59. Gupta, S. C., Patchva, S. & Aggarwal, B. B. Therapeutic roles of curcumin: lessons learned from clinical trials. AAPS J. 15, 195–218 (2013).

    CAS  PubMed  Google Scholar 

  60. Moghadamtousi, S. Z. et al. A review on antibacterial, antiviral, and antifungal activity of curcumin. Biomed. Res. Int. 186864 (2014).

  61. Mahady, G. B. et al. Turmeric (Curcuma longa) and curcumin inhibit the growth of Helicobacter pylori, a group 1 carcinogen. Anticancer Res. 22 4179–4181 (2002).

    CAS  PubMed  Google Scholar 

  62. De, R. et al. Antimicrobial activity of curcumin against Helicobacter pylori isolates from India and during infections in mice. Antimicrob. Agents Chemother. 53, 1592–1597 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Aljamal, A. Effect of turmeric in peptic ulcer and H pylori. Plant Sci. Res. 3, 25–28 (2011).

    Google Scholar 

  64. Di Mario, F. et al. A curcumin-based 1-week triple therapy for eradication of Helicobacter pylori infection: something to learn from failure? Helicobacter 12, 238–243 (2007).

    CAS  PubMed  Google Scholar 

  65. Koosirirat, C. et al. Investigation of the antiinflammatory effect of Curcuma longa in Helicobacter pylori-infected patients. Int. Immunopharmacol. 10, 815–818 (2010).

    CAS  PubMed  Google Scholar 

  66. Patel, R. & Yang, N. Inhibiting hospital associated infection of toxigenic Clostridium difficile using natural spice-turmeric (curcumin). Amer. J. Gastroenterol. 105, S122–S122 (2010).

    Google Scholar 

  67. Sasidharan, N. K. et al. In vitro synergistic effect of curcumin in combination with third generation cephalosporins against bacteria associated with infectious diarrhea. Biomed. Res. Int. 2014, 561456 (2014).

    PubMed  PubMed Central  Google Scholar 

  68. Moghaddam, K. M. et al. The combination effect of curcumin with different antibiotics against Staphylococcus aureus. Int. J. Green Pharm. 3, 141–143 (2009).

    Google Scholar 

  69. Mun, S. H. et al. Synergistic antibacterial effect of curcumin against methicillin-resistant Staphylococcus aureus. Phytotherapy Research 19, 599–604 (2013).

    Google Scholar 

  70. Park, B. S. et al. Curcuma longa L. constituents inhibit sortase A and Staphylococcus aureus cell adhesion to fibronectin. J. Agr. Food Chem. 53, 9005–9009 (2005).

    CAS  Google Scholar 

  71. Aoki, H. et al. Evidence that curcumin suppresses the growth of malignant gliomas in vitro and in vivo through induction of autophagy: role of Akt and extracellular signal-regulated kinase signaling pathways. Mol. Pharmacol. 72, 29–39 (2007).

    CAS  PubMed  Google Scholar 

  72. Gradisar, H. et al. MD-2 as the target of curcumin in the inhibition of response to LPS. J. Leukocyte Biol. 82, 968–974 (2007).

    CAS  PubMed  Google Scholar 

  73. Tu, X.-K. et al. Curcumin inhibits TLR2/4-NF-κB signaling pathway and attenuates brain damage in permanent focal cerebral ischemia in rats. Inflammation 37, 1544–1551 (2014).

    CAS  PubMed  Google Scholar 

  74. Shuto, T. et al. Curcumin decreases toll-like receptor-2 gene expression and function in human monocytes and neutrophils. Biochem. Biophys. Res. Commun. 398, 647–652 (2010).

    CAS  PubMed  Google Scholar 

  75. Tu, C.-T. et al. Curcumin attenuates concanavalin A-induced liver injury in mice by inhibition of Toll-like receptor (TLR) 2, TLR4 and TLR9 expression. Intnl Immunopharmacol. 12, 151–157 (2012).

    CAS  Google Scholar 

  76. Chan, M. M. Inhibition of tumor necrosis factor by curcumin, a phytochemical. Biochem. Pharmacol. 49, 1551–1556 (1995).

    CAS  PubMed  Google Scholar 

  77. Chainani-Wu, N. Safety and anti-inflammatory activity of curcumin: a component of tumeric (Curcuma longa). J. Altern. Compl. Med. 9, 161–168 (2003).

    Google Scholar 

  78. Bengmark, S. Curcumin, an atoxic antioxidant and natural NFκB, cyclooxygenase-2, lipooxygenase, and inducible nitric oxide synthase inhibitor: a shield against acute and chronic diseases. J. Parenteral Enteral Nutr. 30, 45–51 (2006).

    CAS  Google Scholar 

  79. Jain, S. K. et al. Curcumin supplementation lowers TNF-α, IL-6, IL-8, and MCP-1 secretion in high glucose-treated cultured monocytes and blood levels of TNF-α, IL-6, MCP-1, glucose, and glycosylated hemoglobin in diabetic rats. Antioxid. Redox Signal. 11, 241–249 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Hansen, E. et al. A versatile high throughput screening system for the simultaneous identification of anti-inflammatory and neuroprotective compounds. J. Alzheimer's Disease 19, 451–464 (2010).

    CAS  Google Scholar 

  81. Ryan, A. et al. A role for TLR4 in Clostridium difficile infection and the recognition of surface layer proteins. PLoS Pathog. 7, e1002076 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Pothoulakis, C. Effects of Clostridium difficile toxins on epithelial cell barrier. Ann. NY Acad. Sci. 915, 347–356 (2000).

    CAS  PubMed  Google Scholar 

  83. Sintara, K. et al. Curcumin suppresses gastric NF-κB activation and macromolecular leakage in Helicobacter pylori-infected rats. World J. Gastroenterol. 16, 4039–4046 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Brennan, P. & O'Neill, L. A. Inhibition of nuclear factor κB by direct modification in whole cells — mechanism of action of nordihydroguaiaritic acid, curcumin and thiol modifiers. Biochem. Pharmacol. 55, 965–973 (1998).

    CAS  PubMed  Google Scholar 

  85. Steiner, T. S. et al. Faecal lactoferrin, interleukin 1b, and interleukin-8 are elevated in patients with severe Clostridium difficile colitis. Clin. Diagn. Lab. Immunol. 4, 719–722 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Jafari, N. V. et al. Clostridium difficile modulates host innate immunity via toxin-independent and dependent mechanism(s). PLoS ONE 8, e69846 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Rao, K. et al. The systemic inflammatory response to Clostridium difficile infection. PLoS ONE 9, e92578 (2014).

    PubMed  PubMed Central  Google Scholar 

  88. Feghaly, R. et al. Markers of intestinal inflammation, not bacterial burden, correlate with clinical outcomes in Clostridium difficile infection. Clin. Infect. Dis. 56, 1713–1721 (2013).

    PubMed  PubMed Central  Google Scholar 

  89. Basu, P. P. et al. Turmeric enema: a novel therapy for C. difficile colitis (CDAD): A randomized, double blinded, placebo controlled prospective clinical trial. Internat. J. Infectious Diseases 15 (Suppl. 15), S39 (2011).

    Google Scholar 

  90. Sharma, R. A. et al. Pharmacodynamic and pharmacokinetic study of oral curcuma extract in patients with colorectal cancer. Clin. Cancer Res. 7, 1894–1900 (2001).

    CAS  PubMed  Google Scholar 

  91. Lim, G. P. et al. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J. Neurosci. 21, 8370–8377 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Begum, A. N. et al. Curcumin structure function, bioavailability, and efficacy in models of neuroinflammation and Alzheimer's disease. J. Pharmacol. Exp. Ther. 326, 196–208 (2008).

    CAS  PubMed  Google Scholar 

  93. Yang, F. et al. Curcumin inhibits formation of amyloid-β oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J. Biol. Chem. 280, 5892–5901 (2005).

    CAS  PubMed  Google Scholar 

  94. McNaught, J. On the action of cold or lukewarm tea on Bacillus typhosus. J. R. Army Med. Corps 7, 372–373 (1906).

    Google Scholar 

  95. Steinmann, J. et al. Anti-infective properties of epigallocatechin-3-gallate (EGCG), a component of green tea. Br. J. Pharmacol. 168, 1059–1073 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Wolska, K. I., Grzes´, K. & Kurek, A. Synergy between novel antimicrobials and conventional antibiotics or bacteriocins. Pol. J. Microbiol. 61, 95–104 (2012).

    CAS  PubMed  Google Scholar 

  97. Yam, T. S., Hamilton-Miller, J. M. & Shah S. The effect of a component of tea (Camellia sinensis) on methicillin resistance, PBP2′ synthesis, and β-lactamase production in Staphylococcus aureus. J. Antimicrob. Chemother. 42, 211–216 (1998).

    CAS  PubMed  Google Scholar 

  98. Stapleton, P. D. et al. Modulation of β-lactam resistance in Staphylococcus aureus by catechins and gallates. Int. J. Antimicrob. Agents 23, 462–467 (2004).

    CAS  PubMed  Google Scholar 

  99. Zhao, W. et al. Mechanism of synergy between epigallocatechin gallate and β-lactams against methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 45, 1737–1742 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Hu, Z.-Q. et al. Epigallocatechin gallate synergy with ampicillin/sulbactam against 28 clinical isolates of methicillin-resistant Staphylococcus aureus. J. Antimicrob. Chemother. 48, 361–364 (2001).

    CAS  PubMed  Google Scholar 

  101. Hu, Z.-Q. et al. Epigallocatechin gallate synergistically enhances the activity of carbapenems against methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 46, 558–560 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Navarro-Martinez, M. D. et al. Antifolate activity of epigallocatechin gallate against Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 49, 2914–2920 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Lee, H. C. et al. Effect of tea phenolics and their aromatic fecal bacterial metabolites on intestinal microbiota. Res. Microbiol. 157, 876–884 (2006).

    CAS  PubMed  Google Scholar 

  104. Reygaert, W. & Jusufi, I. Green tea as an effective antimicrobial for urinary tract infections caused by Escherichia coli. Front. Microbiol. 4, 162 (2013).

    PubMed  PubMed Central  Google Scholar 

  105. Li, W. et al. A major ingredient of green tea rescues mice from lethal sepsis partly by inhibiting HMGB1. PLoS ONE 2, e1153 (2007).

    PubMed  PubMed Central  Google Scholar 

  106. Zhao, W.-H. et al. Inhibition of penicillinase by epigallocatechin gallate resulting in restoration of antibacterial activity of penicillin against penicillinase-producing Staphylococcus aureus. Antimicrob. Agents Chemother. 46, 2266–2268 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Stapleton, P. D. et al. The β-lactam-resistance modifier (−)-epicatechin gallate alters the architecture of the cell wall of Staphylococcus aureus. Microbiology 153, 2093–2103 (2007).

    CAS  PubMed  Google Scholar 

  108. Grandišar, H. et al. Green tea catechins inhibit bacterial DNA gyrase by interaction with its ATP binding site. J. Med. Chem. 50, 264–271 (2007).

    Google Scholar 

  109. Zhang, Y. M. & Rock, C. O. Evaluation of epigallocatechin gallate and related plant polyphenols as inhibitors of the FabG and FabI reductases of bacterial type II fatty-acid synthesis. J. Biol. Chem. 279, 30994–31001 (2004).

    CAS  PubMed  Google Scholar 

  110. Lee, K. M. et al. Protective mechanism of epigallocatechin-3-gallate against Helicobacter pylori-induced gastric epithelial cytotoxicity via the blockage of TLR-4 signaling. Helicobacter 9, 632–642 (2004).

    CAS  PubMed  Google Scholar 

  111. Zhao, W. H. et al. Inhibition by epigallocatechin gallate (EGCG) of conjugative R plasmid transfer in Escherichia coli. J. Infect. Chemother. 7, 195–197 (2001).

    CAS  PubMed  Google Scholar 

  112. Sudano Roccaro, A. et al. Epigallocatechin-gallate enhances the activity of tetracycline in staphylococci by inhibiting its efflux from bacterial cells. Antimicrob. Agents Chemother. 48, 1968–1973 (2004).

    PubMed  Google Scholar 

  113. Li, W. et al. EGCG stimulates autophagy and reduces cytoplasmic HMGB1 levels in endotoxin-stimulated macrophages. Biochem. Pharmacol. 81, 1152–1163 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Kim, H. S. et al. Epigallocatechin gallate (EGCG) stimulates autophagy in vascular endothelial cells: a potential role for reducing lipid accumulation. J. Biol. Chem. 288, 22693–22705 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Zhou, J. et al. Epigallocatechin-3-gallate (EGCG), a green tea polyphenol, stimulates hepatic autophagy and lipid clearance. PLoS ONE 9, e87161 (2014).

    PubMed  PubMed Central  Google Scholar 

  116. Ullmann, U. et al. A single ascending dose study of epigallocatechin gallate in healthy volunteers. J. Int. Med. Res. 31, 88–101 (2003).

    CAS  PubMed  Google Scholar 

  117. Lambert, J. D. et al. Peracetylation as a means of enhancing in vitro bioactivity and bioavailability of epigallocatechin-3-gallate. Drug Metab. Dispos. 34, 2111–2116 (2006).

    CAS  PubMed  Google Scholar 

  118. Matsumoto, Y. et al. Antibacterial and antifungal activities of new acylated derivatives of epigallocatechin gallate. Front. Microbiol. 3, 53 (2012).

    PubMed  PubMed Central  Google Scholar 

  119. Hutchinson, M. R. et al. Evidence that opioids may have toll like receptor 4 and MD-2 effects. Brain Behav. Immun. 24, 83–95 (2010).

    CAS  PubMed  Google Scholar 

  120. Hutchinson, M. R. et al. Opioid activation of toll-like receptor 4 contributes to drug reinforcement. J. Neurosci. 32, 11187–11200 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Dawson, A. in Medical Toxicology 3rd edn (ed. Dart, R.) 228–230 (Lippincott, Williams and Wilkins, 2004).

    Google Scholar 

  122. Clifton, L. A. et al. Effect of divalent cation removal on the structure of Gram-negative bacterial outer membrane models. Langmuir 31, 404–412 (2015).

    CAS  PubMed  Google Scholar 

  123. Gill, E. E., Franco, O. L. & Hancock, R. E. Antibiotic adjuvants: diverse strategies for controlling drug-resistant pathogens. Chem. Biol. Drug Des. 85, 56–78 (2015).

    CAS  PubMed  Google Scholar 

  124. Chauhan, A. et al. Full and broad-spectrum in vivo eradication of catheter-associated biofilms using gentamicin-EDTA antibiotic lock therapy. Antimicrob. Agents Chemother. 56, 6310–6318 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Deretic, V. Autophagy in immunity and cell-autonomous defense against intracellular microbes. Immunol. Rev. 240, 92–104 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Campoy, E. & Colombo, M. I. Autophagy in intracellular bacterial infection. Biochim. Biophys. Acta 1793, 1465–1477 (2009).

    CAS  PubMed  Google Scholar 

  127. Nakagawa, I. et al. Autophagy defends cells against invading group A Streptococcus. Science 306, 1037–1040 (2004).

    CAS  PubMed  Google Scholar 

  128. Birmingham, C. L. et al. Autophagy controls salmonella infection in response to damage to the salmonella-containing vacuole. J. Biol. Chem. 281, 11374–11383 (2006).

    CAS  PubMed  Google Scholar 

  129. Ogawa, M. et al. Escape of intracellular Shigella from autophagy. Science 307, 727–731 (2005).

    CAS  PubMed  Google Scholar 

  130. Yano, T. et al. Autophagic control of Listeria through intracellular innate immune recognition in drosophila. Nat. Immunol. 9, 908–916 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Gutierrez, M. G. et al. Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell 119, 753–766 (2004).

    CAS  PubMed  Google Scholar 

  132. Amano, A., Nakagawa, I. & Yoshimori, T. Autophagy in innate immunity against intracellular bacteria. J. Biochem. 140, 161–166 (2006).

    CAS  PubMed  Google Scholar 

  133. Vergne, I. et al. Autophagy in immune defense against Mycobacterium tuberculosis. Autophagy 2, 175–178 (2006).

    CAS  PubMed  Google Scholar 

  134. Mostowy, S. Autophagy and bacterial clearance: a not so clear picture. Cell. Microbiol. 15, 395–402 (2013).

    CAS  PubMed  Google Scholar 

  135. Kuballa, P. et al. Autophagy and the immune system. Annu. Rev. Immunol. 30, 611–646 (2012).

    CAS  PubMed  Google Scholar 

  136. Levine, B., Mizushima, N. & Virgin, H. W. Autophagy in immunity and inflammation. Nature 469, 323–335 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Poels, J. et al. Expanding roles for AMP-activated protein kinase in neuronal survival and autophagy. Bioessays 31, 944–952 (2009).

    CAS  PubMed  Google Scholar 

  138. Inoki, K. et al. TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell 126, 955–968 (2006).

    CAS  PubMed  Google Scholar 

  139. Ulgherait, M. et al. AMPK modulates tissue and organismal aging in a non-cell-autonomous manner. Cell Rep. 8, 1767–1780 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Wang, W. H. et al. Aspirin inhibits the growth of Helicobacter pylori and enhances its susceptibility to antimicrobial agents. Gut 52, 490–495 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Price, C. T. et al. The effects of salicylate on bacteria. Internat. J. Biochem. Cell Biol. 32, 1029–1043 (2000).

    CAS  Google Scholar 

  142. Nicolau, D. P. et al. Influence of aspirin on development and treatment of experimental Staphylococcus aureus endocarditis. Antimicrob. Agents Chemother. 39, 1748–1751 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Nicolau, D. P. et al. Reduction of bacterial titers by low-dose aspirin in experimental aortic valve endocarditis. Infect. Immun. 61, 1593–1595 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Dutta, N. K. et al. The anti-inflammatory drug diclofenac retains anti-listerial activity in vivo. Lett. Appl. Microbiol. 47, 106–111 (2008).

    CAS  PubMed  Google Scholar 

  145. Dutta, N. K. et al. Potential management of resistant microbial infections with a novel non-antibiotic: the anti-inflammatory drug diclofenac sodium. Int. J. Antimicrob. Agents 30, 242–249 (2007).

    CAS  PubMed  Google Scholar 

  146. Dutta, N. K. et al. Activity of diclofenac used alone and in combination with streptomycin against Mycobacterium tuberculosis in mice. Int. J. Antimicrob. Agents 30, 336–340 (2007).

    CAS  PubMed  Google Scholar 

  147. Zhang, X. et al. Inhibitory effects of ivermectin on nitric oxide and prostaglandin E2 production in LPS-stimulated RAW 264.7 macrophages. Int. Immunopharmacol. 9, 354–359 (2009).

    CAS  PubMed  Google Scholar 

  148. Bae, H.-B. et al. AMP-activated protein kinase enhances the phagocytic ability of macrophages and neutrophils. FASEB J. 25, 4358–4368 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Singhal, A. et al. Metformin as adjunct antituberculosis therapy. Sci. Transl. Med. 6, 263ra159 (2014).

    PubMed  Google Scholar 

  150. Maeurer, M. & Zumla, A. The host battles drug-resistant tuberculosis. Sci. Transl. Med. 6, 263fs47 (2014).

    PubMed  Google Scholar 

  151. Salahuddin, N. et al. Vitamin D accelerates clinical recovery from tuberculosis: results of the SUCCINCT Study [Supplementary Cholecalciferol in recovery from tuberculosis]. A randomized, placebo-controlled clinical trial of vitamin D supplementation in patients with pulmonary tuberculosis. BMC Infect. Dis. 13, 22 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Anand, P. K. & Kaul, D. Vitamin D3-dependent pathway regulates TACO gene transcription. Biochem. Biophys. Res. Commun. 310, 876–877 (2003).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The author thanks the following for expert discussions on the drugs reviewed: A. Coates (clinical antibiotic resistance and ARB concept); S. Shaunak (clinical antibiotic resistance, TLRs and innate immune system); N. Ktistakis (autophagy); D. Cavalla (drug repurposing); and members of the Science and Technology Advisory Committee of Antibiotics Research UK.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to David Brown.

Ethics declarations

Competing interests

The author declares no competing financial interests.

Related links

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brown, D. Antibiotic resistance breakers: can repurposed drugs fill the antibiotic discovery void?. Nat Rev Drug Discov 14, 821–832 (2015). https://doi.org/10.1038/nrd4675

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd4675

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