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.

Inhaled bacteriophage-loaded polymeric microparticles ameliorate acute lung infections

Abstract

Lung infections associated with pneumonia, or cystic fibrosis caused by Pseudomonas aeruginosa or other bacteria, result in significant morbidity and mortality, in part owing to the development of multidrug resistance, also against last-resort antibiotics. Lytic bacteriophages (that is, viruses that specifically kill bacteria) can reduce lung-associated infections, yet their clinical use is hindered by difficulties in delivering active phages to the deep lung. Here, we show that phage-loaded polymeric microparticles deposit throughout the lung via dry powder inhalation and that they deliver active phages. Phage-loaded microparticles effectively reduced P. aeruginosa infections and the associated inflammation in wild-type and cystic fibrosis transmembrane-conductance-regulator knockout mice, and rescued the mice from pneumonia-associated death. These polymeric microparticles might constitute a clinically translatable therapy for eradicating hospital-acquired lung infections and infections associated with cystic fibrosis.

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

Fig. 1: Porous MPs deliver active phages and are effective against P. aeruginosa.
Fig. 2: Phage-MPs are effective against P. aeruginosa biofilms.
Fig. 3: Dry powder formulations of porous MPs for lung delivery.
Fig. 4: Phage-loaded porous MPs reduce bacteria in the lungs of mice.
Fig. 5: Phage-loaded porous MPs reduce bacteria in the lungs of CFKO mice.
Fig. 6: Phage-MPs significantly reduce infection by a clinically derived strain of bacteria in wild-type mice.
Fig. 7: Pretreatment with phage-MPs does not reduce the functional efficacy of the treatment in clearing bacteria from mouse lungs.

References

  1. Niederman, M. S. Challenges in the management of community-acquired pneumonia: the role of quinolones and moxifloxacin. Clin. Infect. Dis. 41, S158–S166 (2005).

    Article  CAS  Google Scholar 

  2. Klevens, R. M. et al. Estimating health care-associated infections and deaths in U.S. hospitals, 2002. Public Health Rep. 122, 160–166 (2007).

    Article  Google Scholar 

  3. Patient Registry 2015 Annual Data Report (Cystic Fibrosis Foundation, 2015).

  4. Cohen, T. S. & Prince, A. Cystic fibrosis: a mucosal immunodeficiency syndrome. Nat. Med. 18, 509–519 (2012).

    Article  CAS  Google Scholar 

  5. Ciofu, O., Tolker-Nielsen, T., Jensen, P. O., Wang, H. & Hoiby, N. Antimicrobial resistance, respiratory tract infections and role of biofilms in lung infections in cystic fibrosis patients. Adv. Drug Deliv. Rev. 85, 7–23 (2015).

    Article  CAS  Google Scholar 

  6. Magill, S. S. et al. Multistate point-prevalence survey of health care-associated infections. N. Engl. J. Med. 370, 1198–1208 (2014).

    Article  CAS  Google Scholar 

  7. Weiner, L. M. et al. Antimicrobial-resistant pathogens associated with healthcare-associated infections: summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2011–2014. Infect. Control Hosp. Epidemiol. 37, 1288–1301 (2016).

    Article  Google Scholar 

  8. Johansen, H. K., Moskowitz, S. M., Ciofu, O., Pressler, T. & Hoiby, N. Spread of colistin resistant non-mucoid Pseudomonas aeruginosa among chronically infected Danish cystic fibrosis patients. J. Cyst. Fibros. 7, 391–397 (2008).

    Article  Google Scholar 

  9. Gould, I. M. The epidemiology of antibiotic resistance. Int. J. Antimicrob. Agents 32, S2–S9 (2008).

    Article  CAS  Google Scholar 

  10. Gomez, M. I. & Prince, A. Opportunistic infections in lung disease: Pseudomonas infections in cystic fibrosis. Curr. Opin. Pharmacol. 7, 244–251 (2007).

    Article  CAS  Google Scholar 

  11. Ciofi Degli Atti, M. et al. An outbreak of extremely drug-resistant Pseudomonas aeruginosa in a tertiary care pediatric hospital in Italy. BMC Infect. Dis. 14, 494 (2014).

    Article  Google Scholar 

  12. Sprenger, M. & Fukuda, K. Antimicrobial resistance. New mechanisms, new worries. Science 351, 1263–1264 (2016).

    Article  CAS  Google Scholar 

  13. Donlan, R. M. Preventing biofilms of clinically relevant organisms using bacteriophage. Trends Microbiol. 17, 66–72 (2009).

    Article  CAS  Google Scholar 

  14. Servick, K. Drug development. Beleaguered phage therapy trial presses on. Science 352, 1506 (2016).

    Article  CAS  Google Scholar 

  15. Geier, M. R., Trigg, M. E. & Merril, C. R. Fate of bacteriophage lambda in non-immune germ-free mice. Nature 246, 221–223 (1973).

    Article  CAS  Google Scholar 

  16. Bruttin, A. & Brüssow, H. Human volunteers receiving Escherichia coli phage T4 orally: a safety test of phage therapy. Antimicrob. Agents Chemother. 49, 2874–2878 (2005).

    Article  CAS  Google Scholar 

  17. Hughes, K. A., Sutherland, I. W., Clark, J. & Jones, M. V. Bacteriophage and associated polysaccharide depolymerases—novel tools for study of bacterial biofilms. J. Appl. Microbiol. 85, 583–590 (1998).

    Article  CAS  Google Scholar 

  18. Biswas, B. et al. Bacteriophage therapy rescues mice bacteremic from a clinical isolate of vancomycin-resistant Enterococcus faecium. Infect. Immun. 70, 204–210 (2002).

    Article  CAS  Google Scholar 

  19. Chanishvili, N., Chanishvili, T., Tediashvili, M. & Barrow, P. A. Phages and their application against drug-resistant bacteria. J. Chem. Technol. Biotechnol. 76, 689–699 (2001).

    Article  CAS  Google Scholar 

  20. Wright, A., Hawkins, C. H., Änggård, E. E. & Harper, D. R. A controlled clinical trial of a therapeutic bacteriophage preparation in chronic otitis due to antibiotic-resistant Pseudomonas aeruginosa; a preliminary report of efficacy. Clin. Otolaryngol. 34, 349–357 (2009).

    Article  CAS  Google Scholar 

  21. Cho, I. & Blaser, M. J. The human microbiome: at the interface of health and disease. Nat. Rev. Genet. 13, 260–270 (2012).

    Article  CAS  Google Scholar 

  22. Dufour, N., Delattre, R., Ricard, J. D. & Debarbieux, L. The lysis of pathogenic Escherichia coli by bacteriophages releases less endotoxin than by β-lactams. Clin. Infect. Dis. 64, 1582–1588 (2017).

    Article  Google Scholar 

  23. Lu, T. K. & Collins, J. J. Dispersing biofilms with engineered enzymatic bacteriophage. Proc. Natl Acad. Sci. USA 104, 11197–11202 (2007).

    Article  CAS  Google Scholar 

  24. Bikard, D. et al. Exploiting CRISPR–Cas nucleases to produce sequence-specific antimicrobials. Nat. Biotechnol. 32, 1146–1150 (2014).

    Article  CAS  Google Scholar 

  25. Citorik, R. J., Mimee, M. & Lu, T. K. Sequence-specific antimicrobials using efficiently delivered RNA-guided nucleases. Nat. Biotechnol. 32, 1141–1145 (2014).

    Article  CAS  Google Scholar 

  26. Yosef, I., Manor, M., Kiro, R. & Qimron, U. Temperate and lytic bacteriophages programmed to sensitize and kill antibiotic-resistant bacteria. Proc. Natl Acad. Sci. USA 112, 7267–7272 (2015).

    Article  CAS  Google Scholar 

  27. Schooley, R. T. et al. Development and use of personalized bacteriophage-based therapeutic cocktails to treat a patient with a disseminated resistant Acinetobacter baumannii infection. Antimicrob. Agents Chemother. 61, e00954-17 (2017).

    Article  Google Scholar 

  28. Jennes, S. et al. Use of bacteriophages in the treatment of colistin-only-sensitive Pseudomonas aeruginosa septicaemia in a patient with acute kidney injury—a case report. Crit. Care 21, 129 (2017).

    Article  Google Scholar 

  29. Pabary, R. et al. Antipseudomonal bacteriophage reduces infective burden and inflammatory response in murine lung. Antimicrob. Agents Chemother. 60, 744–751 (2016).

    Article  CAS  Google Scholar 

  30. Debarbieux, L. et al. Bacteriophages can treat and prevent Pseudomonas aeruginosa lung infections. J. Infect. Dis. 201, 1096–1104 (2010).

    Article  CAS  Google Scholar 

  31. Morello, E. et al. Pulmonary bacteriophage therapy on Pseudomonas aeruginosa cystic fibrosis strains: first steps towards treatment and prevention. PLoS ONE 6, e16963 (2011).

    Article  CAS  Google Scholar 

  32. Golshahi, L., Seed, K. D., Dennis, J. J. & Finlay, W. H. Toward modern inhalational bacteriophage therapy: nebulization of bacteriophages of Burkholderia cepacia complex. J. Aerosol Med. Pulm. Drug Deliv. 21, 351–360 (2008).

    Article  CAS  Google Scholar 

  33. Hraiech, S., Bregeon, F. & Rolain, J. M. Bacteriophage-based therapy in cystic fibrosis-associated Pseudomonas aeruginosa infections: rationale and current status. Drug Des. Devel. Ther. 9, 3653–3663 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Yang, M. et al. Therapeutic effect of the YH6 phage in a murine hemorrhagic pneumonia model. Res. Microbiol. 166, 633–643 (2015).

    Article  CAS  Google Scholar 

  35. Sahota, J. S. et al. Bacteriophage delivery by nebulization and efficacy against phenotypically diverse Pseudomonas aeruginosa from cystic fibrosis patients. J. Aerosol Med. Pulm. Drug Deliv. 28, 353–360 (2015).

    Article  CAS  Google Scholar 

  36. Saussereau, E. et al. Effectiveness of bacteriophages in the sputum of cystic fibrosis patients. Clin. Microbiol. Infect. 20, O983–O990 (2014).

    Article  CAS  Google Scholar 

  37. Saussereau, E. & Debarbieux, L. Bacteriophages in the experimental treatment of Pseudomonas aeruginosa infections in mice. Adv. Virus Res. 83, 123–141 (2012).

    Article  CAS  Google Scholar 

  38. Alemayehu, D. et al. Bacteriophages phiMR299-2 and phiNH-4 can eliminate Pseudomonas aeruginosa in the murine lung and on cystic fibrosis lung airway cells. MBio 3, e00029–e00012 (2012).

    Article  CAS  Google Scholar 

  39. Roach, D. R. et al. Synergy between the host immune system and bacteriophage is essential for successful phage therapy against an acute respiratory pathogen. Cell Host Microbe 22, 38–47.e34 (2017).

    Article  CAS  Google Scholar 

  40. Golshahi, L., Lynch, K. H., Dennis, J. J. & Finlay, W. H. In vitro lung delivery of bacteriophages KS4-M and PhiKZ using dry powder inhalers for treatment of Burkholderia cepacia complex and Pseudomonas aeruginosa infections in cystic fibrosis. J. Appl. Microbiol. 110, 106–117 (2011).

    Article  CAS  Google Scholar 

  41. Puapermpoonsiri, U., Spencer, J. & Van der Walle, C. F. A freeze-dried formulation of bacteriophage encapsulated in biodegradable microspheres. Eur. J. Pharm. Biopharm. 72, 26–33 (2009).

    Article  CAS  Google Scholar 

  42. Matinkhoo, S., Lynch, K. H., Dennis, J. J., Finlay, W. H. & Vehring, R. Spray-dried respirable powders containing bacteriophages for the treatment of pulmonary infections. J. Pharm. Sci. 100, 5197–5205 (2011).

    Article  CAS  Google Scholar 

  43. Vandenheuvel, D. et al. Feasibility of spray drying bacteriophages into respirable powders to combat pulmonary bacterial infections. Eur. J. Pharm. Biopharm. 84, 578–582 (2013).

    Article  CAS  Google Scholar 

  44. Leung, S. S. Y. et al. Effects of storage conditions on the stability of spray dried, inhalable bacteriophage powders. Int. J. Pharm. 521, 141–149 (2017).

    Article  CAS  Google Scholar 

  45. Meenach, S. A. et al. Design, physicochemical characterization, and optimization of organic solution advanced spray-dried inhalable dipalmitoylphosphatidylcholine (DPPC) and dipalmitoylphosphatidylethanolamine poly(ethylene glycol) (DPPE-PEG) microparticles and nanoparticles for targeted respiratory nanomedicine delivery as dry powder inhalation aerosols. Int. J. Nanomed. 8, 275–293 (2013).

    Google Scholar 

  46. Edwards, D. A. et al. Large porous particles for pulmonary drug delivery. Science 276, 1868–1871 (1997).

    Article  CAS  Google Scholar 

  47. Champion, J. A., Walker, A. & Mitragotri, S. Role of particle size in phagocytosis of polymeric microspheres. Pharm. Res. 25, 1815–1821 (2008).

    Article  CAS  Google Scholar 

  48. Ungaro, F. et al. Insulin-loaded PLGA/cyclodextrin large porous particles with improved aerosolization properties: in vivo deposition and hypoglycaemic activity after delivery to rat lungs. J. Control. Release 135, 25–34 (2009).

    Article  CAS  Google Scholar 

  49. Ungaro, F. et al. Engineering gas-foamed large porous particles for efficient local delivery of macromolecules to the lung. Eur. J. Pharm. Sci. 41, 60–70 (2010).

    Article  CAS  Google Scholar 

  50. Dhanda, D. S., Tyagi, P., Mirvish, S. S. & Kompella, U. B. Supercritical fluid technology based large porous celecoxib-PLGA microparticles do not induce pulmonary fibrosis and sustain drug delivery and efficacy for several weeks following a single dose. J. Control. Release 168, 239–250 (2013).

    Article  CAS  Google Scholar 

  51. d’Angelo, I. et al. Large porous particles for sustained release of a decoy oligonucelotide and poly(ethylenimine): potential for combined therapy of chronic Pseudomonas aeruginosa lung infections. Biomacromolecules 17, 1561–1571 (2016).

    Article  Google Scholar 

  52. De Stefano, D. et al. A decoy oligonucleotide to NF-κB delivered through inhalable particles prevents LPS-induced rat airway inflammation. Am. J. Respir. Cell Mol. Biol. 49, 288–295 (2013).

    Article  CAS  Google Scholar 

  53. Yang, Y. et al. Development of highly porous large PLGA microparticles for pulmonary drug delivery. Biomaterials 30, 1947–1953 (2009).

    Article  CAS  Google Scholar 

  54. Diraviam Dinesh, S. Artificial sputum medium. Protoc. Exch. https://doi.org/10.1038/protex.2010.212 (2010).

  55. Safdar, N., Crnich, C. J. & Maki, D. G. The pathogenesis of ventilator-associated pneumonia: its relevance to developing effective strategies for prevention. Respir. Care 50, 725–739 (2005).

    PubMed  Google Scholar 

  56. Makadia, H. K. & Siegel, S. J. Poly lactic-co-glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers 3, 1377–1397 (2011).

    Article  CAS  Google Scholar 

  57. Zhou, L. et al. Correction of lethal intestinal defect in a mouse model of cystic fibrosis by human CFTR. Science 266, 1705–1708 (1994).

    Article  CAS  Google Scholar 

  58. Poschet, J. F. et al. Molecular basis for defective glycosylation and Pseudomonas pathogenesis in cystic fibrosis lung. Proc. Natl Acad. Sci. USA 98, 13972–13977 (2001).

    Article  CAS  Google Scholar 

  59. Pier, G. B. Role of the cystic fibrosis transmembrane conductance regulator in innate immunity to Pseudomonas aeruginosa infections. Proc. Natl Acad. Sci. USA 97, 8822–8828 (2000).

    Article  CAS  Google Scholar 

  60. Liu, P. V. The roles of various fractions of Pseudomonas aeruginosa in its pathogenesis: II. Effects of lecithinase and protease. J. Infect. Dis. 116, 112–116 (1966).

    Article  CAS  Google Scholar 

  61. Foweraker, J. E., Laughton, C. R., Brown, D. F. & Bilton, D. Phenotypic variability of Pseudomonas aeruginosa in sputa from patients with acute infective exacerbation of cystic fibrosis and its impact on the validity of antimicrobial susceptibility testing. J. Antimicrob. Chemother. 55, 921–927 (2005).

    Article  CAS  Google Scholar 

  62. Winstanley, C., O’Brien, S. & Brockhurst, M. A. Pseudomonas aeruginosa evolutionary adaptation and diversification in cystic fibrosis chronic lung infections. Trends Microbiol. 24, 327–337 (2016).

    Article  CAS  Google Scholar 

  63. Waters, E. M. et al. Phage therapy is highly effective against chronic lung infections with Pseudomonas aeruginosa. Thorax 72, 666–667 (2017).

    Article  Google Scholar 

  64. Singh, N., Vats, A., Sharma, A., Arora, A. & Kumar, A. The development of lower respiratory tract microbiome in mice. Microbiome 5, 61 (2017).

    Article  Google Scholar 

  65. Barfod, K. K. et al. The murine lung microbiome changes during lung inflammation and intranasal vancomycin treatment. Open Microbiol. J. 9, 167–179 (2015).

    Article  CAS  Google Scholar 

  66. Lehman, S. M. & Donlan, R. M. Bacteriophage-mediated control of a two-species biofilm formed by microorganisms causing catheter-associated urinary tract infections in an in vitro urinary catheter model. Antimicrob. Agents Chemother. 59, 1127–1137 (2015).

    Article  Google Scholar 

  67. Adriaenssens, E. M. et al. CIM(®) monolithic anion-exchange chromatography as a useful alternative to CsCl gradient purification of bacteriophage particles. Virology 434, 265–270 (2012).

    Article  CAS  Google Scholar 

  68. Adams, M. H. Bacteriophages (Interscience, New York, 1959).

Download references

Acknowledgements

This work was funded by the National Institutes of Health (R01 AR062920 (to A.J.G.), F30 AR069472 (to C.T.J.) and S10 OD016264 (to A.J.G.) and a research partnership between Children’s Healthcare of Atlanta and the Georgia Institute of Technology. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or the Centers for Disease Control and Prevention. Use of trade names and commercial sources is for identification only and does not imply endorsement by the Public Health Service or the US Department of Health and Human Services. We thank R. Hunt for advice and discussions over the course of the project. We also thank DFE Pharma for providing inhalation grade lactose Respitose ML006. Clinical strains were obtained from the Clinical and Translational Research Core of the CF@LANTA RDP Center, funded by the Cystic Fibrosis Foundation (MCCART15R0). PA103 was a gift from J. Goldberg at Emory University.

Author information

Authors and Affiliations

Authors

Contributions

R.A., C.T.J., R.M.D., N.A.M. and A.J.G conceived and designed the experiments. R.A., C.T.J. and B.R.I. performed all the experiments. R.M.D. provided phage samples and advice on their use. B.R.I. and N.A.M. handled the cystic fibrosis knockout mice breeding, testing and care. The manuscript was written by R.A., N.A.M. and A.J.G. All authors discussed the results and reviewed the manuscript.

Corresponding author

Correspondence to Andrés J. García.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Supplementary Information

Supplementary figures and tables.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Agarwal, R., Johnson, C.T., Imhoff, B.R. et al. Inhaled bacteriophage-loaded polymeric microparticles ameliorate acute lung infections. Nat Biomed Eng 2, 841–849 (2018). https://doi.org/10.1038/s41551-018-0263-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-018-0263-5

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