Loading metrics

Open Access

Open Highlights

Open Highlights present themed summaries of content from across the PLOS journals and other Open Access sources.

See all article types »

Understanding and overcoming antibiotic resistance

* E-mail: [email protected]

Affiliation Public Library of Science, San Francisco, California, United States of America

ORCID logo

  • Lauren A. Richardson

PLOS

Published: August 23, 2017

  • https://doi.org/10.1371/journal.pbio.2003775
  • Reader Comments

Fig 1

Citation: Richardson LA (2017) Understanding and overcoming antibiotic resistance. PLoS Biol 15(8): e2003775. https://doi.org/10.1371/journal.pbio.2003775

Copyright: © 2017 Lauren A. Richardson. This is an open access article distributed under the terms of the Creative Commons Attribution License , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Competing interests: LR is a current paid employee at Public Library of Science.

Provenance: Written by editorial staff; not externally peer reviewed

Antibiotic drugs have revolutionized medicine and made our modern way of life possible. In addition to their essential role in the clinic, antibiotics are used in a huge array of non-medical applications, from promoting growth in livestock, to preserving building materials from contamination, to treating blight in orchards. However, overuse threatens their efficacy due to the promotion and spread of antibiotic resistant bacteria.

Antibiotics target and inhibit essential cellular processes, retarding growth and causing cell death. However, if bacteria are exposed to drugs below the dose required to kill all bacteria in a population (the minimum bactericidal concentration or MBC), they can mutate and resist antibiotic treatment via natural selection for resistance-conferring mutations. These genetic mutations can arise from the adoption of a plasmid encoding a resistance gene or by mutation to the bacterial chromosome itself.

The concern around the increasing prevalence of drug resistant bacteria is compounded by the fact that the discovery of new antibiotics is a fleeting rare event. Most classes of antibiotics on the market were discovered in the mid-to-late 20 th century. Thus, there is a limited arsenal of drugs to fight resistant bacteria, and bacteria can be resistant to multiple drugs at a time.

Given the importance of antibiotics to modern medicine, and the growing apprehension surrounding the threat of resistance, scientists are studying every aspect of antibiotic resistance. This Open Highlight features some of the cutting-edge research from the Open Access corpus on three major areas of focus: the cellular mechanisms of resistance, the evolution and spread of resistance, and techniques for combating resistance.

Mechanisms of Resistance

A common mechanism used by bacteria to minimize the effects of antibiotics is to acquire or increase the expression of drug efflux pumps. As the name implies, these pumps expel drugs from the cytoplasm, limiting their ability to access their target. In a PLOS Pathogens article, researchers investigated how efflux pump expression is regulated in the human pathogen Pseudomonas aeruginosa [ 1 ]. They found that the multifaceted transcription regulator CpxR regulates the expression of the major efflux pump in P . aeruginosa , and is involved in modulating resistance in clinical isolates.

Resistance-conferring mutations can be specific to a particular antibiotic or they can provide protection to multiple—often related—drugs. A recent eLife article described a surprising set of mutations in the genes encoding components of the ribosome of Mycobacterium smegmatis that confer resistance to numerous antibiotics that are not related structurally or mechanistically [ 2 ]. The authors find that these mutations cause extensive changes in the transcriptome and proteome of the bacterium, including alterations to several proteins known to impact resistance. Importantly, these mutations promote further evolution of the bacteria in a multi-drug environment in a drug-specific manner, thus they both provide resistance and spur further development of resistance.

Resistance is not just a property of an individual bacteria, resistance can also be a property of the microbial community. In a PLOS Biology article, scientists show that intracellular expression of an antibiotic-metabolizing enzyme in a non-pathogenic strain of bacteria can provide resistance to the pathogen Streptococcus pneumoniae when the two types of bacteria are grown together, both in vitro and in vivo [ 3 ]. The expression of the metabolizing enzyme in the non-pathogenic strain deactivates the drug in the immediate environment, despite its intracellular localization, allowing for the outgrowth of the drug-sensitive pathogenic strain ( Fig 1 ). This work highlights the importance of considering the microbial context during infectious disease.

thumbnail

  • PPT PowerPoint slide
  • PNG larger image
  • TIFF original image

Still images of a time-lapse experiment show that the green-labeled drug-resistant strain of Staphylococcus aureus provides resistance for the non-labeled drug-sensitive strain of Streptococcus pneumoniae during antibiotic treatment. Image credit: doi: 10.1371/journal.pbio.2000631 .

https://doi.org/10.1371/journal.pbio.2003775.g001

Evolution and spread of resistance

Since antibiotic resistance is the result of natural selection for resistance-conferring mutations, it is important to understand the evolutionary processes underlying this selection. One interesting element to this puzzle is that bacteria acquire resistance to different antibiotics at different rates. In a PLOS Biology article, the authors sought to understand the properties that determine how quickly resistance will evolve [ 4 ]. They identified two properties, resistance variability and dose sensitivity, that could predict the rate of evolution in seven of eight of the drugs.

Another critical element for understanding the evolution of resistance is the cost that resistance-conferring mutations have on bacterial fitness (i.e. growth rate). Most mutations have an associated cost, however bacteria can gain additional mutations, known as compensatory mutations, that offset those costs and help maintain resistance mutations in a population. A PLOS Biology paper describes how multi-drug resistant bacteria compensate, finding that their compensatory evolution is distinct from that of bacteria resistant to single drugs alone, due to the interaction between the resistance mutations [ 5 ].

Resistance to antibiotics is often acquired by the transfer of resistance-conferring genes between bacteria, and this acquisition is usually facilitated by a conjugative plasmid. These plasmids encode the genes necessary for two bacteria to pass the plasmid between them, and they can also encode resistance genes. But, as mentioned above, resistance comes at a cost, and a study published in Science Reports of the compensatory evolution of a large conjugative resistance plasmid showed that evolution follows common paths leading to plasmid stabilization and persistence of resistance [ 6 ].

Methicillin-resistant Staphylococcus aureus (MRSA) is the most common antibiotic resistant infection in humans, and the most frequent mechanism of resistance in MRSA is via the acquisition of mecA ( Fig 2 ). mecA is a member of the penicillin-binding protein family that doesn’t bind β-lactams (like penicillin) effectively and is thus immune to its effects. In a PLOS Genetics article, the scientists map the evolution of mecA from its original role cell wall biosynthesis [ 7 ]. They identify four mechanisms that have led to its new role in resistance, and most importantly, show that it was the use of antibiotics in medicine and in livestock feed that drove this evolution and spread of resistance.

thumbnail

Scanning electron micrograph of a human neutrophil ingesting MRSA (purple). Image credit : NIAID

https://doi.org/10.1371/journal.pbio.2003775.g002

Techniques for combating resistance

While we only have a limited set of antibiotics in our arsenal, there are better ways of dosing and combining drugs to increase efficacy and decrease resistance. One method is using a sequential regimen. Sequential regimens alternate the use of two (or more) drugs over time. The authors of a PLOS Biology article showed that they could design sequential regimens that eliminated bacteria at doses that would normally lead to resistance and treatment failure [ 8 ]. The key goal is to maximize collateral sensitivity–or when one drug sensitizes the bacteria to the second drug–while minimizing cross-resistance–where resistance to one drug confers resistance to the second drug.

A pair of synergistic antibiotics are more effective than the sum of the efficacies of each antibiotic when used alone and their dual action is thought to be more difficult overcome. Unfortunately, finding synergistic pairs is difficult and traditionally requires screening a huge number of drug combinations. In a PLOS Biology article, researchers utilized a technique originally developed for identifying novel antifungals [ 9 ]. The method uses previously generated chemical-genetic datasets and as a proof-of-concept, the researchers identified new synergistic combinations, including one with the classic antiviral, AZT.

Even in single-drug regimens, proper dosing is essential to minimize the development of resistance. Yet, finding the optimal dosing regimen is tricky and requires costly in vivo experiments. By modeling and understanding the kinetics of how the drug and target interaction, an article appearing in PLOS Computational Biology demonstrates that the best time and concentration parameters for an antibiotic dose can be predicted [ 10 ].

A critically important but often overlooked aspect of the treatment of antibiotic-resistant infections is the role of the immune system in clearance. In an article published in PLOS Computational Biology , the authors use mathematical modelling of within-host infection dynamics to understand the interaction between the host immune response and antibiotic treatment [ 11 ]. By comparing a standard fixed dose and duration treatment regime to dynamic regimes that account for pathogen load, they are able to identify treatments that promote synergy between the immune system and the antibiotics.

What if, instead of treating resistant bacteria with more drugs, we could instead make them sensitive to the original drug again? That is the question asked by the authors of a recent PLOS Biology article [ 12 ]. They show that resistant bacteria can be re-sensitized by treating with a specifically designed anti-sense oligonucleotide. This oligonucleotide, a peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO), acts as an antisense mRNA translation inhibitor, and can be designed to target the mRNAs encoding resistance genes. They found that the most effective PPMOs target a constituent of the major drug efflux pump, and treatment with this PPMO can lead to a 2- to 40-fold increase in antibiotic efficacy.

For more detailed reading please see the associated PLOS Collection [ 13 ].

  • View Article
  • PubMed/NCBI
  • Google Scholar
  • 13. In PLOS Collections. Available: http://collections.plos.org/open-highlights-antibiotic-resistance

Prevention of antibiotic resistance - an epidemiological scoping review to identify research categories and knowledge gaps

Affiliation.

  • 1 Department of Epidemiology and Global Health, Umeå University , Umeå, Sweden.
  • PMID: 32475304
  • PMCID: PMC7782542
  • DOI: 10.1080/16549716.2020.1756191

Background: Antibiotics have become the cornerstone for the treatment of infectious diseases and contributed significantly to the dramatic global health development during the last 70 years. Millions of people now survive what were previously life-threatening infections. But antibiotics are finite resources and misuse has led to antibiotic resistance and reduced efficacy within just a few years of introduction of each new antibiotic. The World Health Organization rates antibiotic resistance as a 'global security threat' impacting on global health, food security and development and as important as terrorism and climate change.

Objectives: This paper explores, through a scoping review of the literature published during the past 20 years, the magnitude of peer-reviewed and grey literature that addresses antibiotic resistance and specifically the extent to which "prevention" has been at the core. The ultimate aim is to identify know-do gaps and strategies to prevent ABR.

Methods: The review covers four main data bases, Web of Science, Medline, Scopus and Ebsco searched for 2000-17. The broader research field "antibiotic OR antimicrobial resistance" gave 431,335 hits. Narrowing the search criteria to "Prevention of antibiotic OR antimicrobial resistance" resulted in 1062 remaining titles. Of these, 622 were unique titles. After screening of the 622 titles for relevance, 420 abstracts were read, and of these 282 papers were read in full. An additional 53 references were identified from these papers, and 64 published during 2018 and 2019 were also included. The final scoping review database thus consisted of 399 papers.

Results: A thematic structure emerged when categorizing articles in different subject areas, serving as a proxy for interest expressed from the research community. The research area has been an evolving one with about half of the 399 papers published during the past four years of the study period. Epidemiological modelling needs strengthening and there is a need for more and better surveillance systems, especially in lower- and middle-income countries. There is a wealth of information on the local and national uses and misuses of antibiotics. Educational and stewardship programmes basically lack evidence. Several studies address knowledge of the public and prescribers. The lessons for policy are conveyed in many alarming reports from national and international organizations.

Conclusions: Descriptive rather than theoretical ambitions have characterized the literature. If we want to better understand and explain the antibiotic situation from a behavioural perspective, the required approaches are lacking. A framework for an epidemiological causal web behind ABR is suggested and may serve to identify entry points for potential interventions.

Keywords: Antibiotic resistance; Antimicrobial Resistance; antimicrobial resistance; behaviour; drug resistance; global threat; health policy; prevention.

Publication types

  • Systematic Review
  • Anti-Bacterial Agents / pharmacology*
  • Drug Resistance, Microbial*
  • Global Health
  • World Health Organization
  • Anti-Bacterial Agents

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.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Review Article
  • Published: 30 April 2024

Ecological and evolutionary mechanisms driving within-patient emergence of antimicrobial resistance

  • Matthew J. Shepherd   ORCID: orcid.org/0000-0002-3283-9930 1 ,
  • Taoran Fu   ORCID: orcid.org/0000-0003-4683-2171 1 ,
  • Niamh E. Harrington 2 ,
  • Anastasia Kottara 1 ,
  • Kendall Cagney 2 ,
  • James D. Chalmers 3 ,
  • Steve Paterson   ORCID: orcid.org/0000-0002-1307-2981 2 ,
  • Joanne L. Fothergill 2 &
  • Michael A. Brockhurst   ORCID: orcid.org/0000-0003-0362-820X 1  

Nature Reviews Microbiology ( 2024 ) Cite this article

1171 Accesses

69 Altmetric

Metrics details

  • Antimicrobial resistance
  • Bacterial infection

The ecological and evolutionary mechanisms of antimicrobial resistance (AMR) emergence within patients and how these vary across bacterial infections are poorly understood. Increasingly widespread use of pathogen genome sequencing in the clinic enables a deeper understanding of these processes. In this Review, we explore the clinical evidence to support four major mechanisms of within-patient AMR emergence in bacteria: spontaneous resistance mutations; in situ horizontal gene transfer of resistance genes; selection of pre-existing resistance; and immigration of resistant lineages. Within-patient AMR emergence occurs across a wide range of host niches and bacterial species, but the importance of each mechanism varies between bacterial species and infection sites within the body. We identify potential drivers of such differences and discuss how ecological and evolutionary analysis could be embedded within clinical trials of antimicrobials, which are powerful but underused tools for understanding why these mechanisms vary between pathogens, infections and individuals. Ultimately, improving understanding of how host niche, bacterial species and antibiotic mode of action combine to govern the ecological and evolutionary mechanism of AMR emergence in patients will enable more predictive and personalized diagnosis and antimicrobial therapies.

This is a preview of subscription content, access via your institution

Access options

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

24,99 € / 30 days

cancel any time

Subscribe to this journal

Receive 12 print issues and online access

195,33 € per year

only 16,28 € per issue

Buy this article

  • Purchase on Springer Link
  • Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

research paper on antibiotic resistance

Similar content being viewed by others

research paper on antibiotic resistance

A distinct Fusobacterium nucleatum clade dominates the colorectal cancer niche

research paper on antibiotic resistance

A novel antibiotic class targeting the lipopolysaccharide transporter

research paper on antibiotic resistance

Pseudomonas aeruginosa: pathogenesis, virulence factors, antibiotic resistance, interaction with host, technology advances and emerging therapeutics

O’Neill, J. Tackling drug-resistant infections globally: final report and recommendations. AMR Review https://amr-review.org/sites/default/files/160518_Final%20paper_with%20cover.pdf (2016).

Murray, C. J. et al. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet 399 , 629–655 (2022).

Article   CAS   Google Scholar  

Choudhury, R., Panda, S. & Singh, D. Emergence and dissemination of antibiotic resistance: a global problem. Indian J. Med. Microbiol. 30 , 384–390 (2012).

Article   CAS   PubMed   Google Scholar  

Bougnom, B. P. & Piddock, L. J. V. Wastewater for urban agriculture: a significant factor in dissemination of antibiotic resistance. Environ. Sci. Technol. 51 , 5863–5864 (2017).

Goulas, A. et al. How effective are strategies to control the dissemination of antibiotic resistance in the environment? A systematic review. Environ. Evid. 9 , 4 (2020).

Article   Google Scholar  

Zhu, G. et al. Air pollution could drive global dissemination of antibiotic resistance genes. ISME J. 15 , 270–281 (2021).

Ellabaan, M. M. H., Munck, C., Porse, A., Imamovic, L. & Sommer, M. O. A. Forecasting the dissemination of antibiotic resistance genes across bacterial genomes. Nat. Commun. 12 , 2435 (2021).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Grote, A. & Earl, A. M. Within-host evolution of bacterial pathogens during persistent infection of humans. Curr. Opin. Microbiol. 70 , 102197 (2022).

Didelot, X., Walker, A. S., Peto, T. E., Crook, D. W. & Wilson, D. J. Within-host evolution of bacterial pathogens. Nat. Rev. Microbiol. 14 , 150–162 (2016).

Castro, R. A. D., Borrell, S. & Gagneux, S. The within-host evolution of antimicrobial resistance in Mycobacterium tuberculosis . FEMS Microbiol. Rev. 45 , fuaa071 (2021).

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).

Marvig, R. L., Sommer, L. M., Molin, S. & Johansen, H. K. Convergent evolution and adaptation of Pseudomonas aeruginosa within patients with cystic fibrosis. Nat. Genet. 47 , 57–64 (2015).

Giulieri, S. G. et al. Niche-specific genome degradation and convergent evolution shaping Staphylococcus aureus adaptation during severe infections. eLife 11 , e77195 (2022).

Azarian, T., Ridgway, J. P., Yin, Z. & David, M. Z. Long-term intrahost evolution of methicillin resistant Staphylococcus aureus among cystic fibrosis patients with respiratory carriage. Front. Genet. 10 , 546 (2019).

Li, S., Feng, X., Li, M. & Shen, Z. In vivo adaptive antimicrobial resistance in Klebsiella pneumoniae during antibiotic therapy. Front. Microbiol. 14 , 1159912 (2023).

Article   PubMed   PubMed Central   Google Scholar  

Friedman, N. D., Temkin, E. & Carmeli, Y. The negative impact of antibiotic resistance. Clin. Microbiol. Infect. 22 , 416–422 (2016).

Goossens, H., Ferech, M., Stichele, R. V. & Elseviers, M. Outpatient antibiotic use in Europe and association with resistance: a cross-national database study. Lancet 365 , 579–587 (2005).

Article   PubMed   Google Scholar  

Gustafsson, I. Bacteria with increased mutation frequency and antibiotic resistance are enriched in the commensal flora of patients with high antibiotic usage. J. Antimicrob. Chemother. 52 , 645–650 (2003).

Baquero, F. et al. Evolutionary pathways and trajectories in antibiotic resistance. Clin. Microbiol. Rev. 34 , e00050-19 (2021).

Vanacker, M., Lenuzza, N. & Rasigade, J.-P. The fitness cost of horizontally transferred and mutational antimicrobial resistance in Escherichia coli . Front. Microbiol. 14 , 1186920 (2023).

Melnyk, A. H., Wong, A. & Kassen, R. The fitness costs of antibiotic resistance mutations. Evol. Appl. 8 , 273–283 (2015).

Hendriksen, R. S. et al. Using genomics to track global antimicrobial resistance. Front. Public. Health 7 , 242 (2019).

Darby, E. M. et al. Molecular mechanisms of antibiotic resistance revisited. Nat. Rev. Microbiol. 21 , 280–295 (2023).

McEwen, S. A. & Collignon, P. J. Antimicrobial resistance: a one health perspective. Microbiol. Spectr. 6 , 6.2.10 (2018).

Hiltunen, T., Virta, M. & Laine, A.-L. Antibiotic resistance in the wild: an eco-evolutionary perspective. Philos. Trans. R. Soc. B Biol. Sci. 372 , 20160039 (2017).

Larsson, D. G. J. & Flach, C.-F. Antibiotic resistance in the environment. Nat. Rev. Microbiol. 20 , 257–269 (2022).

Chung, H. et al. Rapid expansion and extinction of antibiotic resistance mutations during treatment of acute bacterial respiratory infections. Nat. Commun. 13 , 1231 (2022). Captures dynamic fluctuations in resistance mutations across 420 P. aeruginosa isolates during acute infection, capturing both spontaneous mutation and dynamic selection of pre-existing resistance at the start of antibiotic selection.

Eklöf, J. et al. Persistence and genetic adaptation of Pseudomonas aeruginosa in patients with chronic obstructive pulmonary disease. Clin. Microbiol. Infect. 28 , 990–995 (2022). Study of P. aeruginosa adaptation in patients with chronic obstructive pulmonary disease, covering 153 isolates from 23 patients across 1 year of infection and identifying multiple spontaneous mutations that conferred resistance to anti-pseudomonal drugs.

Hjort, K. et al. Dynamics of extensive drug resistance evolution of Mycobacterium tuberculosis in a single patient during 9 years of disease and treatment. J. Infect. Dis. 225 , 1011–1020 (2022).

Khademi, S. M. H., Sazinas, P. & Jelsbak, L. Within-host adaptation mediated by intergenic evolution in Pseudomonas aeruginosa . Genome Biol. Evol. 11 , 1385–1397 (2019).

Liao, W. et al. Evolution of tet(A) variant mediating tigecycline resistance in KPC-2-producing Klebsiella pneumoniae during tigecycline treatment. J. Glob. Antimicrob. Resist. 28 , 168–173 (2022).

Lindemann, P. C. et al. Case report: whole-genome sequencing of serially collected Haemophilus influenzae from a patient with common variable immunodeficiency reveals within-host evolution of resistance to trimethoprim–sulfamethoxazole and azithromycin after prolonged treatment with these antibiotics. Front. Cell. Infect. Microbiol. 12 , 896823 (2022).

Long, D. R. et al. Polyclonality, shared strains, and convergent evolution in chronic cystic fibrosis Staphylococcus aureus airway infection. Am. J. Respir. Crit. Care Med. 203 , 1127–1137 (2021).

Sommer, L. M. et al. Bacterial evolution in PCD and CF patients follows the same mutational steps. Sci. Rep. 6 , 28732 (2016).

Aihara, M. et al. Within-host evolution of a Klebsiella pneumoniae clone: selected mutations associated with the alteration of outer membrane protein expression conferred multidrug resistance. J. Antimicrob. Chemother. 76 , 362–369 (2021).

Boulant, T. et al. A 2.5-year within-patient evolution of Pseudomonas aeruginosa isolates with in vivo acquisition of ceftolozane–tazobactam and ceftazidime–avibactam resistance upon treatment. Antimicrob. Agents Chemother. 63 , e01637-19 (2019).

Chen, C.-J., Huang, Y.-C. & Shie, S.-S. Evolution of multi-resistance to vancomycin, daptomycin, and linezolid in methicillin-resistant Staphylococcus aureus causing persistent bacteremia. Front. Microbiol. 11 , 1414 (2020).

Elgrail, M. M. et al. Convergent evolution of antibiotic tolerance in patients with persistent methicillin-resistant Staphylococcus aureus bacteremia. Infect. Immun. 90 , e00001–e00022 (2022).

Gao, W. et al. Large tandem chromosome expansions facilitate niche adaptation during persistent infection with drug-resistant Staphylococcus aureus . Microb. Genomics 1 , e000026 (2015).

Giulieri, S. G. et al. Genomic exploration of sequential clinical isolates reveals a distinctive molecular signature of persistent Staphylococcus aureus bacteraemia. Genome Med. 10 , 65 (2018).

van Hal, S. J. et al. In vivo evolution of antimicrobial resistance in a series of Staphylococcus aureus patient isolates: the entire picture or a cautionary tale? J. Antimicrob. Chemother. 69 , 363–367 (2014).

Mwangi, M. M. et al. Tracking the in vivo evolution of multidrug resistance in Staphylococcus aureus by whole-genome sequencing. Proc. Natl Acad. Sci. USA 104 , 9451–9456 (2007).

Jin, X. et al. Resistance evolution of hypervirulent carbapenem-resistant Klebsiella pneumoniae ST11 during treatment with tigecycline and polymyxin. Emerg. Microbes Infect. 10 , 1129–1136 (2021).

Kao, C.-Y., Chen, J.-W., Liu, T.-L., Yan, J.-J. & Wu, J.-J. Comparative genomics of Escherichia coli sequence type 219 clones from the same patient: evolution of the IncI1 blaCMY-carrying plasmid in vivo. Front. Microbiol. 9 , 1518 (2018).

Vallée, M., Harding, C. & Hall, J. Exploring the in situ evolution of nitrofurantoin resistance in clinically derived uropathogenic Escherichia coli isolates. J. Antimicrob. Chemother. 78 , 373–379 (2023). Case of within-patient spontaneous evolution in E. coli of resistance to nitrofurantoin, an antibiotic to which resistance in E. coli is rare. Resistance confers a 2–10% reduction in doubling time in resistant isolates, indicating fitness trade-offs.

Ye, M. et al. In vivo development of tigecycline resistance in Klebsiella pneumoniae owing to deletion of the ramR ribosomal binding site. Int. J. Antimicrob. Agents 50 , 523–528 (2017).

Hawkey, J. et al. Evolution of carbapenem resistance in Acinetobacter baumannii during a prolonged infection. Microb. Genomics 4 , e000165 (2018).

Dengler Haunreiter, V. et al. In-host evolution of Staphylococcus epidermidis in a pacemaker-associated endocarditis resulting in increased antibiotic tolerance. Nat. Commun. 10 , 1149 (2019).

Ji, S. et al. In-host evolution of daptomycin resistance and heteroresistance in methicillin-resistant Staphylococcus aureus strains from three endocarditis patients. J. Infect. Dis. 221 , S243–S252 (2020).

Bloomfield, S. J. et al. Long-term colonization by Campylobacter jejuni within a human host: evolution, antimicrobial resistance, and adaptation. J. Infect. Dis. 217 , 103–111 (2018).

Low, A. S., MacKenzie, F. M., Gould, I. M. & Booth, I. R. Protected environments allow parallel evolution of a bacterial pathogen in a patient subjected to long-term antibiotic therapy: evolution of a bacterial pathogen. Mol. Microbiol. 42 , 619–630 (2008). Case of E. coli infection of liver cysts, studying the action of the protected cyst environment in facilitating resistance evolution. Spontaneous mutation to the ampC promoter conferred β-lactam resistance. The sheltered environment likely offered protection against the action of antibiotics, alongside invasion or interference by other bacterial lineages.

Yu, X. et al. Effect of short-term antimicrobial therapy on the tolerance and antibiotic resistance of multidrug-resistant Staphylococcus capitis . Infect. Drug Resist. 13 , 2017–2026 (2020).

Notter, J. et al. AmpC hyperproduction in a Cedecea davisae implant-associated bone infection during treatment: a case report and therapeutic implications. BMC Infect. Dis. 22 , 33 (2022).

Colque, C. A. et al. Hypermutator Pseudomonas aeruginosa exploits multiple genetic pathways to develop multidrug resistance during long-term infections in the airways of cystic fibrosis patients. Antimicrob. Agents Chemother. 64 , e02142–19 (2020).

Diaz Caballero, J. et al. Selective sweeps and parallel pathoadaptation drive Pseudomonas aeruginosa evolution in the cystic fibrosis lung. mBio 6 , e00981-15 (2015).

Colque, C. A. et al. Longitudinal evolution of the Pseudomonas -derived cephalosporinase (PDC) structure and activity in a cystic fibrosis patient treated with β-lactams. mBio 13 , e01663-22 (2022). 24-year study of P. aeruginosa cystic fibrosis airway infection. Documents the emergence of a hypermutator lineage and parallel evolution of PDC during ceftazidime therapy.

Zhang, J. et al. Genomic and phenotypic evolution of tigecycline-resistant Acinetobacter baumannii in critically ill patients. Microbiol. Spectr. 10 , e01593-21 (2022).

Sherrard, L. J. et al. Within-host whole genome analysis of an antibiotic resistant Pseudomonas aeruginosa strain sub-type in cystic fibrosis. PLoS ONE 12 , e0172179 (2017).

Alexander, H. K. & MacLean, R. C. Stochastic bacterial population dynamics restrict the establishment of antibiotic resistance from single cells. Proc. Natl Acad. Sci. USA 117 , 19455–19464 (2020).

MacLean, R. C., Hall, A. R., Perron, G. G. & Buckling, A. The population genetics of antibiotic resistance: integrating molecular mechanisms and treatment contexts. Nat. Rev. Genet. 11 , 405–414 (2010).

Wheatley, R. et al. Rapid evolution and host immunity drive the rise and fall of carbapenem resistance during an acute Pseudomonas aeruginosa infection. Nat. Commun. 12 , 2460 (2021).

Mariam, S. H. et al. Dynamics of antibiotic resistant Mycobacterium tuberculosis during long-term infection and antibiotic treatment. PLoS ONE 6 , e21147 (2011). Investigation of M. tuberculosis evolution during a 9-year infection case study of one patient. Resistance mutations progressively accumulated through spontaneous mutation during this period, and evidence of clonal sweeps and co-existence of different resistance mutations are reported.

Foster, P. L. Stress-induced mutagenesis in bacteria. Crit. Rev. Biochem. Mol. Biol. 42 , 373–397 (2007).

Rodríguez-Rojas, A., Oliver, A. & Blázquez, J. Intrinsic and environmental mutagenesis drive diversification and persistence of Pseudomonas aeruginosa in chronic lung infections. J. Infect. Dis. 205 , 121–127 (2012).

Revitt-Mills, S. A. & Robinson, A. Antibiotic-induced mutagenesis: under the microscope. Front. Microbiol. 11 , 585175 (2020).

Wright, E. A., Fothergill, J. L., Paterson, S., Brockhurst, M. A. & Winstanley, C. Sub-inhibitory concentrations of some antibiotics can drive diversification of Pseudomonas aeruginosa populations in artificial sputum medium. BMC Microbiol. 13 , 170 (2013).

Linz, B. et al. A mutation burst during the acute phase of Helicobacter pylori infection in humans and rhesus macaques. Nat. Commun. 5 , 4165 (2014).

Estibariz, I. et al. In vivo genome and methylome adaptation of cag -negative Helicobacter pylori during experimental human infection. mBio 11 , e01803-20 (2020).

Mehta, H. H. et al. The essential role of hypermutation in rapid adaptation to antibiotic stress. Antimicrob. Agents Chemother. 63 , e00744-19 (2019).

Rees, V. E. et al. Characterization of hypermutator Pseudomonas aeruginosa isolates from patients with cystic fibrosis in Australia. Antimicrob. Agents Chemother. 63 , e02538-18 (2019).

Khil, P. P. et al. Dynamic emergence of mismatch repair deficiency facilitates rapid evolution of ceftazidime-avibactam resistance in Pseudomonas aeruginosa acute infection. mBio 10 , e01822-19 (2019).

Gabrielaite, M. et al. Transmission and antibiotic resistance of Achromobacter in cystic fibrosis. J. Clin. Microbiol. 59 , e02911–e02920 (2021).

Viberg, L. T. et al. Within-host evolution of Burkholderia pseudomallei during chronic infection of seven Australasian cystic fibrosis patients. mBio 8 , e00356-17 (2017).

Pompilio, A. et al. Stenotrophomonas maltophilia phenotypic and genotypic diversity during a 10-year colonization in the lungs of a cystic fibrosis patient. Front. Microbiol. 7 , 1551 (2016).

Turrientes, M. C. et al. Polymorphic mutation frequencies of clinical and environmental Stenotrophomonas maltophilia populations. Appl. Environ. Microbiol. 76 , 1746–1758 (2010). Epidemiological data from more than 9,000 patients and genomes of 250 enterobacteria indicate highly frequent patient to patient transmission of pOXA-48 carbapenemase-encoding plasmid and document frequent within-patient transfer of the plasmid horizontally between enterobacterial species.

Iguchi, S., Mizutani, T., Hiramatsu, K. & Kikuchi, K. Rapid acquisition of linezolid resistance in methicillin-resistant Staphylococcus aureus : role of hypermutation and homologous recombination. PLoS ONE 11 , e0155512 (2016).

Jolivet-Gougeon, A. et al. Bacterial hypermutation: clinical implications. J. Med. Microbiol. 60 , 563–573 (2011).

Gottig, S., Gruber, T. M., Stecher, B., Wichelhaus, T. A. & Kempf, V. A. J. In vivo horizontal gene transfer of the carbapenemase OXA-48 during a nosocomial outbreak. Clin. Infect. Dis. 60 , 1808–1815 (2015).

Evans, D. R. et al. Systematic detection of horizontal gene transfer across genera among multidrug-resistant bacteria in a single hospital. eLife 9 , e53886 (2020).

Kociolek, L. K. et al. Whole-genome analysis reveals the evolution and transmission of an MDR DH/NAP11/106 Clostridium difficile clone in a paediatric hospital. J. Antimicrob. Chemother. 73 , 1222–1229 (2018).

Yoshino, M. et al. Stepwise evolution of a Klebsiella pneumoniae clone within a host leading to increased multidrug resistance. mSphere 6 , e00734-21 (2021).

León-Sampedro, R. et al. Pervasive transmission of a carbapenem resistance plasmid in the gut microbiota of hospitalized patients. Nat. Microbiol. 6 , 606–616 (2021).

DelaFuente, J. et al. Within-patient evolution of plasmid-mediated antimicrobial resistance. Nat. Ecol. Evol. 6 , 1980–1991 (2022). Landmark study of within-patient plasmid-mediated AMR evolution, including compensatory mutation and fitness trade-offs to enhance plasmid-borne AMR evolution.

Arnold, B. J., Huang, I.-T. & Hanage, W. P. Horizontal gene transfer and adaptive evolution in bacteria. Nat. Rev. Microbiol. 20 , 206–218 (2022).

Gaibani, P. et al. Dynamic evolution of imipenem/relebactam resistance in a KPC-producing Klebsiella pneumoniae from a single patient during ceftazidime/avibactam-based treatments. J. Antimicrob. Chemother. 77 , 1570–1577 (2022).

Groussin, M. et al. Elevated rates of horizontal gene transfer in the industrialized human microbiome. Cell 184 , 2053–2067.e18 (2021).

Zeng, X. & Lin, J. Factors influencing horizontal gene transfer in the intestine. Anim. Health Res. Rev. 18 , 153–159 (2018).

Feasey, N. A. et al. Drug resistance in Salmonella enterica ser. Typhimurium bloodstream infection, Malawi. Emerg. Infect. Dis. 20 , 1957–1959 (2014).

Weber, R. E. et al. IS26-mediated transfer of bla NDM–1 as the main route of resistance transmission during a polyclonal, multispecies outbreak in a German hospital. Front. Microbiol. 10 , 2817 (2019).

Enault, F. et al. Phages rarely encode antibiotic resistance genes: a cautionary tale for virome analyses. ISME J. 11 , 237–247 (2017).

Billaud, M. et al. Analysis of viromes and microbiomes from pig fecal samples reveals that phages and prophages rarely carry antibiotic resistance genes. ISME Commun. 1 , 55 (2021).

Fernández-Orth, D. et al. Faecal phageome of healthy individuals: presence of antibiotic resistance genes and variations caused by ciprofloxacin treatment. J. Antimicrob. Chemother. 74 , 854–864 (2019).

Sutcliffe, S. G., Shamash, M., Hynes, A. P. & Maurice, C. F. Common oral medications lead to prophage induction in bacterial isolates from the human gut. Viruses 13 , 455 (2021).

Haaber, J., Penadés, J. R. & Ingmer, H. Transfer of antibiotic resistance in Staphylococcus aureus . Trends Microbiol. 25 , 893–905 (2017).

Varga, M. et al. Efficient transfer of antibiotic resistance plasmids by transduction within methicillin-resistant Staphylococcus aureus USA300 clone. FEMS Microbiol. Lett. 332 , 146–152 (2012).

Barrett, R. & Schluter, D. Adaptation from standing genetic variation. Trends Ecol. Evol. 23 , 38–44 (2008).

Trindade, S. et al. Positive epistasis drives the acquisition of multidrug resistance. PLoS Genet. 5 , e1000578 (2009).

Diaz Caballero, J. et al. Mixed strain pathogen populations accelerate the evolution of antibiotic resistance in patients. Nat. Commun. 14 , 4083 (2023). Comparison of clonal origin and mixed-strain P. aeruginosa lung infections in evolution of AMR. Demonstrates that mixed-strain infections display accelerated evolution of AMR within patients through selection of pre-existing resistant lineages.

Trauner, A. et al. The within-host population dynamics of Mycobacterium tuberculosis vary with treatment efficacy. Genome Biol. 18 , 71 (2017).

Ailloud, F. et al. Within-host evolution of Helicobacter pylori shaped by niche-specific adaptation, intragastric migrations and selective sweeps. Nat. Commun. 10 , 2273 (2019).

Kao, C.-Y. et al. Heteroresistance of Helicobacter pylori from the same patient prior to antibiotic treatment. Infect. Genet. Evol. 23 , 196–202 (2014).

Bello Gonzalez, T. D. J. et al. Characterization of Enterococcus isolates colonizing the intestinal tract of intensive care unit patients receiving selective digestive decontamination. Front. Microbiol. 8 , 1596 (2017).

WHO. Global Antimicrobial Resistance and Use Surveillance System (GLASS) Report: 2022 (World Health Organization, 2022).

Vargas, R. et al. In-host population dynamics of Mycobacterium tuberculosis complex during active disease. eLife 10 , e61805 (2021).

Roodgar, M. et al. Longitudinal linked-read sequencing reveals ecological and evolutionary responses of a human gut microbiome during antibiotic treatment. Genome Res. 31 , 1433–1446 (2021).

Li, J. et al. Antibiotic treatment drives the diversification of the human gut resistome. Genomics Proteomics Bioinformatics 17 , 39–51 (2019).

Malik, U. et al. Association between prior antibiotic therapy and subsequent risk of community-acquired infections: a systematic review. J. Antimicrob. Chemother. 73 , 287–296 (2018).

Stracy, M. et al. Minimizing treatment-induced emergence of antibiotic resistance in bacterial infections. Science 375 , 889–894 (2022). A large-scale study (>140,000 patients) indicating that the prevailing mode of in-host AMR for urinary tract and wound infections is immigration of resistant lineages, not spontaneous mutation.

Lieberman, T. D. et al. Parallel bacterial evolution within multiple patients identifies candidate pathogenicity genes. Nat. Genet. 43 , 1275–1280 (2011).

Azarian, T. et al. Intrahost evolution of methicillin-resistant Staphylococcus aureus USA300 among individuals with reoccurring skin and soft-tissue infections. J. Infect. Dis. 214 , 895–905 (2016).

Woods, R. J. et al. The evolution of antibiotic resistance in an incurable and ultimately fatal infection. Evol. Med. Public Health 11 , 163–173 (2023).

Moradigaravand, D. et al. Within-host evolution of Enterococcus faecium during longitudinal carriage and transition to bloodstream infection in immunocompromised patients. Genome Med. 9 , 119 (2017).

Hayward, C., Brown, M. H. & Whiley, H. Hospital water as the source of healthcare-associated infection and antimicrobial-resistant organisms. Curr. Opin. Infect. Dis. 35 , 339–345 (2022).

Ding, B. et al. The predominance of strain replacement among Enterobacteriaceae pairs with emerging carbapenem resistance during hospitalization. J. Infect. Dis. 221 , S215–S219 (2020). Study identifying evolution of resistance in 41 patients treated with carbapenems, where most (90%) K. pneumoniae infections developed resistance through immigration of strains of a different sequence type.

Wheatley, R. M. et al. Gut to lung translocation and antibiotic mediated selection shape the dynamics of Pseudomonas aeruginosa in an ICU patient. Nat. Commun. 13 , 6523 (2022). Case study of meropenem resistance evolution by P. aeruginosa spontaneously in both the lung and gut of a patient, along with translocation of resistant lineages from the gut to the lung.

Dickson, R. P. et al. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat. Microbiol. 1 , 16113 (2016).

Gillings, M. R., Paulsen, I. T. & Tetu, S. G. Genomics and the evolution of antibiotic resistance: genomics and antibiotic resistance. Ann. NY Acad. Sci. 1388 , 92–107 (2017).

Davies, J. & Davies, D. Origins and evolution of antibiotic resistance. Microbiol. Mol. Biol. Rev. 74 , 417–433 (2010).

Palmer, A. C. & Kishony, R. Understanding, predicting and manipulating the genotypic evolution of antibiotic resistance. Nat. Rev. Genet. 14 , 243–248 (2013).

MacLean, R. C. & San Millan, A. The evolution of antibiotic resistance. Science 365 , 1082–1083 (2019).

Baquero, F., Alvarez-Ortega, C. & Martinez, J. L. Ecology and evolution of antibiotic resistance. Environ. Microbiol. Rep. 1 , 469–476 (2009).

Eldholm, V. & Balloux, F. Antimicrobial resistance in Mycobacterium tuberculosis : the odd one out. Trends Microbiol. 24 , 637–648 (2016).

Sun, G. et al. Dynamic population changes in Mycobacterium tuberculosis during acquisition and fixation of drug resistance in patients. J. Infect. Dis. 206 , 1724–1733 (2012).

Eldholm, V. et al. Evolution of extensively drug-resistant Mycobacterium tuberculosis from a susceptible ancestor in a single patient. Genome Biol. 15 , 490 (2014).

Lehtinen, S. et al. Horizontal gene transfer rate is not the primary determinant of observed antibiotic resistance frequencies in Streptococcus pneumoniae . Sci. Adv. 6 , eaaz6137 (2020).

Hall, R. J. et al. Gene-gene relationships in an Escherichia coli pangenome are linked to function and mobility. Microb. Genom. 7 , 000650 (2021).

CAS   PubMed   PubMed Central   Google Scholar  

Rocha, J., Henriques, I., Gomila, M. & Manaia, C. M. Common and distinctive genomic features of Klebsiella pneumoniae thriving in the natural environment or in clinical settings. Sci. Rep. 12 , 10441 (2022).

Matic, I. et al. Highly variable mutation rates in commensal and pathogenic Escherichia coli . Science 277 , 1833–1834 (1997).

Kapel, N., Caballero, J. D. & MacLean, R. C. Localized pmrB hypermutation drives the evolution of colistin heteroresistance. Cell Rep. 39 , 110929 (2022).

Horton, J. S. & Taylor, T. B. Mutation bias and adaptation in bacteria. Microbiology 169 , 001404 (2023).

Boyce, K. J. Mutators enhance adaptive micro-evolution in pathogenic microbes. Microorganisms 10 , 442 (2022).

Luján, A. M. et al. Polymicrobial infections can select against Pseudomonas aeruginosa mutators because of quorum-sensing trade-offs. Nat. Ecol. Evol. 6 , 979–988 (2022).

Rojas, L. J. et al. Genomic heterogeneity underlies multidrug resistance in Pseudomonas aeruginosa : a population-level analysis beyond susceptibility testing. PLoS ONE 17 , e0265129 (2022).

Bianconi, I. et al. Persistence and microevolution of Pseudomonas aeruginosa in the cystic fibrosis lung: a single-patient longitudinal genomic study. Front. Microbiol. 9 , 3242 (2019).

Tueffers, L. et al. Pseudomonas aeruginosa populations in the cystic fibrosis lung lose susceptibility to newly applied β-lactams within 3 days. J. Antimicrob. Chemother. 74 , 2916–2925 (2019).

Coluzzi, C. et al. Chance favors the prepared genomes: horizontal transfer shapes the emergence of antibiotic resistance mutations in core genes. Mol. Biol. Evol. 40 , msad217 (2023).

Papkou, A., Hedge, J., Kapel, N., Young, B. & MacLean, R. C. Efflux pump activity potentiates the evolution of antibiotic resistance across S. aureus isolates. Nat. Commun. 11 , 3970 (2020).

Lopatkin, A. J. et al. Clinically relevant mutations in core metabolic genes confer antibiotic resistance. Science 371 , eaba0862 (2021).

Gifford, D. R. et al. Identifying and exploiting genes that potentiate the evolution of antibiotic resistance. Nat. Ecol. Evol. 2 , 1033–1039 (2018).

Connor, C. H. et al. Multidrug-resistant E. coli encoding high genetic diversity in carbohydrate metabolism genes displace commensal E. coli from the intestinal tract. PLoS Biol. 21 , e3002329 (2023).

Bartell, J. A. et al. Bacterial persisters in long-term infection: emergence and fitness in a complex host environment. PLoS Pathog. 16 , e1009112 (2020).

Reynoso-García, J. et al. A complete guide to human microbiomes: body niches, transmission, development, dysbiosis, and restoration. Front. Syst. Biol. 2 , 951403 (2022).

Schenk, M. F. et al. Population size mediates the contribution of high-rate and large-benefit mutations to parallel evolution. Nat. Ecol. Evol. 6 , 439–447 (2022).

Andersson, D. I. Improving predictions of the risk of resistance development against new and old antibiotics. Clin. Microbiol. Infect. 21 , 894–898 (2015).

Dekaboruah, E., Suryavanshi, M. V., Chettri, D. & Verma, A. K. Human microbiome: an academic update on human body site specific surveillance and its possible role. Arch. Microbiol. 202 , 2147–2167 (2020).

Baron, S. A., Diene, S. M. & Rolain, J.-M. Human microbiomes and antibiotic resistance. Hum. Microbiome J. 10 , 43–52 (2018).

Moura De Sousa, J., Lourenço, M. & Gordo, I. Horizontal gene transfer among host-associated microbes. Cell Host Microbe 31 , 513–527 (2023).

Gumpert, H. et al. Transfer and persistence of a multi-drug resistance plasmid in situ of the infant gut microbiota in the absence of antibiotic treatment. Front. Microbiol. 8 , 1852 (2017).

Baumgartner, M., Bayer, F., Pfrunder-Cardozo, K. R., Buckling, A. & Hall, A. R. Resident microbial communities inhibit growth and antibiotic-resistance evolution of Escherichia coli in human gut microbiome samples. PLoS Biol. 18 , e3000465 (2020).

De Nies, L., Kobras, C. M. & Stracy, M. Antibiotic-induced collateral damage to the microbiota and associated infections. Nat. Rev. Microbiol. 21 , 789–804 (2023).

Bottery, M. J., Pitchford, J. W. & Friman, V.-P. Ecology and evolution of antimicrobial resistance in bacterial communities. ISME J. 15 , 939–948 (2021).

Flores-Mireles, A. L., Walker, J. N., Caparon, M. & Hultgren, S. J. Urinary tract infections: epidemiology, mechanisms of infection and treatment options. Nat. Rev. Microbiol. 13 , 269–284 (2015).

Linde, H.-J. et al. In vivo increase in resistance to ciprofloxacin in Escherichia coli associated with deletion of the C-terminal part of MarR. Antimicrob. Agents Chemother. 44 , 1865–1868 (2000).

Otani, S. & Coopersmith, C. M. Gut integrity in critical illness. J. Intensive Care 7 , 17 (2019).

Witzany, C., Rolff, J., Regoes, R. R. & Igler, C. The pharmacokinetic–pharmacodynamic modelling framework as a tool to predict drug resistance evolution. Microbiology 169 , 001368 (2023).

Stanton, I. C., Murray, A. K., Zhang, L., Snape, J. & Gaze, W. H. Evolution of antibiotic resistance at low antibiotic concentrations including selection below the minimal selective concentration. Commun. Biol. 3 , 467 (2020).

Feliziani, S. et al. Coexistence and within-host evolution of diversified lineages of hypermutable Pseudomonas aeruginosa in long-term cystic fibrosis infections. PLoS Genet. 10 , e1004651 (2014).

Both, A. et al. Distinct clonal lineages and within-host diversification shape invasive Staphylococcus epidermidis populations. PLoS Pathog. 17 , e1009304 (2021).

Lewin, A. et al. Genetic diversification of persistent Mycobacterium abscessus within cystic fibrosis patients. Virulence 12 , 2415–2429 (2021).

Hall, K. M., Pursell, Z. F. & Morici, L. A. The role of the Pseudomonas aeruginosa hypermutator phenotype on the shift from acute to chronic virulence during respiratory infection. Front. Cell. Infect. Microbiol. 12 , 943346 (2022).

Maciá, M. D. et al. Hypermutation is a key factor in development of multiple-antimicrobial resistance in Pseudomonas aeruginosa strains causing chronic lung infections. Antimicrob. Agents Chemother. 49 , 3382–3386 (2005).

Prunier, A. et al. High rate of macrolide resistance in Staphylococcus aureus strains from patients with cystic fibrosis reveals high proportions of hypermutable strains. J. Infect. Dis. 187 , 1709–1716 (2003).

Spellberg, B. & Rice, L. B. Duration of antibiotic therapy: shorter is better. Ann. Intern. Med. 171 , 210 (2019).

Martínez, J. L., Baquero, F. & Andersson, D. I. Predicting antibiotic resistance. Nat. Rev. Microbiol. 5 , 958–965 (2007).

Miller, C. R., Monk, J. M., Szubin, R. & Berti, A. D. Rapid resistance development to three antistaphylococcal therapies in antibiotic-tolerant Staphylococcus aureus bacteremia. PLoS ONE 16 , e0258592 (2021).

Bjarnsholt, T. The role of bacterial biofilms in chronic infections. APMIS Suppl. 121 , 1–58 (2013).

Kolpen, M. et al. Bacterial biofilms predominate in both acute and chronic human lung infections. Thorax 77 , 1015–1022 (2022).

Henriksen, N. N. S. E. et al. Biofilm cultivation facilitates coexistence and adaptive evolution in an industrial bacterial community. NPJ Biofilms Microbiomes 8 , 59 (2022).

Jo, J., Price-Whelan, A. & Dietrich, L. E. P. Gradients and consequences of heterogeneity in biofilms. Nat. Rev. Microbiol. 20 , 593–607 (2022).

Gupta, S., Laskar, N. & Kadouri, D. E. Evaluating the effect of oxygen concentrations on antibiotic sensitivity, growth, and biofilm formation of human pathogens. Microbiol. Insights 9 , MBI.S40767 (2016).

Burmølle, M. et al. Enhanced biofilm formation and increased resistance to antimicrobial agents and bacterial invasion are caused by synergistic interactions in multispecies biofilms. Appl. Environ. Microbiol. 72 , 3916–3923 (2006).

Shewaramani, S. et al. Anaerobically grown Escherichia coli has an enhanced mutation rate and distinct mutational spectra. PLoS Genet. 13 , e1006570 (2017).

Hull, R. C. et al. Antibiotics limit adaptation of drug-resistant Staphylococcus aureus to hypoxia. Antimicrob. Agents Chemother. 66 , e00926-22 (2022).

Madsen, J. S., Burmølle, M., Hansen, L. H. & Sørensen, S. J. The interconnection between biofilm formation and horizontal gene transfer. FEMS Immunol. Med. Microbiol. 65 , 183–195 (2012).

Stalder, T. & Top, E. Plasmid transfer in biofilms: a perspective on limitations and opportunities. NPJ Biofilms Microbiomes 2 , 16022 (2016).

Huo, W. et al. Immunosuppression broadens evolutionary pathways to drug resistance and treatment failure during Acinetobacter baumannii pneumonia in mice. Nat. Microbiol. 7 , 796–809 (2022).

Handel, A., Margolis, E. & Levin, B. R. Exploring the role of the immune response in preventing antibiotic resistance. J. Theor. Biol. 256 , 655–662 (2009).

Dropulic, L. K. & Lederman, H. M. Overview of Infections in the Immunocompromised host. Microbiol. Spectr. 4 , 4.4.43 (2016).

Xu, Y. et al. In vivo gene expression in a Staphylococcus aureus prosthetic joint infection characterized by RNA sequencing and metabolomics: a pilot study. BMC Microbiol. 16 , 80 (2016).

Kolpen, M. et al. Nitric oxide production by polymorphonuclear leucocytes in infected cystic fibrosis sputum consumes oxygen. Clin. Exp. Immunol. 177 , 310–319 (2014).

Dwyer, D. J. et al. Antibiotics induce redox-related physiological alterations as part of their lethality. Proc. Natl Acad. Sci. USA 111 , E2100–E2109 (2014).

Borriello, G. et al. Oxygen limitation contributes to antibiotic tolerance of Pseudomonas aeruginosa in biofilms. Antimicrob. Agents Chemother. 48 , 2659–2664 (2004).

Hajdamowicz, N. H., Hull, R. C., Foster, S. J. & Condliffe, A. M. The impact of hypoxia on the host-pathogen interaction between neutrophils and Staphylococcus aureus . Int. J. Mol. Sci. 20 , 5561 (2019).

Raad, C. et al. Trends in bacterial bloodstream infections and resistance in immuno-compromised patients with febrile neutropenia: a retrospective analysis. Eur. J. Pediatr. 180 , 2921–2930 (2021).

Beaume, M. et al. Rapid adaptation drives invasion of airway donor microbiota by Pseudomonas after lung transplantation. Sci. Rep. 7 , 40309 (2017).

Hughes, D. & Andersson, D. I. Evolutionary trajectories to antibiotic resistance. Annu. Rev. Microbiol. 71 , 579–596 (2017).

Luchen, C. C. et al. Impact of antibiotics on gut microbiome composition and resistome in the first years of life in low- to middle-income countries: a systematic review. PLoS Med. 20 , e1004235 (2023).

Fernández-Calvet, A. et al. The distribution of fitness effects of plasmid pOXA-48 in clinical enterobacteria : this article is part of the microbial evolution collection. Microbiology 169 , 001369 (2023).

Rodríguez-Beltrán, J. et al. Genetic dominance governs the evolution and spread of mobile genetic elements in bacteria. Proc. Natl Acad. Sci. USA 117 , 15755–15762 (2020).

Zhou, H., Beltrán, J. F. & Brito, I. L. Functions predict horizontal gene transfer and the emergence of antibiotic resistance. Sci. Adv. 7 , eabj5056 (2021).

Fragata, I., Blanckaert, A., Dias Louro, M. A., Liberles, D. A. & Bank, C. Evolution in the light of fitness landscape theory. Trends Ecol. Evol. 34 , 69–82 (2019).

Kivisaar, M. Mutation and recombination rates vary across bacterial chromosome. Microorganisms 8 , 25 (2020).

Wei, W. et al. Rapid evolution of mutation rate and spectrum in response to environmental and population-genetic challenges. Nat. Commun. 13 , 4752 (2022).

Butler, M. S., Henderson, I. R., Capon, R. J. & Blaskovich, M. A. T. Antibiotics in the clinical pipeline as of December 2022. J. Antibiot. 76 , 431–473 (2023).

Download references

Acknowledgements

The authors thank members of Microbial Evolution Research Manchester for discussion of the ideas in this manuscript. They gratefully acknowledge funding from the Wellcome Trust (Collaborative Award in Science 220243/Z/20/Z).

Author information

Authors and affiliations.

Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK

Matthew J. Shepherd, Taoran Fu, Anastasia Kottara & Michael A. Brockhurst

Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK

Niamh E. Harrington, Kendall Cagney, Steve Paterson & Joanne L. Fothergill

Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK

James D. Chalmers

You can also search for this author in PubMed   Google Scholar

Contributions

M.J.S. researched data for the article, substantially contributed to discussion of content, wrote the article, and reviewed and edited the article. M.A.B., N.E.H. and J.L.F. substantially contributed to discussion of content, wrote the article, and reviewed and edited the article. A.K., K.C. and S.P. substantially contributed to discussion of content, and reviewed and edited the article. T.F. researched data for the article and substantially contributed to discussion of content. J.D.C. reviewed and edited the article.

Corresponding authors

Correspondence to Matthew J. Shepherd or Michael A. Brockhurst .

Ethics declarations

Competing interests.

The authors declare no competing interests.

Peer review

Peer review information.

Nature Reviews Microbiology thanks Javier de la Fuente, Rachel Wheatley and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Supplementary information

Supplementary information.

An antibiotic is an agent that is used against bacteria and can be classified as bactericidal (kills bacteria) or bacteriostatic (inhibits bacterial growth).

(AMR). The ability of microorganisms — bacteria, viruses, fungi or parasites — to withstand the effects of drugs that would normally inhibit or kill them. This phenomenon occurs when these organisms adapt and develop resistance mechanisms against antimicrobial agents, rendering previously effective treatments ineffective. Clinically, AMR is defined as when the minimum inhibitory concentration of the antimicrobial required to halt growth of a bacterium exceeds the clinical breakpoint, which is the highest concentration of that antimicrobial that can be given to a patient.

The ability of a population of microorganisms to survive transient exposure to a microbicidal agent. It differs from resistance in that the agent remains effective against the microorganism as measured by the minimum inhibitory concentration but requires more prolonged treatment to successfully eliminate the infection.

When a subpopulation of bacteria has a much higher tolerance to an antibiotic than the majority, that population is described as persistent. When the pressure of the antibiotic is removed, this persistent community can re-emerge, leading to recurrent infection despite antibiotic treatment.

Antimicrobial resistance that evolves through adaptation to another antimicrobial agent. This can occur when evolution of resistance to one agent confers resistance to another, typically due to a resistance mechanism that equally affects the action of all drugs within a class or to a nonspecific mechanism such as multidrug efflux pumps.

The reciprocal interactions and feedback between ecological processes and evolutionary changes in populations over short time scales. Ecological shifts promote adaptation of populations to their changing environments, and the resulting evolutionary changes can in turn shape ecological interactions. In the context of infections — the within-patient niche ecology will shape the evolution of antibiotic resistance, which can then affect the ecology of the infection itself through failure to clear the infection, disease progression and loss of sensitive strains and microflora.

The physiological or energetic cost of an advantage in reproductive success (fitness). For example, the fitness benefit of resistance to an antibiotic may come at the cost of a reduced growth rate. Importantly, this trade-off becomes a disadvantage when the antibiotic is absent.

The state in which a genetic variant becomes the only variant present for that specific locus. All individuals within the population share that same allele.

The change in frequency of an allele owing to random chance. The impact of such random effects is stronger at smaller population sizes. Newly generated random mutations present at very low frequency must escape random loss due to genetic drift even if they provide a benefit before becoming established at a higher frequency in the population.

(HGT). The process by which microorganisms may exchange genetic material that bypasses vertical transmission from parent to offspring.

A microbial strain with an unusually high mutation rate, often caused by deficiencies in DNA repair mechanisms. Under selection pressure from an antimicrobial agent, this rapid accumulation of spontaneous genetic mutations may accelerate the process of selection and thus evolution of antimicrobial resistance by increasing the likelihood of a mutation-conferring resistance to occur.

The process by which genetic variations within a population become more prevalent because they confer traits that influence the fitness of the organisms in their environment. In the context of antimicrobial therapy, selection refers to the survival and growth of resistant strains and the loss of sensitive ones during treatment.

An evolutionary event whereby a highly advantageous mutation rapidly increases in frequency owing to strong positive selective pressure. As it does, the genetic diversity in the region of the mutation decreases, creating a detectable signature of reduced allele frequency.

Also known as de novo mutations. Heritable alterations in the genome of a microorganism that arise spontaneously during replication or repair and were not previously present in the population or acquired from external sources of genetic material.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Cite this article.

Shepherd, M.J., Fu, T., Harrington, N.E. et al. Ecological and evolutionary mechanisms driving within-patient emergence of antimicrobial resistance. Nat Rev Microbiol (2024). https://doi.org/10.1038/s41579-024-01041-1

Download citation

Accepted : 13 March 2024

Published : 30 April 2024

DOI : https://doi.org/10.1038/s41579-024-01041-1

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

research paper on antibiotic resistance

VIDEO

  1. Antimicrobial Resistance: Physician's Perspective

  2. #AntimicrobialResistance: How health workers can help slow antimicrobial resistance (part 1)

  3. #AMRSOS: Our mission to prevent antimicrobial resistance

  4. Bacterial soundtracks revealed by graphene membrane

  5. Antibiotic Resistance

  6. Antibiotic Resistance and How Bacteria May be The Solution

COMMENTS

  1. Antibiotic resistance: a rundown of a global crisis

    Introduction. Antibiotic resistance is ancient and the "resistome" is a dynamic and mounting problem. Causes of the global resistome are overpopulation, enhanced global migration, increased use of antibiotics in clinics and animal production, selection pressure, poor sanitation, wildlife spread, and poor sewerage disposal system. 1, 2 Antibiotic treatment is one of the main approaches of ...

  2. Global burden of bacterial antimicrobial resistance in 2019: a

    Discussion. The global burden associated with drug-resistant infections assessed across 88 pathogen-drug combinations in 2019 was an estimated 4·95 million (95% UI 3·62-6·57) deaths, of which 1·27 million (0·911-1·71) deaths were directly attributable to drug resistance.

  3. Molecular mechanisms of antibiotic resistance revisited

    Antibiotic resistance is a global health emergency, with resistance detected to all antibiotics currently in clinical use and only a few novel drugs in the pipeline. ... A Correction to this paper ...

  4. Antibiotic resistance in the environment

    Antibiotic resistance is a global health challenge, involving the transfer of bacteria and genes between humans, animals and the environment. Although multiple barriers restrict the flow of both ...

  5. Antimicrobial resistance

    Microbes can evolve antimicrobial resistance when, for example, populations are subjected to selective pressure through drug use in medicine. Latest Research and Reviews

  6. Global trend of antimicrobial resistance in common bacterial pathogens

    1. Introduction. Since the discovery of penicillin in the 1920s, antimicrobial drugs have been regarded as the panacea to cure bacterial infections and have reduced the global burden of common infectious diseases (Van Boeckel et al., 2014, WHO, 2014); however, the widespread use of antimicrobial drugs is a major driving force for antibiotic resistance (Klein et al., 2018b).

  7. Antibiotic Resistance: Current Perspectives

    Antibiotic resistance is one of the most serious challenges that the world is currently facing. The number of people succumbing to drug-resistant infections is increasing every day, but the rate of drug discovery has failed to match the requisite demands. Most of the currently known antibiotics target the three essential pathways of central dogma. However, bacteria have evolved multiple ...

  8. Understanding and overcoming antibiotic resistance

    In a PLOS Biology article, scientists show that intracellular expression of an antibiotic-metabolizing enzyme in a non-pathogenic strain of bacteria can provide resistance to the pathogen Streptococcus pneumoniae when the two types of bacteria are grown together, both in vitro and in vivo [ 3 ]. The expression of the metabolizing enzyme in the ...

  9. Rising antimicrobial resistance: an evolving epidemic in a pandemic

    While the second wave of the COVID-19 pandemic is sweeping through India, causing unprecedented human tragedy, the prescription of a plethora of unnecessary antibiotics—without regard for the potential for increased antimicrobial resistance—is widespread and unchecked. In The Lancet Microbe, Clark Russell and colleagues1 highlight the overuse of antimicrobials during the first wave of ...

  10. Perspectives on systematic generation of antibiotic resistance with

    Antibiotic resistance is a significant financial burden too for the entire planet as without any effective antimicrobial agents to treat bacterial illnesses, the worldwide economic burden would be around $120 trillion ($3 trillion annually), approximately similar to the current annual US health-care spending (Aslam et al., 2018). 2.5.

  11. (PDF) Antibiotic resistance: The challenges and some emerging

    Background: Antibiotic resistance is currently the most serious global threat to the effective treatment of bacterial infections. Antibiotic resistance has been established to adversely affect ...

  12. Antimicrobial resistance, mechanisms and its clinical significance

    Abstract. Antimicrobial agents play a key role in controlling and curing infectious disease. Soon after the discovery of the first antibiotic, the challenge of antibiotic resistance commenced. Antimicrobial agents use different mechanisms against bacteria to prevent their pathogenesis and they can be classified as bactericidal or bacteriostatic.

  13. Prevention of antibiotic resistance

    But antibiotics are finite resources and misuse has led to antibiotic resistance and reduced efficacy within just a few years of introduction of each new antibiotic. ... Scopus and Ebsco searched for 2000-17. The broader research field "antibiotic OR antimicrobial resistance" gave 431,335 hits. Narrowing the search criteria to "Prevention of ...

  14. Antibiotic resistance

    The emergence of resistant infections caused by these bacteria has led to mortality and morbidity and there is an urgent need to find solutions to combat bacterial resistance [6]. In this paper, the authors discuss some mechanisms of antibiotic resistance such as changing the antibacterial agent's uptake and biofilm formation as well as a wide ...

  15. Antibiotic resistance: The challenges and some emerging strategies for

    Background. Antibiotic resistance is currently the most serious global threat to the effective treatment of bacterial infections. Antibiotic resistance has been established to adversely affect both clinical and therapeutic outcomes, with consequences ranging from treatment failures and the need for expensive and safer alternative drugs to the cost of higher rates of morbidity and mortality ...

  16. (PDF) Antibiotic Resistance

    Antibiotic susceptibility testing against 47 antibiotics showed that the strain was 4 to >500 times more resistant to sulfonamides, quinolones, tetracyclines, macrolides, and the aminoglycoside ...

  17. Ecological and evolutionary mechanisms driving within-patient emergence

    The antimicrobial resistance (AMR) crisis threatens to endanger modern medicine within the next 20-30 years 1, with an estimated 4.95 million deaths associated with bacterial AMR in 2019 (ref. 2

  18. Prevention of antibiotic resistance

    The broader research field "antibiotic OR antimicrobial resistance" gave 431,335 hits. Narrowing the search criteria to "Prevention of antibiotic OR antimicrobial resistance" resulted in 1062 remaining titles. Of these, 622 were unique titles. ... Paper context. Antibiotic resistance is a threat to global security alongside terrorism ...

  19. A Brief History of Antimicrobial Resistance

    Antibiotics are derived from the evolutionary arms race between microbes and their ecological competitors (fellow microbes, fungi, plants, and animals), and, as a result, the emergence of resistance is entirely predictable. As swiftly as we claimed new victories, microbes began evading our latest weapons, altering their cell walls, upregulating ...

  20. Antibiotic resistance: Insights from evolution experiments and

    Abstract. Antibiotic resistance is a growing public health problem. To gain a fundamental understanding of resistance evolution, a combination of systematic experimental and theoretical approaches is required. Evolution experiments combined with next-generation sequencing techniques, laboratory automation, and mathematical modeling are enabling ...

  21. Antimicrobial Resistance and Its Drivers—A Review

    In addition, the Combating Antibiotic Resistant Bacteria Biopharmaceutical Accelerator (CARB-X) offers financial support to companies that are developing novel and potentially effective solutions to antibiotic resistance . However, more research is required to obtain new insights into the fundamental mechanisms of resistance, gene transfer, and ...