Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Nov 18.
Published in final edited form as: Lung. 2024 May 21;202(3):223–232. doi: 10.1007/s00408-024-00700-7

Phage Therapy for Respiratory Infections: Opportunities and Challenges

Arya Khosravi 1,3, Qingquan Chen 1, Arne Echterhof 1, Jonathan L Koff 2, Paul L Bollyky 1
PMCID: PMC11570333  NIHMSID: NIHMS2033666  PMID: 38772946

Abstract

We are entering the post-antibiotic era. Antimicrobial resistance (AMR) is a critical problem in chronic lung infections resulting in progressive respiratory failure and increased mortality. In the absence of emerging novel antibiotics to counter AMR infections, bacteriophages (phages), viruses that infect bacteria, have become a promising option for chronic respiratory infections. However, while personalized phage therapy is associated with improved outcomes in individual cases, clinical trials demonstrating treatment efficacy are lacking, limiting the therapeutic potential of this approach for respiratory infections. In this review, we address the current state of phage therapy for managing chronic respiratory diseases. We then discuss how phage therapy may address major microbiologic obstacles which hinder disease resolution of chronic lung infections with current antibiotic-based treatment practices. Finally, we highlight the challenges that must be addressed for successful phage therapy clinical trials. Through this discussion, we hope to expand on the potential of phages as an adjuvant therapy in chronic lung infections, as well as the microbiologic challenges that need to be addressed for phage therapy to expand beyond personalized salvage therapy.

Keywords: Bacteriophage, Phage therapy, Antimicrobial resistance, Chronic infections

Battling Antimicrobial Resistance (AMR) in Chronic Lung Infections

AMR Infections are a rising global threat, with over 4.9 million associated deaths estimated in 2019 [1]. Lower respiratory tract infections, in particular chronic lung infections, have one the highest burden of AMR infections and are a leading cause of death worldwide [1, 2]. Individuals with defects in structural airways and/or compromised immunity are at heightened risk for AMR chronic infections. This includes people with cystic fibrosis (PwCF), chronic obstructive pulmonary disease (COPD), non-CF bronchiectasis, and lung transplant recipients with chronic rejection [3-6]. Chronic lung infections are marked by episodes of acute exacerbations, distinguished by increased respiratory distress, eventually resulting in progressive respiratory failure and increased mortality [7, 8].

Antibiotic management strategies for chronic respiratory infections include the recurrent use of prophylactic or suppressive antibiotics to prevent exacerbations and the use of targeted antibiotics when exacerbations occur [9-11]. Despite these measures, once established, chronic infections are difficult, if not impossible, to eradicate [12, 13]. Further, recurrent antibiotic exposure often selects for antimicrobial resistant pathogens, limiting the effectiveness of this intervention. As such, chronic lung infections continue to be a major cause of progressive lung failure and death among patients with underlying pulmonary disease.

Rising AMR has Sparked a Renewed Interest in Bacteriophage (Phage) Therapy

Phage therapy (PT) is a potential alternative or adjuvant therapy to conventional antibiotics for managing AMR infections. Lytic phages are viruses that infect bacteria, hijack their cellular machinery to replicate, and eventually lyse their host bacteria to release newly formed viral progeny [14]. In the pre-antibiotic era, phages were used globally to treat infectious diseases [15]. However, the enthusiasm for PT diminished as concerns grew about inconsistent results and reliance on case studies rather than rigorous standardized trials [16]. The development of antibiotics, which early studies hinted at improved outcomes compared to PT, effectively supplanted phages in treating infectious diseases [17, 18]. Presently, insufficient development of new antimicrobial agents to counter the rise of AMR, as well as preclinical studies that demonstrated phage efficacy against drug-resistant infections, has renewed interest in PT [19, 20].

PT is Associated with Positive Outcomes in Individual Cases

In the US, the use of PT is restricted to compassionate use for patients with progressive or fulminant infections recalcitrant to all other FDA-approved therapies. If these criteria are met, FDA approval for PT under the emergency or expanded access investigational new drug (eIND or eaIND, respectively) pathways may be granted [21]. Despite the restricted use, numerous cases, including international studies, have suggested PT to be a safe and effective intervention [22-24].

A retrospective Belgian case series described 100 consecutive cases of phage therapy for difficult-to-treat pulmonary, bone, skin and soft tissue infections [25]. Those authors reported that 77% of cases demonstrated clinical improvement, and 61% achieved eradication of the pathogen in question. While it seems likely that there is under-reporting of unsuccessful cases, together, these data indicate that individualized PT can be successful.

Respiratory infections are the second most common indication for phage therapy [26]. A recent review evaluated 20 cases of respiratory infections utilizing PT, including two clinical trials involving four participants [27]. These cases included ventilator-associated pneumonia (n = 8, 40%), pulmonary infections following solid organ transplantation (n = 6, 30%), and PwCF (n = 4, 20%). The most common pathogens were Acinetobacter baumannii (n = 6, 30%) and Pseudomonas aeruginosa (n = 4, 20%). Among those 20 cases, there were 16 instances of infection resolution (4 of which included eradication of the pathogen) and 4 treatment failures [27]. Additional case series have reported positive results of compassionate phage therapy in PwCF, including treatment for non-tuberculous mycobacteria (NTM) and P. aeruginosa (Table 1) [28-30]. These cases demonstrate PT is likely safe and potentially effective salvage treatment in patients with advanced pulmonary infections resistant to all other approved therapeutics.

Table 1.

Summary of clinical case studies involving phage therapy in patients with pulmonary infections from 2023 to 2019

Article Pathogen/Indication Phage treatment Clinical outcome
Chan 2023 (preprint) [30] P. aeruginosa (MDR n = 6, PDR n = 3) pulmonary infections in CF patients (n = 9) Inh LPS-5 (n = 3)
Inh LPS-5, TIVP-H6 (n = 4)
Inh LPS-5, TIVP-H6, OMKO1 (n = 2)
Clinical improvement with increased ppFEV1 and reduction of bacterial burden n = 9
Haidar, 2023
PMID: 36864824
B. multivorans pneumonia and bacteremia in CF patient (n = 1) Inh Bch7 Clinical failure and death, reduction of bacterial burden
Nick, 2022
PMID:35568033
M. abscessus, MDR P. aeruginosa and MRSA pulmonary infection in CF patient with advanced bronchiectasis (n = 1) IV engineered BPsΔ33HTH_HRM10 and D29_HRMGD40 targeting M. abscessus Clinical improvement and listed for lung transplant, reduction of bacterial burden
Winzigh, 2022
PMID: 36568830
Achromobacter (MDR and PDR) lung colonization in CF patient (n = 1)* Inh Achr-1 and Achr-2 Clinical improvement, reduction of bacterial burden
Rao, 2022
PMID: 34662188
A. baumannii (CRAB) and P. aeruginosa ventilation-associated pneumonia (n = 1) Inh and IV AbW4932ø1, AbW4878ø1 targeting Acinetobacter Clinical improvement and bacterial eradication
Dedrick, 2022
PMID: 35676823
NTM pulmonary infection including M. abscessus (n = 17), M. avium complex (n = 1), BCG (n = 1). Of these patients, 17 had CF.* Inh and IV Muddy (n = 7)
Inh and IV BPsΔ33HTH_HRMGD03 (n = 3)
Inh an IV BPsΔ33HTH_HRM10, Itos (n = 2)
Inh and IV BPsΔ33HTH_HRMGD03, D29_HRMGD40 (n = 1)
Inh and IV BPsΔ33HTH_HRM1, D29_HRMGD40 (n = 1)
Inh and IV BPsΔ33HTH_HRMGD03, Itos (n = 1)
Inh and IV Muddy, D29, FionnbharthΔ43Δ45, Fred313cpmΔ33 (n = 1)
Inh and IV BPsΔ33HTH_HRM10, Muddy, ZoeJΔ45 (n = 2)
Favorable or partial results, bacterial eradication: n = 10

Complex, inconclusive or incomplete responses: n = 5

No evident clinical improvement: n = 4
Gainey, 2020
PMID: 32662948
Achromobacter (MDR and PDR) lung colonization inn pediatric CF patient (n = 1) IV Ax2CJ45ϕ2 Clinical improvement, bacterial eradication
Aslam, 2019
PMID:31207123
B. dolosa (MDR) pulmonary colonization in CF patient followed by a bilateral lung transplant and recurrent B. dolosa pneumonia (n = 1)
P. aeruginosa pneumonia (MDR) in a bilateral lung transplant patient (n = 1)

P. aeruginosa (MDR), M. abscessus lung infection in a post-transplant non CF patient with bronchiectasis (n = 1)
IV BdPF16phi4281

IV 4 different cocktails: AB-PA01, AB-PA01 m1, Navy cocktail 1, Navy cocktail 2
IV AB-PA01 targeting P. aeruginosa
Initial clinical improvement, but died 11 months after transplantation, no bacterial eradication

Clinical improvement and bacterial eradication

Clinical improvement and bacterial eradication
Law, 2019
PMID:31102236
P. aeruginosa (MDR) pulmonary infection in CF patient on waitlist for lung transplant (n = 1) IV AB-PA01 Clinical improvement, underwent bilateral lung transplant successfully

Cases reported in the USA during this time period

Antibiotic use was not reported in cases with asterisks (*)

All other cases were co-treated with antibiotics

CF cystic fibrosis, NTM non tuberculosis mycobacterium, MDR multi-drug-resistant, PDR pan-drug-resistant, CRAB carbapenem resistant A. baumannii, MRSA Methicillin resistant Staphylococcus aureus, BCG Bacillus Calmette-Guérin, PFU plaque forming units, N/A not available, ppFEV1 percent predicted FEV1, IV intravenously injected, Inh inhaled nebulized therapy, AB-PA01 cocktail: Pa193, Pa204, Pa222 and Pa223, AB-PA01 m1 cocktail: Pa193, Pa204, Pa222, Pa223 and Pa176, Navy cocktail 1: Paφ1, PaSKWφ17, and PaSKWφ22, Navy cocktail 2: PaATFφ1 and PaATFφ3

The positive outcomes seen with phage treatment in these studies hint at the potential of PT beyond compassionate use. However, the heterogenicity of cases in these studies, with a range in disease severity, variety of infectious pathogens, lack of standardization or a control group for comparison, and prevalent concomitant use of antibiotics (which is often required for eIND cases), precludes conclusive judgment on the efficacy of PT [25, 31]. To broaden the application of PT beyond compassionate use requires clinical trials to evaluate the safety and effectiveness of PT for respiratory infections. While these trials are currently underway, they have not yet been published.

In the absence of clinical trials to rigorously assess the virtue of PT, we draw insights from in vitro, preclinical, and case studies. By scrutinizing the distinct microbiological challenges to treat chronic lung infections, including AMR, biofilm formation, and polymicrobial disease, we examine how PT addresses these specific obstacles. We also discuss the intrinsic limitations of PT, including widespread phage resistance. Through this framework, we assess the advantages and deficiencies of PT to treat chronic lung infections and pinpoint crucial areas for improvement to expand its application beyond compassionate use.

Widespread Resistance Impedes Pathogen Clearance in Chronic Pulmonary Infections

As discussed above, one of the hallmarks of chronic pulmonary infections is the development of antibiotic resistance. One option may be to substitute antibiotics with PT, which has demonstrated clinical utility [27]. However, phage monotherapy carries the potential of selecting for phage resistance. Another possibility is to combine PT with antibiotics, which, in addition to additive bacterial killing effects, may prevent resistance (antibiotic and phage) through synergistic activity [32]. Indeed, numerous studies have demonstrated in vitro phage-antibiotic synergy (PAS) [33-36]. Evidence of PAS has also been shown in animal models, including evidence that combined therapy re-sensitizes resistant bacteria to antibiotics [37-39]. It has been more challenging to ascertain PAS in clinical cases, as trials comparing phage versus antibiotic monotherapy to combined therapy have not been initiated. However, some clinical reports hint at the potential of PAS. This includes demonstrating that phage-antibiotic combination therapy is associated with the absence of development of antibiotic resistance as well as re-sensitization of AMR strains to antibiotics [22, 31, 40, 41]. Altogether, these studies encapsulate the potential of adjuvant PT to combat AMR pulmonary infections.

There are several examples by which PAS counters AMR. Co-treatment of bacterial cultures with phages and antibiotics reduces selection for antibiotic resistance, compared to mono-therapy alone [42-44]. Further, phage treatment selects for phage resistance which may re-sensitize antibiotic resistant bacteria through an evolutionary tradeoff. This includes downregulation of phage target receptors involved in drug efflux to prevent phage binding and absorption [45, 46]. While this leads to decreased phage predation, it also results in impaired drug efflux and enhanced antibiotic susceptibility [45, 46]. Phage-mediated bacterial membrane stress, including capsule mutatgenesis, may also re-sensitize resistant bacteria by enhancing antibiotic penetration and killing [35, 46, 47]. Reciprocally, antibiotic-induced membrane stress promotes phage-induced killing by enhancing phage attachment, absorption, and virion production [33, 35, 47]. Although these mechanisms through which PAS enhances antimicrobial susceptibility have been consistently demonstrated in vitro, whether they are observed in patient populations remains a critical question.

A more sobering possible consequence of phage-antibiotic combination therapy, however, is antagonism. Phage-antibiotic antagonism (PAA) was observed as early as the 1940s, when Jones et al. reported specific classes of antibiotics that inhibited phage killing in Escherichia coli and Staphylococcus aureus [36]. Multiple reports have since reproduced these findings, which show reduced viral production, delayed lysis time, and impaired bacterial killing when phages are combined with antibiotics [48, 49]. Several mechanisms have been proposed to explain the mechanism of PAA. Phages rely on the molecular machinery of its bacterial host for genome replication, virion production, and cell lysis [48]. Consistent with this, antibiotics that inhibit bacterial DNA gyrase and ribosomes, or that reduce bacteria below the phage proliferation threshold, impair phage production [50-53]. Antibiotics may also promote phage resistance. Varied mechanisms have been described, including antibiotic enhancement of Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR), a form of bacterial immunity that protects against phage predation [54-57]. Further, while selection for phage resistance has been associated with a fitness tradeoff leading to enhanced antibiotic susceptibility, Burmeister et al. revealed that some phage-resistant mutants actually demonstrated enhanced resistance to antibiotics [58]. Given the potential of PAA, staggering treatment of phage and antibiotics, rather than co-administration, has been proposed as a method to limit antagonism. However, this approach has had mixed results [59-61].

The final potential consequence of combined therapy is additive or no effect [47]. Here, phages and antibiotics work (or fail) independent of one another. Screening phages with antibiotics largely demonstrates independent activity [62]. Thus, while PT has a promising potential to reverse AMR, this phenomenon is not universally found. The unpredictable phage-antibiotic interactions impede the empiric use of combination therapy. Instead, similar to antibiograms, which delineate antibiotic susceptibility, optimizing PT may require both a “phagogram” and “synogram”, which will reveal phage susceptibility and antibiotic synergy, respectively, for each bacterial isolate [31]. This, however, reveals a new challenge, in that phage activity demonstrated in vitro may be diminished, if not completely absent, when tested in physiologic conditions [32]. This inconsistency in phage activity recalls Gunther Stent’s early observation “just why bacteriophages, so virulent in their antibacterial action in vitro, proved so impotent in vivo has never been adequately explained” [15].

Formation of Bacterial Biofilms in Chronic Lung Infections Promote Persistent Disease

The CYstic Fibrosis bacterioPHage Study at Yale (CYPHY) is a phase 2 study evaluating the use of an inhaled single phage (YPT-01) phage in PwCF (ClinicalTrials.gov ID NCT04684641). YPT-01 was previously shown to kill P. aeruginosa and select fitness-defective mutants susceptible to antibiotic killing. Patients selected for the trial were each found to have phage susceptible isolates upon in vitro culture. A preliminary review of the data available on ClinicalTrials.gov suggests that while there were no safety concerns, the primary endpoint was not met as PT did not decrease bacterial titers. While these results and any analyses of them have yet to be published, the difference between in vitro and in vivo outcomes is striking.

One factor relevant to in vivo infections, but is not typically assessed by in vitro phage susceptibility studies, is bacterial biofilms. In contrast to planktonic bacterial growth, which is characterized by free-floating bacteria, biofilms are comprised of densely packed microbial cells contained within a protective matrix of extracellular polymeric substances (EPS) [63]. Biofilm matrix substrates serve as an anchor to the respiratory epithelium, mediating bacterial adherence and resistance to mucociliary clearance [64, 65]. This extracellular matrix also shields bacteria from eradication by host immune cells and antibiotics. Indeed, bacteria grown in biofilms may demonstrate 100–1000-fold increased antibiotic resistance relative to planktonic growth conditions [66, 67]. Several mechanisms by which biofilm promotes antibiotic resistance have been proposed. This includes the EPS matrix serving as a protective sieve, impeding antibiotic penetration and directly sequestering antibiotics through electrostatic interactions [68-70]. The densely packed microbial community within the biofilm also facilitates horizontal gene transfer, which includes the spread of AMR genes [68, 69]. The expansion of persister cells, a subpopulation of bacteria selected due to nutrient scarcity and varying oxygen gradient throughout the biofilm, also contributes to persistent infection despite antibiotic treatment [67, 71]. Displaying altered metabolism and delayed growth rates, these cells essentially remain dormant and unaffected by antibiotics. Once antibiotic levels diminish, persister cells can repopulate the biofilm [71]. Patients with chronic biofilm-forming lung infections thus remain colonized despite frequent, intensive antibiotic and airway clearance treatments [12, 13].

Phages, used independently or in conjunction with antibiotics, may resolve biofilm infections [48, 61, 72]. Clinically, PT has successfully managed biofilms among patients with chronic wounds or prosthetic device infections [73]. PT is also associated with clinical improvement among patients susceptible to biofilm-forming chronic lung infections (Table 1) [24, 74, 75]. Multiple mechanisms by which phages promote biofilm killing have been ascribed. Similar to AMR strains, the combination of phages and antibiotics demonstrates synergistic activity against biofilms. Some phages express depolymerases, enzymes that degrade polysaccharides including EPS, mediating biofilm breakdown and penetration of both phages and antibiotics [72, 76]. Further, in contrast to antibiotics, phages demonstrate effective activity against persister cells in vitro [77, 78].

Despite the potential of phages to treat biofilm infections, challenges remain. Similar to the uncertainty of phage-antibiotic combinations in addressing AMR, there are concerns regarding which antibiotic classes and treatment order (combined or sequential) that synergize best with phages to dissolve biofilms [79-82]. Phage depolymerases are substrate selective, requiring screening to identify phages capable of penetrating the biofilm matrix [83, 84]. Biofilms may also sequester phages, inhibiting phage diffusion through the biofilm [85]. While matrix-bound phage remain active against surface microbes, bacteria within the biofilm are protected from phage killing [83, 85, 86]. Phages have been shown to kill persister cells in vitro. However, when tested in nutrient-restricted conditions as seen within biofilms, phage activity against persister cells is lost [87, 88]. In nutrient-deplete conditions, phages may actually undergo a hibernation or pseudolysogeny phase within bacteria, during which phage production is halted until growth conditions improve [87-89]. Recently, a study to identify phages capable of killing deep dormant cells in nutrient depleted growth conditions was undertaken. While the majority of phages demonstrated no activity, one phage was isolated capable of killing dormant P. aeruginosa [90]. No phages capable of killing dormant E. coli, however, were isolated. This finding further emphasizes the need to study phage-antibiotic-biofilm interactions in conditions that reflect the infectious environment. Thus, while there is a promising potential for phages to eradicate persistent lung infections, multiple challenges, including screening phages for anti-biofilm and anti-persister cell activity, remain.

Polymicrobial Lung Infections Pose Significant Challenge to Treatment Approaches

A key obstacle in treating chronic pulmonary infections is its polymicrobial and polyclonal nature. PwCF, in particular, demonstrate diverse airway colonization harboring various species and strains with distinct pathogenicity and antibiotic susceptibility [91, 92]. This diversity complicates both diagnosis and treatment strategies. Antibiotic treatment in these cases is often empiric with broad antimicrobial coverage [8].

Phage specificity to distinct bacterial species, if not strains, limits their empiric use for both mono and polymicrobial infections. Bacterial-phage co-evolution “training” protocols or engineered phages may broaden host range [93, 94]. Unfortunately, these efforts are laborious and time-consuming, resulting in delayed treatment. An alternative approach is using phage cocktails that combine multiple phages to address both polymicrobial infections and limit phage resistance. However, as with phage-antibiotic combinations, phage-phage interactions can either be synergistic or antagonistic, which is difficult to predict [95]. Moreover, the optimum number of phages and whether they should be administered serially or in concert are unclear [96].

Despite these challenges, the use of PT in patients with polymicrobial respiratory infections has demonstrated clinical improvement. In a case series of three patients with polymicrobial MDR pneumonia, a phage cocktail administered by inhalation resulted in clinical improvement and elimination of respiratory pathogens in each case [97]. Moreover, patients with polymicrobial infections who received PT targeting a dominant isolated pathogen also may demonstrate clinical improvement (Table 1) [27, 98]. However, even when targeting a specific pathogen, phage strain selectivity limits empiric use. This includes the treatment of non-tuberculosis mycobacterium (NTM), a major cause of chronic pulmonary infections among susceptible patients. Recent reports have demonstrated success in PT against NTM pulmonary infections, including M. abscessus [28, 40]. However, no lytic phages have been identified that target and kill smooth colony variants of M. abscessus, which make up over 45% of clinical isolates, constraining PT's therapeutic potential [28]. While there is evidence of PT success in polymicrobial infections, the strain specificity of phages is a potential limiting factor for the treatment of chronic infections.

Phage Therapy: A New Solution Comes with Unique Challenges

There is promising evidence that phages may help address challenges in treating AMR and biofilm infections. Despite strain selectivity, PT has demonstrated clinical improvement among patients with polymicrobial infections. There is also an abundant supply of natural phages that can be bio-prospected to target resistant organisms. This process is exponentially faster and cheaper than generating new antibiotics with minimal side effects and off-target consequences.

While phages offer potential solutions to limitations in treating chronic lung infections with antibiotics, it poses new challenges. Similar to antibiotics, bacteria possess multiple, diverse mechanisms to resist phage killing [27, 55, 99]. Efforts to counter this resistance include combining multiple phages into a treatment cocktail [100]. However, the optimal cocktail design to prevent resistance while also avoiding phage-phage antagonism is unclear. Further, despite the use of phage cocktails, resistance may still occur, requiring additional screening and the inclusion of additional phages [22]. Alternatively, phages can be chosen for their potential to select for resistant mutants that are avirulent or have enhanced antibiotic susceptibility [27, 100]. Unfortunately, both approaches require laborious and time-consuming screening.

It is important to note that phage resistance is not universally seen. Clinical and in vitro selection for phage resistance in NTM appears to be rare, which suggests these infections may be well suited for PT [40, 101]. Further, phage resistance may not always indicate treatment failure as it can be associated with enhanced antibiotic susceptibility or compromised pathogen fitness. In a review of 20 cases of PT for respiratory infections, phage resistance was identified in only 30% of cases [27]. However, among patients in which phage resistance was detected, some continued to demonstrate clinical improvement [27, 102]. As such, the clinical impact of phage resistance remains unclear.

Even in the absence of resistance, other factors may hinder the effectiveness of phage treatment. This includes unpredictable pharmacokinetics and pharmacodynamics, uncertainty about dosing and delivery, formulation, and storage. In particular, a key challenge in treating pulmonary biofilm infections is drug delivery. Intravenous phage administration is commonly utilized but has poorly predicted biodistribution. Alternatively, nebulized therapy, in which aerosolized droplets containing phages are inhaled, provides targeted delivery. However, the method of nebulization may compromise phage integrity as well as limit the area of distribution throughout the lung [103, 104]. The development of phage-neutralizing antibodies poses another challenge, which may further impede phage activity [105]. However, neutralizing antibodies have also been detected in successful cases of respiratory infections managed by PT, raising questions about their significance [40].

In addition to these distinct limitations, the lack of successful clinical trials compels a sobering pause. Is PT the solution to chronic lung infections? The current enthusiasm is inspired by success in individual cases; however, these studies are heterogeneous and non-standardized. Will PT demonstrate efficacy when tested in rigorous clinical trials, or will inconsistent results continue to stymie this approach? The preliminary review data from the CYPHY, in which PT for treatment of P. aeruginosa in PwCF did not meet the primary endpoint, indicates that more work needs to be done to validate PT as a viable therapeutic option for pulmonary infections.

Conclusions

The rise of AMR, particularly in patients with chronic pulmonary infections, has revived interest in PT. Numerous case studies have demonstrated phage effectiveness in treating chronic pulmonary infections. However, as discussed in this review, for PT to be broadly adopted beyond salvage therapy, numerous challenges need to be overcome. Yet, even with these advances, there may be a larger potential challenge ahead. In his Nobel address commemorating the discovery of penicillin, Fleming anticipated the emergence of antimicrobial resistance, which he attributed to antibiotic under dosing and misuse. Chronic lung infections are major sources of AMR, partly due to the nature of the disease requiring repeated antibiotic exposure, potentiating antibiotic resistance. Thus, one possible consequence of increased utilization of PT for persistent pulmonary infections would be phage overuse and misuse, as seen with antibiotics, leading to resistance. While the consequences of phage resistance on clinical outcomes is variable, it is reasonable to assume that rising resistance over time will adversely impact treatment. To ensure PT does not follow the same destiny Fleming predicted for antibiotics, guiding principles to navigate the appropriate use of PT will be necessary. Phages have not yet been shown to be a “magic bullet” for eradicating chronic pulmonary infections, yet there is reason for optimism. The recent development of CF modulator therapy has dramatically altered disease progression in PwCF. By correcting the malfunctioning CFTR protein, modulator therapy has resulted in less accumulation of airway mucus, resulting in improved respiratory function, decreased pulmonary exacerbations, and even reduced pulmonary pathogen burden [106]. P. aeruginosa pulmonary infections continue to persist in many of these patients [107, 108]. However, the improved source control achieved by modulator therapy may potentiate pathogen eradication with PT alone or in combination with antibiotics. Finally, if optimized, phage-antibiotic synergy, will fundamentally reshape the potential to effectively and definitively treat chronic pulmonary infections.

Funding

This work was supported by grants R01AI12492093, K24AI166718-01A1 R21GM147838 R01HL148184-01, R01AI12492093 and grants from Stanford SPARK, the Emerson Collective, the Cystic Fibrosis Foundation (CFF) and Doris Duke Charitable Foundation (DDCF).

Footnotes

Declarations

Competing interests The authors declare no competing interests.

References

  • 1.Murray CJL, Ikuta KS, Sharara F et al. (2022) Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. The Lancet 399:629–655. 10.1016/S0140-6736(21)02724-0 [DOI] [Google Scholar]
  • 2.The top 10 causes of death. https://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death. Accessed 17 Nov 2023
  • 3.Drevinek P, Canton R, Johansen HK et al. (2022) New concepts in antimicrobial resistance in cystic fibrosis respiratory infections. J Cyst Fibros 21:937–945. 10.1016/j.jcf.2022.10.005 [DOI] [PubMed] [Google Scholar]
  • 4.Smith D, Gill A, Hall L (2021) Prevalence, pattern, risks factors and consequences of antibiotic resistance in COPD: a systematic review. J Chronic Obstr Pulm Dis. 10.1080/15412555.2021.2000957 [DOI] [Google Scholar]
  • 5.Nosotti M, Tarsia P, Morlacchi LC (2018) Infections after lung transplantation. J Thorac Dis 10:3849–3868. 10.21037/jtd.2018.05.204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Inchingolo R, Pierandrei C, Montemurro G et al. (2021) Antimicrobial resistance in common respiratory pathogens of chronic bronchiectasis patients: a literature review. Antibiot (Basel) 10:326. 10.3390/antibiotics10030326 [DOI] [Google Scholar]
  • 7.Goss CH (2019) Acute pulmonary exacerbation in cystic fibrosis. Semin Respir Crit Care Med 40:792–803. 10.1055/s-0039-1697975 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Flume PA, Mogayzel PJ, Robinson KA et al. (2009) Cystic fibrosis pulmonary guidelines: treatment of pulmonary exacerbations. Am J Respir Crit Care Med 180:802–808. 10.1164/rccm.200812-1845PP [DOI] [PubMed] [Google Scholar]
  • 9.Sanders DB, Bittner RCL, Rosenfeld M et al. (2010) Failure to recover to baseline pulmonary function after cystic fibrosis pulmonary exacerbation. Am J Respir Crit Care Med 182:627–632. 10.1164/rccm.200909-1421OC [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ramsey BW, Pepe MS, Quan JM et al. (1999) Intermittent administration of inhaled tobramycin in patients with cystic fibrosis. N Engl J Med 340:23–30. 10.1056/NEJM199901073400104 [DOI] [PubMed] [Google Scholar]
  • 11.Taccetti G, Francalanci M, Pizzamiglio G et al. (2021) Cystic fibrosis: recent insights INTO inhaled antibiotic treatment and future perspectives. Antibiot (Basel) 10:338. 10.3390/antibiotics10030338 [DOI] [Google Scholar]
  • 12.Langton Hewer SC, Smyth AR (2017) Antibiotic strategies for eradicating Pseudomonas aeruginosa in people with cystic fibrosis. Cochrane Database Syst Rev. 10.1002/14651858.CD004197.pub5 [DOI] [Google Scholar]
  • 13.Sangiovanni S, Morales EI, Fernández-Trujillo L (2021) Inhaled tobramycin for chronic infection with Pseudomonas aeruginosa in non-cystic fibrosis bronchiectasis: a systematic review and meta-analysis. Respir Med 176:106283. 10.1016/j.rmed.2020.106283 [DOI] [PubMed] [Google Scholar]
  • 14.Kortright KE, Chan BK, Koff JL, Turner PE (2019) Phage therapy: a renewed approach to combat antibiotic-resistant bacteria. Cell Host Microbe 25:219–232. 10.1016/j.chom.2019.01.014 [DOI] [PubMed] [Google Scholar]
  • 15.Summers WC (2012) The strange history of phage therapy. Bacteriophage 2:130–133. 10.4161/bact.20757 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Eaton MD, Bayne-Jones S (1934) Bacteriophage therapy: review of the principles and results of the use of bacteriophage in the treatment of infections. J Am Med Assoc 103:1769–1776. 10.1001/jama.1934.72750490003007 [DOI] [Google Scholar]
  • 17.Marcuk LM, Nikiforov VN, Scerbak JF et al. (1971) Clinical studies of the use of bacteriophage in the treatment of cholera. Bull World Health Org 45:77–83 [PMC free article] [PubMed] [Google Scholar]
  • 18.Monsur KA, Rahman MA, Huq F et al. (1970) Effect of massive doses of bacteriophage on excretion of vibrios, duration of diarrhoea and output of stools in acute cases of cholera. Bull World Health Org 42:723–732 [PMC free article] [PubMed] [Google Scholar]
  • 19.Smith HW, Huggins MB (1982) Successful treatment of experimental Escherichia coli infections in mice using phage: its general superiority over antibiotics. J Gen Microbiol 128:307–318. 10.1099/00221287-128-2-307 [DOI] [PubMed] [Google Scholar]
  • 20.Slopek S, Weber-Dabrowska B, Dabrowski M, Kucharewicz-Krukowska A (1987) Results of bacteriophage treatment of suppurative bacterial infections in the years 1981–1986. Arch Immunol Ther Exp (Warsz) 35:569–583 [PubMed] [Google Scholar]
  • 21.McCallin S, Sacher JC, Zheng J, Chan BK (2019) Current state of compassionate phage therapy. Viruses 11:343. 10.3390/v11040343 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Schooley RT, Biswas B, Gill JJ et al. (2017) Development and use of personalized bacteriophage-based therapeutic cocktails to treat a patient with a disseminated resistant Acinetobacter baumannii infection. Antimicrob Agents Chemother. 10.1128/aac.00954-17.10.1128/aac.00954-17 [DOI] [Google Scholar]
  • 23.Chan BK, Turner PE, Kim S et al. (2018) Phage treatment of an aortic graft infected with Pseudomonas aeruginosa. Evol Med Public Health 2018:60–66. 10.1093/emph/eoy005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dedrick RM, Guerrero-Bustamante CA, Garlena RA et al. (2019) Engineered bacteriophages for treatment of a patient with a disseminated drug resistant Mycobacterium abscessus. Nat Med 25:730–733. 10.1038/s41591-019-0437-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pirnay J-P, Djebara S, Steurs G et al. (2023) Retrospective observational analysis of the first one hundred consecutive cases of personalized bacteriophage therapy of difficult-to-treat infections facilitated by a belgian consortium. medRxiv. 10.1101/2023.08.28.23294728 [DOI] [Google Scholar]
  • 26.Strathdee SA, Hatfull GF, Mutalik VK, Schooley RT (2023) Phage therapy: from biological mechanisms to future directions. Cell 186:17–31. 10.1016/j.cell.2022.11.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Mitropoulou G, Koutsokera A, Csajka C et al. (2022) Phage therapy for pulmonary infections: lessons from clinical experiences and key considerations. Eur Respir Rev. 10.1183/16000617.0121-2022 [DOI] [Google Scholar]
  • 28.Dedrick RM, Smith BE, Cristinziano M et al. (2023) Phage therapy of mycobacterium infections: compassionate use of phages in 20 patients with drug-resistant mycobacterial disease. Clin Infect Dis 76:103–112. 10.1093/cid/ciac453 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chan BK, Stanley GL, Kortright KE et al. (2023) Personalized inhaled bacteriophage therapy decreases multidrug-resistant pseudomonas aeruginosa. MedRxiv. 10.1101/2023.01.23.22283996 [DOI] [Google Scholar]
  • 30.Wu N, Dai J, Guo M et al. (2021) Pre-optimized phage therapy on secondary Acinetobacter baumannii infection in four critical COVID-19 patients. Emerg Microbes Infect 10:612–618. 10.1080/22221751.2021.1902754 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Uyttebroek S, Chen B, Onsea J et al. (2022) Safety and efficacy of phage therapy in difficult-to-treat infections: a systematic review. Lancet Infect Dis 22:e208–e220. 10.1016/S1473-3099(21)00612-5 [DOI] [PubMed] [Google Scholar]
  • 32.Gu Liu C, Green SI, Min L et al. (2020) Phage-antibiotic synergy is driven by a unique combination of antibacterial mechanism of action and stoichiometry. MBio. 10.1128/mbio.01462-20 [DOI] [Google Scholar]
  • 33.Comeau AM, Tétart F, Trojet SN et al. (2007) Phage-antibiotic synergy (PAS): β-lactam and quinolone antibiotics stimulate virulent phage growth. PLoS ONE 2:e799. 10.1371/journal.pone.0000799 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen Q, Dharmaraj T, Cai PC et al. (2022) Bacteriophage and bacterial susceptibility, resistance, and tolerance to antibiotics. Pharmaceutics 14:1425. 10.3390/pharmaceutics14071425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bulssico J, PapukashvilI I, Espinosa L et al. (2023) Phage-antibiotic synergy: cell filamentation is a key driver of successful phage predation. PLoS Pathog 19:e1011602. 10.1371/journal.ppat.1011602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Jones D (1945) The effect of antibiotic substances upon bacteriophage 12. J Bacteriol 50:341–348 [Google Scholar]
  • 37.Huff WE, Huff GR, Rath NC et al. (2004) Therapeutic efficacy of bacteriophage and baytril (enrofloxacin) individually and in combination to treat colibacillosis in broilers1. Poult Sci 83:1944–1947. 10.1093/ps/83.12.1944 [DOI] [PubMed] [Google Scholar]
  • 38.Oechslin F, Piccardi P, Mancini S et al. (2017) Synergistic interaction between phage therapy and antibiotics clears Pseudomonas aeruginosa Infection in endocarditis and reduces virulence. J Infect Dis 215:703–712. 10.1093/infdis/jiw632 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Altamirano FLG, Kostoulias X, Subedi D et al. (2022) Phage-antibiotic combination is a superior treatment against Acinetobacter baumannii in a preclinical study. EBioMedicine. 10.1016/j.ebiom.2022.104045 [DOI] [Google Scholar]
  • 40.Nick JA, Dedrick RM, Gray AL et al. (2022) Host and pathogen response to bacteriophage engineered against Mycobacterium abscessus lung infection. Cell 185:1860–1874.e12. 10.1016/j.cell.2022.04.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hahn A, Sami I, Chaney H et al. (2023) Bacteriophage therapy for Pan-drug-resistant Pseudomonas aeruginosa in two persons with cystic fibrosis. J Investig Med High Impact Case Rep 11:23247096231188244. 10.1177/23247096231188243 [DOI] [Google Scholar]
  • 42.Kirby AE (2012) Synergistic action of gentamicin and bacteriophage in a continuous culture population of Staphylococcus aureus. PLoS ONE 7:e51017. 10.1371/journal.pone.0051017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Coulter LB, McLean RJC, Rohde RE, Aron GM (2014) Effect of bacteriophage infection in combination with tobramycin on the emergence of resistance in Escherichia coli and Pseudomonas aeruginosa biofilms. Viruses 6:3778–3786. 10.3390/v6103778 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Valério N, Oliveira C, Jesus V et al. (2017) Effects of single and combined use of bacteriophages and antibiotics to inactivate Escherichia coli. Virus Res 240:8–17. 10.1016/j.virusres.2017.07.015 [DOI] [PubMed] [Google Scholar]
  • 45.Chan BK, Sistrom M, Wertz JE et al. (2016) Phage selection restores antibiotic sensitivity in MDR Pseudomonas aeruginosa. Sci Rep 6:26717. 10.1038/srep26717 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Gordillo Altamirano F, Forsyth JH, Patwa R et al. (2021) Bacteriophage-resistant Acinetobacter baumannii are resensitized to antimicrobials. Nat Microbiol 6:157–161. 10.1038/s41564-020-00830-7 [DOI] [PubMed] [Google Scholar]
  • 47.Kim M, Jo Y, Hwang YJ et al. (2018) Phage-antibiotic synergy via delayed lysis. Appl Environ Microbiol 84:e02085–e2118. 10.1128/AEM.02085-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Abedon ST (2019) Phage-antibiotic combination treatments: antagonistic impacts of antibiotics on the pharmacodynamics of phage th erapy? Antibiotics 8:182. 10.3390/antibiotics8040182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Martin I, Morales S, Alton EWFW, Davies JC (2023) Lytic bacteriophage is a promising adjunct to common antibiotics across cystic fibrosis clinical strains and culture models of Pseudomonas aeruginosa infection. Antibiot (Basel) 12:593. 10.3390/antibiotics12030593 [DOI] [Google Scholar]
  • 50.Payne RJH, Jansen VAA (2002) Evidence for a phage proliferation threshold? J Virol 76:13123–13124. 10.1128/jvi.76.24.13123-13124.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Brock TD, Wooley SO (1963) Streptomycin as an antiviral agent: mode of action. Science 141:1065–1067. 10.1126/science.141.3585.1065 [DOI] [PubMed] [Google Scholar]
  • 52.Kever L, Hardy A, Luthe T et al. (2022) Aminoglycoside antibiotics inhibit phage infection by blocking an early step of the infection cycle. MBio. 10.1128/mbio.00783-22 [DOI] [Google Scholar]
  • 53.Alonso JC, Sarachu AN, Grau O (1981) DNA gyrase inhibitors block development of Bacillus subtilis bacteriophage SP01. J Virol 39:855–860. 10.1128/JVI.39.3.855-860.1981 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Cairns J, Becks L, Jalasvuori M, Hiltunen T (2017) Sublethal streptomycin concentrations and lytic bacteriophage together promote resistance evolution. Philoso Trans Royal Soc B Biol Sci 372:20160040. 10.1098/rstb.2016.0040 [DOI] [Google Scholar]
  • 55.Sorek R, Kunin V, Hugenholtz P (2008) CRISPR — a widespread system that provides acquired resistance against phages in bacteria and archaea. Nat Rev Microbiol 6:181–186. 10.1038/nrmicro1793 [DOI] [PubMed] [Google Scholar]
  • 56.Dimitriu T, Kurilovich E, Łapińska U et al. (2022) Bacteriostatic antibiotics promote CRISPR-Cas adaptive immunity by enabling increased spacer acquisition. Cell Host Microbe 30:31–40.e5. 10.1016/j.chom.2021.11.014 [DOI] [PubMed] [Google Scholar]
  • 57.Pons BJ, Dimitriu T, Westra ER, van Houte S (2023) Antibiotics that affect translation can antagonize phage infectivity by interfering with the deployment of counter-defenses. Proc Natl Acad Sci 120:e2216084120. 10.1073/pnas.2216084120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Burmeister AR, Fortier A, Roush C et al. (2020) Pleiotropy complicates a trade-off between phage resistance and antibiotic resistance. Proc Natl Acad Sci 117:11207–11216. 10.1073/pnas.1919888117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kumaran D, Taha M, Yi Q et al. (2018) Does treatment order matter? investigating the ability of bacteriophage to augment antibiotic activity against Staphylococcus aureus biofilms. Front Microbiol. 10.3389/fmicb.2018.00127 [DOI] [Google Scholar]
  • 60.Torres-Barceló C, Arias-Sánchez FI, Vasse M et al. (2014) A window of opportunity to control the bacterial pathogen Pseudomonas aeruginosa combining antibiotics and phages. PLoS ONE 9:e106628. 10.1371/journal.pone.0106628 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Mukhopadhyay S, Zhang P, To KKW et al. (2023) Sequential treatment effects on phage–antibiotic synergistic application against multi-drug-resistant Acinetobacter baumannii. Int J Antimicrob Agents 62:106951. 10.1016/j.ijantimicag.2023.106951 [DOI] [PubMed] [Google Scholar]
  • 62.Uchiyama J, Shigehisa R, Nasukawa T et al. (2018) Piperacillin and ceftazidime produce the strongest synergistic phage-antibiotic effect in Pseudomonas aeruginosa. Arch Virol 163:1941–1948. 10.1007/s00705-018-3811-0 [DOI] [PubMed] [Google Scholar]
  • 63.Davies D (2003) Understanding biofilm resistance to antibacterial agents. Nat Rev Drug Discov 2:114–122. 10.1038/nrd1008 [DOI] [PubMed] [Google Scholar]
  • 64.Hall-Stoodley L, McCoy KS (2022) Biofilm aggregates and the host airway-microbial interface. Front Cell Infect Microbiol 12:969326. 10.3389/fcimb.2022.969326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Gloag ES, German GK, Stoodley P, Wozniak DJ (2018) Viscoelastic properties of Pseudomonas aeruginosa variant biofilms. Sci Rep 8:9691. 10.1038/s41598-018-28009-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ceri H, Olson ME, Stremick C et al. (1999) The calgary biofilm device: new technology for rapid determination of antibiotic susceptibilities of bacterial biofilms. J Clin Microbiol 37:1771–1776. 10.1128/JCM.37.6.1771-1776.1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Yan J, Bassler BL (2019) Surviving as a community: antibiotic tolerance and persistence in bacterial biofilms. Cell Host Microbe 26:15–21. 10.1016/j.chom.2019.06.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Stewart PS (2002) Mechanisms of antibiotic resistance in bacterial biofilms. Int J Med Microbiol 292:107–113. 10.1078/1438-4221-00196 [DOI] [PubMed] [Google Scholar]
  • 69.Sharma D, Misba L, Khan AU (2019) Antibiotics versus biofilm: an emerging battleground in microbial communities. Antimicrob Resist Infect Control 8:76. 10.1186/s13756-019-0533-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Gordon CA, Hodges NA, Marriott C (1988) Antibiotic interaction and diffusion through alginate and exopolysaccharide of cystic fibrosis-derived Pseudomonas aeruginosa. J Antimicrob Chemother 22:667–674. 10.1093/jac/22.5.667 [DOI] [PubMed] [Google Scholar]
  • 71.Wood TK, Knabel SJ, Kwan BW (2013) Bacterial persister cell formation and dormancy. Appl Environ Microbiol 79:7116–7121. 10.1128/AEM.02636-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Azeredo J, Sutherland IW (2008) The use of phages for the removal of infectious biofilms. Curr Pharm Biotechnol 9:261–266. 10.2174/138920108785161604 [DOI] [PubMed] [Google Scholar]
  • 73.Kovacs CJ, Rapp EM, McKenzie SM et al. (2023) Disruption of biofilm by bacteriophages in clinically relevant settings. Mil Med. 10.1093/milmed/usad385 [DOI] [Google Scholar]
  • 74.Aslam S, Lampley E, Wooten D et al. (2020) Lessons learned from the first 10 consecutive cases of intravenous bacteriophage therapy to treat multidrug-resistant Bacterial infections at a single center in the United States. Open Forum Infect Dis. 10.1093/ofid/ofaa389 [DOI] [Google Scholar]
  • 75.Law N, Logan C, Yung G et al. (2019) Successful adjunctive use of bacteriophage therapy for treatment of multidrug-resistant Pseudomonas aeruginosa infection in a cystic fibrosis patient. Infection 47:665–668. 10.1007/s15010-019-01319-0 [DOI] [PubMed] [Google Scholar]
  • 76.Guo Z, Liu M, Zhang D (2023) Potential of phage depolymerase for the treatment of bacterial biofilms. Virulence 14:2273567. 10.1080/21505594.2023.2273567 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Karthika C, Malligarjunan N, Jothi R et al. (2023) Two novel phages PSPa and APPa inhibit planktonic, sessile and persister populations of Pseudomonas aeruginosa, and mitigate its virulence in Zebrafish model. Sci Rep 13:19033. 10.1038/s41598-023-45313-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Tkhilaishvili T, Lombardi L, Klatt A-B et al. (2018) Bacteriophage Sb-1 enhances antibiotic activity against biofilm, degrades exopolysaccharide matrix and targets persisters of Staphylococcus aureus. Int J Antimicrob Agents 52:842–853. 10.1016/j.ijantimicag.2018.09.006 [DOI] [PubMed] [Google Scholar]
  • 79.Chang RYK, Das T, Manos J et al. (2019) Bacteriophage PEV20 and ciprofloxacin combination treatment enhances removal of Pseudomonas aeruginosa biofilm isolated from cystic fibrosis and wound patients. AAPS J 21:49. 10.1208/s12248-019-0315-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Chaudhry WN, Concepción-Acevedo J, Park T et al. (2017) Synergy and order effects of antibiotics and phages in killing Pseudomonas aeruginosa biofilms. PLoS ONE 12:e0168615. 10.1371/journal.pone.0168615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Dickey J, Perrot V (2019) Adjunct phage treatment enhances the effectiveness of low antibiotic concentration against Staphylococcus aureus biofilms in vitro. PLoS ONE 14:e0209390. 10.1371/journal.pone.0209390 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kebriaei R, Lehman SM, Shah RM et al. (2023) Optimization of phage-antibiotic combinations against Staphylococcus aureus biofilms. Microbiol Spectr 11:e04918–e4922. 10.1128/spectrum.04918-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hanlon GW, Denyer SP, Olliff CJ, Ibrahim LJ (2001) Reduction in exopolysaccharide viscosity as an aid to bacteriophage penetration through Pseudomonas aeruginosa biofilms. Appl Environ Microbiol 67:2746–2753. 10.1128/AEM.67.6.2746-2753.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Sutherland IW (1995) Polysaccharide lyases. FEMS Microbiol Rev 16:323–347. 10.1111/j.1574-6976.1995.tb00179.x [DOI] [PubMed] [Google Scholar]
  • 85.Vidakovic L, Singh PK, Hartmann R et al. (2018) Dynamic biofilm architecture confers individual and collective mechanisms of viral protection. Nat Microbiol 3:26–31. 10.1038/s41564-017-0050-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Bond MC, Vidakovic L, Singh PK et al. (2021) Matrix-trapped viruses can prevent invasion of bacterial biofilms by colonizing cells. Elife 10:e65355. 10.7554/eLife.65355 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Bryan D, El-Shibiny A, Hobbs Z et al. (2016) Bacteriophage T4 infection of stationary Phase E. coli: life after log from a phage perspective. Front Microbiol. 10.3389/fmicb.2016.01391 [DOI] [Google Scholar]
  • 88.Łoś M, Węgrzyn G (2012) Pseudolysogeny. Adv Virus Res 82:339–349. 10.1016/B978-0-12-394621-8.00019-4 [DOI] [PubMed] [Google Scholar]
  • 89.Ripp S, Miller RV (1998) Dynamics of the pseudolysogenic response in slowly growing cells of Pseudomonas aeruginosa. Microbiol (Read) 144:2225–2232. 10.1099/00221287-144-8-2225 [DOI] [Google Scholar]
  • 90.Maffei E, Woischnig A-K, Burkolter MR et al. (2024) Phage paride can kill dormant, antibiotic-tolerant cells of Pseudomonas aeruginosa by direct lytic replication. Nat Commun 15:175. 10.1038/s41467-023-44157-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Mulcahy LR, Burns JL, Lory S, Lewis K (2010) Emergence of Pseudomonas aeruginosa strains producing high levels of persister cells in patients with cystic fibrosis. J Bacteriol 192:6191–6199. 10.1128/JB.01651-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Jorth P, Staudinger BJ, Wu X et al. (2015) Regional isolation drives bacterial diversification within cystic fibrosis lungs. Cell Host Microbe 18:307–319. 10.1016/j.chom.2015.07.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Mapes AC, Trautner BW, Liao KS, Ramig RF (2016) Development of expanded host range phage active on biofilms of multi-drug resistant Pseudomonas aeruginosa. Bacteriophage 6:e1096995. 10.1080/21597081.2015.1096995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Du J, Meile S, Baggenstos J et al. (2023) Enhancing bacteriophage therapeutics through in situ production and release of heterologous antimicrobial effectors. Nat Commun 14:4337. 10.1038/s41467-023-39612-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Schmerer M, Molineux IJ, Bull JJ (2014) Synergy as a rationale for phage therapy using phage cocktails. PeerJ 2:e590. 10.7717/peerj.590 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Haines MEK, Hodges FE, Nale JY et al. (2021) Analysis of selection methods to develop novel phage therapy cocktails against antimicrobial resistant clinical isolates of bacteria. Front Microbiol. 10.3389/fmicb.2021.613529 [DOI] [Google Scholar]
  • 97.Zurabov F, Petrova M, Zurabov A et al. (2023) Adaptive phage therapy for the prevention of recurrent nosocomial pneumonia: novel protocol description and case series. Antibiotics 12:1734. 10.3390/antibiotics12121734 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Chan BK, Stanley G, Modak M et al. (2021) Bacteriophage therapy for infections in CF. Pediatr Pulmonol 56:S4–S9. 10.1002/ppul.25190 [DOI] [PubMed] [Google Scholar]
  • 99.Labrie SJ, Samson JE, Moineau S (2010) Bacteriophage resistance mechanisms. Nat Rev Microbiol 8:317–327. 10.1038/nrmicro2315 [DOI] [PubMed] [Google Scholar]
  • 100.Torres-Barceló C, Turner PE, Buckling A (2022) Mitigation of evolved bacterial resistance to phage therapy. Curr Opin Virol 53:101201. 10.1016/j.coviro.2022.101201 [DOI] [PubMed] [Google Scholar]
  • 101.Dedrick RM, Smith BE, Garlena RA et al. (2021) Mycobacterium abscessus strain morphotype determines phage susceptibility, the repertoire of therapeutically useful phages, and phage resistance. Micro biol. 10.1128/mbio.03431-20 [DOI] [Google Scholar]
  • 102.Aslam S, Courtwright AM, Koval C et al. (2019) Early clinical experience of bacteriophage therapy in 3 lung transplant recipients. Am J Transplant 19:2631–2639. 10.1111/ajt.15503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Wang X, Xie Z, Zhao J et al. (2021) Prospects of inhaled phage therapy for combatting pulmonary infections. Front Cell Infect Microbiol 11:758392. 10.3389/fcimb.2021.758392 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Astudillo A, Leung SSY, Kutter E et al. (2018) Nebulization effects on structural stability of bacteriophage PEV 44. Eur J Pharm Biopharm 125:124–130. 10.1016/j.ejpb.2018.01.010 [DOI] [PubMed] [Google Scholar]
  • 105.Dedrick RM, Freeman KG, Nguyen JA et al. (2021) Potent anti-body-mediated neutralization limits bacteriophage treatment of a pulmonary Mycobacterium abscessus infection. Nat Med 27:1357–1361. 10.1038/s41591-021-01403-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Taylor-Cousar JL, Robinson PD, Shteinberg M, Downey DG (2023) CFTR modulator therapy: transforming the landscape of clinical care in cystic fibrosis. Lancet 402:1171–1184. 10.1016/S0140-6736(23)01609-4 [DOI] [PubMed] [Google Scholar]
  • 107.Ledger EL, Smith DJ, Teh JJ et al. (2024) Impact of CFTR modulation on Pseudomonas aeruginosa infection in people with cystic fibrosis. J Infect Dis. 10.1093/infdis/jiae051 [DOI] [Google Scholar]
  • 108.Nichols DP, Morgan SJ, Skalland M et al. (2023) Pharmacologic improvement of CFTR function rapidly decreases sputum pathogen density but lung infections generally persist. J Clin Invest. 10.1172/JCI167957 [DOI] [Google Scholar]

RESOURCES