Next Article in Journal
Cefiderocol Antimicrobial Susceptibility Testing by Disk Diffusion: Influence of Agar Media and Inhibition Zone Morphology in K. pneumoniae Metallo-β-lactamase
Previous Article in Journal
Antimicrobial Susceptibility Profiles of Staphylococcus aureus Isolates from Domestic Pigeons in Hungary in 2022
Previous Article in Special Issue
Anticandidal Properties of Launaea sarmentosa among the Salt Marsh Plants Collected from Palk Bay and the Gulf of Mannar Coast, Southeastern India
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Combatting Pseudomonas aeruginosa with β-Lactam Antibiotics: A Revived Weapon?

by
Dylan W. Zhao
and
Christopher T. Lohans
*
Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
*
Author to whom correspondence should be addressed.
Antibiotics 2025, 14(5), 526; https://doi.org/10.3390/antibiotics14050526
Submission received: 25 April 2025 / Revised: 15 May 2025 / Accepted: 16 May 2025 / Published: 20 May 2025

Abstract

:
Pseudomonas aeruginosa is a significant threat to public health as an aggressive, opportunistic pathogen. The use of β-lactam antibiotics such as penicillins, cephalosporins, monobactams, and carbapenems remains a front-line treatment against P. aeruginosa. However, the widespread use of β-lactams has led to the emergence of β-lactam-resistant isolates that significantly increase the economic burden and risk of mortality in patients. With the declining productivity of the antibiotic discovery pipeline, research has investigated synergistic agents to revive the use of β-lactam antibiotics against β-lactam-resistant P. aeruginosa. In this review, we summarize the mechanism of β-lactam antibiotics and provide an overview of major mechanisms associated with β-lactam resistance in P. aeruginosa. We then describe the background and use of three promising classes of agents that have shown extensive beneficial effects with β-lactam antibiotics against P. aeruginosa, namely β-lactamase inhibitors, bacteriophages, and antimicrobial peptides. The current understanding of the mechanisms of these synergistic agents is discussed. Lastly, we provide an overview of the current barriers impeding antibiotic development, and offer a glimpse into recent advances of artificial intelligence-based discovery that may serve as a new foundation for antimicrobial discovery and treatment.

Graphical Abstract

1. Introduction

Pseudomonas aeruginosa is a Gram-negative bacillus that is a prevalent cause of nosocomial infections, and is responsible for mortality rates as high as 61% in ventilator-associated pneumonia, post-surgical infections, burn wounds, and cystic fibrosis-associated infections [1,2,3]. Epidemiological studies across international settings emphasize how P. aeruginosa remains a critical, global threat to public health. In a systematic analysis of global mortality from bacterial infections in 2019, P. aeruginosa was one of five pathogens responsible for 54.9% of the 7.7 million deaths analyzed, contributing to 590,000 deaths alone [4]. The 2016 EPINE survey found that Escherichia coli and P. aeruginosa were the most prevalent nosocomial pathogens in Spain, with P. aeruginosa accounting for 10.5% of all infections [5]. From European hospital-acquired infection reports between 2011 and 2021, P. aeruginosa caused approximately 9% of all infections as the fourth most common nosocomial pathogen [6]. In North America, the Centers for Disease Control and Prevention found that P. aeruginosa accounted for 7.1% of all nosocomial infections [3].
Once a person is infected by P. aeruginosa, this bacterium aggressively employs various virulence factors (e.g., toxin secretion systems, outer membrane vesicles, rhamnolipids) to enhance colonization, directly cause tissue damage (especially in the pulmonary tract), and evade the immune response [7,8,9,10,11,12,13,14,15,16,17,18]. As P. aeruginosa infections can rapidly progress towards systemic bacteremia, sepsis, and death [19], having adequate antibiotic regimens available remains imperative. Following treatment guidelines published by the Infectious Diseases Society of America and Health Canada, the use of β-lactam antibiotics (e.g., penicillins, cephalosporins, monobactams, and carbapenems) remains the first line of treatment for P. aeruginosa with recommended agents including ceftazidime, cefepime, piperacillin, meropenem, and imipenem [20]. However, with β-lactam prescriptions representing over 65% of the antibiotic global market [21,22,23], combined with the improper use, disposal, and overall stewardship of antibiotics [24], resistant P. aeruginosa strains have been identified worldwide [25,26,27,28]. Although the extent of β-lactam resistance on a global scale is unknown, a 2-year analysis of multi-drug resistant (MDR) Gram-negative infections in a Greek tertiary hospital revealed 55% of all P. aeruginosa isolates from the intensive care unit were resistant to carbapenems, a β-lactam often reserved as a last resort [29]. As resistant P. aeruginosa infections can cause an approximate two-fold increase in the risk of mortality and a 30% increase in cost relative to susceptible infections, this microorganism remains a significant financial burden and threat to public health [30,31].
Although resistance against β-lactams remains a constant threat, sustained efforts have not only yielded novel antibiotics with a β-lactam scaffold, but have also contributed to the development of novel agents that have been shown to enhance or rescue the efficacy of β-lactam antibiotics. These recent successes not only relieve some pressure on the deteriorating antibiotic discovery pipeline, but also offer a new avenue of therapies to treat MDR P. aeruginosa. As several excellent reviews already exist on combination therapies of β-lactams and other antibiotics [32,33,34,35,36], this review examines the current literature describing three prominent classes of potential adjuvant agents: β-lactamase inhibitors, bacteriophages, and antimicrobial peptides. We begin by describing the mechanism of β-lactam antibiotics and the β-lactam resistance mechanisms that occur in P. aeruginosa. We next describe the mechanism and use of synergistic adjuvants for the rescue of β-lactam activity. Finally, we provide a brief perspective on the future outlook relating to the use of machine learning to develop novel agents to combat β-lactam-resistant P. aeruginosa infections.

2. β-Lactam Antibiotics and Mechanisms of Resistance

2.1. Mechanism and Classification of β-Lactams

β-lactam antibiotics are classified based on their core, four-membered β-lactam ring (Figure 1A), the structural motif that is essential for their bactericidal activity [37]. This scaffold is conserved across all four major classes of β-lactam antibiotics (penicillins, cephalosporins, carbapenems, monobactams) and is fused to an additional heterocycle in all classes with the exception of the monobactams [38]. Subtle changes in the functional groups decorating this core give rise to distinct differences in overall biological activity and interactions with bacterial targets and resistance mechanisms. As the carbapenem subclass is frequently reserved as an antibiotic of last resort [39], resistance against carbapenems remains a significant threat to public health and will be a focus of this review.
To withstand fluctuations in osmotic stress, bacteria rely on the structural integrity of peptidoglycan in the cell wall. This structural integrity is greatly dependent on cross-links between peptide chains in peptidoglycan that are formed by penicillin-binding proteins (PBPs) [21,40]. In Gram-negative bacteria such as P. aeruginosa, peptidoglycan and some of the enzymes that synthesize it are found in the periplasmic space between the outer and cytoplasmic membranes. β-lactam antibiotics in the periplasm interfere with cell wall synthesis by targeting PBPs through a suicide inhibition mechanism (Figure 1B). This is achieved through an acylation reaction between the β-lactam ring and an essential serine residue in the PBP active site, forming a stable acyl–enzyme complex that blocks PBP-catalyzed cross-link formation [41]. Consequently, the bacterial cell wall weakens and becomes increasingly susceptible to osmotic stress, leading to cellular autolysis [21,40].

2.2. β-Lactamase-Mediated Resistance

A prominent resistance mechanism employed by P. aeruginosa and other β-lactam-resistant Gram-negative bacteria is the production of β-lactamases. These enzymes catalyze a hydrolytic reaction that opens the β-lactam ring, rendering the antibiotic unable to inhibit PBPs (Figure 2A). In Gram-negative bacteria such as P. aeruginosa, β-lactamases primarily reside in the periplasmic space. β-lactamases are commonly categorized according to their sequence motifs and mechanisms into classes A, B, C, and D in the Ambler classification system (Table 1) [42]. β-lactamases can also be more broadly differentiated based on their mechanisms of catalysis; the serine β-lactamases (SBLs; Ambler classes A, C, and D) rely on a covalent, two-step hydrolytic mechanism mediated by a nucleophilic serine residue, while metallo-β-lactamases (MBLs; Ambler class B) hydrolyze β-lactams using a water molecule activated by one or more zinc ions. To this point, at least 120 different β-lactamases have been identified in clinical isolates of P. aeruginosa [43], and these enzymes remain clinically significant given their ability to confer high levels of resistance against β-lactams and their horizontal mobility, as their genes are commonly carried on plasmids. β-lactamases are also often categorized based on their substrate specificities, yielding groups such as the penicillinases, cephalosporinases, extended-spectrum β-lactamases (ESBLs), and carbapenemases. As carbapenems are reserved as antibiotics of last resort due to their ability to withstand hydrolysis from most β-lactamases, carbapenemase-producing organisms are widely regarded as one of the greatest threats resulting from antibiotic resistance.
Within the Ambler classification system, hundreds of carbapenemase variants fall under classes A, B, and D [48]. Prevalent carbapenemases within these respective classes include the KPC family (class A; SBLs), the NDM, IMP, and VIM families (class B; MBLs), and the OXA family (class D; SBLs). As many reviews have been published on the Ambler classification, epidemiology, and catalytic spectra of β-lactamases [41,44,45,49,50,51,52], this section will highlight the clinical consequences of SBL- and MBL-producing P. aeruginosa.
The KPC family of SBLs can hydrolyze all classes of β-lactams, while the OXA family, although sharing a similar spectrum of hydrolysis, exhibits significantly reduced activity against carbapenems while still contributing to carbapenem resistance in vivo [53]. These properties complicate the clinical detection of OXA-type carbapenemases and have earned them a reputation of being a “phantom menace” [54,55,56,57]. Many MBLs are carbapenemases that can hydrolyze most β-lactams with the notable exception of monobactams like aztreonam [58]. To date, at least 32 variants of IMP and 23 variants of VIM have been identified in P. aeruginosa [59], and the reported prevalence of these enzymes is as high as 30% among carbapenem-resistant isolates [60]. Due to the broad catalytic activities of the MBLs, the presence of resistance genes to other antibiotics within the same plasmid can lead to pan-resistant phenotypes. Another significant challenge posed by MBLs is that, unlike SBLs, they are not targeted by any of the β-lactamase inhibitors that are currently available for clinical use due to the drastic differences in mechanisms of catalysis between MBLs and SBLs [61].
Clinically, the presence of class A, B, and D carbapenemases has been consistently shown to complicate treatment efficacy and increase P. aeruginosa infection mortality [62,63,64,65,66,67,68,69,70,71,72,73]. A report by Zhang et al. followed the spread of high-risk clones of carbapenemase-producing P. aeruginosa between 2020 and 2022 in Zhejiang, China [62]. Eight out of the 192 isolates were identified as belonging to the high-risk ST463 clone that co-harbored KPC-2 and AFM-1. Due to the presence of both class A and B carbapenemases, all eight isolates exhibited resistance against all β-lactams and β-lactam/β-lactamase inhibitor combinations tested, and were only susceptible to amikacin and intermediately susceptible to colistin [62]. In an analysis of carbapenem-resistant P. aeruginosa isolates from Qatar, almost all isolates (96%) possessed an OXA-family carbapenemase, with 26.7% of isolates carrying an MBL and 4% of isolates producing representatives of all four Ambler classes of β-lactamases [74]. Similar to previous reports, 40% of the MBL-producing isolates were susceptible to aztreonam due to the absence of ESBLs [74]. As a further illustration of the concern associated with the transmissibility of plasmid-encoded β-lactamases, all isolates were even resistant to β-lactam/β-lactamase inhibitor combinations that were not clinically available in Qatar at the time of study [74]. To highlight the morbidity associated with the production of β-lactamases in P. aeruginosa, the pan-resistance of a strain characterized by Costa-Júnior et al. was attributed in part to the production of KPC-2 and VIM-2 alongside the presence of the aminoglycoside resistance gene rmtD1 [75]. Hence, preserving the efficacy of last-resort carbapenems through proper stewardship remains critical for public health in the reduction of highly morbid MDR P. aeruginosa infections [76].

2.3. Target Modification

Changes to the active sites of PBPs can interfere with the ability of β-lactams to bind and acylate the conserved nucleophilic serine residue (Figure 2B). In P. aeruginosa, the transpeptidase PBP3 (encoded by the ftsl gene) catalyzes the formation of peptidoglycan cross-links and is essential for bacterial growth [77]. The loss of β-lactam efficacy in the presence of ftsl mutations, particularly those impacting the active site, has been extensively studied in experimental models and clinical isolates of P. aeruginosa [78,79]. Given that the binding interactions between the different classes of β-lactam antibiotics and the PBP3 active site differ, changes to the active site can confer varying degrees of β-lactam resistance. Moreover, a change to PBP3 can lead to different interactions with β-lactams belonging to the same class. For example, PBP3 variants observed in Pseudomonas isolates from a cystic fibrosis patient treated with β-lactams possessed two notable amino acid substitutions, V465G and A244T, with the former conferring resistance to both aztreonam (monobactam) and cefsulodin (cephalosporin) and the latter was associated with ceftazidime (cephalosporin) and piperacillin (penicillin) resistance [80]. Another study in adult cystic fibrosis patients illustrated that production of the PBP3 R504C variant conferred resistance to ceftazidime and cefsulodin, whereas the P527S variant was associated with resistance to aztreonam, cefepime, ceftazidime, and cefsulodin [81].

2.4. Decreased Membrane Permeability: Role of Porins

Porins are β-barrel proteins that mediate the passive entry of hydrophilic compounds (including β-lactam antibiotics) across the outer membranes of Gram-negative bacteria [82,83]. As such, mutations that cause changes to porin structure or to the number of porins in the outer membrane can protect bacteria from β-lactam activity (Figure 2C). While not all 26 porins identified in P. aeruginosa are involved in β-lactam uptake, those belonging to the OprD and Opd (P/D/H) families have been investigated for their role in β-lactam resistance [84]. In addition to small basic amino acids and peptides, the OprD porin is a crucial mediator in the uptake of carbapenems in P. aeruginosa [85,86,87,88]. Point mutations, premature stop codons, and frameshifts in the oprD gene have been shown to reduce or abolish OprD expression and lead to carbapenem resistance [88]. Of note, mutations to oprD were not observed to impact susceptibility to other β-lactam antibiotics (e.g., penicillins, cephalosporins, and monobactams) [89,90,91]. Regarding the Opd family of porins, the loss of OpdP was shown to reduce susceptibility to meropenem and, in an opdD knockout strain, to imipenem and doripenem as well, supporting that OpdD facilitates the entry of carbapenems in P. aeruginosa [91]. The loss of OpdH was also shown to reduce susceptibility to ceftazidime but not other cephalosporins [89,90,91,92].

2.5. Efflux Pumps

Efflux pumps are transmembrane proteins that can facilitate the expulsion of various substances (including β-lactam antibiotics) from the periplasm across the outer membrane and into the extracellular environment (Figure 2D) [93], with many of the pumps that contribute to antibiotic efflux belonging to the resistance-nodulation-division (RND) family [94]. Structurally, these RND pumps are a tripartite system comprising a periplasmic adaptor protein, a transmembrane inner-membrane transporter, and a protein channel through the outer membrane [95,96]. In P. aeruginosa, efflux pumps MexAB-OprM, MexXY-OprM, MexCD-OprJ, and MexEF-OprN have been shown to contribute to β-lactam resistance, with each exporting a particular selection of β-lactams (Table 2). Unsurprisingly, overexpression of these pumps has been identified in many β-lactam-resistant P. aeruginosa clinical isolates [85,86,97,98,99,100,101,102,103], with one multicenter study in France revealing efflux pump overexpression in 88% of isolates collected from cystic fibrosis patients [85]. Specifically, 71 out of 80 isolates overexpressed at least one or more efflux system, with 65 overexpressing MexXY-OprM, 36 overexpressing MexAB-OprM, and two overexpressing MexCD-OprJ [85]. Within the overexpressing isolates, an additional 29 and five simultaneously overexpressed two or three efflux systems, respectively [85]. The molecular basis of efflux pump overexpression is often attributed to mutations in repressor genes that regulate each respective mex operon. As one example of many [104,105,106,107], overexpression of MexAB-OprM is primarily attributed to loss of function mutations in the DNA-binding region of the MexR repressor protein [108], preventing MexR from repressing the mexAB oprM operon [109,110]. Such mutations were identified in clinical isolates resistant to meropenem, ceftazidime, aztreonam, ticarcillin, and carbenicillin [111,112]. Deletions in repressor genes nalD and nalC also contribute to MexAB-OprM overexpression, although these changes are less prevalent [111,113].

3. Adjuvant Agents: Mechanisms and Use

3.1. β-Lactamase Inhibitors

Since the discovery of clavulanic acid in 1979 as an irreversible inhibitor of certain class A β-lactamases [117], the development of novel β-lactamase inhibitors has received great interest as a method of combatting antibiotic-resistant P. aeruginosa, especially considering the declining productivity of the antibiotic discovery pipeline for treating Gram-negative infections [78,79]. Although these agents do not possess bactericidal effects alone against P. aeruginosa, when combined with a β-lactam, the inhibition of β-lactamases spares the co-prescribed β-lactam antibiotic from hydrolysis [118,119]. By the end of the 20th century, sulbactam and tazobactam (Figure 3) also reached clinical use, although these agents were primarily active against class A and D serine β-lactamases [120,121,122,123,124]. After nearly two decades of research, a novel class of β-lactamase inhibitors based on a bridged diazabicyclooctane (DBO) scaffold emerged, which includes inhibitors such as avibactam and relebactam (Figure 3) [125]. Other research efforts have similarly moved beyond β-lactam-based β-lactamase inhibitors, yielding boronates such as vaborbactam (Figure 3), which has been successfully paired with the carbapenem meropenem [126,127].
Avibactam, relebactam, tazobactam, and vaborbactam share similarities with regard to their inhibitory activities against the β-lactamases found in clinical P. aeruginosa isolates. Out of these four inhibitors, avibactam exhibits the broadest spectrum of activity, possessing the ability to target class A (e.g., TEM, SHV, CTX-M), class C (e.g., chromosomal AmpC), and some class D (e.g., OXA) β-lactamases [124]. Relebactam and tazobactam have been shown to target class A and C β-lactamases, although the latter is less effective than newer compounds, while vaborbactam has been shown to be particularly effective against class A KPC-type β-lactamases [124,128,129,130].
Based on prior epidemiological and in vitro evidence, 61.8–70.2% of β-lactam-resistant P. aeruginosa isolates were susceptible to ceftazidime/avibactam, with meropenem/vaborbactam exhibiting a narrower range of activity targeting 59.0% of isolates [131]. As imipenem/relebactam is a relatively new combination, data specific to β-lactam-resistant P. aeruginosa remain scarce. However, one study illustrated that the addition of relebactam failed to lower the minimum inhibitory concentrations (MICs) of β-lactam-resistant P. aeruginosa isolates below clinical breakpoints, with only 10.3% of tested isolates being classified as susceptible according to European Committee on Antimicrobial Susceptibility Testing criteria and 26.8% by Federal Drug Administration/Clinical and Laboratory Standards Institute criteria [132]. The development of new DBO-based inhibitors continues to be a major focus, with nacubactam (RG6080; Figure 3) and zidebactam (WCK-5107; Figure 3) [133,134] combinations having been shown to lower the MICs of β-lactams between 0.5 and 16 fold against P. aeruginosa isolates [135,136]. New boronates have also been explored, such as QPX7728 (Figure 3), an “ultrabroad” spectrum β-lactamase inhibitor that, when paired with meropenem, cefepime, or ceftolozane, demonstrated potent activity against >90% of all tested clinical P. aeruginosa isolates [137].
Despite the clinical success of β-lactamase inhibitors, it is no surprise to observe resistance emerging in MDR P. aeruginosa strains. Specifically, resistance against β-lactamase inhibitors in P. aeruginosa has occurred via mutations in the ampC gene or overexpression of efflux pumps. Against avibactam, amino acid substitutions such as E247K, G183D, T96I, and ΔG229-E247 in the AmpC β-lactamase significantly impair inhibitor binding while maintaining or enhancing catalytic activity [138,139]. Overexpression of the MexAB-OprM efflux pump has also been shown to facilitate expulsion of avibactam [139]. Tazobactam is similarly impacted by overexpression of the efflux pump MexXY-OprM, and a D245N mutation has been shown to drastically decrease the binding affinity of tazobactam to the P. aeruginosa β-lactamase PDC-315 [139,140]. Vaborbactam resistance has rarely been attributed to target modification-mediated resistance and often results from the production of an MBL, while resistance against relebactam has been documented to occur through the production of class A β-lactamases including GES-2, GES-19, and GES-20 [130,141]. Porin loss, including through the disruption of the oprD gene, has been observed to interfere with the entry of vaborbactam and relebactam as well [130,141].

3.2. Bacteriophages

First pioneered in the 1920s for the treatment of staphylococcal cutaneous furuncles and carbuncles [142], the initial development of phage therapy was largely carried out in the former Soviet Union during the 20th century with minimal penetrance into Western-dominated regions of medicine and research [143,144]. With the continued emergence of MDR bacteria, bacteriophages have been re-emphasized for use as potential synergistic agents in the treatment of MDR infections [145,146,147,148]. Although this therapy primarily uses lytic phages to kill the strains of bacteria that are causing an infection, an abundance of recent research has demonstrated that bacteriophages can also act synergistically and revive the bactericidal effects of antibiotics, including β-lactams.
One proposed mechanism involves the destruction of biofilms, which are complex aggregates of bacteria encased in extracellular polymeric substances (EPS) that include polysaccharides (alginate, etc.), extracellular DNA, proteins, and lipids [149]. They contribute to β-lactam resistance by not only acting as a physical barrier to β-lactam entry, but also by forming an extracellular reservoir of β-lactamases (Figure 4A), as was observed by Dibdin et al. [150]. Specifically, increases in the concentration of extracellular β-lactamases depleted the concentration of active antibiotic, with alterations to the initial β-lactam concentration making no difference in the amount of active antibiotic remaining near the bacterial cells [150]. Wang et al. observed similar results where the pharmacodynamics of ceftazidime-induced lysis changed from a time-dependent model to a concentration model against biofilm-producing P. aeruginosa, when compared to a planktonic strain [151]. This shift towards a concentration model illustrates that the biofilm matrix can sequester and concentrate β-lactamase enzymes, thereby necessitating higher β-lactam concentrations to achieve bactericidal effects.
Bacteriophages can destroy biofilms by encoding or inducing the expression of enzymes that degrade the EPS that make up the matrix of a biofilm, or by inhibiting the synthesis of EPS (Figure 4B) [152]. For example, following treatment of P. aeruginosa PAO1 with non-bactericidal phages KT28 and KTN6, the rate of dye diffusion through a biofilm formed by this bacterium significantly increased, supporting the presence of a phage-encoded polysaccharide hydrolase able to digest glycosidic bonds found in biofilm-associated carbohydrate polymers [153]. An enzyme with similar activity was expressed by P. aeruginosa NCIMB 10548 following bacteriophage F116 infection, degrading over 75% of biofilm material after two hours of exposure [154].
Another study showcased that three phages (ATCC 12175-B1, ATCC 14203-B1, ATCC 14205-B1) produced an 85–95% reduction in P. aeruginosa PAO1 biofilm volume [155]. However, when surviving colonies were isolated and replated, an alginate-producing, slow-growing, mucoid strain was obtained. Sequencing revealed a mutation in the negative regulator of alginate production mucA, thereby conferring an alginate-hyperproducing phenotype notably characterized by an additional outer alginate capsular polysaccharide layer [156]. Further experimentation with the wild-type non-alginate producing PAO1 strain and the mucoid mucA strain yielded data consistent with this observation, whereby the presence of alginate completely abolished phage-dependent biofilm destruction, as the biovolume of wild-type PAO1 decreased by 96% while the biovolume of the mucoid strain remained unaffected. As with any form of antimicrobial intervention, this illustrates that P. aeruginosa can adapt and become less susceptible to biofilm-degrading phages.
Interestingly, alginate-degrading P. aeruginosa-specific bacteriophages have also been identified. In a study by Glonti et al., bacteriophage PT-6 rapidly reduced the viscosity of four different P. aeruginosa alginate polymers by 62–66% within 15 min [157]. This finding was supported by the detection of α,β-unsaturated uronic acid monomers that are produced by alginic acid polysaccharide depolymerases [158,159,160,161,162]. Given that mature P. aeruginosa biofilm development is dependent on quorum sensing, an engineered lactonase-encoding T7 bacteriophage was found to abolish P. aeruginosa biofilm formation by specifically degrading acyl homoserine lactone autoinducers [163]. Other studies using P. aeruginosa-specific bacteriophages have also reported similar reductions in biofilm formation, but their underlying mechanism of degradation remains unknown [162,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178].
Despite the potential utility of biofilm-degrading bacteriophages against P. aeruginosa¸ a major limitation arises from the high level of specificity that occurs between phages and the targets that they recognize on the surfaces of host cells. Heterogeneity between clinical isolates of P. aeruginosa, particularly regarding outer membrane proteins and other surface molecules, can greatly impact phage infectivity and ability to produce biofilm-degrading enzymes. This necessitates the development of broader phage libraries, as well as robust and efficient methods for the preparation of personalized phage cocktails tailored to the particular strain(s) responsible for an infection.
Overall, the use of bacteriophages against P. aeruginosa can aid β-lactam therapy through the disruption of biofilms and the β-lactamase reservoirs they may contain that would otherwise prevent intact β-lactams from reaching their bacterial targets. This biofilm disruption could also enhance the ability of β-lactamase inhibitors to access bacterial cells, improving their potency in the context of an infection. Thus, integrating the use of bacteriophages with β-lactam/β-lactamase inhibitor combinations may provide a more comprehensive strategy to overcome β-lactamase-mediated β-lactam resistance in P. aeruginosa.

3.3. Antimicrobial Peptides

Antimicrobial peptides (AMPs), generally comprising 12–50 amino acid residues, exhibit a broad range of antibacterial activities, and are often produced by insects, birds, fish, and other animals [179]. In humans, AMPs are produced by immune cells during a front-line, innate immune response against invading pathogens. These AMPs are specific to bacteria, and their amphiphilic nature allows them to bind to negatively charged groups on the surfaces of bacterial cells, facilitating their entry into the periplasmic space and the cytoplasm [180,181]. Alongside their selective toxicity, thermal stability, and high water solubility, the ability of AMPs to disrupt the outer membranes of Gram-negative bacteria illustrates their significant potential to be used as adjuvant agents alongside β-lactam antibiotics [182,183].
Although AMPs exhibit different bactericidal mechanisms, they can be broadly classified as agents that either directly target the bacterial membrane or that target other cellular components. AMPs that target the bacterial membrane remain selectively toxic, as the membranes of mammalian cells are primarily composed of net-neutral phospholipids (phosphatidylcholine, phosphatidylethanolamine, sphingomyelins) [184]. These membrane-targeting peptides are believed to act through three distinct mechanisms that have the potential to facilitate β-lactam entry across the outer membrane (Figure 5), as has been extensively reviewed elsewhere [185,186,187,188,189,190,191,192,193,194,195].
Specific to the use of AMPs as an adjuvant therapy against P. aeruginosa, researchers have observed synergistic activity between bacterial, synthetic, and animal-derived AMPs with β-lactam antibiotics. Although there is strong evidence for this synergism, the specific mechanism(s) by which these AMPs exert their synergistic effect is often not fully understood. Nonetheless, the studies described below demonstrate that structurally diverse AMPs from different origins are able to dramatically decrease the amount of β-lactam antibiotic needed to control the growth and biofilm formation of P. aeruginosa.
Jahanigirl et al. examined the effects of nisin and peptide P10 in combination with ceftazidime against colistin-resistant clinical isolates of P. aeruginosa [196]. Nisin is a bacteriocin produced by Lactococcus lactis that inhibits cell wall synthesis by binding and sequestering lipid II, a cell wall precursor necessary for peptidoglycan synthesis [194,197,198,199,200,201], while P10 is a synthetic derivative of mammalian LL-37 that induces bacterial cell wall lysis potentially through the barrel-stave, toroidal pore, or carpet models represented in Figure 5 [202,203,204]. When combined with ceftazidime, the MIC of P10 decreased between 2–8-fold across six P. aeruginosa isolates, with four illustrating synergistic effects (total fractional inhibitory concentrations (ΣFIC) < 0.5) [196]. A similar AMP, P5, was tested against the carbapenem-resistant P. aeruginosa M13513 strain [205] following confirmation of antimicrobial activity in previous studies [206,207]. When combined with meropenem at a concentration of half of the MIC, a 2-log decrease relative to individual treatments (i.e., P5 or meropenem alone) in a time-kill kinetics assay illustrated significant synergism [205]. Moreover, the ability of P5 to permeabilize the outer membrane of P. aeruginosa was demonstrated by the increased uptake of the fluorescent dye 1-N-phenylnapthylamine [205]. Although P5 did also exhibit biofilm-degrading properties, the potency of this activity was relatively low, as a reduction greater than 20% only occurred at P5 concentrations at least two-fold above the MIC [205].
Testing imipenem-resistant P. aeruginosa strains with decreased permeability arising from mutations to oprD, Rudilla et al. observed that treatment with 4 μg/mL of synthetic peptide AMP38 and 4 μg/mL of imipenem abolished bacterial growth compared to individual treatment groups [208]. The authors designed this peptide based on the structure of polymyxin, suggesting that AMP38 binds to lipopolysaccharides in the outer membrane of P. aeruginosa, displacing cations and inducing autolysis through membrane disruption [209]. Data from time-kill kinetics further supported the presence of synergistic activity, with all strains exhibiting an ΣFIC of less than 0.5 [208]. Interestingly, culturing P. aeruginosa PA11636 with AMP38 and imipenem decreased the minimal biofilm concentration from >500 μg/mL (imipenem alone) to 62.5 μg/mL, suggesting that AMP38 could contribute to β-lactam rescue in part through the destruction of biofilm-related β-lactamase reservoirs described above [208].
Contrasting the synthetic adjuvants described above, Shang et al. observed a novel, synergistic mechanism between tryptophan (Trp)-containing AMPs (L1W, L12W) and β-lactam antibiotics against the MDR P. aeruginosa MRPA0108 strain. When concentrations of L1W and L12W four-fold below their MICs were administered with ceftazidime and piperacillin, the authors observed an 8–32-fold and 2–12-fold reduction in the ceftazidime and piperacillin MICs, respectively [210]. Based on further transcriptomic analyses, the underlying synergistic mechanism was attributed to the L1W- and L12W-mediated downregulation of genes associated with β-lactam resistance, including the β-lactamase-encoding ampC gene and efflux pump genes oprM, mexX, and mexA [210]. L1W and L12W were also found to decrease biofilm formation by 33–53% by downregulating key genes involved in biofilm production (pelA, algD, and pslA) [210], illustrating the multifactorial nature of these Trp-containing AMPs in the context of the treatment of β-lactam-resistant P. aeruginosa infections.
Similar to the synthetic P5, P10, and AMP38 peptides, the ocellatin AMP family isolated from sebaceous secretions of the South American frog Leptodactylus labyrinthicus kills bacteria through a membrane-targeting mechanism [211]. When ocellatin PT3 was combined with ceftazidime against MDR P. aeruginosa isolates Pa1-SA2 and Pa4-SA2, Bessa et al. observed a 4–8-fold decrease in MIC with synergistic activity (ΣFIC < 0.5) [212]. Ocellatin PT3 was also able to inhibit biofilm formation in clinical isolate Pa4-SA2, rescuing β-lactam antibiotics by preventing the accumulation of β-lactamases in biofilms [212]. A study by Pandidan et al. further showed highly synergistic effects of melittin, an AMP in bee venom that forms toroidal pores [213], when combined with doripenem and ceftazidime against five P. aeruginosa isolates retrieved from burn patients [214]. The MIC of doripenem decreased between 32–128-fold when combined with melittin, demonstrating synergistic activity [214]. Synergism with ceftazidime was also reported across all five strains, but the MIC of ceftazidime decreased less relative to doripenem (4–64-fold reduction) [214].
Synthetic AMPs and those derived from natural sources show great promise as adjuvants that enhance the activity of β-lactam antibiotics against P. aeruginosa and other bacterial pathogens. However, P. aeruginosa can achieve high levels of β-lactam resistance by limiting the concentration of β-lactams in the periplasm through the simultaneous use of multiple resistance mechanisms (e.g., β-lactamase production, decreased permeability, increased efflux). Future studies could explore the extent to which AMPs impact the periplasmic concentration of β-lactams (and β-lactamase inhibitors) against P. aeruginosa strains that exhibit multiple resistance mechanisms. Potential synergy between AMPs and bacteriophages leading to increased biofilm disruption and outer membrane permeabilization would also be of interest. In addition, the mechanisms by which AMPs disrupt the outer membrane could be investigated further, supporting the rational development of more potent derivatives and providing insights into the propensity for bacterial resistance to develop.

4. Future Directions for Identification of β-Lactam Adjuvant Agents

Over the past few decades, the stagnation of antibiotic discovery has led to a steady decline in the number of approved antibiotics. As antibiotics cannot currently compete with the profitability of other blockbuster drugs (antihypertensive agents, etc.), many pharmaceutical companies, including Novartis, Sanofi, AstraZeneca, and GSK, have largely abandoned their “high risk” antibacterial programs [215,216]. Moreover, the recent bankruptcies of Aradigm, Achaogen, Tetraphase Pharmaceuticals, and Melinta Therapeutics, all of which had gained approval to sell novel antibiotics within the past decade, highlights how a shift towards identifying novel synergistic agents may help relieve the stress placed on the traditional antibiotic discovery pipeline [217]. Predicated on recent technological milestones, the emergence of high-throughput approaches to screen and identify lead synergistic compounds, particularly through artificial intelligence (AI) algorithms, may serve as a new foundation for antimicrobial discovery and treatment.

4.1. AI-Based Identification of Novel Synergistic Agents

Several papers have been written on the development of training algorithms and the current and future projections of AI-based identification of novel antimicrobial agents [218,219,220,221,222]. Above all, these manuscripts highlight a revolutionary advantage AI has over traditional, manual analyses of natural and synthetic molecular libraries—the capacity of AI to generate potential novel antibacterial agents beyond our current knowledgebase of existing compounds simply cannot be matched. A recent example involves the identification of the antibiotic abaucin for the treatment of MDR Acinetobacter baumannii through a mechanism that involves the disruption of lipoprotein trafficking [223]. However, this section will focus on summarizing published examples specific to the design and development of novel agents that may have synergistic activity with β-lactams.
AI has been applied to the identification of all three categories of adjuvants described in this review, as demonstrated by the following examples. In relation to identifying novel β-lactamase inhibitors, Parvaiz et al. utilized Site Identification by Ligand Competitive Saturation technology to generate pharmacophore models and functional group requirements for the β-lactamase CMY-10 active site [224]. Subsequent machine learning-based random forest methods were then used to screen and filter a repository of 700,000 compounds, whereby 74 were subjected to in vitro β-lactamase inhibition assays [224]. Of these, 11 were demonstrated to inhibit CMY-10, with one compound showing significant synergistic activity with the cephalosporin cefixime against MDR clinical isolates of Enterobacter cloacae, E. alvei, and E. agglomerans [224]. A search for related inhibitors yielded an additional 28 compounds, many of which also exhibited synergistic activity against MDR isolates [224].
Focusing on the identification of novel bacteriophages, poor-quality annotations of phage genomes from environmental sources hamper the efficient production of species-specific phages, including those that target P. aeruginosa [225,226]. To tackle this issue, Thung et al. developed STEP3, a computational tool that identifies, distinguishes, and categorizes genomic features in uncharacterized phages [227]. As structural motifs on the phage tail facilitate binding to the bacterial surface, STEP3 can be used to classify proteins with conserved features for researchers to easily pinpoint the target species of a given phage [227]. The integration of data into an ensemble framework also remains robust despite the high evolutionary rates of phage proteins, with prediction accuracy significantly surpassing traditional pairwise sequence matching methods of phage protein identification [228].
In the field of AMPs, Bhadra et al. assessed the accuracy of 19 random forest algorithms in predicting the antibacterial and synergistic activity of unknown peptides (166,791 sequences) based on the amino acid sequences of classified AMPs (3268 sequences). Following their analyses, the model AmPEP produced an accuracy score of 96% and an Area Under the Receiver Operating Characteristic Curve of 0.99, illustrating the high discriminatory power of the model in differentiating hits from false positives in the context of discovering antibacterial AMPs [229]. However, the study did not validate the results through in vitro growth/time-kill kinetics analyses. Nonetheless, with the rapid advancement of algorithms and learning models as illustrated by the three examples presented above, it will be no surprise to witness a significant integration of AI into the framework of antimicrobial drug discovery.

4.2. Challenges Moving Forward

Although the rise of AI-based identification algorithms may serve as a nearly limitless reservoir of new lead compounds for the development of novel synergistic agents and beyond, several roadblocks remain from development to commercialization. Aside from the immense financial burden of drug development, many lead compounds that show promise in in vitro studies are ineffective when tested in vivo. Challenges with reproducibility have also been extensively noted between institutions [230,231,232,233]. As with any synergistic pharmaceutical intervention, ensuring that the pharmacological profile of the combination has no unfavorable outcomes or off-target effects remains a major hurdle. For example, many β-lactam/β-lactamase inhibitor combinations identified in vitro require high concentrations for clinical efficacy, potentially leading to toxicity or other undesired outcomes [234]. Reports of antibacterial and synergistic agents targeting both bacterial and human pathways further illustrate the importance of preclinical screening [235]. Surveying for potential pharmacological contraindications also remains a priority. For example, reserpine, a known synergistic agent to norfloxacin, was shown to directly act as a calcium antagonist on mammalian smooth muscle cells [236]. Compounds in a combinatory formulation may also exhibit differing pharmacodynamics and kinetics, including dissimilar methods of absorption, metabolism, and excretion. In these cases, more work may be required to structurally modify a specific agent in favor of similar pharmacological profiles. For instance, the β-lactamase inhibitor sulbactam in its native form exhibited poor oral absorption before a pivaloyloxymethyl ester functional group was introduced [237].
Beyond these pharmacological challenges, AI-based methods for drug discovery have several inherent limitations. A primary concern lies in model generalizability, as the use of incomplete, inconsistent, or low-quality data to train an algorithm can significantly compromise its performance. For instance, a model may perform poorly when predicting the mechanism of a novel compound if limited or biased training datasets are used. Additionally, while AI models can rapidly identify candidate compounds, validation through in vitro or in vivo experiments is essential, as algorithms may not effectively account for the complex interactions that occur between a pharmaceutical, a patient, and a pathogen. As the integration of AI into biomedical research continues to develop at an incredible pace, its incorporation into existing regulatory frameworks can lag behind, complicating the clinical development pathway and regulatory approval. These limitations, combined with increasing computational resource needs and biases toward known chemical scaffolds, can stifle the discovery of truly novel agents. Addressing these limitations will be crucial to fully realize the potential that AI offers towards the discovery of β-lactam adjuvants and other synergistic antimicrobial agents.
Above all challenges, however, novel synergistic agents still exert a selective pressure against bacteria and are thus not immune to the development of resistance. Relevant to β-lactamase inhibitors, strains harboring mutations that modify the active sites of β-lactamases are naturally favored when inhibitors are used. As one example of many, Alonso-Garcia et al. documented the presence of mutations in the β-lactamase gene blaPDC, which contributed to resistance against cephalosporin/β-lactamase inhibitor combinations and imipenem/relebactam for a P. aeruginosa strain that was responsible for nosocomial meningoventriculitis [139]. Similarly, bacteriophages may be evaded through mutations to genes that encode for bacterial surface proteins, and by the CRISPR/Cas-9 system, which prevents integration of viral DNA during the lysogenic cycle and disrupts viral genome replication and protein production during the lytic cycle [238]. AMPs are not immune either, with studies demonstrating how modifications to cell surface structures (e.g., increased expression of anionic capsular polysaccharides) in P. aeruginosa confer resistance [239,240,241].

5. Conclusions

In conclusion, the exploration of synergistic agents, particularly in tandem with β-lactam antibiotics, marks a critical stride in the ongoing battle against resistant P. aeruginosa. The clinical success of β-lactamase inhibitors is perhaps the greatest and most recent example of this feat, offering novel therapeutic options against β-lactam-resistant strains. While the integration of artificial intelligence offers unprecedented opportunities for identifying novel compounds, the constant threat of resistance, in tandem with financial hurdles and intricate pharmacological considerations, emphasizes the need for cautious optimism. Future work should therefore not only be focused on developing novel antibiotics and synergistic agents but must also continue to address and improve antibiotic stewardship in clinical and agricultural settings.

Author Contributions

Conceptualization, D.W.Z. and C.T.L.; writing—original draft preparation, D.W.Z. and C.T.L.; writing—review and editing, D.W.Z. and C.T.L.; visualization, D.W.Z. and C.T.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
MDRMultidrug resistant
PBPPenicillin-binding protein
SBLSerine β-lactamase
MBLMetallo-β-lactamase
ESBLExtended-spectrum β-lactamase
RNDResistance-nodulation-division
DBODiazabicyclooctane
MICMinimum inhibitory concentration
EPSExtracellular polymeric substances
AMPAntimicrobial peptide
FICFractional inhibitory concentration
TrpTryptophan

References

  1. Magill, S.S.; Edwards, J.R.; Bamberg, W.; Beldavs, Z.G.; Dumyati, G.; Kainer, M.A.; Lynfield, R.; Maloney, M.; McAllister-Hollod, L.; Nadle, J. Multistate point-prevalence survey of health care–associated infections. N. Engl. J. Med. 2014, 370, 1198–1208. [Google Scholar] [CrossRef] [PubMed]
  2. Reynolds, D.; Kollef, M. The Epidemiology and Pathogenesis and Treatment of Pseudomonas aeruginosa Infections: An Update. Drugs 2021, 81, 2117–2131. [Google Scholar] [CrossRef] [PubMed]
  3. Weiner, L.M.; Webb, A.K.; Limbago, B.; Dudeck, M.A.; Patel, J.; Kallen, A.J.; Edwards, J.R.; Sievert, D.M. Antimicrobial-resistant pathogens associated with healthcare-associated infections: Summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2011–2014. Infect. Control Hosp. Epidemiol. 2016, 37, 1288–1301. [Google Scholar] [CrossRef]
  4. Ikuta, K.S.; Swetschinski, L.R.; Aguilar, G.R.; Sharara, F.; Mestrovic, T.; Gray, A.P.; Weaver, N.D.; Wool, E.E.; Han, C.; Hayoon, A.G. Global mortality associated with 33 bacterial pathogens in 2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2022, 400, 2221–2248. [Google Scholar] [CrossRef]
  5. López-Calleja, A.I.; Morales, E.M.; Medina, R.N.; Esgueva, M.F.; Pareja, J.S.; Moya, J.M.G.-L.; Cerón, I.F.; Bayon, J.V.; López, A.R. Antimicrobial activity of ceftolozane-tazobactam against multidrug-resistant and extensively drug-resistant Pseudomonas aeruginosa clinical isolates from a Spanish hospital. Rev. Esp. Quimioter. 2019, 32, 68. [Google Scholar]
  6. Zarb, P.; Coignard, B.; Griskeviciene, J.; Muller, A.; Vankerckhoven, V.; Weist, K.; Goossens, M.M.; Vaerenberg, S.; Hopkins, S.; Catry, B. The European Centre for Disease Prevention and Control (ECDC) pilot point prevalence survey of healthcare-associated infections and antimicrobial use. Eurosurveillance 2012, 17, 20316. [Google Scholar] [CrossRef]
  7. Cooke, A.C.; Nello, A.V.; Ernst, R.K.; Schertzer, J.W. Analysis of Pseudomonas aeruginosa biofilm membrane vesicles supports multiple mechanisms of biogenesis. PLoS ONE 2019, 14, e0212275. [Google Scholar] [CrossRef]
  8. Metruccio, M.M.; Evans, D.J.; Gabriel, M.M.; Kadurugamuwa, J.L.; Fleiszig, S.M. Pseudomonas aeruginosa outer membrane vesicles triggered by human mucosal fluid and lysozyme can prime host tissue surfaces for bacterial adhesion. Front. Microbiol. 2016, 7, 871. [Google Scholar] [CrossRef]
  9. Kadurugamuwa, J.L.; Beveridge, T.J. Bacteriolytic effect of membrane vesicles from Pseudomonas aeruginosa on other bacteria including pathogens: Conceptually new antibiotics. J. Bacteriol. 1996, 178, 2767–2774. [Google Scholar] [CrossRef]
  10. Koeppen, K.; Barnaby, R.; Jackson, A.A.; Gerber, S.A.; Hogan, D.A.; Stanton, B.A. Tobramycin reduces key virulence determinants in the proteome of Pseudomonas aeruginosa outer membrane vesicles. PLoS ONE 2019, 14, e0211290. [Google Scholar] [CrossRef]
  11. He, C.; Zhou, Y.; Liu, F.; Liu, H.; Tan, H.; Jin, S.; Wu, W.; Ge, B. Bacterial nucleotidyl cyclase inhibits the host innate immune response by suppressing TAK1 activation. Infect. Immun. 2017, 85, e00239-17. [Google Scholar] [CrossRef] [PubMed]
  12. Sharma, A.K.; Dhasmana, N.; Dubey, N.; Kumar, N.; Gangwal, A.; Gupta, M.; Singh, Y. Bacterial virulence factors: Secreted for survival. Indian J. Microbiol. 2017, 57, 1–10. [Google Scholar] [CrossRef]
  13. Rasko, D.A.; Sperandio, V. Anti-virulence strategies to combat bacteria-mediated disease. Nat. Rev. Drug Discov. 2010, 9, 117–128. [Google Scholar] [CrossRef]
  14. Hauser, A.R. The type III secretion system of Pseudomonas aeruginosa: Infection by injection. Nat. Rev. Microbiol. 2009, 7, 654–665. [Google Scholar] [CrossRef]
  15. Bagayoko, S.; Leon-Icaza, S.A.; Pinilla, M.; Hessel, A.; Santoni, K.; Péricat, D.; Bordignon, P.-J.; Moreau, F.; Eren, E.; Boyancé, A. Host phospholipid peroxidation fuels ExoU-dependent cell necrosis and supports Pseudomonas aeruginosa-driven pathology. PLoS Path. 2021, 17, e1009927. [Google Scholar] [CrossRef]
  16. Finck-Barbançon, V.; Goranson, J.; Zhu, L.; Sawa, T.; Wiener-Kronish, J.P.; Fleiszig, S.M.; Wu, C.; Mende-Mueller, L.; Frank, D.W. ExoU expression by Pseudomonas aeruginosa correlates with acute cytotoxicity and epithelial injury. Mol. Microbiol. 1997, 25, 547–557. [Google Scholar] [CrossRef]
  17. Zhao, F.; Wang, Q.; Zhang, Y.; Lei, L. Anaerobic biosynthesis of rhamnolipids by Pseudomonas aeruginosa: Performance, mechanism and its application potential for enhanced oil recovery. Microb. Cell Factories 2021, 20, 103. [Google Scholar] [CrossRef]
  18. Eder, K.; Vizler, C.; Kusz, E.; Karcagi, I.; Glavinas, H.; Balogh, G.E.; Vigh, L.; Duda, E.; Gyorfy, Z. The role of lipopolysaccharide moieties in macrophage response to Escherichia coli. Biochem. Biophys. Res. Commun. 2009, 389, 46–51. [Google Scholar] [CrossRef]
  19. Gellatly, S.L.; Hancock, R.E. Pseudomonas aeruginosa: New insights into pathogenesis and host defenses. Pathog. Dis. 2013, 67, 159–173. [Google Scholar] [CrossRef]
  20. Banerjee, D.; Stableforth, D. The treatment of respiratory pseudomonas infection in cystic fibrosis: What drug and which way? Drugs 2000, 60, 1053–1064. [Google Scholar] [CrossRef]
  21. Bush, K.; Bradford, P.A. β-Lactams and β-lactamase inhibitors: An overview. Cold Spring Harb. Perspect. Med. 2016, 6, a025247. [Google Scholar] [CrossRef] [PubMed]
  22. Thakuria, B.; Lahon, K. The Beta Lactam Antibiotics as an Empirical Therapy in a Developing Country: An Update on Their Current Status and Recommendations to Counter the Resistance against Them. J. Clin. Diagn. Res. 2013, 7, 1207–1214. [Google Scholar] [CrossRef] [PubMed]
  23. Klein, E.Y.; Van Boeckel, T.P.; Martinez, E.M.; Pant, S.; Gandra, S.; Levin, S.A.; Goossens, H.; Laxminarayan, R. Global increase and geographic convergence in antibiotic consumption between 2000 and 2015. Proc. Natl. Acad. Sci. USA 2018, 115, E3463–E3470. [Google Scholar] [CrossRef] [PubMed]
  24. Bankar, N.J.; Ugemuge, S.; Ambad, R.S.; Hawale, D.V.; Timilsina, D.R. Implementation of Antimicrobial Stewardship in the Healthcare Setting. Cureus 2022, 14, e26664. [Google Scholar] [CrossRef]
  25. Poole, K. Pseudomonas aeruginosa: Resistance to the max. Front. Microbiol. 2011, 2, 65. [Google Scholar] [CrossRef]
  26. Breidenstein, E.B.; de la Fuente-Núñez, C.; Hancock, R.E. Pseudomonas aeruginosa: All roads lead to resistance. Trends Microbiol. 2011, 19, 419–426. [Google Scholar] [CrossRef]
  27. Cholley, P.; Thouverez, M.; Hocquet, D.; van der Mee-Marquet, N.; Talon, D.; Bertrand, X. Most multidrug-resistant Pseudomonas aeruginosa isolates from hospitals in eastern France belong to a few clonal types. J. Clin. Microbiol. 2011, 49, 2578–2583. [Google Scholar] [CrossRef]
  28. Treepong, P.; Kos, V.N.; Guyeux, C.; Blanc, D.S.; Bertrand, X.; Valot, B.; Hocquet, D. Global emergence of the widespread Pseudomonas aeruginosa ST235 clone. Clin. Microbiol. Infect. 2018, 24, 258–266. [Google Scholar] [CrossRef]
  29. Feretzakis, G.; Loupelis, E.; Sakagianni, A.; Skarmoutsou, N.; Michelidou, S.; Velentza, A.; Martsoukou, M.; Valakis, K.; Petropoulou, S.; Koutalas, E. A 2-year single-centre audit on antibiotic resistance of Pseudomonas aeruginosa, Acinetobacter baumannii and Klebsiella pneumoniae strains from an intensive care unit and other wards in a general public hospital in Greece. Antibiotics 2019, 8, 62. [Google Scholar] [CrossRef]
  30. Matos, E.C.O.d.; Andriolo, R.B.; Rodrigues, Y.C.; Lima, P.D.L.d.; Carneiro, I.C.d.R.S.; Lima, K.V.B. Mortality in patients with multidrug-resistant Pseudomonas aeruginosa infections: A meta-analysis. Rev. Soc. Bras. Med. Trop. 2018, 51, 415–420. [Google Scholar] [CrossRef]
  31. Nathwani, D.; Raman, G.; Sulham, K.; Gavaghan, M.; Menon, V. Clinical and economic consequences of hospital-acquired resistant and multidrug-resistant Pseudomonas aeruginosa infections: A systematic review and meta-analysis. Antimicrob. Resist. Infect. Control. 2014, 3, 32. [Google Scholar] [CrossRef] [PubMed]
  32. Tängdén, T. Combination antibiotic therapy for multidrug-resistant Gram-negative bacteria. Ups. J. Med. Sci. 2014, 119, 149–153. [Google Scholar] [CrossRef] [PubMed]
  33. Worthington, R.J.; Melander, C. Combination approaches to combat multidrug-resistant bacteria. Trends Biotechnol. 2013, 31, 177–184. [Google Scholar] [CrossRef]
  34. Ahmed, A.; Azim, A.; Gurjar, M.; Baronia, A.K. Current concepts in combination antibiotic therapy for critically ill patients. Indian J. Crit. Care Med. 2014, 18, 310–314. [Google Scholar] [CrossRef]
  35. Pletz, M.W.; Hagel, S.; Forstner, C. Who benefits from antimicrobial combination therapy? Lancet Infect. Dis. 2017, 17, 677–678. [Google Scholar] [CrossRef]
  36. Wang, N.; Luo, J.; Deng, F.; Huang, Y.; Zhou, H. Antibiotic combination therapy: A strategy to overcome bacterial resistance to aminoglycoside antibiotics. Front. Pharmacol. 2022, 13, 839808. [Google Scholar] [CrossRef]
  37. Kim, D.; Kim, S.; Kwon, Y.; Kim, Y.; Park, H.; Kwak, K.; Lee, H.; Lee, J.H.; Jang, K.M.; Kim, D.; et al. Structural Insights for β-Lactam Antibiotics. Biomol. Ther. 2023, 31, 141–147. [Google Scholar] [CrossRef]
  38. Dalhoff, A.; Janjic, N.; Echols, R. Redefining penems. Biochem. Pharmacol. 2006, 71, 1085–1095. [Google Scholar] [CrossRef]
  39. Papp-Wallace, K.M.; Endimiani, A.; Taracila, M.A.; Bonomo, R.A. Carbapenems: Past, present, and future. Antimicrob. Agents Chemother. 2011, 55, 4943–4960. [Google Scholar] [CrossRef]
  40. Flores-Kim, J.; Dobihal, G.S.; Fenton, A.; Rudner, D.Z.; Bernhardt, T.G. A switch in surface polymer biogenesis triggers growth-phase-dependent and antibiotic-induced bacteriolysis. Elife 2019, 8, e44912. [Google Scholar] [CrossRef]
  41. Mora-Ochomogo, M.; Lohans, C.T. β-Lactam antibiotic targets and resistance mechanisms: From covalent inhibitors to substrates. RSC Med. Chem. 2021, 12, 1623–1639. [Google Scholar] [CrossRef] [PubMed]
  42. Ambler, R.P. The structure of β-lactamases. Philos. Trans. R. Soc. London. B Biol. Sci. 1980, 289, 321–331. [Google Scholar]
  43. Zhao, W.-H.; Hu, Z.-Q. β-lactamases identified in clinical isolates of Pseudomonas aeruginosa. Crit. Rev. Microbiol. 2010, 36, 245–258. [Google Scholar] [CrossRef]
  44. Tooke, C.L.; Hinchliffe, P.; Bragginton, E.C.; Colenso, C.K.; Hirvonen, V.H.A.; Takebayashi, Y.; Spencer, J. β-Lactamases and β-Lactamase Inhibitors in the 21st Century. J. Mol. Biol. 2019, 431, 3472–3500. [Google Scholar] [CrossRef]
  45. Bush, K. Bench-to-bedside review: The role of β-lactamases in antibiotic-resistant Gram-negative infections. Crit. Care 2010, 14, 224. [Google Scholar] [CrossRef]
  46. Jacoby George, A. AmpC β-Lactamases. Clin. Microbiol. Rev. 2009, 22, 161–182. [Google Scholar] [CrossRef]
  47. Evans Benjamin, A.; Amyes Sebastian, G.B. OXA β-Lactamases. Clin. Microbiol. Rev. 2014, 27, 241–263. [Google Scholar] [CrossRef]
  48. Naas, T.; Oueslati, S.; Bonnin, R.A.; Dabos, M.L.; Zavala, A.; Dortet, L.; Retailleau, P.; Iorga, B.I. Beta-lactamase database (BLDB)–structure and function. J. Enzyme Inhib. Med. Chem. 2017, 32, 917–919. [Google Scholar] [CrossRef]
  49. Bonomo, R.A. β-Lactamases: A Focus on Current Challenges. Cold Spring Harb. Perspect. Med. 2017, 7, a025239. [Google Scholar] [CrossRef]
  50. Bush, K.; Bradford, P.A. Epidemiology of β-lactamase-producing pathogens. Clin. Microbiol. Rev. 2020, 33, e00047-19. [Google Scholar] [CrossRef]
  51. Bush, K.; Bradford, P.A. Interplay between β-lactamases and new β-lactamase inhibitors. Nat. Rev. Microbiol. 2019, 17, 295–306. [Google Scholar] [CrossRef] [PubMed]
  52. Castanheira, M.; Simner, P.J.; Bradford, P.A. Extended-spectrum β-lactamases: An update on their characteristics, epidemiology and detection. JAC-Antimicrob. Resist. 2021, 3, dlab092. [Google Scholar] [CrossRef]
  53. Walther-Rasmussen, J.; Høiby, N. OXA-type carbapenemases. J. Antimicrob. Chemother. 2006, 57, 373–383. [Google Scholar] [CrossRef]
  54. Medeiros, A.A. Evolution and dissemination of β-lactamases accelerated by generations of β-lactam antibiotics. Clin. Infect. Dis. 1997, 24, S19–S45. [Google Scholar] [CrossRef]
  55. Walther-Rasmussen, J.; Høiby, N. Class A carbapenemases. J. Antimicrob. Chemother. 2007, 60, 470–482. [Google Scholar] [CrossRef]
  56. Poirel, L.; Potron, A.; Nordmann, P. OXA-48-like carbapenemases: The phantom menace. J. Antimicrob. Chemother. 2012, 67, 1597–1606. [Google Scholar] [CrossRef]
  57. Boyd, S.E.; Holmes, A.; Peck, R.; Livermore, D.M.; Hope, W. OXA-48-like β-lactamases: Global epidemiology, treatment options, and development pipeline. Antimicrob. Agents Chemother. 2022, 66, e00216–e00222. [Google Scholar] [CrossRef]
  58. Halat, D.H.; Moubareck, C.A. The Intriguing Carbapenemases of Pseudomonas aeruginosa: Current Status, Genetic Profile, and Global Epidemiology. Yale J. Biol. Med. 2022, 95, 507. [Google Scholar]
  59. Potron, A.; Poirel, L.; Nordmann, P. Emerging broad-spectrum resistance in Pseudomonas aeruginosa and Acinetobacter baumannii: Mechanisms and epidemiology. Int. J. Antimicrob. Agents 2015, 45, 568–585. [Google Scholar] [CrossRef] [PubMed]
  60. Walsh, T.R.; Toleman, M.A.; Poirel, L.; Nordmann, P. Metallo-β-lactamases: The quiet before the storm? Clin. Microbiol. Rev. 2005, 18, 306–325. [Google Scholar] [CrossRef]
  61. Ju, L.C.; Cheng, Z.; Fast, W.; Bonomo, R.A.; Crowder, M.W. The Continuing Challenge of Metallo-β-Lactamase Inhibition: Mechanism Matters. Trends Pharmacol. Sci. 2018, 39, 635–647. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, P.; Wu, W.; Wang, N.; Feng, H.; Wang, J.; Wang, F.; Zhang, Y.; Chen, H.; Yang, Q.; Jiang, Y. Pseudomonas aeruginosa High-Risk Sequence Type 463 Co-Producing KPC-2 and AFM-1 Carbapenemases, China, 2020–2022. Emerging Infect. Dis. 2023, 29, 2136. [Google Scholar] [CrossRef] [PubMed]
  63. Peña, C.; Suarez, C.; Gozalo, M.; Murillas, J.; Almirante, B.; Pomar, V.; Aguilar, M.; Granados, A.; Calbo, E.; Rodríguez-Baño, J. Prospective multicenter study of the impact of carbapenem resistance on mortality in Pseudomonas aeruginosa bloodstream infections. Antimicrob. Agents Chemother. 2012, 56, 1265–1272. [Google Scholar] [CrossRef]
  64. Reyes, J.; Komarow, L.; Chen, L.; Ge, L.; Hanson, B.M.; Cober, E.; Herc, E.; Alenazi, T.; Kaye, K.S.; Garcia-Diaz, J. Global epidemiology and clinical outcomes of carbapenem-resistant Pseudomonas aeruginosa and associated carbapenemases (POP): A prospective cohort study. Lancet Microbe 2023, 4, e159–e170. [Google Scholar] [CrossRef]
  65. Thaden, J.T.; Park, L.P.; Maskarinec, S.A.; Ruffin, F.; Fowler, V.G., Jr.; Van Duin, D. Results from a 13-year prospective cohort study show increased mortality associated with bloodstream infections caused by Pseudomonas aeruginosa compared to other bacteria. Antimicrob. Agents Chemother. 2017, 61, e02671-16. [Google Scholar] [CrossRef]
  66. Yuan, Q.; Guo, L.; Li, B.; Zhang, S.; Feng, H.; Zhang, Y.; Yu, M.; Hu, H.; Chen, H.; Yang, Q. Risk factors and outcomes of inpatients with carbapenem-resistant Pseudomonas aeruginosa bloodstream infections in China: A 9-year trend and multicenter cohort study. Front. Microbiol. 2023, 14, 1137811. [Google Scholar] [CrossRef]
  67. Liu, Q.; Li, X.; Li, W.; Du, X.; He, J.-Q.; Tao, C.; Feng, Y. Influence of carbapenem resistance on mortality of patients with Pseudomonas aeruginosa infection: A meta-analysis. Sci. Rep. 2015, 5, 11715. [Google Scholar] [CrossRef]
  68. Kang, C.-I.; Kim, S.-H.; Kim, H.-B.; Park, S.-W.; Choe, Y.-J.; Oh, M.-d.; Kim, E.-C.; Choe, K.-W. Pseudomonas aeruginosa bacteremia: Risk factors for mortality and influence of delayed receipt of effective antimicrobial therapy on clinical outcome. Clin. Infect. Dis. 2003, 37, 745–751. [Google Scholar] [CrossRef]
  69. Kang, C.-I.; Kim, S.-H.; Park, W.B.; Lee, K.-D.; Kim, H.-B.; Kim, E.-C.; Oh, M.-d.; Choe, K.-W. Bloodstream infections caused by antibiotic-resistant gram-negative bacilli: Risk factors for mortality and impact of inappropriate initial antimicrobial therapy on outcome. Antimicrob. Agents Chemother. 2005, 49, 760–766. [Google Scholar] [CrossRef]
  70. Cosgrove, S.E. The relationship between antimicrobial resistance and patient outcomes: Mortality, length of hospital stay, and health care costs. Clin. Infect. Dis. 2006, 42, S82–S89. [Google Scholar] [CrossRef]
  71. Kadri, S.S.; Lai, Y.L.; Warner, S.; Strich, J.R.; Babiker, A.; Ricotta, E.E.; Demirkale, C.Y.; Dekker, J.P.; Palmore, T.N.; Rhee, C. Inappropriate empirical antibiotic therapy for bloodstream infections based on discordant in-vitro susceptibilities: A retrospective cohort analysis of prevalence, predictors, and mortality risk in US hospitals. Lancet Infect. Dis. 2021, 21, 241–251. [Google Scholar] [CrossRef] [PubMed]
  72. Chumbita, M.; Puerta-Alcalde, P.; Yáñez, L.; Angeles Cuesta, M.; Chinea, A.; Español-Morales, I.; Fernandez-Abellán, P.; Gudiol, C.; González-Sierra, P.; Rojas, R. High rate of inappropriate antibiotics in patients with hematologic malignancies and Pseudomonas aeruginosa bacteremia following international guideline recommendations. Microbiol. Spectr. 2023, 11, e00674-23. [Google Scholar] [CrossRef] [PubMed]
  73. Suarez, C.; Pena, C.; Gavalda, L.; Tubau, F.; Manzur, A.; Dominguez, M.A.; Pujol, M.; Gudiol, F.; Ariza, J. Influence of carbapenem resistance on mortality and the dynamics of mortality in Pseudomonas aeruginosa bloodstream infection. Int. J. Infect. Dis. 2010, 14, e73–e78. [Google Scholar] [CrossRef]
  74. Sid Ahmed, M.A.; Khan, F.A.; Sultan, A.A.; Söderquist, B.; Ibrahim, E.B.; Jass, J.; Omrani, A.S. β-lactamase-mediated resistance in MDR-Pseudomonas aeruginosa from Qatar. Antimicrob. Resist. Infect. Control. 2020, 9, 170. [Google Scholar] [CrossRef]
  75. Costa-Júnior, S.D.; da Silva, A.M.C.M.; Niedja da Paz Pereira, J.; da Costa Lima, J.L.; Cavalcanti, I.M.F.; Maciel, M.A.V. Emergence of rmtD1 gene in clinical isolates of Pseudomonas aeruginosa carrying bla KPC and/or bla VIM-2 genes in Brazil. Braz. J. Microbiol. 2021, 52, 1959–1965. [Google Scholar] [CrossRef]
  76. Murray, C.J.; Ikuta, K.S.; Sharara, F.; Swetschinski, L.; Aguilar, G.R.; Gray, A.; Han, C.; Bisignano, C.; Rao, P.; Wool, E. Global burden of bacterial antimicrobial resistance in 2019: A systematic analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef]
  77. Chen, W.; Zhang, Y.-M.; Davies, C. Penicillin-binding protein 3 is essential for growth of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2017, 61, e01651-16. [Google Scholar] [CrossRef]
  78. Falagas, M.E.; Mavroudis, A.D.; Vardakas, K.Z. The antibiotic pipeline for multi-drug resistant gram negative bacteria: What can we expect? Expert Rev. Anti Infect. Ther. 2016, 14, 747–763. [Google Scholar] [CrossRef]
  79. Theuretzbacher, U.; Gottwalt, S.; Beyer, P.; Butler, M.; Czaplewski, L.; Lienhardt, C.; Moja, L.; Paul, M.; Paulin, S.; Rex, J.H. Analysis of the clinical antibacterial and antituberculosis pipeline. Lancet Infect. Dis. 2019, 19, e40–e50. [Google Scholar] [CrossRef]
  80. Diaz Caballero, J.; Clark, S.T.; Coburn, B.; Zhang, Y.; Wang, P.W.; Donaldson, S.L.; Tullis, D.E.; Yau, Y.C.; Waters, V.J.; Hwang, D.M. Selective sweeps and parallel pathoadaptation drive Pseudomonas aeruginosa evolution in the cystic fibrosis lung. mBio 2015, 6, e00981-15. [Google Scholar] [CrossRef]
  81. Clark, S.T.; Sinha, U.; Zhang, Y.; Wang, P.W.; Donaldson, S.L.; Coburn, B.; Waters, V.J.; Yau, Y.C.; Tullis, D.E.; Guttman, D.S. Penicillin-binding protein 3 is a common adaptive target among Pseudomonas aeruginosa isolates from adult cystic fibrosis patients treated with β-lactams. Int. J. Antimicrob. Agents 2019, 53, 620–628. [Google Scholar] [CrossRef] [PubMed]
  82. Nikaido, H. Molecular basis of bacterial outer membrane permeability revisited. Microbiol. Mol. Biol. Rev. 2003, 67, 593–656. [Google Scholar] [CrossRef] [PubMed]
  83. Pagès, J.-M.; James, C.E.; Winterhalter, M. The porin and the permeating antibiotic: A selective diffusion barrier in Gram-negative bacteria. Nat. Rev. Microbiol. 2008, 6, 893–903. [Google Scholar] [CrossRef] [PubMed]
  84. Chevalier, S.; Bouffartigues, E.; Bodilis, J.; Maillot, O.; Lesouhaitier, O.; Feuilloley, M.G.; Orange, N.; Dufour, A.; Cornelis, P. Structure, function and regulation of Pseudomonas aeruginosa porins. FEMS Microbiol. Rev. 2017, 41, 698–722. [Google Scholar] [CrossRef]
  85. Llanes, C.; Pourcel, C.; Richardot, C.; Plésiat, P.; Fichant, G.; Cavallo, J.-D.; Mérens, A.; Group, G.S.; Vu-Thien, H.; Leclercq, R. Diversity of β-lactam resistance mechanisms in cystic fibrosis isolates of Pseudomonas aeruginosa: A French multicentre study. J. Antimicrob. Chemother. 2013, 68, 1763–1771. [Google Scholar] [CrossRef]
  86. Giske, C.G.; Buarø, L.; Sundsfjord, A.; Wretlind, B. Alterations of porin, pumps, and penicillin-binding proteins in carbapenem resistant clinical isolates of Pseudomonas aeruginosa. Microb. Drug Resist. 2008, 14, 23–30. [Google Scholar] [CrossRef]
  87. Li, H.; Luo, Y.-F.; Williams, B.J.; Blackwell, T.S.; Xie, C.-M. Structure and function of OprD protein in Pseudomonas aeruginosa: From antibiotic resistance to novel therapies. Int. J. Med. Microbiol. 2012, 302, 63–68. [Google Scholar] [CrossRef]
  88. Ochs, M.M.; McCusker, M.P.; Bains, M.; Hancock, R.E. Negative regulation of the Pseudomonas aeruginosa outer membrane porin OprD selective for imipenem and basic amino acids. Antimicrob. Agents Chemother. 1999, 43, 1085–1090. [Google Scholar] [CrossRef]
  89. Iyer, R.; Sylvester, M.A.; Velez-Vega, C.; Tommasi, R.; Durand-Reville, T.F.; Miller, A.A. Whole-cell-based assay to evaluate structure permeation relationships for carbapenem passage through the Pseudomonas aeruginosa porin OprD. ACS Infect. Dis. 2017, 3, 310–319. [Google Scholar] [CrossRef]
  90. Ude, J.; Tripathi, V.; Buyck, J.M.; Söderholm, S.; Cunrath, O.; Fanous, J.; Claudi, B.; Egli, A.; Schleberger, C.; Hiller, S. Outer membrane permeability: Antimicrobials and diverse nutrients bypass porins in Pseudomonas aeruginosa. Proc. Natl. Acad. Sci. USA 2021, 118, e2107644118. [Google Scholar] [CrossRef]
  91. Isabella, V.M.; Campbell, A.J.; Manchester, J.; Sylvester, M.; Nayar, A.S.; Ferguson, K.E.; Tommasi, R.; Miller, A.A. Toward the rational design of carbapenem uptake in Pseudomonas aeruginosa. Chem. Biol. 2015, 22, 535–547. [Google Scholar] [CrossRef] [PubMed]
  92. Tamber, S.; Maier, E.; Benz, R.; Hancock, R.E. Characterization of OpdH, a Pseudomonas aeruginosa porin involved in the uptake of tricarboxylates. J. Bacteriol. 2007, 189, 929–939. [Google Scholar] [CrossRef]
  93. Kabra, R.; Chauhan, N.; Kumar, A.; Ingale, P.; Singh, S. Efflux pumps and antimicrobial resistance: Paradoxical components in systems genomics. Prog. Biophys. Mol. Biol. 2019, 141, 15–24. [Google Scholar] [CrossRef]
  94. Nikaido, H.; Takatsuka, Y. Mechanisms of RND multidrug efflux pumps. Biochim. Biophys. Acta (BBA)-Proteins Proteom. 2009, 1794, 769–781. [Google Scholar] [CrossRef]
  95. Fernando, D.M.; Kumar, A. Resistance-nodulation-division multidrug efflux pumps in gram-negative bacteria: Role in virulence. Antibiotics 2013, 2, 163–181. [Google Scholar] [CrossRef]
  96. Alav, I.; Kobylka, J.; Kuth, M.S.; Pos, K.M.; Picard, M.; Blair, J.M.; Bavro, V.N. Structure, assembly, and function of tripartite efflux and type 1 secretion systems in gram-negative bacteria. Chem. Rev. 2021, 121, 5479–5596. [Google Scholar] [CrossRef]
  97. del Barrio-Tofiño, E.; López-Causapé, C.; Cabot, G.; Rivera, A.; Benito, N.; Segura, C.; Montero, M.M.; Sorlí, L.; Tubau, F.; Gómez-Zorrilla, S. Genomics and susceptibility profiles of extensively drug-resistant Pseudomonas aeruginosa isolates from Spain. Antimicrob. Agents Chemother. 2017, 61, e01589-17. [Google Scholar] [CrossRef]
  98. Lee, J.-Y.; Ko, K.S. OprD mutations and inactivation, expression of efflux pumps and AmpC, and metallo-β-lactamases in carbapenem-resistant Pseudomonas aeruginosa isolates from South Korea. Int. J. Antimicrob. Agents 2012, 40, 168–172. [Google Scholar] [CrossRef]
  99. Moubareck, C.A.; Halat, D.H.; Akkawi, C.; Nabi, A.; AlSharhan, M.A.; AlDeesi, Z.O.; Peters, C.C.; Celiloglu, H.; Sarkis, D.K. Role of outer membrane permeability, efflux mechanism, and carbapenemases in carbapenem-nonsusceptible Pseudomonas aeruginosa from Dubai hospitals: Results of the first cross-sectional survey. Int. J. Infect. Dis. 2019, 84, 143–150. [Google Scholar] [CrossRef]
  100. Köhler, T.; Michéa-Hamzehpour, M.; Henze, U.; Gotoh, N.; Kocjancic Curty, L.; Pechère, J.C. Characterization of MexE–MexF–OprN, a positively regulated multidrug efflux system of Pseudomonas aeruginosa. Mol. Microbiol. 1997, 23, 345–354. [Google Scholar] [CrossRef]
  101. Moyá, B.; Beceiro, A.; Cabot, G.; Juan, C.; Zamorano, L.; Alberti, S.; Oliver, A. Pan-β-lactam resistance development in Pseudomonas aeruginosa clinical strains: Molecular mechanisms, penicillin-binding protein profiles, and binding affinities. Antimicrob. Agents Chemother. 2012, 56, 4771–4778. [Google Scholar] [CrossRef] [PubMed]
  102. Pan, Y.-p.; Xu, Y.-h.; Wang, Z.-x.; Fang, Y.-p.; Shen, J.-l. Overexpression of MexAB-OprM efflux pump in carbapenem-resistant Pseudomonas aeruginosa. Arch. Microbiol. 2016, 198, 565–571. [Google Scholar] [CrossRef] [PubMed]
  103. Chalhoub, H.; Pletzer, D.; Weingart, H.; Braun, Y.; Tunney, M.M.; Elborn, J.S.; Rodriguez-Villalobos, H.; Plésiat, P.; Kahl, B.C.; Denis, O. Mechanisms of intrinsic resistance and acquired susceptibility of Pseudomonas aeruginosa isolated from cystic fibrosis patients to temocillin, a revived antibiotic. Sci. Rep. 2017, 7, 40208. [Google Scholar] [CrossRef]
  104. Masuda, N.; Sakagawa, E.; Ohya, S.; Gotoh, N.; Tsujimoto, H.; Nishino, T. Substrate specificities of MexAB-OprM, MexCD-OprJ, and MexXY-oprM efflux pumps in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2000, 44, 3322–3327. [Google Scholar] [CrossRef]
  105. Frimodt-Møller, J.; Rossi, E.; Haagensen, J.A.J.; Falcone, M.; Molin, S.; Johansen, H.K. Mutations causing low level antibiotic resistance ensure bacterial survival in antibiotic-treated hosts. Sci. Rep. 2018, 8, 12512. [Google Scholar] [CrossRef]
  106. Masuda, N.; Sakagawa, E.; Ohya, S.; Gotoh, N.; Tsujimoto, H.; Nishino, T. Contribution of the MexX-MexY-OprM efflux system to intrinsic resistance in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2000, 44, 2242–2246. [Google Scholar] [CrossRef]
  107. Gomis-Font, M.A.; Pitart, C.; del Barrio-Tofiño, E.; Zboromyrska, Y.; Cortes-Lara, S.; Mulet, X.; Marco, F.; Vila, J.; López-Causapé, C.; Oliver, A. Emergence of Resistance to Novel Cephalosporin–β-lactamase Inhibitor Combinations through the Modification of the Pseudomonas aeruginosa MexCD-OprJ Efflux Pump. Antimicrob. Agents Chemother. 2021, 65, e00089-21. [Google Scholar] [CrossRef]
  108. Evans, K.; Adewoye, L.; Poole, K. MexR repressor of the mexAB-oprM multidrug efflux operon of Pseudomonas aeruginosa: Identification of MexR binding sites in the mexA-mexR intergenic region. J. Bacteriol. 2001, 183, 807–812. [Google Scholar] [CrossRef]
  109. Saito, K.; Eda, S.; Maseda, H.; Nakae, T. Molecular mechanism of MexR-mediated regulation of MexAB–OprM efflux pump expression in Pseudomonas aeruginosa. FEMS Microbiol. Lett. 2001, 195, 23–28. [Google Scholar] [CrossRef]
  110. Poole, K.; Tetro, K.; Zhao, Q.; Neshat, S.; Heinrichs, D.E.; Bianco, N. Expression of the multidrug resistance operon mexA-mexB-oprM in Pseudomonas aeruginosa: mexR encodes a regulator of operon expression. Antimicrob. Agents Chemother. 1996, 40, 2021–2028. [Google Scholar] [CrossRef]
  111. Sobel, M.L.; Hocquet, D.; Cao, L.; Plesiat, P.; Poole, K. Mutations in PA3574 (nalD) lead to increased MexAB-OprM expression and multidrug resistance in laboratory and clinical isolates of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2005, 49, 1782–1786. [Google Scholar] [CrossRef] [PubMed]
  112. Saito, K.; Yoneyama, H.; Nakae, T. nalB-type mutations causing the overexpression of the MexAB-OprM efflux pump are located in the mexR gene of the Pseudomonas aeruginosa chromosome. FEMS Microbiol. Lett. 1999, 179, 67–72. [Google Scholar] [CrossRef]
  113. Morita, Y.; Cao, L.; Gould, V.C.; Avison, M.B.; Poole, K. nalD encodes a second repressor of the mexAB-oprM multidrug efflux operon of Pseudomonas aeruginosa. J. Bacteriol. 2006, 188, 8649–8654. [Google Scholar] [CrossRef]
  114. Köhler, T.; Michea-Hamzehpour, M.; Epp, S.F.; Pechere, J.-C. Carbapenem activities against Pseudomonas aeruginosa: Respective contributions of OprD and efflux systems. Antimicrob. Agents Chemother. 1999, 43, 424–427. [Google Scholar] [CrossRef]
  115. Sobel, M.L.; Neshat, S.; Poole, K. Mutations in PA2491 (mexS) promote MexT-dependent mexEF-oprN expression and multidrug resistance in a clinical strain of Pseudomonas aeruginosa. J. Bacteriol. 2005, 187, 1246–1253. [Google Scholar] [CrossRef]
  116. Köhler, T.; Epp, S.F.; Curty, L.K.; Pechère, J.-C. Characterization of MexT, the regulator of the MexE-MexF-OprN multidrug efflux system of Pseudomonas aeruginosa. J. Bacteriol. 1999, 181, 6300–6305. [Google Scholar] [CrossRef]
  117. Reading, C.; Cole, M. Clavulanic acid: A beta-lactamase-inhibiting beta-lactam from Streptomyces clavuligerus. Antimicrob. Agents Chemother. 1977, 11, 852–857. [Google Scholar] [CrossRef]
  118. Charnas, R.L.; Fisher, J.; Knowles, J.R. Chemical studies on the inactivation of Escherichia coli RTEM β-lactamase by clavulanic acid. Biochemistry 1978, 17, 2185–2189. [Google Scholar] [CrossRef]
  119. Charnas, R.L.; Knowles, J.R. Inactivation of radiolabeled RTEM. beta.-lactamase from Escherichia coli by clavulanic acid and 9-deoxyclavulanic acid. Biochemistry 1981, 20, 3214–3219. [Google Scholar] [CrossRef]
  120. Bush, K. A resurgence of β-lactamase inhibitor combinations effective against multidrug-resistant Gram-negative pathogens. Int. J. Antimicrob. Agents 2015, 46, 483–493. [Google Scholar] [CrossRef]
  121. Yigit, H.; Queenan, A.M.; Rasheed, J.K.; Biddle, J.W.; Domenech-Sanchez, A.; Alberti, S.; Bush, K.; Tenover, F.C. Carbapenem-resistant strain of Klebsiella oxytoca harboring carbapenem-hydrolyzingβ-lactamase KPC-2. Antimicrob. Agents Chemother. 2003, 47, 3881–3889. [Google Scholar] [CrossRef] [PubMed]
  122. Woodford, N.; Tierno, P.M., Jr.; Young, K.; Tysall, L.; Palepou, M.-F.I.; Ward, E.; Painter, R.E.; Suber, D.F.; Shungu, D.; Silver, L.L. Outbreak of Klebsiella pneumoniae producing a new carbapenem-hydrolyzing class A β-lactamase, KPC-3, in a New York medical center. Antimicrob. Agents Chemother. 2004, 48, 4793–4799. [Google Scholar] [CrossRef]
  123. Moland, E.S.; Hong, S.G.; Thomson, K.S.; Larone, D.H.; Hanson, N.D. Klebsiella pneumoniae isolate producing at least eight different β-lactamases, including AmpC and KPC β-lactamases. Antimicrob. Agents Chemother. 2007, 51, 800. [Google Scholar] [CrossRef]
  124. Yahav, D.; Giske, C.G.; Grāmatniece, A.; Abodakpi, H.; Tam, V.H.; Leibovici, L. New β-lactam–β-lactamase inhibitor combinations. Clin. Microbiol. Rev. 2020, 34. [Google Scholar] [CrossRef]
  125. Coleman, K. Diazabicyclooctanes (DBOs): A potent new class of non-β-lactam β-lactamase inhibitors. Curr. Opin. Microbiol. 2011, 14, 550–555. [Google Scholar] [CrossRef]
  126. Rubino, C.M.; Bhavnani, S.M.; Loutit, J.S.; Morgan, E.E.; White, D.; Dudley, M.N.; Griffith, D.C. Phase 1 study of the safety, tolerability, and pharmacokinetics of vaborbactam and meropenem alone and in combination following single and multiple doses in healthy adult subjects. Antimicrob. Agents Chemother. 2018, 62, e02228-17. [Google Scholar] [CrossRef]
  127. Hecker, S.J.; Reddy, K.R.; Totrov, M.; Hirst, G.C.; Lomovskaya, O.; Griffith, D.C.; King, P.; Tsivkovski, R.; Sun, D.; Sabet, M. Discovery of a cyclic boronic acid β-lactamase inhibitor (RPX7009) with utility vs. class A serine carbapenemases. J. Med. Chem. 2015, 58, 3682–3692. [Google Scholar] [CrossRef]
  128. Lee, S.Y.; Gill, C.M.; Nicolau, D.P. Activity of novel β-lactam/β-lactamase inhibitor combinations against serine carbapenemase-producing carbapenem-resistant Pseudomonas aeruginosa. J. Antimicrob. Chemother. 2023, 78, 2795–2800. [Google Scholar] [CrossRef]
  129. Ruiz, V.H.; Gill, C.M.; Nicolau, D.P. Assessing the in vivo impact of novel β-lactamase inhibitors on the efficacy of their partner β-lactams against serine carbapenemase-producing Pseudomonas aeruginosa using human-simulated exposures. J. Antimicrob. Chemother. 2024, 79, 546–551. [Google Scholar] [CrossRef]
  130. González-Pinto, L.; Alonso-García, I.; Blanco-Martín, T.; Camacho-Zamora, P.; Fraile-Ribot, P.A.; Outeda-García, M.; Lasarte-Monterrubio, C.; Guijarro-Sánchez, P.; Maceiras, R.; Moya, B.; et al. Impact of chromosomally encoded resistance mechanisms and transferable β-lactamases on the activity of cefiderocol and innovative β-lactam/β-lactamase inhibitor combinations against Pseudomonas aeruginosa. J. Antimicrob. Chemother. 2024, 79, 2591–2597. [Google Scholar] [CrossRef]
  131. Carvalhaes, C.G.; Shortridge, D.; Sader, H.S.; Castanheira, M. Activity of Meropenem-Vaborbactam against Bacterial Isolates Causing Pneumonia in Patients in U.S. Hospitals during 2014 to 2018. Antimicrob. Agents Chemother. 2020, 64, e02177-19. [Google Scholar] [CrossRef] [PubMed]
  132. Mushtaq, S.; Meunier, D.; Vickers, A.; Woodford, N.; Livermore, D.M. Activity of imipenem/relebactam against Pseudomonas aeruginosa producing ESBLs and carbapenemases. J. Antimicrob. Chemother. 2021, 76, 434–442. [Google Scholar] [CrossRef] [PubMed]
  133. Livermore, D.M.; Mushtaq, S.; Warner, M.; Woodford, N. Activity of OP0595/β-lactam combinations against Gram-negative bacteria with extended-spectrum, AmpC and carbapenem-hydrolysing β-lactamases. J. Antimicrob. Chemother. 2015, 70, 3032–3041. [Google Scholar] [CrossRef] [PubMed]
  134. Livermore, D.M.; Warner, M.; Mushtaq, S. Activity of MK-7655 combined with imipenem against Enterobacteriaceae and Pseudomonas aeruginosa. J. Antimicrob. Chemother. 2013, 68, 2286–2290. [Google Scholar] [CrossRef]
  135. Le Terrier, C.; Nordmann, P.; Poirel, L. In vitro activity of aztreonam in combination with newly developed β-lactamase inhibitors against MDR Enterobacterales and Pseudomonas aeruginosa producing metallo-β-lactamases. J. Antimicrob. Chemother. 2023, 78, 101–107. [Google Scholar] [CrossRef]
  136. Thomson, K.S.; AbdelGhani, S.; Snyder, J.W.; Thomson, G.K. Activity of cefepime-zidebactam against multidrug-resistant (MDR) Gram-negative pathogens. Antibiotics 2019, 8, 32. [Google Scholar] [CrossRef]
  137. Lomovskaya, O.; Rubio-Aparicio, D.; Nelson, K.; Sun, D.; Tsivkovski, R.; Castanheira, M.; Lindley, J.; Loutit, J.; Dudley, M. In Vitro Activity of the Ultrabroad-Spectrum Beta-Lactamase Inhibitor QPX7728 in Combination with Multiple Beta-Lactam Antibiotics against Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2021, 65, e00210-21. [Google Scholar] [CrossRef]
  138. Slater, C.L.; Winogrodzki, J.; Fraile-Ribot, P.A.; Oliver, A.; Khajehpour, M.; Mark, B.L. Adding Insult to Injury: Mechanistic Basis for How AmpC Mutations Allow Pseudomonas aeruginosa To Accelerate Cephalosporin Hydrolysis and Evade Avibactam. Antimicrob. Agents Chemother. 2020, 64, e00894-20. [Google Scholar] [CrossRef]
  139. Alonso-García, I.; Vázquez-Ucha, J.C.; Lasarte-Monterrubio, C.; González-Mayo, E.; Lada-Salvador, P.; Vela-Fernández, R.; Aja-Macaya, P.; Guijarro-Sánchez, P.; Rumbo-Feal, S.; Muíño-Andrade, M.; et al. Simultaneous and divergent evolution of resistance to cephalosporin/β-lactamase inhibitor combinations and imipenem/relebactam following ceftazidime/avibactam treatment of MDR Pseudomonas aeruginosa infections. J. Antimicrob. Chemother. 2023, 78, 1195–1200. [Google Scholar] [CrossRef]
  140. Ruedas-López, A.; Alonso-García, I.; Lasarte-Monterrubio, C.; Guijarro-Sánchez, P.; Gato, E.; Vázquez-Ucha, J.C.; Vallejo, J.A.; Fraile-Ribot, P.A.; Fernández-Pérez, B.; Velasco, D.; et al. Selection of AmpC β-Lactamase Variants and Metallo-β-Lactamases Leading to Ceftolozane/Tazobactam and Ceftazidime/Avibactam Resistance during Treatment of MDR/XDR Pseudomonas aeruginosa Infections. Antimicrob. Agents Chemother. 2022, 66, e0206721. [Google Scholar] [CrossRef]
  141. Hujer, A.M.; Bethel, C.R.; Taracila, M.A.; Marshall, S.H.; Rojas, L.J.; Winkler, M.L.; Painter, R.E.; Domitrovic, T.N.; Watkins, R.R.; Abdelhamed, A.M.; et al. Imipenem/Relebactam Resistance in Clinical Isolates of Extensively Drug Resistant Pseudomonas aeruginosa: Inhibitor-Resistant β-Lactamases and Their Increasing Importance. Antimicrob. Agents Chemother. 2022, 66, e0179021. [Google Scholar] [CrossRef] [PubMed]
  142. Bruynoghe, R.; Maisin, J. Essais de thérapeutique au moyen du bacteriophage. CR Soc. Biol. 1921, 85, 1120–1121. [Google Scholar]
  143. Stone, R. Stalin’s forgotten cure. Science 2002, 298, 728–731. [Google Scholar] [CrossRef]
  144. Kutateladze, Á.; Adamia, R. Phage therapy experience at the Eliava Institute. Med. Mal. Infect. 2008, 38, 426–430. [Google Scholar] [CrossRef]
  145. Abedon, S.T.; Kuhl, S.J.; Blasdel, B.G.; Kutter, E.M. Phage treatment of human infections. Bacteriophage 2011, 1, 66–85. [Google Scholar] [CrossRef]
  146. Schooley, R.T.; Biswas, B.; Gill, J.J.; Hernandez-Morales, A.; Lancaster, J.; Lessor, L.; Barr, J.J.; Reed, S.L.; Rohwer, F.; Benler, S. Development and use of personalized bacteriophage-based therapeutic cocktails to treat a patient with a disseminated resistant Acinetobacter baumannii infection. Antimicrob. Agents Chemother. 2017, 61, e00954-17. [Google Scholar] [CrossRef]
  147. El Haddad, L.; Harb, C.P.; Gebara, M.A.; Stibich, M.A.; Chemaly, R.F. A systematic and critical review of bacteriophage therapy against multidrug-resistant ESKAPE organisms in humans. Clin. Infect. Dis. 2019, 69, 167–178. [Google Scholar] [CrossRef]
  148. Dedrick, R.M.; Guerrero-Bustamante, C.A.; Garlena, R.A.; Russell, D.A.; Ford, K.; Harris, K.; Gilmour, K.C.; Soothill, J.; Jacobs-Sera, D.; Schooley, R.T. Engineered bacteriophages for treatment of a patient with a disseminated drug-resistant Mycobacterium abscessus. Nat. Med. 2019, 25, 730–733. [Google Scholar] [CrossRef]
  149. Thi, M.T.T.; Wibowo, D.; Rehm, B.H.A. Pseudomonas aeruginosa Biofilms. Int. J. Mol. Sci. 2020, 21, 8671. [Google Scholar] [CrossRef]
  150. Dibdin, G.H.; Assinder, S.J.; Nichols, W.W.; Lambert, P.A. Mathematical model of β-lactam penetration into a biofilm of Pseudomonas aeruginosa while undergoing simultaneous inactivation by released β-lactamases. J. Antimicrob. Chemother. 1996, 38, 757–769. [Google Scholar] [CrossRef]
  151. Wang, H.; Ciofu, O.; Yang, L.; Wu, H.; Song, Z.; Oliver, A.; Høiby, N. High β-lactamase levels change the pharmacodynamics of β-lactam antibiotics in Pseudomonas aeruginosa biofilms. Antimicrob. Agents Chemother. 2013, 57, 196–204. [Google Scholar]
  152. Knecht, L.E.; Veljkovic, M.; Fieseler, L. Diversity and function of phage encoded depolymerases. Front. Microbiol. 2020, 10, 2949. [Google Scholar] [CrossRef]
  153. Danis-Wlodarczyk, K.; Olszak, T.; Arabski, M.; Wasik, S.; Majkowska-Skrobek, G.; Augustyniak, D.; Gula, G.; Briers, Y.; Jang, H.B.; Vandenheuvel, D. Characterization of the newly isolated lytic bacteriophages KTN6 and KT28 and their efficacy against Pseudomonas aeruginosa biofilm. PLoS ONE 2015, 10, e0127603. [Google Scholar]
  154. Hanlon, G.W.; Denyer, S.P.; Olliff, C.J.; Ibrahim, L.J. Reduction in exopolysaccharide viscosity as an aid to bacteriophage penetration through Pseudomonas aeruginosa biofilms. Appl. Environ. Microbiol. 2001, 67, 2746–2753. [Google Scholar] [CrossRef]
  155. Henriksen, K.; Rørbo, N.; Rybtke, M.L.; Martinet, M.G.; Tolker-Nielsen, T.; Høiby, N.; Middelboe, M.; Ciofu, O.P. aeruginosa flow-cell biofilms are enhanced by repeated phage treatments but can be eradicated by phage–ciprofloxacin combination: —Monitoring the phage–P. aeruginosa biofilms interactions. Pathog. Dis. 2019, 77, ftz011. [Google Scholar] [CrossRef]
  156. Franklin, M.J.; Nivens, D.E.; Weadge, J.T.; Howell, P.L. Biosynthesis of the Pseudomonas aeruginosa extracellular polysaccharides, alginate, Pel, and Psl. Front. Microbiol. 2011, 2, 167. [Google Scholar] [CrossRef]
  157. Glonti, T.; Chanishvili, N.; Taylor, P. Bacteriophage-derived enzyme that depolymerizes the alginic acid capsule associated with cystic fibrosis isolates of Pseudomonas aeruginosa. J. Appl. Microbiol. 2010, 108, 695–702. [Google Scholar] [CrossRef]
  158. Ertesvåg, H. Alginate-modifying enzymes: Biological roles and biotechnological uses. Front. Microbiol. 2015, 6, 523. [Google Scholar] [CrossRef]
  159. Yan, J.; Mao, J.; Xie, J. Bacteriophage polysaccharide depolymerases and biomedical applications. Biodrugs 2014, 28, 265–274. [Google Scholar] [CrossRef]
  160. Wong, T.Y.; Preston, L.A.; Schiller, N.L. Alginate lyase: Review of major sources and enzyme characteristics, structure-function analysis, biological roles, and applications. Annu. Rev. Microbiol. 2000, 54, 289–340. [Google Scholar] [CrossRef]
  161. Osawa, T.; Matsubara, Y.; Muramatsu, T.; Kimura, M.; Kakuta, Y. Crystal structure of the alginate (poly α-L-guluronate) lyase from Corynebacterium sp. at 1.2 Å resolution. J. Mol. Biol. 2005, 345, 1111–1118. [Google Scholar] [CrossRef] [PubMed]
  162. Adnan, M.; Shah, M.R.A.; Jamal, M.; Jalil, F.; Andleeb, S.; Nawaz, M.A.; Pervez, S.; Hussain, T.; Shah, I.; Imran, M. Isolation and characterization of bacteriophage to control multidrug-resistant Pseudomonas aeruginosa planktonic cells and biofilm. Biologicals 2020, 63, 89–96. [Google Scholar] [CrossRef] [PubMed]
  163. Pei, R.; Lamas-Samanamud, G.R. Inhibition of biofilm formation by T7 bacteriophages producing quorum-quenching enzymes. Appl. Environ. Microbiol. 2014, 80, 5340–5348. [Google Scholar] [CrossRef] [PubMed]
  164. Latz, S.; Krüttgen, A.; Häfner, H.; Buhl, E.M.; Ritter, K.; Horz, H.-P. Differential effect of newly isolated phages belonging to PB1-like, phiKZ-like and LUZ24-like viruses against multi-drug resistant Pseudomonas aeruginosa under varying growth conditions. Viruses 2017, 9, 315. [Google Scholar] [CrossRef]
  165. Alves, D.R.; Perez-Esteban, P.; Kot, W.; Bean, J.; Arnot, T.; Hansen, L.; Enright, M.C.; Jenkins, A.T.A. A novel bacteriophage cocktail reduces and disperses P seudomonas aeruginosa biofilms under static and flow conditions. Microb. Biotechnol. 2016, 9, 61–74. [Google Scholar] [CrossRef]
  166. Shafique, M.; Alvi, I.A.; Abbas, Z.; ur Rehman, S. Assessment of biofilm removal capacity of a broad host range bacteriophage JHP against Pseudomonas aeruginosa. APMIS 2017, 125, 579–584. [Google Scholar] [CrossRef]
  167. Lehman, S.M.; Donlan, R.M. Bacteriophage-mediated control of a two-species biofilm formed by microorganisms causing catheter-associated urinary tract infections in an in vitro urinary catheter model. Antimicrob. Agents Chemother. 2015, 59, 1127–1137. [Google Scholar] [CrossRef]
  168. Pires, D.; Sillankorva, S.; Faustino, A.; Azeredo, J. Use of newly isolated phages for control of Pseudomonas aeruginosa PAO1 and ATCC 10145 biofilms. Res. Microbiol. 2011, 162, 798–806. [Google Scholar] [CrossRef]
  169. Knezevic, P.; Obreht, D.; Curcin, S.; Petrusic, M.; Aleksic, V.; Kostanjsek, R.; Petrovic, O. Phages of Pseudomonas aeruginosa: Response to environmental factors and in vitro ability to inhibit bacterial growth and biofilm formation. J. Appl. Microbiol. 2011, 111, 245–254. [Google Scholar] [CrossRef]
  170. Fu, W.; Forster, T.; Mayer, O.; Curtin, J.J.; Lehman, S.M.; Donlan, R.M. Bacteriophage cocktail for the prevention of biofilm formation by Pseudomonas aeruginosa on catheters in an in vitro model system. Antimicrob. Agents Chemother. 2010, 54, 397–404. [Google Scholar] [CrossRef]
  171. Jamal, M.; Andleeb, S.; Jalil, F.; Imran, M.; Nawaz, M.A.; Hussain, T.; Ali, M.; Das, C.R. Isolation and characterization of a bacteriophage and its utilization against multi-drug resistant Pseudomonas aeruginosa-2995. Life Sci. 2017, 190, 21–28. [Google Scholar] [CrossRef] [PubMed]
  172. Kwiatek, M.; Parasion, S.; Rutyna, P.; Mizak, L.; Gryko, R.; Niemcewicz, M.; Olender, A.; Łobocka, M. Isolation of bacteriophages and their application to control Pseudomonas aeruginosa in planktonic and biofilm models. Res. Microbiol. 2017, 168, 194–207. [Google Scholar] [CrossRef]
  173. Fong, S.A.; Drilling, A.; Morales, S.; Cornet, M.E.; Woodworth, B.A.; Fokkens, W.J.; Psaltis, A.J.; Vreugde, S.; Wormald, P.-J. Activity of bacteriophages in removing biofilms of Pseudomonas aeruginosa isolates from chronic rhinosinusitis patients. Front. Cell. Infect. Microbiol. 2017, 7, 418. [Google Scholar] [CrossRef]
  174. Jeon, J.; Yong, D. Two novel bacteriophages improve survival in Galleria mellonella infection and mouse acute pneumonia models infected with extensively drug-resistant Pseudomonas aeruginosa. Appl. Environ. Microbiol. 2019, 85, e02900–e02918. [Google Scholar] [CrossRef]
  175. Holguín, A.V.; Rangel, G.; Clavijo, V.; Prada, C.; Mantilla, M.; Gomez, M.C.; Kutter, E.; Taylor, C.; Fineran, P.C.; Barrios, A.F.G. Phage ΦPan70, a putative temperate phage, controls Pseudomonas aeruginosa in planktonic, biofilm and burn mouse model assays. Viruses 2015, 7, 4602–4623. [Google Scholar] [CrossRef]
  176. Alemayehu, D.; Casey, P.; McAuliffe, O.; Guinane, C.; Martin, J.; Shanahan, F.; Coffey, A.; Ross, R.; Hill, C. Bacteriophages MR299-2 and NH-4 can eliminate Pseudomonas aeruginosa in the murine lung and on cystic fibrosis lung airway cells. mBio 2012, 3, e00029-12. [Google Scholar] [CrossRef]
  177. Forti, F.; Roach, D.R.; Cafora, M.; Pasini, M.E.; Horner, D.S.; Fiscarelli, E.V.; Rossitto, M.; Cariani, L.; Briani, F.; Debarbieux, L. Design of a broad-range bacteriophage cocktail that reduces Pseudomonas aeruginosa biofilms and treats acute infections in two animal models. Antimicrob. Agents Chemother. 2018, 62, e02573-17. [Google Scholar] [CrossRef]
  178. Basu, S.; Agarwal, M.; Nath, G. An In vivo Wound Model Utilizing Bacteriophage Therapy of Pseudomonas aeruginosa Biofilms. Ostomy Wound Manag. 2015, 61, 16–23. [Google Scholar]
  179. Ageitos, J.M.; Sánchez-Pérez, A.; Calo-Mata, P.; Villa, T.G. Antimicrobial peptides (AMPs): Ancient compounds that represent novel weapons in the fight against bacteria. Biochem. Pharmacol. 2017, 133, 117–138. [Google Scholar] [CrossRef]
  180. Savini, F.; Loffredo, M.; Troiano, C.; Bobone, S.; Malanovic, N.; Eichmann, T.; Caprio, L.; Canale, V.; Park, Y.; Mangoni, M. Binding of an antimicrobial peptide to bacterial cells: Interaction with different species, strains and cellular components. Biochim. Biophys. Acta (BBA)-Biomembr. 2020, 1862, 183291. [Google Scholar] [CrossRef]
  181. Bahar, A.A.; Ren, D. Antimicrobial peptides. Pharmaceuticals 2013, 6, 1543–1575. [Google Scholar] [CrossRef] [PubMed]
  182. Li, T.; Liu, Q.; Wang, D.; Li, J. Characterization and antimicrobial mechanism of CF-14, a new antimicrobial peptide from the epidermal mucus of catfish. Fish Shellfish Immunol. 2019, 92, 881–888. [Google Scholar] [CrossRef] [PubMed]
  183. Luo, Y.; Song, Y. Mechanism of Antimicrobial Peptides: Antimicrobial, Anti-Inflammatory and Antibiofilm Activities. Int. J. Mol. Sci. 2021, 22, 11401. [Google Scholar] [CrossRef]
  184. Bobone, S.; Stella, L. Selectivity of Antimicrobial Peptides: A Complex Interplay of Multiple Equilibria. Adv. Exp. Med. Biol. 2019, 1117, 175–214. [Google Scholar] [CrossRef]
  185. Harder, J.; Schröder, J.-M. Antimicrobial Peptides: Role in Human Health and Disease; Springer: Berlin/Heidelberg, Germany, 2015. [Google Scholar]
  186. Pushpanathan, M.; Gunasekaran, P.; Rajendhran, J. Antimicrobial peptides: Versatile biological properties. Int. J. Pept. 2013, 2013, 675391. [Google Scholar] [CrossRef]
  187. Yeaman, M.R.; Yount, N.Y. Mechanisms of antimicrobial peptide action and resistance. Pharmacol. Rev. 2003, 55, 27–55. [Google Scholar] [CrossRef]
  188. Matsuzaki, K.; Murase, O.; Fujii, N.; Miyajima, K. Translocation of a channel-forming antimicrobial peptide, magainin 2, across lipid bilayers by forming a pore. Biochemistry 1995, 34, 6521–6526. [Google Scholar] [CrossRef]
  189. Matsuzaki, K.; Murase, O.; Fujii, N.; Miyajima, K. An antimicrobial peptide, magainin 2, induced rapid flip-flop of phospholipids coupled with pore formation and peptide translocation. Biochemistry 1996, 35, 11361–11368. [Google Scholar] [CrossRef]
  190. Lee, T.-H.; N Hall, K.; Aguilar, M.-I. Antimicrobial peptide structure and mechanism of action: A focus on the role of membrane structure. Curr. Top. Med. Chem. 2016, 16, 25–39. [Google Scholar] [CrossRef]
  191. Kumar, P.; Kizhakkedathu, J.N.; Straus, S.K. Antimicrobial peptides: Diversity, mechanism of action and strategies to improve the activity and biocompatibility in vivo. Biomolecules 2018, 8, 4. [Google Scholar] [CrossRef]
  192. Shai, Y. Mode of action of membrane active antimicrobial peptides. Pept. Sci. Orig. Res. Biomol. 2002, 66, 236–248. [Google Scholar] [CrossRef] [PubMed]
  193. Brogden, K.A. Antimicrobial peptides: Pore formers or metabolic inhibitors in bacteria? Nat. Rev. Microbiol. 2005, 3, 238–250. [Google Scholar] [CrossRef]
  194. Lohner, K. Membrane-active antimicrobial peptides as template structures for novel antibiotic agents. Curr. Top. Med. Chem. 2017, 17, 508–519. [Google Scholar] [CrossRef]
  195. Clement, N.R.; Gould, J.M. Pyranine (8-hydroxy-1, 3, 6-pyrenetrisulfonate) as a probe of internal aqueous hydrogen ion concentration in phospholipid vesicles. Biochemistry 1981, 20, 1534–1538. [Google Scholar] [CrossRef]
  196. Jahangiri, A.; Neshani, A.; Mirhosseini, S.A.; Ghazvini, K.; Zare, H.; Sedighian, H. Synergistic effect of two antimicrobial peptides, Nisin and P10 with conventional antibiotics against extensively drug-resistant Acinetobacter baumannii and colistin-resistant Pseudomonas aeruginosa isolates. Microb. Pathog. 2021, 150, 104700. [Google Scholar] [CrossRef]
  197. Field, D.; Seisling, N.; Cotter, P.D.; Ross, R.P.; Hill, C. Synergistic nisin-polymyxin combinations for the control of Pseudomonas biofilm formation. Front. Microbiol. 2016, 7, 1713. [Google Scholar] [CrossRef]
  198. Thomas, V.M.; Brown, R.M.; Ashcraft, D.S.; Pankey, G.A. Synergistic effect between nisin and polymyxin B against pandrug-resistant and extensively drug-resistant Acinetobacter baumannii. Int. J. Antimicrob. Agents 2019, 53, 663–668. [Google Scholar] [CrossRef]
  199. Naghmouchi, K.; Baah, J.; Hober, D.; Jouy, E.; Rubrecht, C.; Sané, F.; Drider, D. Synergistic effect between colistin and bacteriocins in controlling Gram-negative pathogens and their potential to reduce antibiotic toxicity in mammalian epithelial cells. Antimicrob. Agents Chemother. 2013, 57, 2719–2725. [Google Scholar] [CrossRef]
  200. de Leeuw, E.; Li, C.; Zeng, P.; Li, C.; Diepeveen-de Buin, M.; Lu, W.-Y.; Breukink, E.; Lu, W. Functional interaction of human neutrophil peptide-1 with the cell wall precursor lipid II. FEBS Lett. 2010, 584, 1543–1548. [Google Scholar] [CrossRef]
  201. Münch, D.; Sahl, H.-G. Structural variations of the cell wall precursor lipid II in Gram-positive bacteria—Impact on binding and efficacy of antimicrobial peptides. Biochim. Biophys. Acta (BBA)-Biomembr. 2015, 1848, 3062–3071. [Google Scholar] [CrossRef]
  202. Xhindoli, D.; Pacor, S.; Benincasa, M.; Scocchi, M.; Gennaro, R.; Tossi, A. The human cathelicidin LL-37—A pore-forming antibacterial peptide and host-cell modulator. Biochim. Biophys. Acta (BBA)-Biomembr. 2016, 1858, 546–566. [Google Scholar] [CrossRef] [PubMed]
  203. Ridyard, K.E.; Overhage, J. The potential of human peptide LL-37 as an antimicrobial and anti-biofilm agent. Antibiotics 2021, 10, 650. [Google Scholar] [CrossRef] [PubMed]
  204. Haisma, E.M.; de Breij, A.; Chan, H.; van Dissel, J.T.; Drijfhout, J.W.; Hiemstra, P.S.; El Ghalbzouri, A.; Nibbering, P.H. LL-37-derived peptides eradicate multidrug-resistant Staphylococcus aureus from thermally wounded human skin equivalents. Antimicrob. Agents Chemother. 2014, 58, 4411–4419. [Google Scholar] [CrossRef] [PubMed]
  205. Martinez, M.; Gonçalves, S.; Felício, M.R.; Maturana, P.; Santos, N.C.; Semorile, L.; Hollmann, A.; Maffía, P.C. Synergistic and antibiofilm activity of the antimicrobial peptide P5 against carbapenem-resistant Pseudomonas aeruginosa. Biochim. Biophys. Acta (BBA)-Biomembr. 2019, 1861, 1329–1337. [Google Scholar] [CrossRef]
  206. Hollmann, A.; Martínez, M.; Noguera, M.E.; Augusto, M.T.; Disalvo, A.; Santos, N.C.; Semorile, L.; Maffía, P.C. Role of amphipathicity and hydrophobicity in the balance between hemolysis and peptide–membrane interactions of three related antimicrobial peptides. Colloids Surf. B. Biointerfaces 2016, 141, 528–536. [Google Scholar] [CrossRef]
  207. Faccone, D.; Veliz, O.; Corso, A.; Noguera, M.; Martínez, M.; Payes, C.; Semorile, L.; Maffía, P.C. Antimicrobial activity of de novo designed cationic peptides against multi-resistant clinical isolates. Eur. J. Med. Chem. 2014, 71, 31–35. [Google Scholar] [CrossRef]
  208. Rudilla, H.; Fusté, E.; Cajal, Y.; Rabanal, F.; Vinuesa, T.; Viñas, M. Synergistic antipseudomonal effects of synthetic peptide AMP38 and carbapenems. Molecules 2016, 21, 1223. [Google Scholar] [CrossRef]
  209. Wu, M.; Maier, E.; Benz, R.; Hancock, R.E. Mechanism of interaction of different classes of cationic antimicrobial peptides with planar bilayers and with the cytoplasmic membrane of Escherichia coli. Biochemistry 1999, 38, 7235–7242. [Google Scholar] [CrossRef]
  210. Shang, D.; Han, X.; Du, W.; Kou, Z.; Jiang, F. Trp-containing antibacterial peptides impair quorum sensing and biofilm development in multidrug-resistant Pseudomonas aeruginosa and exhibit synergistic effects with antibiotics. Front. Microbiol. 2021, 12, 611009. [Google Scholar] [CrossRef]
  211. Gusmão, K.A.; Santos, D.M.d.; Santos, V.M.; Cortés, M.E.; Reis, P.V.; Santos, V.L.; Piló-Veloso, D.; Verly, R.M.; Lima, M.E.d.; Resende, J.M. Ocellatin peptides from the skin secretion of the South American frog Leptodactylus labyrinthicus (Leptodactylidae): Characterization, antimicrobial activities and membrane interactions. J. Venom. Anim. Tox. incl. Trop. Dis. 2017, 23, 4. [Google Scholar] [CrossRef]
  212. Bessa, L.J.; Eaton, P.; Dematei, A.; Placido, A.; Vale, N.; Gomes, P.; Delerue-Matos, C.; SA Leite, J.R.; Gameiro, P. Synergistic and antibiofilm properties of ocellatin peptides against multidrug-resistant Pseudomonas aeruginosa. Future Microbiol. 2018, 13, 151–163. [Google Scholar] [CrossRef] [PubMed]
  213. Pandidan, S.; Mechler, A. Nano-viscosimetry analysis of the membrane disrupting action of the bee venom peptide melittin. Sci. Rep. 2019, 9, 10841. [Google Scholar] [CrossRef] [PubMed]
  214. Akbari, R.; Hakemi-Vala, M.; Pashaie, F.; Bevalian, P.; Hashemi, A.; Pooshang Bagheri, K. Highly synergistic effects of melittin with conventional antibiotics against multidrug-resistant isolates of Acinetobacter baumannii and Pseudomonas aeruginosa. Microb. Drug Resist. 2019, 25, 193–202. [Google Scholar] [CrossRef] [PubMed]
  215. Stoner, I. Biopharma has abandoned antibiotic development. Here’s why we did too. Endpoints News, 13 December 2019. [Google Scholar]
  216. Geneva Pharmaceuticals v. GlaxoSmithKline; Court of Appeals, Federal Circuit: Washington, DC, USA, 2003; p. 1373.
  217. Mullard, A. Achaogen bankruptcy highlights antibacterial development woes. Nat. Rev. Drug Discov. 2019, 18, 411–412. [Google Scholar] [CrossRef]
  218. de la Fuente-Nunez, C. Antibiotic discovery with machine learning. Nat. Biotechnol. 2022, 40, 833–834. [Google Scholar] [CrossRef]
  219. Stokes, J.M.; Yang, K.; Swanson, K.; Jin, W.; Cubillos-Ruiz, A.; Donghia, N.M.; MacNair, C.R.; French, S.; Carfrae, L.A.; Bloom-Ackermann, Z. A deep learning approach to antibiotic discovery. Cell 2020, 180, 688–702.e613. [Google Scholar] [CrossRef]
  220. Jukič, M.; Bren, U. Machine learning in antibacterial drug design. Front. Pharmacol. 2022, 13, 864412. [Google Scholar] [CrossRef]
  221. Liu, G.; Stokes, J.M. A brief guide to machine learning for antibiotic discovery. Curr. Opin. Microbiol. 2022, 69, 102190. [Google Scholar] [CrossRef]
  222. David, L.; Brata, A.M.; Mogosan, C.; Pop, C.; Czako, Z.; Muresan, L.; Ismaiel, A.; Dumitrascu, D.I.; Leucuta, D.C.; Stanculete, M.F.; et al. Artificial Intelligence and Antibiotic Discovery. Antibiotics 2021, 10, 1376. [Google Scholar] [CrossRef]
  223. Liu, G.; Catacutan, D.B.; Rathod, K.; Swanson, K.; Jin, W.; Mohammed, J.C.; Chiappino-Pepe, A.; Syed, S.A.; Fragis, M.; Rachwalski, K. Deep learning-guided discovery of an antibiotic targeting Acinetobacter baumannii. Nat. Chem. Biol. 2023, 19, 1342–1350. [Google Scholar] [CrossRef]
  224. Parvaiz, N.; Ahmad, F.; Yu, W.; MacKerell, A.D., Jr.; Azam, S.S. Discovery of beta-lactamase CMY-10 inhibitors for combination therapy against multi-drug resistant Enterobacteriaceae. PLoS ONE 2021, 16, e0244967. [Google Scholar] [CrossRef] [PubMed]
  225. McNair, K.; Aziz, R.K.; Pusch, G.D.; Overbeek, R.; Dutilh, B.E.; Edwards, R. Phage genome annotation using the RAST pipeline. Bacteriophages Methods Protoc. 2018, 3, 231–238. [Google Scholar]
  226. McNair, K.; Zhou, C.; Dinsdale, E.A.; Souza, B.; Edwards, R.A. PHANOTATE: A novel approach to gene identification in phage genomes. Bioinformatics 2019, 35, 4537–4542. [Google Scholar] [CrossRef]
  227. Thung, T.Y.; White, M.E.; Dai, W.; Wilksch, J.J.; Bamert, R.S.; Rocker, A.; Stubenrauch, C.J.; Williams, D.; Huang, C.; Schittelhelm, R. Component parts of bacteriophage virions accurately defined by a machine-learning approach built on evolutionary features. Msystems 2021, 6, e00242-21. [Google Scholar] [CrossRef]
  228. Wang, J.; Dai, W.; Li, J.; Xie, R.; Dunstan, R.A.; Stubenrauch, C.; Zhang, Y.; Lithgow, T. PaCRISPR: A server for predicting and visualizing anti-CRISPR proteins. Nucleic Acids Res. 2020, 48, W348–W357. [Google Scholar] [CrossRef]
  229. Bhadra, P.; Yan, J.; Li, J.; Fong, S.; Siu, S.W. AmPEP: Sequence-based prediction of antimicrobial peptides using distribution patterns of amino acid properties and random forest. Sci. Rep. 2018, 8, 1697. [Google Scholar] [CrossRef]
  230. Karp, N.A. Reproducible preclinical research-Is embracing variability the answer? PLoS Biol. 2018, 16, e2005413. [Google Scholar] [CrossRef]
  231. Dirnagl, U.; Duda, G.N.; Grainger, D.W.; Reinke, P.; Roubenoff, R. Reproducibility, relevance and reliability as barriers to efficient and credible biomedical technology translation. Adv. Drug Deliv. Rev. 2022, 182, 114118. [Google Scholar] [CrossRef]
  232. Samuel, S.; König-Ries, B. Understanding experiments and research practices for reproducibility: An exploratory study. PeerJ 2021, 9, e11140. [Google Scholar] [CrossRef]
  233. Hillary, F.G.; Medaglia, J.D. What the replication crisis means for intervention science. Int. J. Psychophysiol. 2020, 154, 3–5. [Google Scholar] [CrossRef]
  234. Papp-Wallace, K.M.; Bethel, C.R.; Caillon, J.; Barnes, M.D.; Potel, G.; Bajaksouzian, S.; Rutter, J.D.; Reghal, A.; Shapiro, S.; Taracila, M.A. Beyond piperacillin-tazobactam: Cefepime and AAI101 as a potent β-lactam− β-lactamase inhibitor combination. Antimicrob. Agents Chemother. 2019, 63, e00105-19. [Google Scholar] [CrossRef] [PubMed]
  235. Dalhoff, A. Are antibacterial effects of non-antibiotic drugs random or purposeful because of a common evolutionary origin of bacterial and mammalian targets? Infection 2021, 49, 569–589. [Google Scholar] [CrossRef]
  236. Casteels, R.; Login, I. Reserpine has a direct action as a calcium antagonist on mammalian smooth muscle cells. J. Physiol. 1983, 340, 403–414. [Google Scholar] [CrossRef]
  237. Campoli-Richards, D.M.; Brogden, R.N. Sulbactam/ampicillin: A review of its antibacterial activity, pharmacokinetic properties, and therapeutic use. Drugs 1987, 33, 577–609. [Google Scholar] [CrossRef]
  238. Cady Kyle, C.; Bondy-Denomy, J.; Heussler Gary, E.; Davidson Alan, R.; O’Toole George, A. The CRISPR/Cas Adaptive Immune System of Pseudomonas aeruginosa Mediates Resistance to Naturally Occurring and Engineered Phages. J. Bacteriol. 2012, 194, 5728–5738. [Google Scholar] [CrossRef]
  239. Gooderham, W.J.; Gellatly, S.L.; Sanschagrin, F.; McPhee, J.B.; Bains, M.; Cosseau, C.; Levesque, R.C.; Hancock, R.E. The sensor kinase PhoQ mediates virulence in Pseudomonas aeruginosa. Microbiology 2009, 155, 699–711. [Google Scholar] [CrossRef]
  240. McPhee, J.B.; Lewenza, S.; Hancock, R.E. Cationic antimicrobial peptides activate a two-component regulatory system, PmrA-PmrB, that regulates resistance to polymyxin B and cationic antimicrobial peptides in Pseudomonas aeruginosa. Mol. Microbiol. 2003, 50, 205–217. [Google Scholar] [CrossRef]
  241. Llobet, E.; Tomas, J.M.; Bengoechea, J.A. Capsule polysaccharide is a bacterial decoy for antimicrobial peptides. Microbiology 2008, 154, 3877–3886. [Google Scholar] [CrossRef]
Figure 1. Overview of β-lactam structure and activity. (A) Representative chemical structures of β-lactams belonging to the penicillin, cephalosporin, and carbapenem subclasses. The core β-lactam ring is indicated in red. (B) β-lactam antibiotics can cross the Gram-negative outer membrane through porin proteins embedded in the membrane, thereby accessing the periplasmic space that contains PBPs. The bactericidal effect of β-lactams against P. aeruginosa involves the inhibition of PBP3, which normally catalyzes the formation of peptide cross-links in peptidoglycan. Inhibition of PBP3 and the subsequent decreased formation of peptidoglycan cross-links significantly weakens the cell wall and induces autolysis. Created in BioRender (https://www.biorender.com/).
Figure 1. Overview of β-lactam structure and activity. (A) Representative chemical structures of β-lactams belonging to the penicillin, cephalosporin, and carbapenem subclasses. The core β-lactam ring is indicated in red. (B) β-lactam antibiotics can cross the Gram-negative outer membrane through porin proteins embedded in the membrane, thereby accessing the periplasmic space that contains PBPs. The bactericidal effect of β-lactams against P. aeruginosa involves the inhibition of PBP3, which normally catalyzes the formation of peptide cross-links in peptidoglycan. Inhibition of PBP3 and the subsequent decreased formation of peptidoglycan cross-links significantly weakens the cell wall and induces autolysis. Created in BioRender (https://www.biorender.com/).
Antibiotics 14 00526 g001
Figure 2. Overview of β-lactam resistance mechanisms in P. aeruginosa. (A) β-lactamase enzymes in the periplasmic space degrade β-lactams, preventing them from inhibiting PBPs such as PBP3. (B) Certain mutations to the ftsl gene modify the structure of PBP3 such that β-lactam antibiotics are no longer able to bind and interfere with peptidoglycan synthesis. (C) Mutations to the genes encoding for porins such as OprD and OpdP can decrease the entry of β-lactams into the periplasmic space by reducing the porin content in the outer membrane or by modifying the structures of porins. (D) Increased expression of efflux pumps such as MexAB-OprM allows P. aeruginosa to expel β-lactam antibiotics from the periplasm, preventing them from interacting with PBPs. Created in BioRender.
Figure 2. Overview of β-lactam resistance mechanisms in P. aeruginosa. (A) β-lactamase enzymes in the periplasmic space degrade β-lactams, preventing them from inhibiting PBPs such as PBP3. (B) Certain mutations to the ftsl gene modify the structure of PBP3 such that β-lactam antibiotics are no longer able to bind and interfere with peptidoglycan synthesis. (C) Mutations to the genes encoding for porins such as OprD and OpdP can decrease the entry of β-lactams into the periplasmic space by reducing the porin content in the outer membrane or by modifying the structures of porins. (D) Increased expression of efflux pumps such as MexAB-OprM allows P. aeruginosa to expel β-lactam antibiotics from the periplasm, preventing them from interacting with PBPs. Created in BioRender.
Antibiotics 14 00526 g002
Figure 3. Representative β-lactamase inhibitors. Chemical structures of selected β-lactamase inhibitors that are in current use or under clinical development, organized according to their scaffolds as β-lactams, diazabicyclooctanes, and boronates.
Figure 3. Representative β-lactamase inhibitors. Chemical structures of selected β-lactamase inhibitors that are in current use or under clinical development, organized according to their scaffolds as β-lactams, diazabicyclooctanes, and boronates.
Antibiotics 14 00526 g003
Figure 4. Synergistic use of bacteriophages with β-lactams against P. aeruginosa. (A) Biofilms produced by P. aeruginosa act as a physical barrier to β-lactams and serve as reservoirs for the accumulation of β-lactamases. These enzymes can inactivate β-lactam antibiotics before they enter the bacterial cells embedded in the biofilm. (B) Degradation of P. aeruginosa biofilms by bacteriophages can revive the efficacy of β-lactams by dispersing reservoirs of extracellular β-lactamases. Created in BioRender.
Figure 4. Synergistic use of bacteriophages with β-lactams against P. aeruginosa. (A) Biofilms produced by P. aeruginosa act as a physical barrier to β-lactams and serve as reservoirs for the accumulation of β-lactamases. These enzymes can inactivate β-lactam antibiotics before they enter the bacterial cells embedded in the biofilm. (B) Degradation of P. aeruginosa biofilms by bacteriophages can revive the efficacy of β-lactams by dispersing reservoirs of extracellular β-lactamases. Created in BioRender.
Antibiotics 14 00526 g004
Figure 5. Mechanisms of AMP-mediated membrane disruption that allow for enhanced β-lactam entry. (A) In the barrel-stave model, the insertion of multiple AMPs into a membrane forms a protein-based transmembrane channel. (B) Although the toroidal pore model also involves the insertion of AMPs into the membrane, here, the peptides induce bending of lipid molecules such that a pore lined with lipid head groups is created. (C) The carpet model differs from the other two models described, as the AMPs primarily interact with the lipid head groups rather than inserting directly into the membrane; this interaction increases the permeability of the membrane and can cause the membrane to disintegrate and form micelles. Created in BioRender.
Figure 5. Mechanisms of AMP-mediated membrane disruption that allow for enhanced β-lactam entry. (A) In the barrel-stave model, the insertion of multiple AMPs into a membrane forms a protein-based transmembrane channel. (B) Although the toroidal pore model also involves the insertion of AMPs into the membrane, here, the peptides induce bending of lipid molecules such that a pore lined with lipid head groups is created. (C) The carpet model differs from the other two models described, as the AMPs primarily interact with the lipid head groups rather than inserting directly into the membrane; this interaction increases the permeability of the membrane and can cause the membrane to disintegrate and form micelles. Created in BioRender.
Antibiotics 14 00526 g005
Table 1. Overview of β-lactamase classification and activity.
Table 1. Overview of β-lactamase classification and activity.
Ambler ClassRepresentative Members Found in P. aeruginosaSubstrate Scope and Relevance
A (SBLs)KPC-2, GES-2Certain members of this class can degrade all β-lactam subclasses; includes ESBLs and carbapenemases [44]
B (MBLs)NDM-1, IMP-1, VIM-2Most noted for carbapenemase activity; some members degrade penicillins and cephalosporins; do not protect against monobactams [45]
C (SBLs)AmpC, PDC-1Generally effective against penicillins and cephalosporins; some have ESBL activity; limited ability to degrade carbapenems [46]
D (SBLs)OXA-10, OXA-48Degrade certain penicillins and cephalosporins; some members have ESBL or carbapenemase activity [47]
Table 2. Efflux pump-mediated β-lactam resistance in P. aeruginosa.
Table 2. Efflux pump-mediated β-lactam resistance in P. aeruginosa.
Efflux Systemβ-Lactam Substrates
MexAB-OprMAll tested β-lactams, * excluding imipenem [104,114]
MexXY-OprMAll tested β-lactams, * excluding carbenicillin, sulbenicillin, cefsulodin, ceftazidime, moxalactam, flomoxef, aztreonam, and imipenem [104]
MexCD-OprJAll tested β-lactams, * excluding carbenicillin, sulbenicillin, cefsulodin, ceftazidime, moxalactam, aztreonam, and imipenem [104]
MexEF-OprNImipenem [115,116]
* Penicillin G, cloxacillin, nafcillin, amoxicillin, piperacillin, carbenicillin, sulbenicillin, cefamandole, cefuroxime, cefoperazone, cefotaxime, ceftizoxime, ceftriaxone, cefsulodin, ceftazidime, cefpirome, cefepime, cefozopran, cefoselis, cefoxitin, moxalactam, flomoxef, imipenem, and meropenem.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhao, D.W.; Lohans, C.T. Combatting Pseudomonas aeruginosa with β-Lactam Antibiotics: A Revived Weapon? Antibiotics 2025, 14, 526. https://doi.org/10.3390/antibiotics14050526

AMA Style

Zhao DW, Lohans CT. Combatting Pseudomonas aeruginosa with β-Lactam Antibiotics: A Revived Weapon? Antibiotics. 2025; 14(5):526. https://doi.org/10.3390/antibiotics14050526

Chicago/Turabian Style

Zhao, Dylan W., and Christopher T. Lohans. 2025. "Combatting Pseudomonas aeruginosa with β-Lactam Antibiotics: A Revived Weapon?" Antibiotics 14, no. 5: 526. https://doi.org/10.3390/antibiotics14050526

APA Style

Zhao, D. W., & Lohans, C. T. (2025). Combatting Pseudomonas aeruginosa with β-Lactam Antibiotics: A Revived Weapon? Antibiotics, 14(5), 526. https://doi.org/10.3390/antibiotics14050526

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop