Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1949 2023-11-14 11:13:07 |
2 format correct Meta information modification 1949 2023-11-15 08:38:40 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Giovagnorio, F.; De Vito, A.; Madeddu, G.; Parisi, S.G.; Geremia, N. Treatment for Pseudomonas aeruginosa Infections. Encyclopedia. Available online: https://encyclopedia.pub/entry/51530 (accessed on 07 July 2024).
Giovagnorio F, De Vito A, Madeddu G, Parisi SG, Geremia N. Treatment for Pseudomonas aeruginosa Infections. Encyclopedia. Available at: https://encyclopedia.pub/entry/51530. Accessed July 07, 2024.
Giovagnorio, Federico, Andrea De Vito, Giordano Madeddu, Saverio Giuseppe Parisi, Nicholas Geremia. "Treatment for Pseudomonas aeruginosa Infections" Encyclopedia, https://encyclopedia.pub/entry/51530 (accessed July 07, 2024).
Giovagnorio, F., De Vito, A., Madeddu, G., Parisi, S.G., & Geremia, N. (2023, November 14). Treatment for Pseudomonas aeruginosa Infections. In Encyclopedia. https://encyclopedia.pub/entry/51530
Giovagnorio, Federico, et al. "Treatment for Pseudomonas aeruginosa Infections." Encyclopedia. Web. 14 November, 2023.
Treatment for Pseudomonas aeruginosa Infections
Edit

Pseudomonas aeruginosa is a ubiquitous Gram-negative bacterium renowned for its resilience and adaptability across diverse environments, including clinical settings, where it emerges as a formidable pathogen. Notorious for causing nosocomial infections, P. aeruginosa presents a significant challenge due to its intrinsic and acquired resistance mechanisms.

Pseudomonas aeruginosa resistance combination therapy TOL-TZB CAZ-AVI FDC

1. Introduction

Pseudomonas aeruginosa, a ubiquitous Gram-negative bacterium from the Pseudomonadaceae family, has garnered considerable attention for its resilience and adaptability in many environments. Notably, it bears a relatively larger genome, ranging from 5.5 to 7 Mbp, giving it a remarkable metabolic versatility and enabling it to thrive and adapt to diverse environmental shifts, thereby contributing to its survival in varied habitats, including water, soil, and associations with animals [1][2]. Furthermore, the bacteria’s widespread presence in water sources, such as tap water and hand soap dispensers, has been associated with hospital outbreaks, indicating a pressing need for stringent hygiene measures and environmental control [3][4]. P. aeruginosa genotypes analysis has identified household environments, such as sinks and nebulizers, as potential sources of infection, necessitating vigilant monitoring and sanitation [3][4].
While P. aeruginosa’s environmental tenacity is noteworthy, its role as an opportunistic pathogen has raised significant concerns, particularly in hospital settings [5]. It is notorious for causing nosocomial infections and ventilator-associated pneumonia, primarily affecting immunocompromised individuals, severe burn victims, and patients with underlying health conditions such as cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) [6][7][8][9][10]. In particular, P. aeruginosa’s ability to form biofilms, coupled with its intrinsic, acquired, and adaptive resistance mechanisms, has rendered it a formidable adversary in the clinical setting [11][12][13][14]. It exhibits resistance to many antibiotics, including aminoglycosides, fluoroquinolones, and β-lactams, through mechanisms such as low outer membrane permeability, the expression of efflux pumps, and the production of antibiotic-inactivating enzymes [11][15][16][17]. Acquired resistance through horizontal gene transfer and mutations, along with adaptive resistance exemplified by biofilm formation and the emergence of persister cells, further complicate its treatment landscape [18].
Globally, the incidence of multidrug-resistant (MDR.) P. aeruginosa has exhibited an alarming upswing, posing considerable challenges to public health and clinical treatment [19][20]. The European Centre for Disease Prevention and Control (ECDC) has reported a varying prevalence of MDR P. aeruginosa, with some regions witnessing a heightened occurrence, underscoring the geographical disparity and the necessity for region-specific interventions [21]. The global landscape is similarly marked by a heterogeneous distribution of MDR strains, emphasizing the criticality of continuous surveillance and adaptive strategies to curb the spread of resistance [22]. The escalating challenge posed by P. aeruginosa has been accentuated by the World Health Organization (WHO), which has categorized carbapenem-resistant strains as being in critical need of new antibiotics [23]. The emergence of these strains has been associated with elevated morbidity and mortality, amplifying the urgency for innovative and effective therapeutic approaches [24]. These strategies include novel antibiotics and non-antibiotic therapeutic options, including phage therapy, nanoparticle application, and quorum sensing inhibition [25][26][27][28][29][30][31]. These strategies aim to augment or substitute conventional antibiotic treatments in addressing the rising tide of antibiotic resistance.

2. New Treatment Options

Managing P. aeruginosa infections has always been a clinical challenge due to its intrinsic and acquired resistance mechanisms. Traditionally, the primary treatment approach was monotherapy with antipseudomonal agents such as FQ, PIP-TZB, or carbapenems [32][33][34]. However, the emergence of MDR strains has prompted a shift towards combination therapy [35]. Due to their synergistic effects, common combinations include β-lactam agents with aminoglycosides or FQ [36]. Another reason for using combination therapy is to counter the bacteria’s ability to form biofilms, which are inherently resistant to many antibiotics. It is also crucial to consider the phenomenon of inducible resistance, where the bacteria can upregulate specific resistance genes in response to certain antibiotics [37]. The different types of resistance mechanisms will be further discussed.
The rise of MDR strains has necessitated the development of new antibiotics. Notable additions to the therapeutic arsenal include TOL-TZB, CAZ-AVI, IMI-REL, and FDC.
TOL-TZB (Zerbaxa®) is a novel antibiotic combination with a next-generation cephalosporin and TZB, a suicidal BLI [38]. This combination has an enhanced affinity for PBPs and potent activity against P. aeruginosa, including ESBL and AmpC-producing strains [39]. Clinical studies have emphasized its efficacy against Pseudomonas infections, especially in complicated urinary tract (cUTI) and intra-abdominal infections (cIAI) [40]. Caston et al. treated 20 infections caused by P. aeruginosa MDR with TOL-TZB, which included 12 cases of septic shock, 6 cases of pneumonia, 1 case of otomastoiditis, and 1 Central Line-associate Bloodstream Infection (CLABSI), reporting a clinical success rate of 75% [41]. Gallagher et al. treated 205 infections caused by P. aeruginosa MDR, primarily pneumonia (59%), and reported a clinical success rate of 74%. Interestingly, they highlighted the importance of the prompt administration of TOL-TZB (within four days), identifying it as a predictor of clinical success [42]. This observation aligns with previous studies that associated a delay in initiating effective antibacterial therapy with increased mortality in serious bacterial infections, including HAP [43][44].
An observational study involving 200 patients compared the outcomes of a TOL-TZB-based regimen versus polymyxin or aminoglycoside-based therapy [45]. In this study, a favorable clinical outcome was observed in 81% of patients in the TOL-TZB arm versus 61% of patients in the polymyxin- or aminoglycoside-based regimen arm. This difference was statistically significant.
Recently, an ASPECT-NP study involving patients with nosocomial pneumonia caused by Gram-negative pathogens was published. Pneumonia is the most frequent healthcare-associated infection acquired in the Intensive Care Unit (ICU), with high mortality rates [46][47][48]. Hospital-acquired pneumonia (HAP) can be distinguished as ventilator-associated pneumonia (VAP) or hospital-acquired ventilator-requiring pneumonia (vHAP) [49]. Within HAP, vHAP has the highest mortality [46][50].
The ASPECT-NP trial demonstrated the noninferiority of TOL-TZB to MEM for treating vHAP and VAP in both the primary endpoint of 28-day all-cause mortality and the secondary endpoint of clinical cure at the test-of-cure visit, respectively [51]. In the subgroup of vHAP, the ASPECT-NP trial also demonstrated lower mortality in the arm of patients treated with TOL-TZB compared with the arm of patients treated with MEM [51]. This result was further evaluated with multivariable analysis by Kollef et al., confirming the protective effect of TOL-TZB in this special subgroup population [52], although additional studies are needed for confirmation of these findings.
The distinct pharmacological profile of TOL-TZB, marked by enhanced activity against P. aeruginosa, makes it a valuable option where resistant Pseudomonas strains are prevalent or suspected [53][54].
CAZ-AVI (Zavicefta®) combines CAZ, a third-generation cephalosporin with potent antipseudomonal activity, and AVI, a non-BL/BLI [55]. AVI effectively neutralizes a broad spectrum of β-lactamases, including carbapenemases like KPC, OXA-48 [56], and GES [49]. Horcajada et al. reported in vitro susceptibility to CAZ-AVI ranging from 66% to 86% for MDR P. aeruginosa strains collected from all over the world [57].
In the literature, some studies have highlighted the efficacy of CAZ-AVI in treating various infections due to MDR P. aeruginosa, from cUTIs to HAP [58][59][60]. However, clinical trials involving CAZ-AVI treatments are scarce. Stone et al. reported pooled data from five Randomized Controlled Trials (RCTs), showing a favorable clinical outcome in patients treated with CAZ-AVI versus patients treated with more traditional treatment, albeit within the limitations of the study [61].
Nevertheless, the literature concerning the real-world use of CAZ-AVI is spreading. Several studies have been published, underlying positive outcomes in favor of CAZ-AVI in treating MDR P. aeruginosa infections [62].
The Infectious Diseases Society of America (IDSA) indicates CAZ-AVI as one of the drugs of choice in the treatment of MDR and difficult-to-treat (DTR)-P. aeruginosa strains, both in urinary tract infections and outside urinary tract infections, when tested susceptible [63].
FDC (Fetcroja®) is a siderophore cephalosporin with a unique penetration mechanism. It utilizes the iron-transport mechanism to penetrate bacterial cells, making it active against a broad spectrum of Gram-negative microorganisms, including CRP [64][65]. It is also effective against MBL. Hacket et al. demonstrated in vitro its activity against most MDR P. aeruginosa (99.2%), including those resistant to CAZ-AVI and TOL-TZB [66]. Lasarte-Monterrubio et al. evaluated the in vitro activity of FDC (and other novel antibiotic combinations) against strains of P. aeruginosa specifically resistant to CAZ-AVI and TOL-TZB [67], showing that FDC was the most active agent.
The SIDERO surveillance program, conducted between 2014 and 2019, showed a susceptibility rate to FDC of CRP strains of 99.8%, according to CLSI breakpoints [68].
The randomized APESK-cUTI demonstrated the non-inferiority of FDC versus IMP-REL in the treatment of complicated urinary tract infections in hospitalized patients for the primary endpoint of composite microbiological eradication and clinical cure at the test-of-cure visit [69].
In the APESK-NP study evaluating the efficacy and safety of FDC for the treatment of nosocomial Gram-negative pneumonia, FDC demonstrated non-inferiority to the MEM treatment. Of all the cases, P. aeruginosa represented the second most common pathogen in the FDC arm. Clinical success was achieved in 67% of pneumonia cases caused by P. aeruginosa, without statistical difference from the pneumonia cases caused by P. aeruginosa in the MEM arm [70].
However, further studies on its efficacy in real life are needed to better assess its effectiveness.
IMP-REL (Recarbrio®) combines IMP, a carbapenem approved in 1985, and REL, a BLI that enhances IMP’s activity by protecting it from enzymatic degradation. This combination is active against class A and class C β-lactamases. On the other hand, it is ineffective against OXA-48 and MBLs [71].
The SMART study, a surveillance study conducted in several countries across the world, assessed the susceptibility of IMP-REL [72]. Furthermore, Lob et al. showed that the addition of REL restored the activity of IMP against P. aeruginosa strains resistant to IMP alone.
Mushtaq et al. collected P. aeruginosa clinical strains producing ESBL and Carbapenemases, showing an 80.5% susceptibility for IMP-REL [73]. In this study, the main mechanism of resistance was the production of beta-lactamases not susceptible to inhibition by REL, such as MBLs, OXAs, and GES.
In the RESTORE-IMI clinical trial, IMP-REL demonstrated greater efficacy than COL/MEM against P. aeruginosa infection (81% vs. 63%). However, this difference was not statistically significant due to the small sample size [74].
The RESTORE-IMI 2 clinical trial, a randomized, double-blind controlled trial, was conducted to assess the efficacy of IMP-REL in adult patients with HAP/VAP [75]. IMP-REL was non-inferior to the comparator (PIP-TZB) for both endpoints (day 28 all-cause mortality and favorable clinical response at early follow-up). In this study, P. aeruginosa was the second most abundant pathogen isolated.
IDSA guidelines recommend IMP-REL as one of the drugs of choice in the treatment of cUTIs and infections outside the urinary tract due to DTR P. aeruginosa strains when tested as susceptible [63].
In addition, promising antibiotics are coming into the pipeline, such as cefepime/enmetazobactam, cefepime/zidebactam, cefepime/taniborbactam, and plazomicine.
These FEP-based antibiotics had broad-spectrum activity against Gram-positive and Gram-negative bacteria. The distinguishing factor is the specific BLI paired with FEP to shield it from enzymatic degradation. While all four inhibitors fall under the category of next-generation BLI, Enmetazobactam is classified as a penicillanic acid sulfone, Taniborbactam as a boronic acid derivative, and Zidebactam as a diazabicyclooctane [76]. Their effectiveness is still being evaluated, but the clinical trials have yielded promising results [77][78][79]. However, no real-life studies compare these three molecules [80]. J Vázquez-Ucha et al. assessed the in vitro efficacy of these molecules, finding that cefepime/zidebactam was the most potent combination against carbapenemase-producing Enterobacterales, followed by cefepime/taniborbactam and cefepime/enmetazobactam. Furthermore, Moya et al. highlighted that zidebactam alone has significant activity against P. aeruginosa. This effect is due to the inhibition of PBP2, leading to the creation of spheroplasts, the disruption of the outer membrane, and, as a result, protection against common membrane-bound resistance mechanisms exhibited by P. aeruginosa [76].
Plazomicin is a next-generation aminoglycoside antibiotic synthetically derived from sisomicin [81]. It works by inhibiting bacterial protein synthesis, leading to dose-dependent bactericidal activity. One of its significant advantages is its activity against bacterial strains harboring clinically relevant AMEs [82]. However, it is worth noting that plazomicin is not active against bacterial isolates expressing ribosomal methyltransferases, which can lead to aminoglycoside resistance [82]. Focusing on P. aeruginosa, plazomicin has shown promising data [83]. The antibiotic demonstrates synergistic activity when combined with other agents like FEP, Doripenem, IMP, or PIP/TZB [84].

References

  1. Moradali, M.F.; Ghods, S.; Rehm, B.H.A. Pseudomonas aeruginosa Lifestyle: A Paradigm for Adaptation, Survival, and Persistence. Front. Cell. Infect. Microbiol. 2017, 7, 39.
  2. Lewenza, S.; Abboud, J.; Poon, K.; Kobryn, M.; Humplik, I.; Bell, J.R.; Mardan, L.; Reckseidler-Zenteno, S. Pseudomonas aeruginosa Displays a Dormancy Phenotype during Long-Term Survival in Water. PLoS ONE 2018, 13, e0198384.
  3. Hutchins, C.F.; Moore, G.; Thompson, K.A.; Webb, J.; Walker, J.T. Contamination of Hospital Tap Water: The Survival and Persistence of Pseudomonas aeruginosa on Conventional and “antimicrobial” Outlet Fittings. J. Hosp. Infect. 2017, 97, 156–161.
  4. Lanini, S.; D’Arezzo, S.; Puro, V.; Martini, L.; Imperi, F.; Piselli, P.; Montanaro, M.; Paoletti, S.; Visca, P.; Ippolito, G. Molecular Epidemiology of a Pseudomonas aeruginosa Hospital Outbreak Driven by a Contaminated Disinfectant-Soap Dispenser. PLoS ONE 2011, 6, e17064.
  5. Kelsey, M. Pseudomonas in Augmented Care: Should We Worry? J. Antimicrob. Chemother. 2013, 68, 2697–2700.
  6. Lieberman, D.; Lieberman, D. Pseudomonal Infections in Patients with COPD: Epidemiology and Management. Am. J. Respir. Med. 2003, 2, 459–468.
  7. Davies, J.C. Pseudomonas aeruginosa in Cystic Fibrosis: Pathogenesis and Persistence. Paediatr. Respir. Rev. 2002, 3, 128–134.
  8. Holder, I.A.P. aeruginosa Burn Infections: Pathogenesis and Treatment. In Pseudomonas aeruginosa as an Opportunistic Pathogen; Springer: Berlin/Heidelberg, Germany, 1993; pp. 275–295.
  9. Ramírez-Estrada, S.; Borgatta, B.; Rello, J. Pseudomonas aeruginosa Ventilator-Associated Pneumonia Management. Infect. Drug Resist. 2016, 9, 7.
  10. Yang, Y.W.; Jiang, Y.Z.; Hsu, C.M.; Chen, L.W. Pseudomonas aeruginosa Ventilator-Associated Pneumonia Induces Lung Injury through TNF-α/c-Jun NH2-Terminal Kinase Pathways. PLoS ONE 2017, 12, e0169267.
  11. Pang, Z.; Raudonis, R.; Glick, B.R.; Lin, T.J.; Cheng, Z. Antibiotic Resistance in Pseudomonas aeruginosa: Mechanisms and Alternative Therapeutic Strategies. Biotechnol. Adv. 2019, 37, 177–192.
  12. Okamoto, K.; Gotoh, N.; Nishino, T. Pseudomonas aeruginosa Reveals High Intrinsic Resistance to Penem Antibiotics: Penem Resistance Mechanisms and Their Interplay. Antimicrob. Agents Chemother. 2001, 45, 1964.
  13. Poole, K. Pseudomonas aeruginosa: Resistance to the Max. Front. Microbiol. 2011, 2, 65.
  14. Thi, M.T.T.; Wibowo, D.; Rehm, B.H.A. Pseudomonas aeruginosa Biofilms. Int. J. Mol. Sci. 2020, 21, 8671.
  15. Langendonk, R.F.; Neill, D.R.; Fothergill, J.L. The Building Blocks of Antimicrobial Resistance in Pseudomonas Aeruginosa: Implications for Current Resistance-Breaking Therapies. Front. Cell Infect. Microbiol. 2021, 11.
  16. Angus, B.L.; Carey, A.M.; Caron, D.A.; Kropinski, A.M.; Hancock, R.E. Outer Membrane Permeability in Pseudomonas aeruginosa: Comparison of a Wild-Type with an Antibiotic-Supersusceptible Mutant. Antimicrob. Agents Chemother. 1982, 21, 299–309.
  17. Janda, J.M.; Bottone, E.J. Pseudomonas aeruginosa Enzyme Profiling: Predictor of Potential Invasiveness and Use as an Epidemiological Tool. J. Clin. Microbiol. 1981, 14, 55.
  18. Michaelis, C.; Grohmann, E. Horizontal Gene Transfer of Antibiotic Resistance Genes in Biofilms. Antibiotics 2023, 12, 328.
  19. Peng, Y.; Shi, J.; Bu, T.; Li, Y.; Ye, X.; Chen, X.; Yao, Z. Alarming and Increasing Prevalence of Multidrug-Resistant Pseudomonas aeruginosa among Healthcare-Associated Infections in China: A Meta-Analysis of Cross-Sectional Studies. J. Glob. Antimicrob. Resist. 2015, 3, 155–160.
  20. Yang, A.F.; Huang, V.; Samaroo-Campbell, J.; Augenbraun, M. Multi-Drug Resistant Pseudomonas Aeruginosa: A 2019–2020 Single Center Retrospective Case Control Study. Infect. Prev. Pract. 2023, 5, 100296.
  21. ECDC. Antimicrobial Resistance Surveillance in Europe; ECDC: Solna, Sweden, 2022.
  22. Reyes, J.; Komarow, L.; Chen, L.; Ge, L.; Hanson, B.M.; Cober, E.; Herc, E.; Alenazi, T.; Kaye, K.S.; Garcia-Diaz, J.; et al. Global Epidemiology and Clinical Outcomes of Carbapenem-Resistant Pseudomonas aeruginosa and Associated Carbapenemases (POP): A Prospective Cohort Study. Lancet Microbe 2023, 4, e159–e170.
  23. Tacconelli, E.; Carrara, E.; Savoldi, A.; Harbarth, S.; Mendelson, M.; Monnet, D.L.; Pulcini, C.; Kahlmeter, G.; Kluytmans, J.; Carmeli, Y.; et al. Discovery, Research, and Development of New Antibiotics: The WHO Priority List of Antibiotic-Resistant Bacteria and Tuberculosis. Lancet Infect. Dis. 2018, 18, 318–327.
  24. Sathe, N.; Beech, P.; Croft, L.; Suphioglu, C.; Kapat, A.; Athan, E. Pseudomonas aeruginosa: Infections and Novel Approaches to Treatment “Knowing the Enemy” the Threat of Pseudomonas aeruginosa and Exploring Novel Approaches to Treatment. Infect. Med. 2023, 2, 178–194.
  25. Duplantier, M.; Lohou, E.; Sonnet, P. Quorum Sensing Inhibitors to Quench P. aeruginosa Pathogenicity. Pharmaceuticals 2021, 14, 1262.
  26. Salomoni, R.; Léo, P.; Montemor, A.F.; Rinaldi, B.G.; Rodrigues, M.F.A. Antibacterial Effect of Silver Nanoparticles in Pseudomonas aeruginosa. Nanotechnol. Sci. Appl. 2017, 10, 115–121.
  27. Köhler, T.; Luscher, A.; Falconnet, L.; Resch, G.; McBride, R.; Mai, Q.A.; Simonin, J.L.; Chanson, M.; Maco, B.; Galiotto, R.; et al. Personalized Aerosolised Bacteriophage Treatment of a Chronic Lung Infection Due to Multidrug-Resistant Pseudomonas aeruginosa. Nat. Commun. 2023, 14, 3629.
  28. Vaitekenas, A.; Tai, A.S.; Ramsay, J.P.; Stick, S.M.; Kicic, A. Pseudomonas aeruginosa Resistance to Bacteriophages and Its Prevention by Strategic Therapeutic Cocktail Formulation. Antibiotics 2021, 10, 145.
  29. Losito, A.R.; Raffaelli, F.; Del Giacomo, P.; Tumbarello, M. New Drugs for the Treatment of Pseudomonas aeruginosa Infections with Limited Treatment Options: A Narrative Review. Antibiotics 2022, 11, 579.
  30. Kunz Coyne, A.J.; El Ghali, A.; Holger, D.; Rebold, N.; Rybak, M.J. Therapeutic Strategies for Emerging Multidrug-Resistant Pseudomonas aeruginosa. Infect. Dis. Ther. 2022, 11, 661.
  31. Marino, A.; Stracquadanio, S.; Campanella, E.; Munafò, A.; Gussio, M.; Ceccarelli, M.; Bernardini, R.; Nunnari, G.; Cacopardo, B. Intravenous Fosfomycin: A Potential Good Partner for Cefiderocol. Clinical Experience and Considerations. Antibiotics 2022, 12, 49.
  32. Gribble, M.J.; Chow, A.W.; Naiman, S.C.; Smith, J.A.; Bowie, W.R.; Sacks, S.L.; Grossman, L.; Buskard, N.; Growe, G.H.; Plenderleith, L.H. Prospective Randomized Trial of Piperacillin Monotherapy versus Carboxypenicillin-Aminoglycoside Combination Regimens in the Empirical Treatment of Serious Bacterial Infections. Antimicrob. Agents Chemother. 1983, 24, 388.
  33. Landman, D.; Bratu, S.; Kochar, S.; Panwar, M.; Trehan, M.; Doymaz, M.; Quale, J. Evolution of Antimicrobial Resistance among Pseudomonas aeruginosa, Acinetobacter Baumannii and Klebsiella Pneumoniae in Brooklyn, NY. J. Antimicrob. Chemother. 2007, 60, 78–82.
  34. Burgess, D.S.; Nathisuwan, S. Cefepime, Piperacillin/Tazobactam, Gentamicin, Ciprofloxacin, and Levofloxacin Alone and in Combination against Pseudomonas aeruginosa. Diagn. Microbiol. Infect. Dis. 2002, 44, 35–41.
  35. Paul, M.; Leibovici, L.; Maki, D.G.; Safdar, N. Combination Antibiotic Therapy for Pseudomonas aeruginosa Bacteraemia. Lancet Infect. Dis. 2005, 5, 192–193.
  36. Davis, B.D. Bactericidal Synergism between Beta-Lactams and Aminoglycosides: Mechanism and Possible Therapeutic Implications. Rev. Infect. Dis. 1982, 4, 237–245.
  37. Breidenstein, E.B.M.; de la Fuente-Núñez, C.; Hancock, R.E.W. Pseudomonas aeruginosa: All Roads Lead to Resistance. Trends Microbiol. 2011, 19, 419–426.
  38. Cluck, D.; Lewis, P.; Stayer, B.; Spivey, J.; Moorman, J. Ceftolozane-Tazobactam: A New-Generation Cephalosporin. Am. J. Health Syst. Pharm. 2015, 72, 2135–2146.
  39. Bassetti, M.; Vena, A.; Giacobbe, D.R.; Falcone, M.; Tiseo, G.; Giannella, M.; Pascale, R.; Meschiari, M.; Digaetano, M.; Oliva, A.; et al. Ceftolozane/Tazobactam for Treatment of Severe ESBL-Producing Enterobacterales Infections: A Multicenter Nationwide Clinical Experience (CEFTABUSE II Study). Open Forum. Infect. Dis. 2020, 7, ofaa139.
  40. Zhanel, G.G.; Chung, P.; Adam, H.; Zelenitsky, S.; Denisuik, A.; Schweizer, F.; Lagacé-Wiens, P.R.S.; Rubinstein, E.; Gin, A.S.; Walkty, A.; et al. Ceftolozane/Tazobactam: A Novel Cephalosporin/β-Lactamase Inhibitor Combination with Activity against Multidrug-Resistant Gram-Negative Bacilli. Drugs 2014, 74, 31–51.
  41. Castón, J.J.; De la Torre, Á.; Ruiz-Camps, I.; Sorlí, M.L.; Torres, V.; Torre-Cisneros, J. Salvage Therapy with Ceftolozane-Tazobactam for Multidrug-Resistant Pseudomonas aeruginosa Infections. Antimicrob. Agents Chemother. 2017, 61, 02136-16.
  42. Gallagher, J.C.; Satlin, M.J.; Elabor, A.; Saraiya, N.; McCreary, E.K.; Molnar, E.; El-Beyrouty, C.; Jones, B.M.; Dixit, D.; Heil, E.L.; et al. Ceftolozane-Tazobactam for the Treatment of Multidrug-Resistant Pseudomonas aeruginosa Infections: A Multicenter Study. Open Forum. Infect. Dis. 2018, 5, ofy280.
  43. Albin, O.R.; Henig, O.; Patel, T.S.; Valley, T.S.; Pogue, J.M.; Petty, L.A.; Mills, J.P.; Brancaccio, A.; Martin, E.T.; Kaye, K.S. Clinical Implications of Microbiologic Treatment Failure in the Setting of Clinical Cure of Bacterial Pneumonia. Clin. Infect. Dis. 2020, 71, 3033–3041.
  44. Bonine, N.G.; Berger, A.; Altincatal, A.; Wang, R.; Bhagnani, T.; Gillard, P.; Lodise, T. Impact of Delayed Appropriate Antibiotic Therapy on Patient Outcomes by Antibiotic Resistance Status from Serious Gram-Negative Bacterial Infections. Am. J. Med. Sci. 2019, 357, 103–110.
  45. Pogue, J.M.; Kaye, K.S.; Veve, M.P.; Patel, T.S.; Gerlach, A.T.; Davis, S.L.; Puzniak, L.A.; File, T.M.; Olson, S.; Dhar, S.; et al. Ceftolozane/Tazobactam vs Polymyxin or Aminoglycoside-Based Regimens for the Treatment of Drug-Resistant Pseudomonas aeruginosa. Clin. Infect. Dis. 2020, 71, 304–310.
  46. Talbot, G.H.; Das, A.; Cush, S.; Dane, A.; Wible, M.; Echols, R.; Torres, A.; Cammarata, S.; Rex, J.H.; Powers, J.H.; et al. Evidence-Based Study Design for Hospital-Acquired Bacterial Pneumonia and Ventilator-Associated Bacterial Pneumonia. J. Infect. Dis. 2019, 219, 1536–1544.
  47. Torres, A.; Niederman, M.S.; Chastre, J.; Ewig, S.; Fernandez-Vandellos, P.; Hanberger, H.; Kollef, M.; Li Bassi, G.; Luna, C.M.; Martin-Loeches, I.; et al. International ERS/ESICM/ESCMID/ALAT Guidelines for the Management of Hospital-Acquired Pneumonia and Ventilator-Associated Pneumonia. Eur. Respir. J. 2017, 50, 1700582.
  48. Kalil, A.C.; Metersky, M.L.; Klompas, M.; Muscedere, J.; Sweeney, D.A.; Palmer, L.B.; Napolitano, L.M.; O’Grady, N.P.; Bartlett, J.G.; Carratalà, J.; et al. Management of Adults with Hospital-Acquired and Ventilator-Associated Pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin. Infect. Dis. 2016, 63, e61–e111.
  49. Cantón, R.; Jehl, F.; Rossolini, G.M.; Soriano, A.; Tascini, C.; Viaggi, B. Mdr Infections in Critically Ill Patients. From the Microlab Bench to the Patient’s Bedside; EDIMES: Pavia, Italy, 2022.
  50. Esperatti, M.; Ferrer, M.; Theessen, A.; Liapikou, A.; Valencia, M.; Saucedo, L.M.; Zavala, E.; Welte, T.; Torres, A. Nosocomial Pneumonia in the Intensive Care Unit Acquired by Mechanically Ventilated versus Nonventilated Patients. Am. J. Respir. Crit. Care Med. 2010, 182, 1533–1539.
  51. Kollef, M.H.; Nováček, M.; Kivistik, Ü.; Réa-Neto, Á.; Shime, N.; Martin-Loeches, I.; Timsit, J.-F.; Wunderink, R.G.; Bruno, C.J.; Huntington, J.A.; et al. Ceftolozane-Tazobactam versus Meropenem for Treatment of Nosocomial Pneumonia (ASPECT-NP): A Randomised, Controlled, Double-Blind, Phase 3, Non-Inferiority Trial. Lancet Infect. Dis. 2019, 19, 1299–1311.
  52. Kollef, M.H.; Timsit, J.-F.; Martin-Loeches, I.; Wunderink, R.G.; Huntington, J.A.; Jensen, E.H.; Yu, B.; Bruno, C.J. Outcomes in Participants with Failure of Initial Antibacterial Therapy for Hospital-Acquired/Ventilator-Associated Bacterial Pneumonia Prior to Enrollment in the Randomized, Controlled Phase 3 ASPECT-NP Trial of Ceftolozane/Tazobactam versus Meropenem. Crit. Care 2022, 26, 373.
  53. Bergas, A.; Albasanz-Puig, A.; Fernández-Cruz, A.; Machado, M.; Novo, A.; van Duin, D.; Garcia-Vidal, C.; Hakki, M.; Ruiz-Camps, I.; del Pozo, J.L.; et al. Real-Life Use of Ceftolozane/Tazobactam for the Treatment of Bloodstream Infection Due to Pseudomonas aeruginosa in Neutropenic Hematologic Patients: A Matched Control Study (ZENITH Study). Microbiol. Spectr. 2022, 10, e02292-21.
  54. Montesinos, I.L.; Luisa Sorlí, M.M.; Horcajada, J.P. Ceftolozane-Tazobactam: When, How and Why Using It? Rev. Esp. Quim. 2021, 34 (Suppl. 1), 35–37.
  55. Zhong, H.; Zhao, X.Y.; Zhang, Z.L.; Gu, Z.C.; Zhang, C.; Gao, Y.; Cui, M. Evaluation of the Efficacy and Safety of Ceftazidime/Avibactam in the Treatment of Gram-Negative Bacterial Infections: A Systematic Review and Meta-Analysis. Int. J. Antimicrob. Agents 2018, 52, 443–450.
  56. Fröhlich, C.; Sørum, V.; Thomassen, A.M.; Johnsen, P.J.; Leiros, H.-K.S.; Samuelsen, Ø. OXA-48-Mediated Ceftazidime-Avibactam Resistance Is Associated with Evolutionary Trade-Offs. mSphere 2019, 4, e00024-19.
  57. Horcajada, J.P.; Montero, M.; Oliver, A.; Sorlí, L.; Luque, S.; Gómez-Zorrilla, S.; Benito, N.; Grau, S. Epidemiology and Treatment of Multidrug-Resistant and Extensively Drug-Resistant Pseudomonas aeruginosa Infections. Clin. Microbiol. Rev. 2019, 32, 00031-19.
  58. Carmeli, Y.; Armstrong, J.; Laud, P.J.; Newell, P.; Stone, G.; Wardman, A.; Gasink, L.B. Ceftazidime-Avibactam or Best Available Therapy in Patients with Ceftazidime-Resistant Enterobacteriaceae and Pseudomonas aeruginosa Complicated Urinary Tract Infections or Complicated Intra-Abdominal Infections (REPRISE): A Randomised, Pathogen-Directed, Phase 3 Study. Lancet Infect. Dis. 2016, 16, 661–673.
  59. Mazuski, J.E.; Wagenlehner, F.; Torres, A.; Carmeli, Y.; Chow, J.W.; Wajsbrot, D.; Stone, G.G.; Irani, P.; Bharucha, D.; Cheng, K.; et al. Clinical and Microbiological Outcomes of Ceftazidime-Avibactam Treatment in Adults with Gram-Negative Bacteremia: A Subset Analysis from the Phase 3 Clinical Trial Program. Infect. Dis. Ther. 2021, 10, 2399.
  60. Marino, A.; Campanella, E.; Stracquadanio, S.; Calvo, M.; Migliorisi, G.; Nicolosi, A.; Cosentino, F.; Marletta, S.; Spampinato, S.; Prestifilippo, P.; et al. Ceftazidime/Avibactam and Meropenem/Vaborbactam for the Management of Enterobacterales Infections: A Narrative Review, Clinical Considerations, and Expert Opinion. Antibiotics 2023, 12, 1521.
  61. Stone, G.G.; Newell, P.; Gasink, L.B.; Broadhurst, H.; Wardman, A.; Yates, K.; Chen, Z.; Song, J.; Chow, J.W. Clinical Activity of Ceftazidime/Avibactam against MDR Enterobacteriaceae and Pseudomonas aeruginosa: Pooled Data from the Ceftazidime/Avibactam Phase III Clinical Trial Programme. J. Antimicrob. Chemother. 2018, 73, 2519–2523.
  62. Daikos, G.L.; da Cunha, C.A.; Rossolini, G.M.; Stone, G.G.; Baillon-Plot, N.; Tawadrous, M.; Irani, P. Review of Ceftazidime-Avibactam for the Treatment of Infections Caused by Pseudomonas aeruginosa. Antibiotics 2021, 10, 1126.
  63. Tamma, P.D.; Aitken, S.L.; Bonomo, R.A.; Mathers, A.J.; van Duin, D.; Clancy, C.J. Infectious Diseases Society of America 2022 Guidance on the Treatment of Extended-Spectrum β-Lactamase Producing Enterobacterales (ESBL-E), Carbapenem-Resistant Enterobacterales (CRE), and Pseudomonas aeruginosa with Difficult-to-Treat Resistance (DTR-P. aeruginosa). Clin. Infect. Dis. 2022, 75, 187–212.
  64. Wu, J.Y.; Srinivas, P.; Pogue, J.M. Cefiderocol: A Novel Agent for the Management of Multidrug-Resistant Gram-Negative Organisms. Infect. Dis. Ther. 2020, 9, 17.
  65. Ybanez Garcia, L.; Manzano Lorenzo, R.; Martinez Segasti, F.; Martinez Sesmero, J.M. Real-Life Experience with Cefiderocol for the Treatment of Difficult-to-Treat Gram-Negative Infections. Infect. Dis. Trop. Med. 2023, 9, e1157.
  66. Hackel, M.A.; Tsuji, M.; Yamano, Y.; Echols, R.; Karlowsky, J.A.; Sahm, D.F. In Vitro Activity of the Siderophore Cephalosporin, Cefiderocol, against Carbapenem-Nonsusceptible and Multidrug-Resistant Isolates of Gram-Negative Bacilli Collected Worldwide in 2014 to 2016. Antimicrob. Agents Chemother. 2018, 62, 01968-17.
  67. Lasarte-Monterrubio, C.; Fraile-Ribot, P.A.; Vázquez-Ucha, J.C.; Cabot, G.; Guijarro-Sánchez, P.; Alonso-García, I.; Rumbo-Feal, S.; Galán-Sánchez, F.; Beceiro, A.; Arca-Suárez, J.; et al. Activity of Cefiderocol, Imipenem/Relebactam, Cefepime/Taniborbactam and Cefepime/Zidebactam against Ceftolozane/Tazobactam- and Ceftazidime/Avibactam-Resistant Pseudomonas aeruginosa. J. Antimicrob. Chemother. 2022, 77, 2809–2815.
  68. Karlowsky, J.A.; Hackel, M.A.; Takemura, M.; Yamano, Y.; Echols, R.; Sahm, D.F. In Vitro Susceptibility of Gram-Negative Pathogens to Cefiderocol in Five Consecutive Annual Multinational SIDERO-WT Surveillance Studies, 2014 to 2019. Antimicrob. Agents Chemother. 2022, 66, e01990-21.
  69. Portsmouth, S.; van Veenhuyzen, D.; Echols, R.; Machida, M.; Ferreira, J.C.A.; Ariyasu, M.; Tenke, P.; Nagata, T. Den Cefiderocol versus Imipenem-Cilastatin for the Treatment of Complicated Urinary Tract Infections Caused by Gram-Negative Uropathogens: A Phase 2, Randomised, Double-Blind, Non-Inferiority Trial. Lancet Infect. Dis. 2018, 18, 1319–1328.
  70. Wunderink, R.G.; Matsunaga, Y.; Ariyasu, M.; Clevenbergh, P.; Echols, R.; Kaye, K.S.; Kollef, M.; Menon, A.; Pogue, J.M.; Shorr, A.F.; et al. Cefiderocol versus High-Dose, Extended-Infusion Meropenem for the Treatment of Gram-Negative Nosocomial Pneumonia (APEKS-NP): A Randomised, Double-Blind, Phase 3, Non-Inferiority Trial. Lancet Infect. Dis. 2021, 21, 213–225.
  71. Reig, S.; Le Gouellec, A.; Bleves, S. What Is New in the Anti–Pseudomonas aeruginosa Clinical Development Pipeline Since the 2017 WHO Alert? Front. Cell. Infect. Microbiol. 2022, 12, 862.
  72. Lob, S.H.; Hackel, M.A.; Kazmierczak, K.M.; Young, K.; Motyl, M.R.; Karlowsky, J.A.; Sahm, D.F. In Vitro Activity of Imipenem-Relebactam against Gram-Negative ESKAPE Pathogens Isolated by Clinical Laboratories in the United States in 2015 (Results from the SMART Global Surveillance Program). Antimicrob. Agents Chemother. 2017, 61, 02209-16.
  73. 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.
  74. Motsch, J.; De Oliveira, C.U.M.; Stus, V.; Kö Ksal, I.; Lyulko, O.; Boucher, H.W.; Kaye, K.S.; File, T.M.; Brown, M.L.; Khan, I.; et al. RESTORE-IMI 1: A Multicenter, Randomized, Double-Blind Trial Comparing Efficacy and Safety of Imipenem/Relebactam vs Colistin Plus Imipenem in Patients with Imipenem-Nonsusceptible Bacterial Infections. Clin. Infect. Dis. 2020, 70, 1799–1808.
  75. Titov, I.; Wunderink, R.G.; Roquilly, A.; Rodríguez Gonzalez, D.; David-Wang, A.; Boucher, H.W.; Kaye, K.S.; Losada, M.C.; Du, J.; Tipping, R.; et al. A Randomized, Double-Blind, Multicenter Trial Comparing Efficacy and Safety of Imipenem/Cilastatin/Relebactam Versus Piperacillin/Tazobactam in Adults wth Hospital-Acquired or Ventilator-Associated Bacterial Pneumonia (RESTORE-IMI 2 Study). Clin. Infect. Dis. 2021, 73, e4539–e4548.
  76. Isler, B.; Harris, P.; Stewart, A.G.; Paterson, D.L. An Update on Cefepime and Its Future Role in Combination with Novel β-Lactamase Inhibitors for MDR Enterobacterales and Pseudomonas aeruginosa. J. Antimicrob. Chemother. 2021, 76, 550–560.
  77. ClinicalTrials.gov Safety and Efficacy Study of Cefepime-AAI101 in the Treatment of Complicated Urinary Tract Infections. Available online: https://clinicaltrials.gov/study/NCT03687255?intr=cefepime%2Fenmetazobactam&rank=4#publications (accessed on 11 October 2023).
  78. ClinicalTrials.gov Safety and Efficacy Study of Cefepime/VNRX-5133 in Patients with Complicated Urinary Tract Infections. Available online: https://clinicaltrials.gov/study/NCT03840148?intr=cefepime-taniborbactam&rank=2#publications (accessed on 11 October 2023).
  79. ClinicalTrials.gov Study of Cefepime-Zidebactam (FEP-ZID) in Complicated Urinary Tract Infection (CUTI) or Acute Pyelonephritis (AP). Available online: https://clinicaltrials.gov/study/NCT04979806 (accessed on 11 October 2023).
  80. Vázquez-Ucha, J.C.; Lasarte-Monterrubio, C.; Guijarro-Sánchez, P.; Oviaño, M.; Álvarez-Fraga, L.; Alonso-García, I.; Arca-Suárez, J.; Bou, G.; Beceiro, A. Assessment of Activity and Resistance Mechanisms to Cefepime in Combination with the Novel β-Lactamase Inhibitors Zidebactam, Taniborbactam, and Enmetazobactam against a Multicenter Collection of Carbapenemase-Producing Enterobacterales. Antimicrob. Agents Chemother. 2022, 66, e01676-21.
  81. Eljaaly, K.; Alharbi, A.; Alshehri, S.; Ortwine, J.K.; Pogue, J.M. Plazomicin: A Novel Aminoglycoside for the Treatment of Resistant Gram-Negative Bacterial Infections. Drugs 2019, 79, 243–269.
  82. Golkar, T.; Bassenden, A.V.; Maiti, K.; Arya, D.P.; Schmeing, T.M.; Berghuis, A.M. Structural Basis for Plazomicin Antibiotic Action and Resistance. Commun. Biol. 2021, 4, 729.
  83. Wagenlehner, F.M.E.; Cloutier, D.J.; Komirenko, A.S.; Cebrik, D.S.; Krause, K.M.; Keepers, T.R.; Connolly, L.E.; Miller, L.G.; Friedland, I.; Dwyer, J.P. Once-Daily Plazomicin for Complicated Urinary Tract Infections. J. Urol. 2019, 202, 641–642.
  84. Thwaites, M.; Hall, D.; Stoneburner, A.; Shinabarger, D.; Serio, A.W.; Krause, K.M.; Marra, A.; Pillar, C. Activity of Plazomicin in Combination with Other Antibiotics against Multidrug-Resistant Enterobacteriaceae. Diagn. Microbiol. Infect. Dis. 2018, 92, 338–345.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 249
Revisions: 2 times (View History)
Update Date: 15 Nov 2023
1000/1000
Video Production Service