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 -- 2921 2022-11-02 10:30:31 |
2 format + 1 word(s) 2922 2022-11-03 02:44:26 |

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.
Zakhour, J.;  Sharara, S.L.;  Hindy, J.;  Haddad, S.F.;  Kanj, S.S. Antimicrobial Treatment of Pseudomonas aeruginosa Severe Sepsis. Encyclopedia. Available online: https://encyclopedia.pub/entry/32529 (accessed on 27 July 2024).
Zakhour J,  Sharara SL,  Hindy J,  Haddad SF,  Kanj SS. Antimicrobial Treatment of Pseudomonas aeruginosa Severe Sepsis. Encyclopedia. Available at: https://encyclopedia.pub/entry/32529. Accessed July 27, 2024.
Zakhour, Johnny, Sima L. Sharara, Joya-Rita Hindy, Sara F. Haddad, Souha S. Kanj. "Antimicrobial Treatment of Pseudomonas aeruginosa Severe Sepsis" Encyclopedia, https://encyclopedia.pub/entry/32529 (accessed July 27, 2024).
Zakhour, J.,  Sharara, S.L.,  Hindy, J.,  Haddad, S.F., & Kanj, S.S. (2022, November 02). Antimicrobial Treatment of Pseudomonas aeruginosa Severe Sepsis. In Encyclopedia. https://encyclopedia.pub/entry/32529
Zakhour, Johnny, et al. "Antimicrobial Treatment of Pseudomonas aeruginosa Severe Sepsis." Encyclopedia. Web. 02 November, 2022.
Antimicrobial Treatment of Pseudomonas aeruginosa Severe Sepsis
Edit

Pseudomonas aeruginosa is a pathogen often encountered in a healthcare setting. It has consistently ranked among the most frequent pathogens seen in nosocomial infections, particularly bloodstream and respiratory tract infections. Aside from having intrinsic resistance to many antibiotics, it rapidly acquires resistance to novel agents. Given the high mortality of pseudomonal infections generally, and pseudomonal sepsis particularly, and with the rise of resistant strains, treatment can be very challenging for the clinician.

Pseudomonas aeruginosa severe sepsis antibiotics antimicrobial resistance

1. Empirical Antimicrobial Therapy

EAT in sepsis should consider the patient’s allergies, comorbidities, the primary site of infection, prior antibiotic exposure, as well as local susceptibility patterns [1]. AMR should be highly suspected if there is recent admission to a hospital unit where prevalence of MDR-PA is greater than 20% or if the patient has received antipseudomonal beta-lactam antimicrobials within the past three months [2]. Although some studies reported trends towards decreased resistance of P. aeruginosa [3], low and middle income countries (LMICs) still suffer from a high burden of AMR [4]. The CDC reports that 32,600 cases of MDR-PA infections occurred in patients hospitalized in the United States in 2017, resulting in 2700 deaths [5]. For P. aeruginosa, MDR is defined as resistance to at least one agent in three or more antibiotic classes, extensive drug resistance (XDR) is defined as resistance to at least one agent in all but two or fewer antibiotic classes, and pan-drug resistance (PDR) is non-susceptibility to all agents [6]. Most recently, it was suggested to label MDR-PA as difficult to treat (DTR) when it is resistant to piperacillin-tazobactam, ceftazidime, cefepime, aztreonam, meropenem, imipenem-cilastatin, ciprofloxacin and levofloxacin [7][8].
Combination therapy in P. aeruginosa is often used to decrease the risk of inadequate EAT by combining drugs with multiple mechanisms of action. In a recently published multicenter retrospective study including 1017 neutropenic patients with P. aeruginosa bacteremic pneumonia, inappropriate EAT was given to 23% of patients and was associated with infection with MDR-PA. Additionally, inappropriate EAT was associated with increased 30-day mortality while appropriate EAT was independently associated with improved survival [9]. No consensus has been reached regarding the use of empirical combination versus monotherapy in P. aeruginosa sepsis, mainly due to the lack of robust prospective studies to provide strong levels of evidence [10]. In fact, a prospective study found no differences in outcomes between patients who received empirical combination antibiotics when compared to monotherapy [11]. A meta-analysis that included 1721 patients showed no difference in mortality among patients with pseudomonal infections who were treated empirically with beta-lactam monotherapy or combination therapy with the addition of an aminoglycoside (AG) or a fluoroquinolone (FQ) [12]. Furthermore, a Cochrane research that included 69 randomized controlled trials (RCTs) with a total of 7863 patients comparing beta lactam monotherapy and combination with an AG in the management of sepsis showed no difference in mortality in the P. aeruginosa subgroup analysis and significantly increased nephrotoxicity with combination therapy [13]. Moreover, a post hoc analysis of 593 patients with P. aeruginosa bacteremia showed no benefit of empiric combination therapy [14] and another meta-analysis of 4980 patients showed no difference in mortality, microbiological, or clinical cure when using empirical combination vs. monotherapy for patients with P. aeruginosa BSI or pneumonia [15]. On the other hand, a retrospective cohort study by Micek et al. including 305 patients with P. aeruginosa BSI showed that using combination therapy while awaiting for identification and susceptibility testing decreased the risk of inadequate EAT from 79.4% to 65.5% (p-value = 0.011). Additionally, mortality was significantly higher in patients who received inappropriate EAT (30.7% versus 17.8%, p-value = 0.018). In that study, inappropriate EAT, respiratory failure and septic shock were found to be independent risk factors for in-hospital mortality [16]. A recent meta-analysis of four studies that evaluated all-cause mortality (total of 148 patients), showed a significant decrease in mortality with combination therapy for severe infections caused by P. aeruginosa (OR 0.31, 95% CI 0.1–0.97, p-value = 0.045) [17].
Given the rise of AMR and the risk of inadequate EAT, combination empiric therapy should be highly considered in cases of severe sepsis [18]. The Surviving Sepsis campaign recommends combination empirical therapy during acute illness [19]. Two different mechanisms of action are preferred, typically a backbone beta-lactam (conventional or novel depending on risk of AMR) combined with an AG or a FQ [2][20]. Although one study suggested better outcomes when FQ was used instead of AG as a second agent [21], the choice of agent should be guided by local susceptibility patterns [2]. A retrospective cross-sectional analysis of blood and respiratory P. aeruginosa isolates from patients admitted to the ICU found that the combination with the highest susceptibility was piperacillin-tazobactam combined with an AG, while the combination with the lowest susceptibility was a carbapenem combined with a FQ [22]. Additionally, isolates were found to have less resistance to combinations with AG than those with FQ. A pharmacokinetic/pharmacodynamic (PK/PD) prospective randomized controlled trial suggested that a higher dose of amikacin (25 mg/kg) for patients with severe sepsis and at risk for P. aeruginosa infection was more likely to achieve an MIC that is closest to the EUCAST susceptibility breakpoint than standard dosing (15 mg/kg) [23]. Above all, the choice of empiric antimicrobial regimen should consider the potential for co-resistance to multiple first-line agents. For instance, a multinational microbiological study including 1783 isolates of MDR-PA from patients with P. aeruginosa BSI reported that co-resistance to many first-line antipseudomonal agents was very common, especially between piperacillin-tazobactam, meropenem and ceftazidime. Among antimicrobials that were included in the study, only Ceftolozane-tazobactam (C/T), a novel beta-lactam-beta-lactamase inhibitor combination, achieved significant additional activity against strains that exhibited resistance to one of the first-line agents [24]. Those findings suggest that C/T may be considered for empirical therapy if local rates of PA resistance to first-line agents is high. If combination empiric therapy is used, it is highly recommended prompt de-escalation once there is clinical improvement and susceptibility results are available. Additionally, although 40% of P. aeruginosa BSIs will have an unidentifiable origin, prompt source control is recommended when possible to improve patient outcomes [25][26].

2. Targeted Therapy for P. aeruginosa Sepsis

2.1. P. aeruginosa Sensitive to First Line Antipseudomonal Agents

P. aeruginosa is intrinsically resistant to several antibiotics due to the low permeability of its outer membrane, expression of various efflux pumps, and the production of antibiotic-inactivating enzymes such as inducible cephalosporinases. First-line beta-lactam agents for P. aeruginosa coverage include beta-lactam/beta-lactamase-inhibitor combinations (BL/BLI) (piperacillin-tazobactam and ticarcillin-clavulanate) and cephalosporins with antipseudomonal activity (ceftazidime, cefepime, and cefoperazone). Cefepime is the most commonly used beta-lactam antibiotic for P. aeruginosa [27]. Fluoroquinolones (ciprofloxacin and levofloxacin) remain currently the only oral treatment options for quinolone-sensitive P. aeruginosa. However, ciprofloxacin is superior to levofloxacin given the higher risk of emergence of quinolone-resistant P. aeruginosa with the use of levofloxacin [28]. Additionally, older FQ are less effective in acidic environments like UTIs [29]. Newer FQ such as finafloxacin and delafloxacin offer more activity in acidic environments but are yet to be widely available [30].
Second line agents for P. aeruginosa sepsis are carbapenems, including meropenem, imipenem, and doripenem. Meropenem is often preferred over imipenem given the latter’s higher propensity to induce resistance during treatment [31]. Doripenem was shown to be more active in vitro against P. aeruginosa compared to meropenem and imipenem but this has not been proven in clinical studies [32][33]. Nonetheless, cephalosporins should be favored over carbapenems when applicable due to more potent activity and narrower spectrum [34] as well as less propensity to select for future resistance [35].
Other agents include the monobactam class (aztreonam) which can be used as an alternative for patients with penicillin allergy. Gentamicin, tobramycin, and amikacin are all AG that can be active against P. aeruginosa but are not indicated as monotherapy except for UTIs, as they are associated with higher mortality rates [36]. In the case of severe sepsis, the pathophysiological shifts may lead to an increased volume of distribution and augmented renal clearance and may lead to suboptimal AG concentrations and potentially poorer outcomes [37]. For optimal coverage, researchers prefer tobramycin or amikacin over gentamicin [38]. Otherwise, plazomicin, a newer AG, was shown to be less effective and is currently only indicated in the treatment of UTIs [39].
Emergence of resistance during the course of treatment is a serious concern. Such is the case of a cohort of 271 patients with various P. aeruginosa infections receiving antipseudomonal antimicrobial therapy, where emergent resistance was reported in up to 10% of cases [35]. Additionally, standard susceptibility testing may not be as accurate in identifying resistance when hospitalization duration increases. This is likely due to development of resistance or acquisition of drug-resistant hospital-acquired strains, especially with prolonged stay in the ICU. Studies have indicated that initial antibiograms become unreliable as a predictor of susceptibility of P. aeruginosa after 1–2 weeks of hospitalization, particularly in the ICU, with a significant increase in MIC for multiple anti-pseudomonal agents [40][41]. Among conventional treatment agents, imipenem was the most likely to cause resistance emergence and ceftazidime was the least likely [35].

2.2. P. aeruginosa Resistant to First Line Therapy

P. aeruginosa can develop resistance through multiple mechanisms including selection of chromosomal mutations or horizontal acquisition of broad-spectrum resistance genes. Many resistance mechanisms are involved and include beta-lactamase production, AG-modifying enzymes, efflux pumps, porin loss, and various target site modifications [34]. Treatment options for CRPA is challenging given the variety of resistance mechanisms like the production of carbapenemases of different classes, outer membrane protein modification (OprD) or efflux pumps (MexAB, MexXY). Novel anti-pseudomonal drugs have been developed in response to this challenge to address the increase in resistance, which has been reported in up to 54% of nosocomial P. aeruginosa infections [42][43][44]. These include novel BL-BLI like C/T, ceftazidime/avibactam (CAZ/AVI), and imipenem-cilastatin/relebactam (IMI/REL) or novel cephalosporins like cefiderocol [1].
C/T has potent intrinsic anti-pseudomonal activity owing to its greater affinity to all essential penicillin-binding proteins (PBP) including PBP1b, PBP1c, and PBP3. Based on RCTs, the US Food and Drug association (FDA) and the European Medicines Agency (EMA) have approved the use of C/T in complicated intra-abdominal infections (IAIs), UTIs, and hospital-acquired pneumonia including ventilator-associated pneumonia (VAP) [45]. Subset analysis in the clinical trials showed that patients with P. aeruginosa had a favorable outcome compared to carbapenems in the HAP trial [46], carbapenems combined with metronidazole in the complicated IAI trial [47], and levofloxacin in the complicated UTIs trial [48]. CAZ/AVI is a novel BL/BLI combination approved by the FDA and the EMA for treatment of complicated UTIs, IAIs and infections with Gram negative resistant pathogens [49][50]. C/T and CAZ/AVI have been considered key therapeutic agents against resistant P. aeruginosa strains. However, since the commercialization of these agents, there has been emergence of resistance of P. aeruginosa following therapy, particularly with highly cephalosporin-resistant conferring mutations [51]. Data for treatment associated resistance in novel agents is still inconclusive, but it appears that the highest risk is with C/T and CAZ/AVI with common cross resistance to both agents [52][53]. Currently, real-world data is scarce and does not show superiority of an agent compared to another and therefore the choice of antimicrobial therapy should be based on the susceptibility profile which can vary according to the local epidemiology with regional variability [54]. Since the introduction of C/T, case reports and case series of MDR P. aeruginosa infections treated with C/T have demonstrated the clinical efficacy of this formulation, including its use to treat infections in critically ill patients, and those with cystic fibrosis [55][56]. The European Society of Clinical Microbiology and Infectious Diseases (ESCMID) recent guidelines for the treatment of infections caused by MDR Gram-negative bacilli suggest treatment with C/T as the single first choice for severe pseudomonal infections like severe sepsis [10]. As for the Infectious Diseases Society of America (IDSA), their guidelines recommend treatment with either C/T, CAZ/AVI, or imipenem/relebactam for infections with DTR P. aeruginosa outside the urinary tract [7]. In fact, although experience with CAZ/AVI in the management of P. aeruginosa infections is more limited, adding avibactam to ceftazidime has shown success in lowering MICs of many XDR P. aeruginosa isolates [57]. When considering XDR P. aeruginosa, an important concept is that although C/T is more likely to be active than CAZ/AVI, there are some C/T-resistant strains that can be susceptible to CAZ/AVI [58]. Therefore, in vitro susceptibilities to both agents should be obtained whenever possible.
Cefiderocol is a novel siderophore cephalosporin that can overcome efflux pumps. The IDSA recommends this agent as an alternative therapy when other novel BL/BLI agents are unavailable or if there is resistance or intolerance. A recent RCT compared the outcomes of patients with infections due to carbapenem-resistant bacteria treated with cefiderocol or best available therapy (BAT) [59]. Although the mortality rate was higher in the cefiderocol arm, the number of patients with P. aeruginosa infection was small, and increased mortality was only observed in patients with a mono- or polymicrobial infection including Acinetobacter baumannii. Furthermore, the results suggest that cefiderocol performed as well as BAT, but was not associated with significantly decreased mortality or reduced adverse events like what was reported from studies on newer BL/BLIs [60][61]. A recently published study of P. aeruginosa isolates resistant to C/T and CAZ/AVI concluded that cefiderocol was the most active agent against these isolates, with only one resistant clinical isolate (R504C substitution in PBP3) [51]. Imipenem-cilastatin/relebactam was also active against all isolates except two that carried the VIM-20 carbapenemase. In the same study, newer combinations such as cefepime/zidebactam and cefepime/taniborbactam displayed activity against most of the isolates, but resistance was observed in some strains with PBP3 amino acid substitutions and those that overexpressed mexAB-oprM or mexXY efflux pumps.
Evidence for the combination IMI/REL is derived from the RESTORE-IMI 1, a randomized controlled phase 3 trial, comparing IMI/REL to a combination of colistin and imipenem for patients with Gram-negative infections of which 77% were due to P. aeruginosa. There was a trend of lower mortality in the arm that was treated with the novel agent compared to combination therapy, but a significantly lower rate of adverse effects and nephrotoxicity [60]. In the subgroup analysis, patients with pneumonia as well as those with renal insufficiency had a higher mortality perhaps owing to lower concentration achieved with the given doses (86). As for meropenem/vaborbactam, it is not recommended given that the addition of vaborbactam was not found to restore susceptibility to meropenem-resistant strains [7].
Despite great outcomes associated with novel agents, these therapies remain inactive against most metallo-beta-lactamase-(MBL)-producing P. aeruginosa strains. The monobactam aztreonam is unique by demonstrating stability to hydrolysis by MBLs and may maintain activity against MBL-producing P. aeruginosa [62]. Many strains of MBL-producing P. aeruginosa will also contain mechanisms of resistance against aztreonam, such as increased expression of pseudomonas derived cephalosporinases (PDCs). Nevertheless, aztreonam is an attractive option in combination with CAZ/AVI for the treatment of infections caused by MBL-producing P. aeruginosa [63]. Cefiderocol has also shown activity against all carbapenemase classes including MBL but more clinical evidence is needed [64].
As for polymyxins (polymyxin B and colistin), the IDSA’s latest guidelines for the treatment of DTR P. aeruginosa recommends against their use when novel options with less nephrotoxicity are available [7]. However, given the increase in resistance rates and scarcity of novel antibiotics in LMICs, colistin has been increasingly used. Studies have shown that colistin can be used as salvage therapy when options are limited [65], and that it can be associated with a lower expected incidence of nephrotoxicity than previously expected. However, renal function should be closely monitored during therapy with appropriate dose adjustments.
Fosfomycin is an interesting choice for DTR P. aeruginosa given that it retains activity against some XDR and PDR strains which may be useful especially in critically ill patients with severe sepsis [66]. A case series including 48 critically ill patients, of whom 17 had infection with MDR-PA and 10 with severe sepsis, evaluated the efficacy of intravenous fosfomycin mainly in combination with colistin. Patients who received fosfomycin were found to have an all-cause 28-day mortality of 37.5%. Additionally, adverse events were minor and included nausea and reversible hypokalemia while resistance emergence to fosfomycin was found in only 3 patients [67]. Another retrospective study comparing outcomes between patients with CRPA pneumonia receiving a combination of doripenem and colistin or doripenem and fosfomycin found similar outcomes between both groups; however, results should be cautiously interpreted given the small size of the study’s population (49 patients) [67]. It should be noted that intravenous fosfomycin should not be given as monotherapy except in cases of uncomplicated UTI; otherwise, it should be given in combination with other agents for bacteremia, nosocomial pneumonia and complicated skin and soft tissue infection to avoid emergence of resistance [18].
While definitive combination therapy may exert a synergistic effect and possibly reduce the emergence of resistance, it can also result in increased side effects and unnecessary costs [34]. It has been previously discussed the lack of rigorous evidence regarding the efficacy and safety of empirical combination therapy. Similarly, the evidence concerning the efficacy and safety of definitive combination antimicrobial therapy is still inconclusive and guidelines are yet to make a specific recommendation regarding combination therapy once susceptibility results are available. A retrospective study of 187 patients with P. aeruginosa BSI found that there was significant decrease in mortality in patients treated with definitive combination therapy compared to monotherapy by multivariate analysis (HR 0.30, 95% CI 0.13–0.71, p = 0.006) [68]. On the other hand, many studies have found no differences in outcomes between patients who received definitive combination vs. monotherapy [11]. For example, a retrospective study including 183 patients with P. aeruginosa VAP found similar outcomes with combination vs. monotherapy [69]. Furthermore, a meta-analysis found no difference in mortality between combination and monotherapy for patients with P. aeruginosa infections [12]. Hence, a single agent (preferably a beta-lactam based on susceptibility profile) should be used for definitive antimicrobial therapy since continuing combination therapy is unlikely to have any added value once susceptibilities are available [20].

References

  1. Ibrahim, D.; Jabbour, J.-F.; Kanj, S.S. Current choices of antibiotic treatment for Pseudomonas aeruginosa infections. Curr. Opin. Infect. Dis. 2020, 33, 464–473.
  2. Bassetti, M.; Vena, A.; Russo, A.; Croxatto, A.; Calandra, T.; Guery, B. Rational approach in the management of Pseudomonas aeruginosa infections. Curr. Opin. Infect. Dis. 2018, 31, 578–586.
  3. Jernigan, J.A.; Hatfield, K.M.; Wolford, H.; Nelson, R.E.; Olubajo, B.; Reddy, S.C.; McCarthy, N.; Paul, P.; McDonald, L.C.; Kallen, A.; et al. Multidrug-Resistant Bacterial Infections in U.S. Hospitalized Patients, 2012–2017. N. Engl. J. Med. 2020, 382, 1309–1319.
  4. Rosenthal, V.D.; Al-Abdely, H.M.; El-Kholy, A.A.; AlKhawaja, S.A.A.; Leblebicioglu, H.; Mehta, Y.; Rai, V.; Hung, N.V.; Kanj, S.S.; Salama, M.F.; et al. International Nosocomial Infection Control Consortium report, data summary of 50 countries for 2010-2015: Device-associated module. Am. J. Infect. Control 2016, 44, 1495–1504.
  5. 2019 Antibiotic Resistance Threats Report. Available online: https://www.cdc.gov/drugresistance/biggest-threats.html (accessed on 31 July 2022).
  6. Magiorakos, A.-P.; Srinivasan, A.; Carey, R.B.; Carmeli, Y.; Falagas, M.E.; Giske, C.G.; Harbarth, S.; Hindler, J.F.; Kahlmeter, G.; Olsson-Liljequist, B.; et al. Multidrug-resistant, extensively drug-resistant and pandrug-resistant bacteria: An international expert proposal for interim standard definitions for acquired resistance. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2012, 18, 268–281.
  7. Tamma, P.D.; Aitken, S.L.; Bonomo, R.A.; Mathers, A.J.; van Duin, D.; Clancy, C.J. IDSA Guidance on the Treatment of Antimicrobial-Resistant Gram-Negative Infections: Version 1.0; IDSA: Arlington, VA, USA, 2022; Available online: https://www.idsociety.org/practice-guideline/amr-guidance/ (accessed on 31 July 2022).
  8. Kadri, S.S.; Adjemian, J.; Lai, Y.L.; Spaulding, A.B.; Ricotta, E.; Prevots, D.R.; Palmore, T.N.; Rhee, C.; Klompas, M.; Dekker, J.P.; et al. Difficult-To-Treat Resistance in Gram-Negative Bacteremia at 173 US Hospitals: Retrospective Cohort Analysis of Prevalence, Predictors, and Outcome of Resistance to All First-Line Agents. Clin. Infect. Dis. 2018, 67, 1803–1814. Available online: https://pubmed.ncbi.nlm.nih.gov/30052813/ (accessed on 28 September 2022).
  9. Albasanz-Puig, A.; Durà-Miralles, X.; Laporte-Amargós, J.; Mussetti, A.; Ruiz-Camps, I.; Puerta-Alcalde, P.; Abdala, E.; Oltolini, C.; Akova, M.; Montejo, J.M.; et al. Effect of Combination Antibiotic Empirical Therapy on Mortality in Neutropenic Cancer Patients with Pseudomonas aeruginosa Pneumonia. Microorganisms 2022, 10, 733.
  10. Paul, M.; Carrara, E.; Retamar, P.; Tängdén, T.; Bitterman, R.; Bonomo, R.A.; de Waele, J.; Daikos, G.L.; Akova, M.; Harbarth, S.; et al. European Society of Clinical Microbiology and Infectious Diseases (ESCMID) guidelines for the treatment of infections caused by multidrug-resistant Gram-negative bacilli (endorsed by European society of intensive care medicine). Clin. Microbiol. Infect. 2022, 28, 521–547.
  11. Díaz-Cañestro, M.; Periañez, L.; Mulet, X.; Martin-Pena, M.L.; Fraile-Ribot, P.A.; Ayestarán, I.; Colomar, A.; Nuñez, B.; Maciá, M.; Novo, A.; et al. Ceftolozane/tazobactam for the treatment of multidrug resistant Pseudomonas aeruginosa: Experience from the Balearic Islands. Eur. J. Clin. Microbiol. Infect. Dis. 2018, 37, 2191–2200.
  12. Vardakas, K.Z.; Tansarli, G.S.; Bliziotis, I.A.; Falagas, M.E. β-Lactam plus aminoglycoside or fluoroquinolone combination versus β-lactam monotherapy for Pseudomonas aeruginosa infections: A meta-analysis. Int. J. Antimicrob. Agents 2013, 41, 301–310.
  13. Paul, M.; Lador, A.; Grozinsky-Glasberg, S.; Leibovici, L. Beta lactam antibiotic monotherapy versus beta lactam-aminoglycoside antibiotic combination therapy for sepsis. Cochrane Database Syst. Rev. 2014, 2014, CD003344.
  14. Peña, C.; Suarez, C.; Ocampo-Sosa, A.; Murillas, J.; Almirante, B.; Pomar, V.; Aguilar, M.; Granados, A.; Calbo, E.; Rodríguez-Baño, J.; et al. Effect of Adequate Single-Drug vs Combination Antimicrobial Therapy on Mortality in Pseudomonas aeruginosa Bloodstream Infections: A Post Hoc Analysis of a Prospective Cohort. In Clinical Infectious Diseases; Oxford Academic: Oxford, UK, 2013; Available online: https://academic.oup.com/cid/article/57/2/208/313652 (accessed on 31 July 2022).
  15. Onorato, L.; Macera, M.; Calò, F.; Cirillo, P.; Di Caprio, G.; Coppola, N. Beta-lactam monotherapy or combination therapy for bloodstream infections or pneumonia due to Pseudomonas aeruginosa: A meta-analysis. Int. J. Antimicrob. Agents 2022, 59, 106512.
  16. Micek, S.T.; Lloyd, A.E.; Ritchie, D.J.; Reichley, R.M.; Fraser, V.J.; Kollef, M.H. Pseudomonas aeruginosa bloodstream infection: Importance of appropriate initial antimicrobial treatment. Antimicrob. Agents Chemother. 2005, 49, 1306–1311.
  17. Fiore, M.; Corrente, A.; Pace, M.C.; Alfieri, A.; Simeon, V.; Ippolito, M.; Giarratano, A.; Cortegiani, A. Ceftolozane-Tazobactam Combination Therapy Compared to Ceftolozane-Tazobactam Monotherapy for the Treatment of Severe Infections: A Systematic Review and Meta-Analysis; Antibiotics: Basel, Switzerland, 2021. Available online: https://pubmed.ncbi.nlm.nih.gov/33467508/ (accessed on 31 July 2022).
  18. Al Salman, J.; Al Dabal, L.; Bassetti, M.; Alfouzan, W.A.; Al Maslamani, M.; Alraddadi, B.; Elhoufi, A.; Enani, M.; Khamis, F.A.; Mokkadas, E.; et al. Management of infections caused by WHO critical priority Gram-negative pathogens in Arab countries of the Middle East: A consensus paper. Int. J. Antimicrob. Agents 2020, 56, 106104.
  19. Rhodes, A.; Evans, L.E.; Alhazzani, W.; Levy, M.M.; Antonelli, M.; Ferrer, R.; Kumar, A.; Sevransky, J.E.; Sprung, C.L.; Nunnally, M.E.; et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Crit. Care Med. 2017, 45, 486–552.
  20. Bassetti, M.; Vena, A.; Croxatto, A.; Righi, E.; Guery, B. How to manage Pseudomonas aeruginosa infections. Drugs Context 2018, 7, 212527.
  21. Paulsson, M.; Granrot, A.; Ahl, J.; Tham, J.; Resman, F.; Riesbeck, K.; Månsson, F. Antimicrobial combination treatment including ciprofloxacin decreased the mortality rate of Pseudomonas aeruginosa bacteraemia: A retrospective cohort study. Eur. J. Clin. Microbiol. Infect. Dis. 2017, 36, 1187–1196.
  22. Puzniak, L.; DePestel, D.D.; Srinivasan, A.; Ye, G.; Murray, J.; Merchant, S.; DeRyke, C.A.; Gupta, V. A Combination Antibiogram Evaluation for Pseudomonas aeruginosa in Respiratory and Blood Sources from Intensive Care Unit (ICU) and Non-ICU Settings in U.S. Hospitals. Antimicrob. Agents Chemother. 2019, 63, e02564-18.
  23. De Winter, S.; Wauters, J.; Meersseman, W.; Verhaegen, J.; Van Wijngaerden, E.; Peetermans, W.; Annaert, P.; Verelst, S.; Spriet, I. Higher versus standard amikacin single dose in emergency department patients with severe sepsis and septic shock: A randomised controlled trial. Int. J. Antimicrob. Agents 2018, 51, 562–570.
  24. Moise, P.A.; Gonzalez, M.; Alekseeva, I.; Lopez, D.; Akrich, B.; DeRyke, C.A.; Chen, W.-T.; Pavia, J.; Palermo, B.; Hackel, M.; et al. Collective assessment of antimicrobial susceptibility among the most common Gram-negative respiratory pathogens driving therapy in the ICU. JAC-Antimicrob. Resist. 2021, 3, dlaa129.
  25. 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.; et al. Multistate point-prevalence survey of health care-associated infections. N. Engl. J. Med. 2014, 370, 1198–1208.
  26. Tofas, P.; Samarkos, M.; Piperaki, E.-T.; Kosmidis, C.; Triantafyllopoulou, I.-D.; Kotsopoulou, M.; Pantazatou, A.; Perlorentzou, S.; Poulli, A.; Vagia, M.; et al. Pseudomonas aeruginosa bacteraemia in patients with hematologic malignancies: Risk factors, treatment and outcome. Diagn. Microbiol. Infect. Dis. 2017, 88, 335–341.
  27. Britt, N.S.; Ritchie, D.J.; Kollef, M.H.; Burnham, C.A.; Durkin, M.J.; Hampton, N.B.; Micek, S.T. Importance of Site of Infection and Antibiotic Selection in the Treatment of Carbapenem-Resistant Pseudomonas aeruginosa Sepsis. Antimicrob. Agents Chemother. 2018, 62, e02400-17.
  28. Kaye, K.S.; Kanafani, Z.A.; Dodds, A.E.; Engemann, J.J.; Weber, S.G.; Carmeli, Y. Differential Effects of Levofloxacin and Ciprofloxacin on the Risk for Isolation of Quinolone-Resistant Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2006, 50, 2192–2196.
  29. McKeage, K. Finafloxacin: First global approval. Drugs 2015, 75, 687–693.
  30. Kocsis, B.; Gulyás, D.; Szabó, D. Delafloxacin, Finafloxacin, and Zabofloxacin: Novel Fluoroquinolones in the Antibiotic Pipeline. Antibiotics 2021, 10, 1506.
  31. Gimeno, C.; Cantón, R.; García, A.; Gobernado, M. Grupo Español de Estudio de Doripenem . Rev. Espanola Quimioter. Publicacion Of. Soc. Espanola Quimioter. 2010, 23, 144–152.
  32. Pillar, C.M.; Torres, M.K.; Brown, N.P.; Shah, D.; Sahm, D.F. In vitro activity of doripenem, a carbapenem for the treatment of challenging infections caused by gram-negative bacteria, against recent clinical isolates from the United States. Antimicrob. Agents Chemother. 2008, 52, 4388–4399.
  33. Korten, V.; Söyletir, G.; Yalçın, A.N.; Oğünç, D.; Dokuzoğuz, B.; Esener, H.; Ulusoy, S.; Tünger, A.; Aygen, B.; Sümerkan, B.; et al. Comparative evaluation of in vitro activities of carbapenems against gram-negative pathogens: Turkish data of COMPACT study. Mikrobiyol. Bul. 2011, 45, 197–209.
  34. Kanj, S.S.; Kanafani, Z.A. Current Concepts in Antimicrobial Therapy Against Resistant Gram-Negative Organisms: Extended-Spectrum β-Lactamase–Producing Enterobacteriaceae, Carbapenem-Resistant Enterobacteriaceae, and Multidrug-Resistant Pseudomonas aeruginosa. Mayo Clin. Proc. 2011, 86, 250–259.
  35. Carmeli, Y.; Troillet, N.; Eliopoulos, G.M.; Samore, M.H. Emergence of antibiotic-resistant Pseudomonas aeruginosa: Comparison of risks associated with different antipseudomonal agents. Antimicrob. Agents Chemother. 1999, 43, 1379–1382.
  36. Phe, K.; Bowers, D.R.; Babic, J.T.; Tam, V.H. Outcomes of empiric aminoglycoside monotherapy for Pseudomonas aeruginosa bacteremia. Diagn. Microbiol. Infect. Dis. 2019, 93, 346–348.
  37. Varghese, J.M.; Roberts, J.A.; Lipman, J. Antimicrobial Pharmacokinetic and Pharmacodynamic Issues in the Critically Ill with Severe Sepsis and Septic Shock-Critical Care Clinics; Elsevier: Amsterdam, The Netherlands, 2011; Available online: https://www.criticalcare.theclinics.com/article/S0749-0704(10)00068-0/fulltext (accessed on 16 September 2022).
  38. Kluge, R.M.; Standiford, H.C.; Tatem, B.; Young, V.M.; Greene, W.H.; Schimpff, S.C.; Calia, F.M.; Hornick, R.B. Comparative activity of tobramycin, amikacin, and gentamicin alone and with carbenicillin against Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 1974, 6, 442–446.
  39. Zhanel, G.G.; Lawson, C.D.; Zelenitsky, S.; Findlay, B.; Schweizer, F.; Adam, H.; Walkty, A.; Rubinstein, E.; Gin, A.S.; Hoban, D.J.; et al. Comparison of the next-generation aminoglycoside plazomicin to gentamicin, tobramycin and amikacin. Expert Rev. Anti Infect. Ther. 2012, 10, 459–473.
  40. Anderson, D.J.; Miller, B.; Marfatia, R.; Drew, R. Ability of an antibiogram to predict Pseudomonas aeruginosa susceptibility to targeted antimicrobials based on hospital day of isolation. Infect. Control Hosp. Epidemiol. 2012, 33, 589–593.
  41. Riou, M.; Carbonnelle, S.; Avrain, L.; Mesaros, N.; Pirnay, J.-P.; Bilocq, F.; De Vos, D.; Simon, A.; Piérard, D.; Jacobs, F.; et al. In vivo development of antimicrobial resistance in Pseudomonas aeruginosa strains isolated from the lower respiratory tract of Intensive Care Unit patients with nosocomial pneumonia and receiving antipseudomonal therapy. Int. J. Antimicrob. Agents 2010, 36, 513–522.
  42. De Matos, E.C.O.; De Matos, H.J.; da Conceição, M.L.; Rodrigues, Y.C.; Carneiro, I.C.D.R.S.; Lima, K.V.B. Clinical and microbiological features of infections caused by Pseudomonas aeruginosa in patients hospitalized in intensive care units. Rev. Soc. Bras. Med. Trop. 2016, 49, 305–311.
  43. Peng, Y.; Bi, J.; Shi, J.; Li, Y.; Ye, X.; Chen, X.; Yao, Z. Multidrug-resistant Pseudomonas aeruginosa infections pose growing threat to health care–associated infection control in the hospitals of Southern China: A case-control surveillance study. Am. J. Infect. Control 2014, 42, 1308–1311.
  44. Jeong, S.J.; Yoon, S.S.; Bae, I.K.; Jeong, S.H.; Kim, J.M.; Lee, K. Risk factors for mortality in patients with bloodstream infections caused by carbapenem-resistant Pseudomonas aeruginosa: Clinical impact of bacterial virulence and strains on outcome. Diagn. Microbiol. Infect. Dis. 2014, 80, 130–135.
  45. EMA. Zerbaxa. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/zerbaxa (accessed on 28 September 2022).
  46. 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.
  47. Solomkin, J.; Hershberger, E.; Miller, B.; Popejoy, M.; Friedland, I.; Steenbergen, J.; Yoon, M.; Collins, S.; Yuan, G.; Barie, P.S.; et al. Ceftolozane/Tazobactam Plus Metronidazole for Complicated Intra-abdominal Infections in an Era of Multidrug Resistance: Results From a Randomized, Double-Blind, Phase 3 Trial (ASPECT-cIAI). Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2015, 60, 1462–1471.
  48. Wagenlehner, L.M.; Umeh, O.; Steenbergen, J.; Yuan, G.; Darouiche, R.O. Ceftolozane-TAZOBACTAM Compared with Levofloxacin in the Treatment of Complicated Urinary-Tract Infections, Including Pyelonephritis: A Randomised, Double-Blind, Phase 3 Trial (ASPECT-cUTI). Lancet 2015, 385, 1949–1956. Available online: https://pubmed.ncbi.nlm.nih.gov/25931244/ (accessed on 28 September 2022).
  49. AVYCAZ. Safely and Effectively. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/206494s005,s006lbl.pdf (accessed on 28 September 2022).
  50. EMA. Zavicefta. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/zavicefta (accessed on 29 September 2022).
  51. 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.
  52. Skoglund, E.; Abodakpi, H.; Rios, R.; Diaz, L.; De La Cadena, E.; Dinh, A.Q.; Ardila, J.; Miller, W.R.; Munita, J.M.; Arias, C.A.; et al. In Vivo Resistance to Ceftolozane/Tazobactam in Pseudomonas aeruginosa Arising by AmpC- and Non-AmpC-Mediated Pathways. Case Rep. Infect. Dis. 2018, 2018, 9095203.
  53. Tamma, P.D.; Beisken, S.; Bergman, Y.; Posch, A.E.; Avdic, E.; Sharara, S.L.; Cosgrove, S.E.; Simner, P.J. Modifiable Risk Factors for the Emergence of Ceftolozane-tazobactam Resistance. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2021, 73, e4599–e4606.
  54. Sid Ahmed, M.A.; Hamid, J.M.; Husain, A.A.; Hadi, H.A.; Skariah, S.; Sultan, A.A.; Ibrahim, E.B.; Al Khal, A.L.; Soderquist, B.; Jass, J.; et al. Clinical outcomes, molecular epidemiology and resistance mechanisms of multidrug-resistant Pseudomonas aeruginosa isolated from bloodstream infections from Qatar. Ann. Med. 2021, 53, 2345–2353.
  55. Garazzino, S.; Altieri, E.; Silvestro, E.; Pruccoli, G.; Scolfaro, C.; Bignamini, E. Ceftolozane/Tazobactam for Treating Children With Exacerbations of Cystic Fibrosis Due to Pseudomonas aeruginosa: A Review of Available Data. Front. Pediatr. 2020, 8, 173.
  56. Pfaller, M.A.; Shortridge, D.; Harris, K.A.; Garrison, M.W.; DeRyke, C.A.; DePestel, D.D.; Moise, P.A.; Sader, H.S. Ceftolozane-tazobactam activity against clinical isolates of Pseudomonas aeruginosa from ICU patients with pneumonia: United States, 2015–2018. Int. J. Infect. Dis. IJID Off. Publ. Int. Soc. Infect. Dis. 2021, 112, 321–326.
  57. Nichols, W.W.; Stone, G.G.; Newell, P.; Broadhurst, H.; Wardman, A.; MacPherson, M.; Yates, K.; Riccobene, T.; Critchley, I.A.; Das, S. Ceftazidime-Avibactam Susceptibility Breakpoints against Enterobacteriaceae and Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2018, 62, e02590-17.
  58. Teo, J.Q.-M.; Lim, J.C.; Tang, C.Y.; Lee, S.J.-Y.; Tan, S.H.; Sim, J.H.-C.; Ong, R.T.-H.; Kwa, A.L.-H. Ceftolozane/Tazobactam Resistance and Mechanisms in Carbapenem-Nonsusceptible Pseudomonas aeruginosa. mSphere 2021, 6, e01026-20.
  59. Bassetti, M.; Echols, R.; Matsunaga, Y.; Ariyasu, M.; Doi, Y.; Ferrer, R.; Lodise, T.P.; Naas, T.; Niki, Y.; Paterson, D.L.; et al. Efficacy and safety of cefiderocol or best available therapy for the treatment of serious infections caused by carbapenem-resistant Gram-negative bacteria (CREDIBLE-CR): A randomised, open-label, multicentre, pathogen-focused, descriptive, phase 3 trial. Lancet Infect. Dis. 2021, 21, 226–240.
  60. Motsch, J.; Murta de Oliveira, C.; Stus, V.; Köksal, 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. Off. Publ. Infect. Dis. Soc. Am. 2020, 70, 1799–1808.
  61. 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. Off. Publ. Infect. Dis. Soc. Am. 2020, 71, 304–310.
  62. Karlowsky, J.A.; Kazmierczak, K.M.; de Jonge, B.L.M.; Hackel, M.A.; Sahm, D.F.; Bradford, P.A. In Vitro Activity of Aztreonam-Avibactam against Enterobacteriaceae and Pseudomonas aeruginosa Isolated by Clinical Laboratories in 40 Countries from 2012 to 2015. Antimicrob. Agents Chemother. 2017, 61, e00472-17. Available online: https://pubmed.ncbi.nlm.nih.gov/28630192/ (accessed on 22 September 2022).
  63. Lee, M.; Abbey, T.; Biagi, M.; Wenzler, E. Activity of aztreonam in combination with ceftazidime–avibactam against serine- and metallo-β-lactamase–producing Pseudomonas aeruginosa. Diagn. Microbiol. Infect. Dis. 2021, 99, 115227. Available online: https://www.sciencedirect.com/science/article/pii/S0732889320306040 (accessed on 22 September 2022).
  64. Zhanel, G.G.; Golden, A.R.; Zelenitsky, S.; Wiebe, K.; Lawrence, C.K.; Adam, H.J.; Idowu, T.; Domalaon, R.; Schweizer, F.; Zhanel, M.A.; et al. Cefiderocol: A Siderophore Cephalosporin with Activity Against Carbapenem-Resistant and Multidrug-Resistant Gram-Negative Bacilli. Drugs 2019, 79, 271–289. Available online: https://link.springer.com/article/10.1007/s40265-019-1055-2 (accessed on 7 September 2022).
  65. Kallel, H.; Bahloul, M.; Hergafi, L.; Akrout, M.; Ketata, W.; Chelly, H.; Hamida, C.B.; Rekik, N.; Hammami, A.; Bouaziz, M. Colistin as a salvage therapy for nosocomial infections caused by multidrug-resistant bacteria in the ICU. Int. J. Antimicrob. Agents 2006, 28, 366–369.
  66. Hanberger, H.; Giske, C.G.; Ciamarellou, H. When and How to Cover for Resistant Gram-Negative Bacilli in Severe Sepsis and Septic Shock. Curr. Infect. Dis. Rep. 2011, 13, 416. Available online: https://link.springer.com/article/10.1007/s11908-011-0200-1 (accessed on 29 September 2022).
  67. Pontikis, K.; Karaiskos, I.; Bastani, S.; Dimopoulos, G.; Kalogirou, M.; Katsiari, M.; Oikonomou, A.; Poulakou, G.; Roilides, E.; Giamarellou, H. Outcomes of critically ill intensive care unit patients treated with fosfomycin for infections due to pandrug-resistant and extensively drug-resistant carbapenemase-producing Gram-negative bacteria. Int. J. Antimicrob. Agents 2014, 43, 52–59.
  68. Tschudin-Sutter, S.; Fosse, N.; Frei, R.; Widmer, A.F. Combination therapy for treatment of Pseudomonas aeruginosa bloodstream infections. PLoS ONE 2018, 13, e0203295.
  69. Garnacho-Montero, J.; Sa-Borges, M.; Sole-Violan, J.; Barcenilla, F.; Escoresca-Ortega, A.; Ochoa, M.; Cayuela, A.; Rello, J. Optimal management therapy for Pseudomonas aeruginosa ventilator-associated pneumonia: An observational, multicenter study comparing monotherapy with combination antibiotic therapy. Crit. Care Med. 2007, 35, 1888–1895.
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: 495
Revisions: 2 times (View History)
Update Date: 03 Nov 2022
1000/1000
Video Production Service