ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Principles of antimicrobial therapy of Pseudomonas aeruginosa infections

Principles of antimicrobial therapy of Pseudomonas aeruginosa infections
Literature review current through: Jan 2024.
This topic last updated: Feb 06, 2023.

INTRODUCTION — Pseudomonas aeruginosa is a key gram-negative aerobic bacillus in the differential diagnosis of a number of infections. This organism is important because it is often antibiotic resistant and can cause severe hospital-acquired infections associated with a high mortality rate, especially in immunocompromised hosts.

The principles of antimicrobial treatment of infections caused by P. aeruginosa will be reviewed here. Discussion on the epidemiology, pathogenesis, clinical manifestations, diagnosis, and treatment of specific types of pseudomonal infections can be found elsewhere:

(See "Epidemiology, microbiology, and pathogenesis of Pseudomonas aeruginosa infection".)

(See "Pseudomonas aeruginosa pneumonia".)

(See "Pseudomonas aeruginosa bacteremia and endocarditis".)

(See "Pseudomonas aeruginosa skin and soft tissue infections".)

(See "Pseudomonas aeruginosa infections of the eye, ear, urinary tract, gastrointestinal tract, and central nervous system".)

Treatment issues related to P. aeruginosa lung infection in patients with cystic fibrosis are also discussed separately. (See "Cystic fibrosis: Antibiotic therapy for chronic pulmonary infection", section on 'Pseudomonas aeruginosa' and "Cystic fibrosis: Antibiotic therapy for pulmonary exacerbations".)

GENERAL PRINCIPLES OF TREATMENT — The following principles apply to the management of P. aeruginosa infections:

The risk of antibiotic resistance, both intrinsic and acquired, is an important consideration when selecting empiric or directed therapy. (See 'Antimicrobial resistance' below.)

Monotherapy is generally adequate, although combination therapy is indicated in certain high-risk patients and in severe infections. (See 'Role of combination antimicrobial therapy' below.)

Prompt initiation of antimicrobial therapy is important, as delayed therapy correlates with increased mortality.

Source control is important. For example, infected catheters and other implanted devices should be removed, abscesses should be drained, and obstructions should be relieved.

ANTIMICROBIAL RESISTANCE — P. aeruginosa is intrinsically resistant to numerous antibiotics and can acquire resistance to other agents during therapy. Some strains are multidrug resistant (ie, they are resistant to three or more classes of antibiotics) [1]. These features limit the choices of antibiotics for P. aeruginosa.

Definitions — A joint initiative by the European Centre for Disease Prevention and Control (ECDC) and the United States Centers for Disease Control and Prevention (CDC) in 2011 proposed specific definitions for characterizing drug resistance in organisms that cause many health care-associated infections [2]. The following definitions were established, based on the extent of resistance to antibiotic classes to which Pseudomonas does not have intrinsic resistance (table 1):

Multidrug-resistant – Isolate is non-susceptible to at least one agent in three or more antibiotic classes.

Extensively drug-resistant – Isolate is non-susceptible to at least one agent in all but two or fewer antibiotic classes.

Pandrug-resistant – Isolate is non-susceptible to all agents.

Epidemiology of resistance — The global burden of antimicrobial resistance in clinical isolates of P. aeruginosa is high. However, national surveillance data should be interpreted with caution because local resistance rates are highly variable and closely linked with local antimicrobial usage patterns and patient type.

Data collected from over 4500 hospitals in the United States' National Healthcare Safety Network from 2011 to 2014 revealed the following rates of multidrug resistance among P. aeruginosa isolates [3]:

Ventilator-associated pneumonia – 20 percent

Central line-associated blood stream infection – 18 percent

Catheter-associated urinary tract infection – 18 percent

Surgical site infection – 4 percent

A subsequent study analyzing data from 890 hospitals in the United States reported that the rate of multidrug-resistant P. aeruginosa infections among hospitalized patients decreased by 30 percent between 2012 and 2017 (from 13.10 to 9.43 cases per 10,000 hospitalizations) [4].

Rates of resistance are higher in resource-limited than in resource-rich settings. In a 2010 to 2015 study of P. aeruginosa device-associated bacteremia isolates collected from over 700 intensive care units (ICUs) in 50 resource-limited countries, rates of resistance to amikacin and imipenem were 30 and 44 percent, respectively [5]. In contrast, corresponding resistance rates in ICUs in the United States were 10 and 26 percent.

Antimicrobial resistance among P. aeruginosa is associated with increased length of hospitalization and increased mortality [6]. Risk factors for infection with resistant P. aeruginosa isolates include the following [7-11]:

ICU stay

Bedridden status

Presence of invasive devices

Prior use of certain antibiotics, including broad-spectrum cephalosporins, aminoglycosides, carbapenems, fluoroquinolones

Diabetes mellitus

Surgery

Nosocomial outbreaks of resistant pseudomonal infections have been reported in healthcare facilities around the world [12-15]. Outbreaks have also been associated with contaminated medical devices, such as endoscopes and transesophageal probes [11,16]. In 2023, a nation-wide outbreak in the United States of extensively drug-resistant P. aeruginosa, resistant to all antipseudomonal agents including ceftolozane-tazobactam and ceftazidime-avibactam as well as polymixins, was found to be associated with contaminated over-the-counter artificial tears [10].

Prior use of carbapenem, including ertapenem (which does not have antipseudomonal activity), has been repeatedly identified as a risk factor for carbapenem resistance [17-19]. Additional risk factors include male gender, ICU stay, and urinary bladder catheterization longer than seven days.

Emergence of antibiotic resistance during therapy for P. aeruginosa infections, resulting in increased rates of morbidity and mortality and higher costs, is a well-known problem. As an example, in a study involving 3393 P. aeruginosa isolates from ICU and non-ICU settings in a tertiary-care center, standard antibiograms were less able to predict susceptibility as the duration of hospitalization increased [20]. Similarly, in an observational study, resistant strains of P. aeruginosa emerged in 10.2 percent of 271 cases during treatment with four individual antipseudomonal agents [21]. Ceftazidime was associated with the lowest risk of emergent resistance, and imipenem with the highest risk. Emergence of resistance during therapy is a challenge with newer agents that have activity against Pseudomonas; this issue is discussed elsewhere. (See 'Emergence of resistance to novel agents' below.)

Acquired resistance is mediated by various mechanisms, including degrading enzymes, reduced permeability, and active efflux via mutation or acquisition of exogenous resistance determinants [22,23]. (See "Epidemiology, microbiology, and pathogenesis of Pseudomonas aeruginosa infection".)

ANTIBIOTICS WITH ANTIPSEUDOMONAL ACTIVITY — A number of antimicrobial agents have activity against P. aeruginosa isolates (table 2).

Intravenous antibiotics

Dosing strategies — Due to high levels of intrinsic and acquired resistance among pseudomonal isolates, higher doses of traditional antibiotics are often required to achieve sufficient levels.

Some beta-lactams are often administered as extended infusions (eg, over two to four hours) to maximize the duration that drug concentrations exceed the minimum inhibitory concentration (MIC) of the organism [24-29]. Although clinical data are limited, administration of prolonged infusions for certain beta-lactams and carbapenems is appropriate, especially for critically ill patients or for isolates with elevated MICs to the agent. (See "Prolonged infusions of beta-lactam antibiotics".)

Dosing for antipseudomonal antibiotics is listed in the table (table 2).

Antibiotic options — Multiple antibiotics from different classes are available to treat pseudomonal infections. Certain first-line agents are preferred due to extensive clinical experience, clinical trial results, and antibiotic stewardship considerations.

First-line agents — The following antibiotics, grouped by class, can usually be used alone as single agents if in vitro testing shows they are susceptible. The selection of individual agents from this group depends on factors such as the site of the infection, local resistance rates of P. aeruginosa, prior culture results, the patient's history of allergies, and the agents' availability in the local hospital formulary.

Dosages for intermittent or prolonged infusion is outlined in the tables (table 2).

Antipseudomonal penicillins in combination with a beta-lactamase inhibitor include:

Piperacillin-tazobactam

Although piperacillin has antipseudomonal activity, the addition of tazobactam inhibits AmpC-type beta-lactamases and results in a lower MIC than piperacillin alone.

Ticarcillin-clavulanate (not available in the United States or Canada)

Cephalosporins with antipseudomonal activity include:

Ceftazidime

Cefoperazone (not available in the United States)

Cefepime

Monobactam:

Aztreonam

Fluoroquinolones:

Ciprofloxacin

Levofloxacin has no advantage over ciprofloxacin for infections due to P. aeruginosa since its additional spectrum of coverage is usually unnecessary and potentially harmful. Levofloxacin is primarily indicated for treatment of respiratory tract infections when additional empiric P. aeruginosa coverage is warranted and in rare situations such as a culture-positive polymicrobial infection that includes susceptible strains of streptococci and P. aeruginosa. We do not advise using other quinolone agents such as moxifloxacin for treatment of P. aeruginosa.

Carbapenems:

Meropenem

In vitro studies have shown that carbapenem MICs are lower with meropenem than with imipenem [30]. It is unknown whether this in vitro potency difference translates into any difference in clinical outcomes. However, meropenem is preferred over imipenem because imipenem has a higher propensity to induce resistance during treatment [21]. Nevertheless, all carbapenems have been associated with emergent resistance during therapy; thus we reserve their use for the treatment of P. aeruginosa infections resistant to other agents or in polymicrobial infections.

Role of aminoglycosides — Aminoglycosides (tobramycin, gentamicin, amikacin) are active against P. aeruginosa but are generally not used as single agents because of inadequate clinical efficacy at most sites. As examples, aminoglycosides should not be used as monotherapy for pneumonia because they perform poorly in an acidic environment, and aminoglycosides used as monotherapy for bacteremia are associated with high mortality rates [31]. Instead, aminoglycosides are frequently used in combination with other antibiotics for empiric therapy, pending susceptibility results or for the treatment of select serious infections.

Aminoglycosides can be used as a single agent for the treatment of lower urinary tract infections (eg, cystitis).

When using an aminoglycoside as part of therapy for an infection with a high risk of P. aeruginosa, we favor tobramycin over gentamicin, as it has greater intrinsic antipseudomonal activity; however, it may not be widely available.

The dosing of aminoglycosides is discussed in detail elsewhere. (See "Dosing and administration of parenteral aminoglycosides".)

Antibiotics reserved for multidrug-resistant isolates — Several novel agents have activity against P. aeruginosa, but are generally reserved for serious infections with isolates resistant to other agents because of cost and antibiotic stewardship concerns (and, for polymyxins, because of toxicity) (see 'Antibiotic selection' below). Doses are listed in the table (table 2).

Novel beta-lactam-beta-lactamase inhibitor combinations:

Ceftolozane-tazobactam [32,33]

Ceftazidime-avibactam [34-37]

Novel cephalosporin:

Cefiderocol [38-40]

Novel carbapenem-beta-lactamase combination:

Imipenem-cilastatin-relebactam

Polymyxins:

Polymyxin B

Colistin

Polymyxins are generally used as part of a combination regimen when treating Pseudomonas infection. Dosing of polymyxin B and colistin are discussed in detail elsewhere. (See "Polymyxins: An overview", section on 'Dosing and administration'.)

Other potential therapeutic options are still in development [41].

Oral antibiotics — Fluoroquinolones are the only antibiotic class available as an oral formulation that is reliably active against P. aeruginosa. As with other classes of antibiotics, higher doses of certain fluoroquinolones (eg, ciprofloxacin) are often used to treat Pseudomonas.

For infections due to P. aeruginosa alone, we suggest ciprofloxacin instead of levofloxacin because levofloxacin's additional spectrum of coverage is usually unnecessary; dosages are found in the table (table 2). Prulifloxacin (600 mg once daily) is another oral fluoroquinolone available in some European countries.

We suggest not using other quinolone agents (eg, moxifloxacin) for treatment of P. aeruginosa. We also suggest not using fosfomycin as a single agent due to increased risk of baseline resistance and rapid emergence of resistance while on therapy.

Alternative formulations — Certain antipseudomonal antibiotics are also available in inhaled, topical, intravitreal, intraventricular, and impregnated cement formulations for specific indications.

Inhaled antibiotics with activity against P. aeruginosa are sometimes used as an adjunct to treatment of pneumonia due to drug-resistant P. aeruginosa and to prevent exacerbations of bronchiectasis (eg, in the setting of cystic fibrosis) [42]. Inhaled administration has the theoretic advantage of targeting drug levels to bronchial secretions in patients with pneumonia while reducing potential systemic side effects. (See "Pseudomonas aeruginosa pneumonia" and "Cystic fibrosis: Antibiotic therapy for chronic pulmonary infection", section on 'Inhaled antibiotics' and "Bronchiectasis in adults: Maintaining lung health", section on 'Inhaled antibiotics'.)

Other antibiotic formulations are discussed elsewhere. (See "Bacterial endophthalmitis", section on 'Intravitreal antibiotics' and "Prosthetic joint infection: Treatment", section on 'Resection arthroplasty with reimplantation' and "Health care-associated meningitis and ventriculitis in adults: Treatment and prognosis", section on 'Intrathecal and intraventricular therapy'.)

ROLE OF COMBINATION ANTIMICROBIAL THERAPY — One of the most controversial management questions involves the use of combination versus monotherapy for serious infections due to P. aeruginosa. High-quality data informing the decision are lacking.

Empiric combination therapy — Combination therapy is used by many clinicians for empiric coverage of known or suspected pseudomonal infections, and is usually discontinued once susceptibility results become available.

Indications — We suggest the use of two agents from different classes with in vitro activity against P. aeruginosa for empiric treatment of serious infections known or suspected to be caused by P. aeruginosa when there is high risk of antimicrobial resistance or in hosts for whom inappropriate antibiotic therapy would likely be associated with an especially high mortality. Such circumstances include the following:

When signs of severe sepsis or septic shock are present

Neutropenic patients with bacteremia

Burn patients (who have a high incidence of multidrug-resistant P. aeruginosa infections) with serious infections

In other settings where the incidence of resistance to the chosen antibiotic class is high (eg, >10 to 15 percent)

In other circumstances, empiric treatment using only one antipseudomonal agent is appropriate.

Our rationale for using empiric combination therapy in the settings listed above is to increase the likelihood that at least one of the agents in the combination is active against the infecting strain of Pseudomonas. Further discussion of the rationale for combination therapy is discussed below. (See 'Rationale' below.)

Selection of agents — The choice of agents for empiric therapy depends on numerous factors, such as the site and severity of the infection, local resistance rates of P. aeruginosa, prior culture susceptibility results, allergies, and the availability of specific agents in the local hospital formulary.

If empiric combination therapy is used, two agents with different mechanisms of action should generally be chosen based on prior patient isolates and local resistance rates. In general, we suggest a beta-lactam as the first agent and an aminoglycoside as the second, provided there are no contraindications to the use of either. The role and selection of aminoglycosides is discussed above. (See 'Role of aminoglycosides' above.)

Aminoglycosides should be avoided in patients with renal insufficiency or in those hospitalized in institutions with a high percentage of P. aeruginosa resistant to aminoglycosides. In such cases, we use a fluoroquinolone as a second agent. One study suggested better outcomes when using a fluoroquinolone rather than an aminoglycoside as the second agent [43], but further data are warranted before routinely favoring a fluoroquinolone over an aminoglycoside in this setting.

Rationale — For empiric therapy, the rationale for using combination therapy is detailed below:

Ensure receipt of an active agent — The best rationale for the use of combination therapy is to provide initial broad spectrum of activity when there is risk for multidrug-resistant P. aeruginosa so that if the P. aeruginosa is resistant to one agent, it may be susceptible to the other. Delays in active antimicrobial therapy of P. aeruginosa infections have been associated with increased mortality, and the role of a second agent may thus be to cover possible resistant pathogens when resistance rates to the primary agent are high [1,44-48]. Delay in treatment has been linked to increased mortality even when a patient is considered clinically stable at the time of initial evaluation [49].

This is a similar rationale to that used to support empiric combination therapy for gram-negative bacillary bacteremia in other settings. (See "Gram-negative bacillary bacteremia in adults", section on 'Indications and rationale for combination therapy'.)

In one retrospective study of patients with ventilator-associated pneumonia due to P. aeruginosa, empiric combination therapy was associated with a higher likelihood that appropriate antibiotic therapy was initially used, although an impact on mortality was not demonstrated [50]. A multinational, retrospective study of 294 episodes of P. aeruginosa bacteremic pneumonia in neutropenic individuals found that appropriate empiric combination therapy was associated with improved overall survival (adjusted hazard ratio [aHR] 0.46; 95% CI 0.27-0.78) [51].

Possible synergistic activity — A commonly cited reason for use of combination therapy is the potential for synergistic activity against P. aeruginosa with two agents, which in turn may result in better outcomes than single-drug therapy. However, there is no compelling evidence that two agents offer improved survival outcomes for treating P. aeruginosa infections.

Data on the use of combination versus single-drug therapy for P. aeruginosa infections are mixed, but overall, the best current evidence suggests that there is no additional benefit of a second active agent [52-62].

In a meta-analysis of 64 randomized trials that compared beta-lactam monotherapy with combination therapy with a beta-lactam and an aminoglycoside for over 7500 immunocompetent patients with sepsis of all-cause (bacteremia did not need to be present), there was no survival advantage with combination therapy [60]. A lack of additional benefit was also observed in the subset of 426 patients with P. aeruginosa infection.

In a meta-analysis of two randomized trials and 15 observational studies that included patients with documented gram-negative bacteremia, many of whom were immunocompromised, combination therapy was not associated with a mortality benefit overall [61]. However, among the subgroup of patients with P. aeruginosa bacteremia, there was a mortality benefit associated with combination therapy (odds ratio [OR] 0.50; 95% CI 0.30-0.79). Of note, several of the studies in the meta-analysis used aminoglycosides as a single agent in the monotherapy group, which is considered inadequate therapy [63].

Observational studies have also not supported an association between empiric or definitive combination therapy (generally with a beta-lactam plus an aminoglycoside or fluoroquinolone) and improved mortality [62,64]. In one study of patients with P. aeruginosa bacteremia, definitive combination therapy was associated with reduced 30-day mortality when ciprofloxacin was the second agent, but not when tobramycin was the second agent [43].

For the most part, these results are in contrast to a frequently quoted retrospective study analyzing data collected more than 20 years ago, which concluded that combination therapy was associated with a reduced mortality rate compared with monotherapy for P. aeruginosa bacteremia (27 versus 47 percent) [52]. However, questions about the validity of these results have arisen because of a number of methodologic problems, including the lack of randomization, the choice of therapy (which may have been influenced by the severity of illness), and the observation that monotherapy in the majority of patients consisted of an aminoglycoside, which is associated with a mortality as high as 70 to 80 percent when used for the treatment of P. aeruginosa bacteremia [53].

Although not all studies showed clear evidence for a survival benefit with the use of combination therapy, it is possible that it offers other clinical benefits. As an example, studies comparing combination therapy with monotherapy in pulmonary infections in patients with cystic fibrosis have shown no differences in outcome, but the density of bacteria was lower in the sputum, and the time to the next pulmonary infection requiring hospitalization was more prolonged in patients receiving combination therapy [65,66].

Prevent emergent resistance — There is limited clinical evidence evaluating the ability of combination therapy to prevent the emergence of antimicrobial resistance in patients with P. aeruginosa bacteremia.

Emergence of resistance during therapy for P. aeruginosa infections can occur, resulting in increased rates of morbidity and mortality and higher costs. However, most studies of human P. aeruginosa infections have been underpowered to evaluate whether combination therapy can prevent emergent resistance. An observational study reported that resistant P. aeruginosa emerged in 10.2 percent of 271 cases during treatment with four individual antipseudomonal agents [21]. Ceftazidime was associated with the lowest risk and imipenem with the highest risk. Addition of an aminoglycoside did not alter this risk.

This is in contrast to animal and in vitro studies that have suggested that combination therapy may alter this risk. As an example, combination therapy with two antipseudomonal antimicrobial agents limited the risk of emergence of resistant pseudomonal strains compared with monotherapy in an animal model of pseudomonal peritonitis [67,68]. Additionally, an in vitro study suggested that levofloxacin and imipenem might be an effective combination for preventing the emergence of resistance during treatment of P. aeruginosa infections [69].

Drawbacks — The main drawbacks to combination therapy include additional cost and toxicity. As an example, in a meta-analysis of 64 randomized trials that compared beta-lactam monotherapy with combination therapy with a beta-lactam and an aminoglycoside for sepsis, combination therapy was associated with a significant increase in nephrotoxicity [60].

Directed combination therapy — Definitive therapy can be tailored to the results of susceptibility tests once they are available. Definitive therapy with a single active agent is appropriate for most infections, as there are no convincing clinical data that demonstrate a mortality benefit to combination therapy. (See 'Possible synergistic activity' above and 'Prevent emergent resistance' above and 'Drawbacks' above.)

Nevertheless, combination therapy is often used in situations in which the risk of emergent resistance or significant morbidity or mortality is high. For example, combination therapy is often used to treat P. aeruginosa endocarditis and bacteremia in high-risk hosts. (See "Pseudomonas aeruginosa bacteremia and endocarditis".)

Initiation of a second antipseudomonal agent for directed therapy may also be reasonable in infections that initially fail or are slow to respond to a single active agent, although there are few clinical, in vitro, or experimental data to support this practice.

In light of these uncertainties, we recommend consultation with an expert in treating such infections (when possible) if combination therapy is used for a multidrug-resistant organism. (See 'Management of multidrug-resistant organisms' below.)

MANAGEMENT OF MULTIDRUG-RESISTANT ORGANISMS — Infections due to multidrug-resistance P. aeruginosa are a growing clinical problem and should be managed with the assistance of an expert in the treatment of such infections.

Susceptibility testing — For infections due to P. aeruginosa resistant to first-line agents (including piperacillin, cephalosporins, and carbapenems), we check susceptibility to novel agents, including ceftazidime-avibactam, ceftolozane-tazobactam, cefiderocol, and imipenem-cilastatin-relebactam, as well as polymyxins (colistin or polymyxin B).

For patients previously infected with a highly resistant isolate who present with a sepsis-like picture, we suggest repeating antibiotic susceptibility testing for novel agents because emergence of resistance during therapy has been reported. (See 'Emergence of resistance to novel agents' below.)

Antibiotic selection — For infections due to P. aeruginosa resistant to first-line agents (including piperacillin, cephalosporins, and carbapenems), we suggest monotherapy with either ceftolozane-tazobactam or ceftazidime-avibactam if the isolate is susceptible to one of these. If neither of these can be used, cefiderocol or imipenem-cilastatin-relebactam are alternatives. Polymyxins are typically reserved for infections when there are no other options.

Our recommendations are largely consistent with the Infectious Diseases Society of America (IDSA) guidance on management of antimicrobial-resistant gram-negative infections, which also favor monotherapy if in vitro susceptibility to one of these agents is confirmed and list ceftolozane-tazobactam, ceftazidime-avibactam, and imipenem-cilastatin-relebactam as preferred treatment options for difficult-to-treat P. aeruginosa infections [70]. Cefiderocol is another preferred agent for urinary tract infections in the IDSA guidance, but for infections outside the urinary tract, it is an alternative agent reserved for patients who cannot use other options. European guidelines generally match IDSA guidance, except ceftolozane-tazobactam is the only preferred agent in the European guidelines, which consider the other regimens to have insufficient evidence [71].

Preferred agents — For infections resistant to first-line agents, we suggest one of the following antibiotics, if available. Dosages are listed in the table (table 2).

For patients who were recently treated with one of these agents and present with a sepsis-like syndrome, we suggest choosing a different novel agent, at least until culture and susceptibility data are available. If ceftolozane-tazobactam was used previously, we suggest also avoiding ceftazidime-avibactam due to cross-resistance between these agents. (See 'Emergence of resistance to novel agents' below.)

Ceftolozane-tazobactam – Several observational studies have described successful use of ceftolozane-tazobactam [72-75]. In a retrospective study of 200 patients with multidrug-resistant P. aeruginosa infections (52 percent with ventilator-associated pneumonia), receipt of ceftolozane-tazobactam was associated with higher rates of clinical cure (adjusted odds ratio [aOR] 2.6) and lower rates of acute kidney injury (aOR 0.8) compared with aminoglycoside or polymyxin-containing regimens; in-hospital mortality was no different [75]. In another study of 205 patients, ceftolozane-tazobactam was associated with clinical success in 74 percent; receiving ceftolozane-tazobactam earlier, within four days of culture results, was associated with better outcomes [72].

Ceftazidime-avibactam – In a pooled analysis of results from randomized trials of patients with ceftazidime-resistant infections, ceftazidime-avibactam resulted in comparable clinical cures compared with the best-available therapy (usually a carbapenem); among 56 patients with P. aeruginosa treated with ceftazidime-avibactam in this study, the microbiologic response rate was 57 percent [76].

There are no data comparing the two agents directly. Ceftolozane-tazobactam is generally regarded as having more potent activity [77], but the choice between them should depend on the susceptibility pattern of the infecting isolate, as there are regional differences in susceptibility to the two agents. In a study of 42 isolates of non-carbapenemase-producing, carbapenem-resistant P. aeruginosa isolates from South Korean hospitals, 95 percent were susceptible to ceftolozane-tazobactam compared with 71 percent to ceftazidime-avibactam [78]. In contrast, in a study of isolates from the Middle East, where many of the isolates produced class A, C, and D beta-lactamases (which are inhibited by avibactam but not tazobactam), susceptibility rates were slightly higher to ceftazidime-avibactam [79].

Alternative agents — If one of the preferred agents is not an option, cefiderocol and imipenem-cilastatin-relebactam are novel agents that are acceptable alternatives. There is even less clinical experience with these agents. Dosages are listed in the table (table 2).

Cefiderocol – Most carbapenem-resistant isolates remain susceptible to cefiderocol, a novel siderophore cephalosporin [10,80]. In a trial of patients with complicated urinary tract infection, cefiderocol resulted in similar clinical and microbiologic cure rates for the few infections caused by P. aeruginosa compared with imipenem; however, the study did not include carbapenem-resistant organisms [38]. The IDSA does not list cefiderocol as a preferred agent for infections outside of the urinary tract because it was associated with higher all-cause mortality compared with best available therapy (typically a polymyxin-containing regimen) for non-urinary tract infections caused by carbapenem-resistant bacteria; only 28 P. aeruginosa infections were included in the trial [40,81].

Imipenem-cilastatin-relebactam – The addition of relebactam to imipenem-cilastatin restores activity against some P. aeruginosa isolates that are resistant to imipenem. In a trial of patients with serious infection caused by imipenem-resistant organisms (70 percent of which were P. aeruginosa), there was a trend toward lower 28-day mortality rates with imipenem-cilastatin-relebactam compared with imipenem-cilastatin plus colistin [82].

Other agents — If the above antibiotics are not available, effective options are limited. Dosages are listed in the table (table 2).

Polymyxins – We only use polymyxins (colistin or polymyxin B) when no other options exist because of their potential for toxicity. If a polymyxin is used for therapy of infections other than cystitis, we typically use it with a second susceptible agent. For cystitis, colistin can be used as monotherapy, but polymyxin B should be avoided due to suboptimal urinary levels. Care must be taken when dosing colistin and polymyxin B, as formulations differ between United States and European products, and each has distinct dosing recommendations. (See "Polymyxins: An overview", section on 'Dosing and administration'.)

Polymyxins have been used with some success in observational studies and case series [83-89]. As an example, in a study of 22 patients with metallo-beta-lactamase-producing Pseudomonas infections who were treated with intravenous colistin, 67 percent had a partial or complete response [87]. However, two-thirds developed at least mild nephrotoxicity.

Aminoglycosides – We typically do not use these agents as monotherapy for infections other than cystitis. For uncomplicated blood stream infections for which source control has been achieved, we sometimes use aminoglycosides when no other options are possible due to resistance or lack of availability. Careful monitoring of dosing, drug levels, and side effects should be assured in these cases. More information regarding the role of aminoglycosides is discussed above. (See 'Role of aminoglycosides' above.)

Antibiotics that should not be used – Many novel antibiotics are not recommended for Pseudomonas infections due to high probability of resistance. These include fosfomycin, tigecycline, and eravacycline. Meropenem-vaborbactam does not provide additional activity beyond meropenem alone and is not active against meropenem-resistant isolates.

Adjunctive inhaled antibiotics for pneumonia — Although we do not routinely use inhaled antibiotics for the treatment of P. aeruginosa pneumonia susceptible for first-line agents, they may be effective as an adjunct to intravenous therapy in cases of infection due to multidrug-resistant strains. This is discussed in detail elsewhere. (See "Pseudomonas aeruginosa pneumonia", section on 'Inhaled antibiotics for selected patients'.)

Emergence of resistance to novel agents — Reports of emergence of resistance during or following therapy with novel agents are accumulating. The frequency of emergent resistance with these agents is difficult to ascertain due to limited data, most of which is extrapolated from treatment trials.

Small observational studies have reported resistance to ceftolozane-tazobactam in 50 to 100 percent of patients who had positive cultures after being treated with the agent for at least 72 hours [90,91]. Of additional concern, cross-resistance between ceftolozane-tazobactam and ceftazidime-avibactam occurs because of similar mechanisms of resistance; in one study of 28 patients treated with ceftolozane-tazobactam, ceftazidime-avibactam resistance occurred in 86 percent of isolates previously susceptible to the agent [91].

Reports of resistance after exposure to imipenem-relebactam or cefiderocol for pseudomonal infections are less common, but these agents are newer than ceftolozane-tazobactam and ceftazidime-avibactam. Concern is warranted for all novel agents until further data emerge.

Pandrug-resistant isolates — Isolates that are resistant to first-line agents, novel beta-lactam-beta-lactamase inhibitors, and cefiderocol present particular therapeutic challenges.

Combination therapy – For extensively resistant organisms, we use combination therapy with the agent that has the lowest MIC combined with either a sensitive aminoglycoside or polymyxin B, if possible [24].

For isolates resistant to all available antibiotics, some clinicians have used combinations of drugs that separately have little or no activity against the isolate [92,93]. There are minimal clinical data to support such combination therapy. If combination therapy is used for treatment of organisms with extreme or unusual multidrug resistance patterns, it should be done in consultation with an expert in treating such infections whenever possible.

In a single clinical series of 64 patients with nosocomial pulmonary infections due to a highly resistant P. aeruginosa susceptible only to colistin, treatment with the combination of cefepime and amikacin was associated with survival in 44 (69 percent) [94]. These agents were the least inactive antibiotics by MIC determination and had demonstrated synergy in vitro. Dual beta-lactam therapy [95] and combinations with fosfomycin [96-98] have also been described with some success.

Combination regimens of other common antibiotics have been found to have enhanced activity against multidrug-resistant P. aeruginosa in vitro, but overall clinical data are extremely limited [93,99-107]. The mechanisms for the enhanced activity are unknown for most combinations.

Investigational approaches

Phage therapy – Phage therapy has been evaluated for P. aeruginosa endovascular infections in case reports and animal studies [108,109]. The frequency and significance of acquired resistance to phage treatment are uncertain issues. (See "Pseudomonas aeruginosa bacteremia and endocarditis".)

Antibiotics in development – Murepavadin is a promising antibacterial agent with potent activity against carbapenem-resistant and colistin-resistant P. aeruginosa [110]. It is being evaluated for P. aeruginosa ventilator-associated pneumonia [111].

TREATMENT IN CHILDREN — The general principles of antipseudomonal therapy in children are similar to those in adults, with the main exception that antimicrobial dosing is weight based. Additionally, the use of fluoroquinolones in children warrants specific consideration.

There have been reports of tendon inflammation and/or rupture with fluoroquinolone antibiotics in all ages; risk may be increased with concurrent steroids and in solid organ transplant recipients. In some pediatric studies, an increased risk of reversible adverse events involving joints or surrounding tissues, often due to reports of arthralgia, has been observed [112,113]. However, no compelling published evidence supports the occurrence of sustained injury to developing bones or joints in children treated with available fluoroquinolone agents [114,115]. Use of a fluoroquinolone is a consideration for gram-negative infections caused by isolates susceptible to these agents. Specifically, this includes treatment for infections when caused by P. aeruginosa, including chronic suppurative otitis media, malignant otitis externa, bony infections associated with nail wound puncture, and complicated urinary tract infection. The risks and benefits should be considered if a fluoroquinolone is prescribed in a child younger than 18 years of age. (See "Chronic suppurative otitis media (CSOM): Treatment, complications, and prevention", section on 'Treatment failure' and "Malignant (necrotizing) external otitis" and "Pseudomonas aeruginosa skin and soft tissue infections", section on 'Infection following nail puncture' and "Urinary tract infections in infants older than one month and children less than two years: Acute management, imaging, and prognosis" and "Fluoroquinolones", section on 'Children'.)

SUMMARY AND RECOMMENDATIONS

Antibiotic resistancePseudomonas aeruginosa is intrinsically resistant to a number of antibiotics and can acquire resistance during therapy. These features limit the choices of antibiotics for P. aeruginosa. (See 'Antimicrobial resistance' above.)

Antibiotic dosing strategies – Certain agents require higher dosing when used for presumptive or known P. aeruginosa infections, and prolonged infusions optimizes pharmacokinetics for some agents (table 2). (See 'Dosing strategies' above and "Prolonged infusions of beta-lactam antibiotics".)

First-line antibiotics – Multiple antibiotics from different classes are available to treat pseudomonal infections. (See 'Antibiotics with antipseudomonal activity' above.)

Intravenous options – Intravenous options include piperacillin-tazobactam, ceftazidime, cefepime, aztreonam, fluoroquinolones, and carbapenems. Aminoglycosides are generally not used as single agents because of inadequate clinical efficacy at most sites. Doses are discussed in the table (table 2). (See 'Antibiotics with antipseudomonal activity' above.)

Oral options – Fluoroquinolones are the only class of antibiotics with an oral formulation that is reliably active against P. aeruginosa. Doses are discussed in the table (table 2). (See 'Oral antibiotics' above.)

Combination therapy – There is no convincing clinical evidence that using two active agents instead of one leads to improved outcomes. However, using two agents for empiric therapy may increase the likelihood that an active agent will be used for a potentially resistant organism, and this, in turn, is associated with better outcomes for serious infections. (See 'Role of combination antimicrobial therapy' above.)

For patients who have sepsis or septic shock, have neutropenia and bacteremia, have severe burns, or are in a setting where the incidence of resistance to the chosen antibiotic class is high (eg, >10 to 15 percent), we suggest empiric therapy with a combination of two antipseudomonal agents (Grade 2C). The two agents should be from different antibiotic classes (eg, a beta-lactam with an aminoglycoside or a quinolone). (See 'Indications' above and 'Selection of agents' above.)

For patients without any of these additional risk factors for mortality or resistant organisms, we suggest empiric treatment with a single antipseudomonal agent (Grade 2B).

Directed therapy – Once antimicrobial susceptibility data are available, we typically use a single active antipseudomonal agent. The rare exceptions when we might continue combination include neutropenia and bacteremia, endocarditis, failure to improve on therapy, and multidrug resistance. (See 'Directed combination therapy' above.)

Management of multidrug-resistant organisms – For P. aeruginosa isolates resistant to first-line agents (including carbapenems), we suggest ceftolozane-tazobactam or ceftazidime-avibactam, as long as the isolate is susceptible (Grade 2C). Multidrug-resistant P. aeruginosa should be managed with the assistance of an expert in the treatment of such infections. (See 'Management of multidrug-resistant organisms' above.)

Treatment of children – The general principles of antipseudomonal therapy in children are similar to those in adults, with the main exception that antimicrobial dosing is weight based. (See 'Treatment in children' above.)

  1. Kanj SS, Kanafani ZA. Current concepts in antimicrobial therapy against resistant gram-negative organisms: extended-spectrum beta-lactamase-producing Enterobacteriaceae, carbapenem-resistant Enterobacteriaceae, and multidrug-resistant Pseudomonas aeruginosa. Mayo Clin Proc 2011; 86:250.
  2. Magiorakos AP, Srinivasan A, Carey RB, 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 2012; 18:268.
  3. Weiner LM, Webb AK, Limbago B, et al. 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.
  4. Jernigan JA, Hatfield KM, Wolford H, et al. Multidrug-Resistant Bacterial Infections in U.S. Hospitalized Patients, 2012-2017. N Engl J Med 2020; 382:1309.
  5. Rosenthal VD, Al-Abdely HM, El-Kholy AA, 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.
  6. Thaden JT, Park LP, Maskarinec SA, et al. Increased mortality associated with bloodstream infections caused by Pseudomonas aeruginosa as compared to other bacteria: Results of a 13-year prospective cohort study. Antimicrob Agents Chemother 2017.
  7. Aloush V, Navon-Venezia S, Seigman-Igra Y, et al. Multidrug-resistant Pseudomonas aeruginosa: risk factors and clinical impact. Antimicrob Agents Chemother 2006; 50:43.
  8. Nakamura A, Miyake K, Misawa S, et al. Meropenem as predictive risk factor for isolation of multidrug-resistant Pseudomonas aeruginosa. J Hosp Infect 2013; 83:153.
  9. Tartof SY, Kuntz JL, Chen LH, et al. Development and Assessment of Risk Scores for Carbapenem and Extensive β-Lactam Resistance Among Adult Hospitalized Patients With Pseudomonas aeruginosa Infection. JAMA Netw Open 2018; 1:e183927.
  10. Health Alert Network. Outbreak of extensively drug-resistant Pseudomonas aeruginosa associated with artificial tears. Centers for Disease Control and Prevention. Feb 1, 2023. Available at: https://emergency.cdc.gov/han/2023/han00485.asp (Accessed on February 02, 2023).
  11. Seki M, Machida H, Yamagishi Y, et al. Nosocomial outbreak of multidrug-resistant Pseudomonas aeruginosa caused by damaged transesophageal echocardiogram probe used in cardiovascular surgical operations. J Infect Chemother 2013; 19:677.
  12. Pagani L, Colinon C, Migliavacca R, et al. Nosocomial outbreak caused by multidrug-resistant Pseudomonas aeruginosa producing IMP-13 metallo-beta-lactamase. J Clin Microbiol 2005; 43:3824.
  13. Catho G, Martischang R, Boroli F, et al. Outbreak of Pseudomonas aeruginosa producing VIM carbapenemase in an intensive care unit and its termination by implementation of waterless patient care. Crit Care 2021; 25:301.
  14. Mudau M, Jacobson R, Minenza N, et al. Outbreak of multi-drug resistant Pseudomonas aeruginosa bloodstream infection in the haematology unit of a South African Academic Hospital. PLoS One 2013; 8:e55985.
  15. Bilavsky E, Pfeffer I, Tarabeia J, et al. Outbreak of multidrug-resistant Pseudomonas aeruginosa infection following urodynamic studies traced to contaminated transducer. J Hosp Infect 2013; 83:344.
  16. Fraser TG, Reiner S, Malczynski M, et al. Multidrug-resistant Pseudomonas aeruginosa cholangitis after endoscopic retrograde cholangiopancreatography: failure of routine endoscope cultures to prevent an outbreak. Infect Control Hosp Epidemiol 2004; 25:856.
  17. Tuon FF, Gortz LW, Rocha JL. Risk factors for pan-resistant Pseudomonas aeruginosa bacteremia and the adequacy of antibiotic therapy. Braz J Infect Dis 2012; 16:351.
  18. Cohen MJ, Block CS, Moses AE, Nir-Paz R. Exposure to ertapenem is possibly associated with Pseudomonas aeruginosa antibiotic resistance. Clin Microbiol Infect 2014; 20:O188.
  19. Djordjevic Z, Folic M, Ruzic Zecevic D, et al. Risk factors for carbapenem-resistant Pseudomonas aeruginosa infection in a tertiary care hospital in Serbia. J Infect Dev Ctries 2013; 7:686.
  20. Anderson DJ, 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.
  21. Carmeli Y, Troillet N, Eliopoulos GM, Samore MH. Emergence of antibiotic-resistant Pseudomonas aeruginosa: comparison of risks associated with different antipseudomonal agents. Antimicrob Agents Chemother 1999; 43:1379.
  22. Endimiani A, Luzzaro F, Pini B, et al. Pseudomonas aeruginosa bloodstream infections: risk factors and treatment outcome related to expression of the PER-1 extended-spectrum beta-lactamase. BMC Infect Dis 2006; 6:52.
  23. Rossolini GM, Mantengoli E. Treatment and control of severe infections caused by multiresistant Pseudomonas aeruginosa. Clin Microbiol Infect 2005; 11 Suppl 4:17.
  24. Infectious Diseases Society of America. IDSA guidance on the treatment of antimicrobial-resistant gram-negative infections: Version 1.0. https://www.idsociety.org/practice-guideline/amr-guidance/#toc-1 (Accessed on July 06, 2022).
  25. Lodise TP Jr, Lomaestro B, Drusano GL. Piperacillin-tazobactam for Pseudomonas aeruginosa infection: clinical implications of an extended-infusion dosing strategy. Clin Infect Dis 2007; 44:357.
  26. Taccone FS, Cotton F, Roisin S, et al. Optimal meropenem concentrations to treat multidrug-resistant Pseudomonas aeruginosa septic shock. Antimicrob Agents Chemother 2012; 56:2129.
  27. Kuti JL, Dandekar PK, Nightingale CH, Nicolau DP. Use of Monte Carlo simulation to design an optimized pharmacodynamic dosing strategy for meropenem. J Clin Pharmacol 2003; 43:1116.
  28. Chastre J, Wunderink R, Prokocimer P, et al. Efficacy and safety of intravenous infusion of doripenem versus imipenem in ventilator-associated pneumonia: a multicenter, randomized study. Crit Care Med 2008; 36:1089.
  29. Falagas ME, Tansarli GS, Ikawa K, Vardakas KZ. Clinical outcomes with extended or continuous versus short-term intravenous infusion of carbapenems and piperacillin/tazobactam: a systematic review and meta-analysis. Clin Infect Dis 2013; 56:272.
  30. Pillar CM, Torres MK, Brown NP, et al. 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.
  31. Phe K, Bowers DR, Babic JT, Tam VH. Outcomes of empiric aminoglycoside monotherapy for Pseudomonas aeruginosa bacteremia. Diagn Microbiol Infect Dis 2019; 93:346.
  32. Wagenlehner FM, Umeh O, Steenbergen J, et al. 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.
  33. Solomkin J, Hershberger E, Miller B, 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 2015; 60:1462.
  34. Carmeli Y, Armstrong J, Laud PJ, et al. 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.
  35. Mazuski JE, Gasink LB, Armstrong J, et al. Efficacy and Safety of Ceftazidime-Avibactam Plus Metronidazole Versus Meropenem in the Treatment of Complicated Intra-abdominal Infection: Results From a Randomized, Controlled, Double-Blind, Phase 3 Program. Clin Infect Dis 2016; 62:1380.
  36. Wagenlehner FM, Sobel JD, Newell P, et al. Ceftazidime-avibactam Versus Doripenem for the Treatment of Complicated Urinary Tract Infections, Including Acute Pyelonephritis: RECAPTURE, a Phase 3 Randomized Trial Program. Clin Infect Dis 2016; 63:754.
  37. Torres A, Zhong N, Pachl J, et al. Ceftazidime-avibactam versus meropenem in nosocomial pneumonia, including ventilator-associated pneumonia (REPROVE): a randomised, double-blind, phase 3 non-inferiority trial. Lancet Infect Dis 2018; 18:285.
  38. Portsmouth S, van Veenhuyzen D, Echols R, et al. 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.
  39. United States National Institutes of Health. Clinical Study of S-649266 for the Treatment of Nosocomial Pneumonia Caused by Gram-negative Pathogens (APEKS-NP). https://clinicaltrials.gov/ct2/show/NCT03032380 (Accessed on January 07, 2020).
  40. Bassetti M, Echols R, Matsunaga Y, 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.
  41. Sader HS, Castanheira M, Mendes RE, et al. Antimicrobial Activity of High-Proportion Cefepime-Tazobactam (WCK 4282) against a Large Number of Gram-Negative Isolates Collected Worldwide in 2014. Antimicrob Agents Chemother 2017; 61.
  42. Máiz L, Girón RM, Olveira C, et al. Inhaled antibiotics for the treatment of chronic bronchopulmonary Pseudomonas aeruginosa infection in cystic fibrosis: systematic review of randomised controlled trials. Expert Opin Pharmacother 2013; 14:1135.
  43. Paulsson M, Granrot A, Ahl J, et al. Antimicrobial combination treatment including ciprofloxacin decreased the mortality rate of Pseudomonas aeruginosa bacteraemia: a retrospective cohort study. Eur J Clin Microbiol Infect Dis 2017.
  44. Tamma PD, Cosgrove SE, Maragakis LL. Combination therapy for treatment of infections with gram-negative bacteria. Clin Microbiol Rev 2012; 25:450.
  45. Kang CI, Kim SH, Kim HB, et al. 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.
  46. Micek ST, Lloyd AE, Ritchie DJ, et al. Pseudomonas aeruginosa bloodstream infection: importance of appropriate initial antimicrobial treatment. Antimicrob Agents Chemother 2005; 49:1306.
  47. Park SY, Park HJ, Moon SM, et al. Impact of adequate empirical combination therapy on mortality from bacteremic Pseudomonas aeruginosa pneumonia. BMC Infect Dis 2012; 12:308.
  48. Bowers DR, Liew YX, Lye DC, et al. Outcomes of appropriate empiric combination versus monotherapy for Pseudomonas aeruginosa bacteremia. Antimicrob Agents Chemother 2013; 57:1270.
  49. El Solh AA, Alhajhusain A. Update on the treatment of Pseudomonas aeruginosa pneumonia. J Antimicrob Chemother 2009; 64:229.
  50. Deconinck L, Meybeck A, Patoz P, et al. Impact of combination therapy and early de-escalation on outcome of ventilator-associated pneumonia caused by Pseudomonas aeruginosa. Infect Dis (Lond) 2017; 49:396.
  51. Albasanz-Puig A, Durà-Miralles X, Laporte-Amargós J, et al. Effect of Combination Antibiotic Empirical Therapy on Mortality in Neutropenic Cancer Patients with Pseudomonas aeruginosa Pneumonia. Microorganisms 2022; 10.
  52. Hilf M, Yu VL, Sharp J, et al. Antibiotic therapy for Pseudomonas aeruginosa bacteremia: outcome correlations in a prospective study of 200 patients. Am J Med 1989; 87:540.
  53. Bodey GP, Jadeja L, Elting L. Pseudomonas bacteremia. Retrospective analysis of 410 episodes. Arch Intern Med 1985; 145:1621.
  54. Chatzinikolaou I, Abi-Said D, Bodey GP, et al. Recent experience with Pseudomonas aeruginosa bacteremia in patients with cancer: Retrospective analysis of 245 episodes. Arch Intern Med 2000; 160:501.
  55. Kuikka A, Valtonen VV. Factors associated with improved outcome of Pseudomonas aeruginosa bacteremia in a Finnish university hospital. Eur J Clin Microbiol Infect Dis 1998; 17:701.
  56. Leibovici L, Paul M, Poznanski O, et al. Monotherapy versus beta-lactam-aminoglycoside combination treatment for gram-negative bacteremia: a prospective, observational study. Antimicrob Agents Chemother 1997; 41:1127.
  57. Siegman-Igra Y, Ravona R, Primerman H, Giladi M. Pseudomonas aeruginosa bacteremia: an analysis of 123 episodes, with particular emphasis on the effect of antibiotic therapy. Int J Infect Dis 1998; 2:211.
  58. Chamot E, Boffi El Amari E, Rohner P, Van Delden C. Effectiveness of combination antimicrobial therapy for Pseudomonas aeruginosa bacteremia. Antimicrob Agents Chemother 2003; 47:2756.
  59. Pizzo PA, Hathorn JW, Hiemenz J, et al. A randomized trial comparing ceftazidime alone with combination antibiotic therapy in cancer patients with fever and neutropenia. N Engl J Med 1986; 315:552.
  60. Paul M, Benuri-Silbiger I, Soares-Weiser K, Leibovici L. Beta lactam monotherapy versus beta lactam-aminoglycoside combination therapy for sepsis in immunocompetent patients: systematic review and meta-analysis of randomised trials. BMJ 2004; 328:668.
  61. Safdar N, Handelsman J, Maki DG. Does combination antimicrobial therapy reduce mortality in Gram-negative bacteraemia? A meta-analysis. Lancet Infect Dis 2004; 4:519.
  62. Vardakas KZ, Tansarli GS, Bliziotis IA, Falagas ME. 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.
  63. Paul M, Leibovici L. Combination antibiotic therapy for Pseudomonas aeruginosa bacteraemia. Lancet Infect Dis 2005; 5:192.
  64. Peña C, Suarez C, Ocampo-Sosa A, 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. Clin Infect Dis 2013; 57:208.
  65. McLaughlin FJ, Matthews WJ Jr, Strieder DJ, et al. Clinical and bacteriological responses to three antibiotic regimens for acute exacerbations of cystic fibrosis: ticarcillin-tobramycin, azlocillin-tobramycin, and azlocillin-placebo. J Infect Dis 1983; 147:559.
  66. Smith AL, Doershuk C, Goldmann D, et al. Comparison of a beta-lactam alone versus beta-lactam and an aminoglycoside for pulmonary exacerbation in cystic fibrosis. J Pediatr 1999; 134:413.
  67. Michéa-Hamzehpour M, Pechère JC, Marchou B, Auckenthaler R. Combination therapy: a way to limit emergence of resistance? Am J Med 1986; 80:138.
  68. Michéa-Hamzehpour M, Auckenthaler R, Regamey P, Pechère JC. Resistance occurring after fluoroquinolone therapy of experimental Pseudomonas aeruginosa peritonitis. Antimicrob Agents Chemother 1987; 31:1803.
  69. Lister PD, Wolter DJ. Levofloxacin-imipenem combination prevents the emergence of resistance among clinical isolates of Pseudomonas aeruginosa. Clin Infect Dis 2005; 40 Suppl 2:S105.
  70. Tamma PD, Aitken SL, Bonomo RA, et al. Infectious Diseases Society of America Guidance on the Treatment of Antimicrobial Resistant Gram-Negative Infections. https://www.idsociety.org/practice-guideline/amr-guidance/ (Accessed on October 12, 2020).
  71. Paul M, Carrara E, Retamar P, 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.
  72. Gallagher JC, Satlin MJ, Elabor A, et al. Ceftolozane-Tazobactam for the Treatment of Multidrug-Resistant Pseudomonas aeruginosa Infections: A Multicenter Study. Open Forum Infect Dis 2018; 5:ofy280.
  73. Haidar G, Philips NJ, Shields RK, et al. Ceftolozane-Tazobactam for the Treatment of Multidrug-Resistant Pseudomonas aeruginosa Infections: Clinical Effectiveness and Evolution of Resistance. Clin Infect Dis 2017; 65:110.
  74. Munita JM, Aitken SL, Miller WR, et al. Multicenter Evaluation of Ceftolozane/Tazobactam for Serious Infections Caused by Carbapenem-Resistant Pseudomonas aeruginosa. Clin Infect Dis 2017.
  75. Pogue JM, Kaye KS, Veve MP, et al. Ceftolozane/Tazobactam vs Polymyxin or Aminoglycoside-based Regimens for the Treatment of Drug-resistant Pseudomonas aeruginosa. Clin Infect Dis 2020; 71:304.
  76. Stone GG, Newell P, Gasink LB, et al. 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.
  77. Moise P, DeRyke CA, Gonzalez M, et al. 1603. Comparison of Ceftolozane/Tazobactam, Ceftazidime/Avibactam, and Meropenem/Vaborbactam Activity Against P. aeruginosa: A Multicenter Evaluation. Open Forum Infect Dis 2021; 7:S797.
  78. Wi YM, Greenwood-Quaintance KE, Schuetz AN, et al. Activity of Ceftolozane-Tazobactam against Carbapenem-Resistant, Non-Carbapenemase-Producing Pseudomonas aeruginosa and Associated Resistance Mechanisms. Antimicrob Agents Chemother 2018; 62.
  79. Sid Ahmed MA, Abdel Hadi H, Hassan AAI, et al. Evaluation of in vitro activity of ceftazidime/avibactam and ceftolozane/tazobactam against MDR Pseudomonas aeruginosa isolates from Qatar. J Antimicrob Chemother 2019; 74:3497.
  80. Kazmierczak KM, Tsuji M, Wise MG, et al. In vitro activity of cefiderocol, a siderophore cephalosporin, against a recent collection of clinically relevant carbapenem-non-susceptible Gram-negative bacilli, including serine carbapenemase- and metallo-β-lactamase-producing isolates (SIDERO-WT-2014 Study). Int J Antimicrob Agents 2019; 53:177.
  81. Shinongi Inc. FDA antimicrobial drugs advisory committee: Cefiderocol briefing document. Oct 16, 2019. https://www.fda.gov/media/131705/download (Accessed on October 12, 2020).
  82. Motsch J, Murta de Oliveira C, Stus V, 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.
  83. Furtado GH, d'Azevedo PA, Santos AF, et al. Intravenous polymyxin B for the treatment of nosocomial pneumonia caused by multidrug-resistant Pseudomonas aeruginosa. Int J Antimicrob Agents 2007; 30:315.
  84. Kallel H, Hergafi L, Bahloul M, et al. Safety and efficacy of colistin compared with imipenem in the treatment of ventilator-associated pneumonia: a matched case-control study. Intensive Care Med 2007; 33:1162.
  85. Rios FG, Luna CM, Maskin B, et al. Ventilator-associated pneumonia due to colistin susceptible-only microorganisms. Eur Respir J 2007; 30:307.
  86. Florescu DF, Qiu F, McCartan MA, et al. What is the efficacy and safety of colistin for the treatment of ventilator-associated pneumonia? A systematic review and meta-regression. Clin Infect Dis 2012; 54:670.
  87. Sabuda DM, Laupland K, Pitout J, et al. Utilization of colistin for treatment of multidrug-resistant Pseudomonas aeruginosa. Can J Infect Dis Med Microbiol 2008; 19:413.
  88. Levin AS, Barone AA, Penço J, et al. Intravenous colistin as therapy for nosocomial infections caused by multidrug-resistant Pseudomonas aeruginosa and Acinetobacter baumannii. Clin Infect Dis 1999; 28:1008.
  89. Linden PK, Kusne S, Coley K, et al. Use of parenteral colistin for the treatment of serious infection due to antimicrobial-resistant Pseudomonas aeruginosa. Clin Infect Dis 2003; 37:e154.
  90. Rubio AM, Kline EG, Jones CE, et al. In Vitro Susceptibility of Multidrug-Resistant Pseudomonas aeruginosa following Treatment-Emergent Resistance to Ceftolozane-Tazobactam. Antimicrob Agents Chemother 2021; 65.
  91. Tamma PD, Beisken S, Bergman Y, et al. Modifiable Risk Factors for the Emergence of Ceftolozane-tazobactam Resistance. Clin Infect Dis 2021; 73:e4599.
  92. Rahal JJ. Novel antibiotic combinations against infections with almost completely resistant Pseudomonas aeruginosa and Acinetobacter species. Clin Infect Dis 2006; 43 Suppl 2:S95.
  93. Kmeid JG, Youssef MM, Kanafani ZA, Kanj SS. Combination therapy for Gram-negative bacteria: what is the evidence? Expert Rev Anti Infect Ther 2013; 11:1355.
  94. Dubois V, Arpin C, Melon M, et al. Nosocomial outbreak due to a multiresistant strain of Pseudomonas aeruginosa P12: efficacy of cefepime-amikacin therapy and analysis of beta-lactam resistance. J Clin Microbiol 2001; 39:2072.
  95. Jiao Y, Moya B, Chen MJ, et al. Comparable Efficacy and Better Safety of Double β-Lactam Combination Therapy versus β‑Lactam plus Aminoglycoside in Gram-Negative Bacteria in Randomized, Controlled Trials. Antimicrob Agents Chemother 2019; 63.
  96. Pontikis K, Karaiskos I, Bastani S, et al. 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.
  97. Apisarnthanarak A, Mundy LM. Carbapenem-resistant Pseudomonas aeruginosa pneumonia with intermediate minimum inhibitory concentrations to doripenem: combination therapy with high-dose, 4-h infusion of doripenem plus fosfomycin versus intravenous colistin plus fosfomycin. Int J Antimicrob Agents 2012; 39:271.
  98. Dinh A, Salomon J, Bru JP, Bernard L. Fosfomycin: efficacy against infections caused by multidrug-resistant bacteria. Scand J Infect Dis 2012; 44:182.
  99. Zuravleff JJ, Yu VL, Yee RB. Ticarcillin-tobramycin-rifampin: in vitro synergy of the triplet combination against Pseudomonas aeruginosa. J Lab Clin Med 1983; 101:896.
  100. Urena MT, Barasoain I, Espinosa M, et al. Evaluation of different antibiotic actions combined with rifampicin. In vitro synergism against Pseudomonas and Proteus. Chemotherapy 1975; 21:82.
  101. Fish DN, Choi MK, Jung R. Synergic activity of cephalosporins plus fluoroquinolones against Pseudomonas aeruginosa with resistance to one or both drugs. J Antimicrob Chemother 2002; 50:1045.
  102. Gunderson BW, Ibrahim KH, Hovde LB, et al. Synergistic activity of colistin and ceftazidime against multiantibiotic-resistant Pseudomonas aeruginosa in an in vitro pharmacodynamic model. Antimicrob Agents Chemother 2003; 47:905.
  103. Saiman L, Chen Y, Gabriel PS, Knirsch C. Synergistic activities of macrolide antibiotics against Pseudomonas aeruginosa, Burkholderia cepacia, Stenotrophomonas maltophilia, and Alcaligenes xylosoxidans isolated from patients with cystic fibrosis. Antimicrob Agents Chemother 2002; 46:1105.
  104. Timurkaynak F, Can F, Azap OK, et al. In vitro activities of non-traditional antimicrobials alone or in combination against multidrug-resistant strains of Pseudomonas aeruginosa and Acinetobacter baumannii isolated from intensive care units. Int J Antimicrob Agents 2006; 27:224.
  105. Asuphon O, Montakantikul P, Houngsaitong J, et al. Optimizing intravenous fosfomycin dosing in combination with carbapenems for treatment of Pseudomonas aeruginosa infections in critically ill patients based on pharmacokinetic/pharmacodynamic (PK/PD) simulation. Int J Infect Dis 2016; 50:23.
  106. Lingscheid T, Tobudic S, Poeppl W, et al. In vitro activity of doripenem plus fosfomycin against drug-resistant clinical blood isolates. Pharmacology 2013; 91:214.
  107. Samonis G, Maraki S, Karageorgopoulos DE, et al. Synergy of fosfomycin with carbapenems, colistin, netilmicin, and tigecycline against multidrug-resistant Klebsiella pneumoniae, Escherichia coli, and Pseudomonas aeruginosa clinical isolates. Eur J Clin Microbiol Infect Dis 2012; 31:695.
  108. Oechslin F, Piccardi P, Mancini S, et al. Synergistic Interaction Between Phage Therapy and Antibiotics Clears Pseudomonas Aeruginosa Infection in Endocarditis and Reduces Virulence. J Infect Dis 2017; 215:703.
  109. Chan BK, Turner PE, Kim S, et al. Phage treatment of an aortic graft infected with Pseudomonas aeruginosa. Evol Med Public Health 2018; 2018:60.
  110. Locher HH, Chiquet P, Brun S, et al. P1836 Activity of murepavadin against colistin-resistant Pseudomonas aeruginosa clinical isolates. Abstract, 29th ECCMID, Amsterdam, Netherlands, April 2019. https://www.escmid.org/escmid_publications/escmid_elibrary/material/?mid=68529 (Accessed on January 07, 2020).
  111. United States National Institutes of Health. Pharmacokinetics, Safety and Efficacy of POL7080 in Patients With Ventilator Associated Pseudomonas Aeruginosa Pneumonia. https://clinicaltrials.gov/ct2/show/NCT02096328 (Accessed on January 07, 2020).
  112. Adefurin A, Sammons H, Jacqz-Aigrain E, Choonara I. Ciprofloxacin safety in paediatrics: a systematic review. Arch Dis Child 2011; 96:874.
  113. Noel GJ, Bradley JS, Kauffman RE, et al. Comparative safety profile of levofloxacin in 2523 children with a focus on four specific musculoskeletal disorders. Pediatr Infect Dis J 2007; 26:879.
  114. Jackson MA, Schutze GE, COMMITTEE ON INFECTIOUS DISEASES. The Use of Systemic and Topical Fluoroquinolones. Pediatrics 2016; 138.
  115. American Academy of Pediatrics. Antimicrobial agents and related therapy. In: Red Book: 2018-2021 Report of the Committee on Infectious Diseases, 31st ed, Kimberlin DW, Brady MT, Jackson MA, Long SS (Eds), American Academy of Pediatrics, Itasca, IL 2018. p.903.
Topic 3135 Version 41.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟