Next Article in Journal
Rapid Monitoring of Viable Genetically Modified Escherichia coli Using a Cell-Direct Quantitative PCR Method Combined with Propidium Monoazide Treatment
Next Article in Special Issue
Antibiotic Resistance Rates for Helicobacter pylori in Rural Arizona: A Molecular-Based Study
Previous Article in Journal
Gut Microbiome and Associated Metabolites Following Bariatric Surgery and Comparison to Healthy Controls
Previous Article in Special Issue
Antimicrobial Resistance Pattern, Pathogenicity and Molecular Properties of Hypervirulent Klebsiella pneumonia (hvKp) among Hospital-Acquired Infections in the Intensive Care Unit (ICU)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antimicrobial Stewardship on Patients with Neutropenia: A Narrative Review Commissioned by Microorganisms

1
Infectious Diseases Department, Hospital de Braga, 4710-243 Braga, Portugal
2
Pharmaceuticals Department, Hospital de Braga, 4710-243 Braga, Portugal
3
Infectious Diseases Department, Centro Hospitalar do Tâmega e Sousa, 4564-007 Penafiel, Portugal
4
Infectious Diseases and Clinical Microbiology Department, Ankara Yıldırım Beyazıt University, 06760 Ankara, Turkey
5
Infectious Diseases Department, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Brazil
6
Clinical Research in Pneumonia & Sepsis (CRIPS), Vall d’Hebron Institute of Research (VHIR), 08035 Barcelona, Spain
7
FOREVA Research Pôle, Centre Hôpitalaire Universitaire de Nîmes, 30900 Nîmes, France
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(5), 1127; https://doi.org/10.3390/microorganisms11051127
Submission received: 27 March 2023 / Revised: 21 April 2023 / Accepted: 24 April 2023 / Published: 26 April 2023

Abstract

:

Highlights

  • “Right first time” and achieving source control is crucial to optimize antibiotic management in neutropenic patients.
  • The fear of missed pathogens and the risk of emergent resistant organisms require detailed knowledge of local patterns of susceptibility and a multidisciplinary team.
  • A consistent pattern of safety requires shortening duration of therapy.
  • Rapid diagnostic tools contribute to improve overall empiric antibiotic use, which should be a priority in neutropenic patients.
  • A 5D approach is a core strategy to ensure improving antibiotic use
  • Emphasis to a personalized prescription of antibiotics is required.

Abstract

The emergence of antibiotic resistance poses a global health threat. High-risk patients such as those with neutropenia are particularly vulnerable to opportunistic infections, sepsis, and multidrug-resistant infections, and clinical outcomes remain the primary concern. Antimicrobial stewardship (AMS) programs should mainly focus on optimizing antibiotic use, decreasing adverse effects, and improving patient outcomes. There is a limited number of published studies assessing the impact of AMS programs on patients with neutropenia, where early appropriate antibiotic choice can be the difference between life and death. This narrative review updates the current advances in strategies of AMS for bacterial infections among high-risk patients with neutropenia. Diagnosis, drug, dose, duration, and de-escalation (5D) are the core variables among AMS strategies. Altered volumes of distribution can make standard dose regimens inadequate, and developing skills towards a personalized approach represents a major advance in therapy. Intensivists should partner antibiotic stewardship programs to improve patient care. Assembling multidisciplinary teams with trained and dedicated professionals for AMS is a priority.

1. Introduction

According to the World Health Organization (Geneva, Switzerland), the overuse and misuse of antibiotics is reflected in the emergence of resistance, posing a threat to global health security. Infections with multidrug-resistant (MDR) microorganisms lead to increased mortality, need for medical assistance, and treatment-related cost.
Infections are the most common cause of death in patients with malignancy [1]. Unfortunately, the emergence of MDR pathogens threatens the efficacy of treatment and the survival of patients that rely on antibiotics for treatment success. Patients with neutropenia are particularly vulnerable to infections; the risk of infection increases with severity and duration of neutropenia [2,3]. The incidence of febrile neutropenia varies according to malignancy. Patients with hematological malignancies have around 80% incidence compared to 10–50% in those receiving neoplastic therapy for solid tumors [4].
Since its first use in 1996 by John E. McGowan Jr. and Dale N. Gerding [5], the term “Antimicrobial Stewardship” (AMS) has become increasingly common in recent years. The objective of this definition is to highlight antimicrobials as a precious and nonrenewable resource, using a concept that encompasses rational use, and moving away from the concept of cost containment, which had prevailed until then. AMS programs enhance patient care by improving the use of antibiotic therapy, decreasing adverse effects, and improving patient outcomes [6,7,8,9,10,11]. There is a limited number of studies published regarding AMS programs on patients with neutropenia. This narrative review updates the current strategies of AMS applied to high-risk patients with neutropenia for bacterial infections, and suggests a five-point approach for optimizing antibiotic therapy in high-risk patients with neutropenia.

2. The Epidemiologic Change

Infections in patients with neutropenia are increasingly challenging, considering recent changes in the resistance patterns of microorganisms. Prolonged neutropenia subsequently leads to the prescription of extended and repeated courses of broad-spectrum antibiotics, contributing to selective pressure on susceptible bacteria and the increase of MDR organisms.
Patients with severe immunosuppression have damaged intestinal mucosal surfaces, which facilitates bacterial translocation across intestinal mucosa, resulting in potentially life-threatening infections [12,13].
Patients with malignancy are susceptible to develop infection, particularly when colonized with MDR pathogens [14]. Isolates from bloodstream infections in patients with hematological malignancies from recent studies are reported in Table 1.
Catheter-related, lung, and skin and soft tissue infections are among the most reported [15,16,17]. Gram-positive infections represent an important burden in these patients, particularly staphylococci [18]. Methicillin-resistance is common among coagulase-negative staphylococci and variable for S. aureus (19.2–80%) [15,17,19]; additionally, vancomycin intermediate S. aureus (VISA) and vancomycin resistant Enterococcus spp. may increase rates of treatment failure [20,21]. Cattaneo et al. reported a higher incidence of Gram-positive bloodstream infections during the induction phase of chemotherapy (where catheter-related infections may play a larger role), whereas Gram-negative infections were more common during the consolidation phase [15]. Moreover, enterococcal infections were more frequent in patients receiving antibiotic prophylaxis (9.7% vs. 2.6%, p = 0.016) [15]—a pathogen which is not adequately covered by quinolones, a class of antibiotics commonly used as prophylaxis. Infections due to penicillin-resistant Streptococcus pneumoniae have also been associated with higher mortality [22].
Of particular concern is the increasing number of invasive Gram-negative bacilli infections, which are associated with higher mortality rates [23]. Importantly, 30-day mortality is higher for MDR Gram-negative infections across studies, even when compared to infections due to resistant Gram-positive bacteria (such as MRSA and VRE) [15,17]. Beta-lactamase-producing Enterobacterales (ESBL-E) and MDR Pseudomonas aeruginosa are increasing; carbapenem-resistant Acinetobacter baumanii and carbapenemase-producing Enterobacterales (CRE) are spreading globally, with some centers reporting carbapenem-resistance rates of 10% in Enterobacterales, and up to 79% in nonfermenters [17,19].
Table 1. Epidemiology and outcomes of bloodstream infections in patients with hematological malignancies in European centers.
Table 1. Epidemiology and outcomes of bloodstream infections in patients with hematological malignancies in European centers.
ReferenceCountryYearsStudy TypePopulationPathogensResistance Type30-Day Mortality
Cattaneo C et al., 2016 [15]Italy2012–2014Prospective, multicentricALL and AML (n = 239, 433 BSI episodes)GPB 44.8%: CoNS 25.4%, S. aureus 4.4%, Enterococcus spp. 2.6%Methicillin-resistance: CoNS 77.6%, MRSA 80%8.5%; higher for CRE + MDR-Pa: 29.6 vs. 6.1% (p < 0.01)
GNB 38.4%: Enterobacterales 28.1%, P. aeruginosa 10.5%,
Polymicrobial 15.7%, fungi 1.1%
ESBL-E 23.2%
During
Induction phase: GBP 50.9%, GNB 31.9%, polymicrobial 13.8%, fungi 3.4%
CRE 9%
During consolidation phase: GPB 38.9%, GNB 46.8%, polymicrobial 14.3%, fungi 0%MDR-Pa 27%
Stoma I et al., 2016 [19]Belarus2013–2015Prospective, single centerHSCT (n = 360, 135 BSI episodes)GPB 34.9%: S. aureus 17%, CoNS 5.2%, Enterococci 3%Methicillin-resistance: CoNS 27.8%, MRSA 65.2%31.1%; higher for carbapenem-resistant nonfermenters (OR 5.46 (95% CI 1.33–20.7), p = 0.0126)
GNB 64.4%: Enterobacterales 43.7%, Nonfermenters 21.5%ESBL-E 40.7%, CRE 11.9%, Carbapenem-resistant nonfermenters 79.3%
Scheich S et al., 2018 [16]Germany2008–2016Retrospective, single centerHM w/GNB BSI (n = 109)Only Gram-negative episodes evaluated:
Enterobacterales 73.4%; Nonfermenters 26.6%
MDR pathogens * 19.4%: P. aeruginosa 37.5%, E. coli 34.4%, K. pneumoniae 25%23.2%; higher for MDR vs. non-MDR GNB: 44.1% vs. 14.4% (p < 0.001)
Ali R et al., 2020 [17]Turkey2006–2016Retrospective, single centerHM (n = 552, 950 BSI episodes)GPB 48.3%:
CoNS 37.8%, S. aureus 2.5%, Enterococcus spp. 5%
Methicillin-resistance: CoNS 84.6%, MRSA 19.2%17.7%; higher for MDR-GNB infections: ESBL: 27.5% vs. 11.7% (p < 0.01), carbapenem-resistance: 65.4% vs. 14.8% (p < 0.01), MDR: 68.2% vs. 20.4% (p < 0.01)
VRE 21.6%
GNB 42.4%: E. coli 19.4%, Klebsiella spp. 11.8%, Pseudomonas spp. 5.5%, Acinetobacter baumanii 3.5%ESBL-E 39.4%
CRE 9.8%
Carbapenem-resistant nonfermenters 32.6%
Weber S et al., 2021 [24]Germany2006–2019Retrospective, single centerHM, other hematological disorders, SOT (n = 391, 637 BSI episodes)Common skin contaminants (coagulase-negative Staphylococcus spp., Bacillus spp., Corynebacterium spp., Cutibacterium spp., and Micrococcus spp.) 24.8%;
Escherichia spp. 19%; Enterococcus spp. 13%
VRE 10%Higher for carbapenem-resistant GNB infections: 62.5% vs. 4.7–18.7%
MDR GNB 6.8%
Carbapenem-resistant GNB 2.5%
Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myelogenous leukemia; BSI, bloodstream infection; CoNS, coagulase-negative staphylococci; CRE, carbapenem-resistant Enterobacterales; ESBL-E, extended spectrum beta-lactamase-producing Enterobacterales; GNB, Gram-negative bacteria; GPB, Gram-positive bacteria; HM, hematological malignancy; HSCT, hematopoietic stem cell transplantation; MDR, multidrug-resistant; MDR-Pa, multidrug-resistant Pseudomonas aeruginosa; SOT, solid organ transplant. * MDR was defined as resistance against at least three out of four antibiotic classes.

3. Antimicrobial Stewardship Programs in Patients with Neutropenia

AMS programs have several objectives, such as cost reduction, therapeutic optimization, and particularly, promoting actions that ultimately contribute to decreased antibiotic resistance rates [7].
In 2007, the Infectious Diseases Society of America (IDSA) defined AMS as ‘coordinated interventions designed to improve and measure the appropriate use of antimicrobial agents by promoting the selection of the optimal antimicrobial drug regimen including dosing, duration of therapy and route of administration’ [25]. In 2017, Dyar et al. [26] suggested that the term AMS could be defined as “A coherent set of actions which promote using antimicrobials responsibly”.
The literature on AMS in patients with malignancy is scarce by limited representation or exclusion of these patients in most studies, even though these patients are more vulnerable to MDR infections, and are more often exposed to broad-spectrum antibiotics. Antibiotic optimization is crucial to reduce morbidity and mortality rates in these patients. AMS general interventions such as formulary review, antibiotic restriction, and audit and feedback are effective in patients with malignancy [27,28,29,30].
A survey applied on solid-organ transplant centers in Switzerland exemplifies the lack of AMS implementation in immunosuppressed patients, with only a 29% response [31]. On the other hand, around 70% of intensive care units have an AMS program [32].
Most hospitals do not have dedicated staff working in antimicrobial stewardship [25,33], representing a gap that needs to be fulfilled. Health professionals working in this field must be well-trained and empowered to implement stewardship interventions on their hospitals. Therapeutic optimization is only achieved when a multidisciplinary team is involved. Multidisciplinary teams with infectious disease specialists, microbiologists, intensivists, oncologists, hematologists, pharmacists, and other front-line professionals are essential. It is important to involve nurses and pharmacists in strategies aiming to reduce unnecessary use of antibiotics [5,9]. AMS programs have a major role in minimizing the impact of multidrug-resistant bacteria, based on the implementation of a more conscious antibiotic prescription (e.g., according to local epidemiology, narrow spectrum, patient history) and the isolation of patients when in the presence of MDR bacteria.

4. AMS Strategies to Optimize Antibiotic Therapy

The emergence of resistance in pathogenic bacteria is a serious public health problem; still, prescribers misuse and over-use antibiotics [25,33,34,35,36]. It is difficult to change prescription behaviour; interventions such as antibiotic education, local clinical practice guidelines, audit and feedback, or antibiotic restriction have been shown to decrease antibiotic consumption in patients with febrile neutropenia without impacting the length of stay or mortality [27,28,29,30], and should be attempted. Which intervention to choose must be decided based on the local behaviour of prescribers.
This narrative review suggests a five-point approach for optimizing antibiotic therapy in high-risk patients with neutropenia (Table 2).

4.1. Diagnosis

The risk of development and the severity of infections are determined by a complex interplay between the pathogen and its virulence, the degree of impaired immunity, and of the host and the related cancer treatment.
Early diagnosis, at a stage when signs and symptoms may be absent and the site of infection may not be evident, is challenging. In patients with neutropenia undergoing chemotherapy, fever may be the only symptom that indicates bloodstream infection, which can result in severe sepsis, septic shock, and death [2,37].

4.1.1. Risk Assessment

Antibiotic decision is based on risk assessment, and high-risk patients, with prolonged (>7 days duration), profound (<100 cells/mm3) neutropenia and/or comorbidities, should receive IV broad-spectrum empirical antibiotic treatment [37,38,39,40,41]. The risk assessment should also account for site of infection, clinical manifestations (such as hypotension), local epidemiological data, and history of previous infection/colonization by MDR microorganisms or previous antimicrobial use. Identifying pathogen carriage has an important role. Several studies report > 30% of any multidrug-resistant pathogen colonization in patients with hematological malignancies [16,42,43,44]; epidemiology varies between countries and centers. For instance, in an American study of 312 patients with acute myeloid leukemia, of which 40.9% were colonized with MDR organisms, 74.4% were vancomycin-resistant Enterococci (VRE), while ESBL-E and CRE represented 20% and 13.3%, respectively [42]. Conversely, in a Spanish study of 250 patients with hematological malignancies, of which 3.7% were colonized, most isolates were Gram-negative bacteria (89.9%), of which ESBL-E, carbapenem-resistant Klebsiella spp., and MDR Pseudomonas aeruginosa represented 24.1%, 0.7%, and 24.8%, respectively; Gram-positive bacteria accounted for 10.1% of isolates [44].
Importantly, some studies reported a considerable concordance between BSI isolates and colonization, as high as 80% [15,44]. Micozzi et al. compared the outcomes of BSI in patients with hematological malignancies who were KPC carriers between two periods (2012–2013 vs. 2017–2018): 68% of 27 patients in the first period developed BSI compared to 11% of 88 patients in the second period, following the use of empirical active antibiotic therapy against KPC (namely, ceftazidime–avibactam ± tigecycline and/or gentamycin) at the onset of febrile neutropenia. Similarly, inhospital mortality decreased (50% vs. 6%, p < 0.01) [45].
Screening for MDR organisms, particularly CRE and MRSA, as part of institutional policy provides early identification of carriers, and may help guide empirical antibiotic therapy decisions during febrile neutropenia episodes. The appropriate methodology and frequency of screening should be guided by local epidemiology, laboratory turnaround time, testing availability, and cost-effectiveness [46,47].

4.1.2. Diagnostic Tools

When clinical diagnosis is difficult and complex, there is an increased risk of antimicrobial overuse. In this sense, the microbiology laboratory has a major role on AMS providing aetiological diagnosis. Morency-Potvin et al. highlighted the role of microbiology laboratories based on AMS strategies [48]. A close collaboration between microbiologist and the physician is the basis for a successful AMS implementation. Cumulative antimicrobial susceptibility reports, cascade or selective reporting, additional messages to enhance microbiology reports, rapid diagnostic testing, and rapid antimicrobial susceptibility testing are examples of diagnostic antimicrobial stewardship.

4.2. Conventional Microbiology

Conventional microbiology tests are the gold standard on diagnosis, yet may require up to 24 h for pathogen identification, and further 24–48 h for antibiotic susceptibility profiles. Timely identification and susceptibility results are the cornerstone for antibiotic optimization. Prescribers rely on susceptibility results; selective antimicrobial susceptibility reports should be used to promote the use of narrower-spectrum antibiotics [6,48].

4.3. Biomarkers

Biomarkers may help identify bacterial infections, limiting unnecessary antibiotic use. C-reactive protein and procalcitonin have been investigated as potential biomarkers for sepsis, considering that levels are increased in bacterial sepsis, but these should not be used alone to decide the diagnosis of bacterial infection [49,50,51]. Other biomarkers, such as proadrenomoduline, might add value to the diagnosis process to differentiate bacterial infection from a nonbacterial inflammatory response. The development of new therapies such, as T-cell CARS and TIL [52], emphasizes the importance of differentiating between the cytokine storm and true bacterial infection.

4.4. Rapid Diagnostic Testing and Rapid Antimicrobial Susceptibility Testing

Currently, the focus is on the use of rapid tests; results are obtained in a few hours with microorganism identification and susceptibility tests. Matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry, usually used to identify pathogens [53], can be used for phenotypic resistance profiles, such as beta-lactam-resistance, from positive blood cultures [54]. VITEK-2 is an automated system for identification in around 3 h, and 18 h for susceptibility testing [55].
Other automated systems using a modified fluorescent in situ hybridization (FISH) assay for identification and microscopy for analysing bacterial growth rates and minimum inhibitory concentration (MIC) values extrapolated from positive blood cultures can provide results in <7 h [56].
Rapid phenotypic antimicrobial susceptibility testing (AST) is a recent EUCAST (European Committee on Antimicrobial Susceptibility Testing) method with an impact on AMS. Rapid AST improves antibiotic optimization in patients with Gram-negative sepsis without mortality differences [57]. Kim et al. [58] represented hematological patients in a randomized control trial, and MDR bacteremia had early optimal target antimicrobial therapy.
The constant change in the incidence of multidrug-resistance Gram-negative bacteria in patients with neutropenia requires rapid diagnostic tests for resistance identification; extended-spectrum B-lactamases and carbapenemases rapid identification, based on biochemical or fluorescence identification, have high sensitivities and specificities, are easy to perform, and provide results in 10–40 min [59,60]; prescribers will optimize antibiotics while waiting for the susceptibility test report.

4.5. Molecular Biology

Novel molecular assays for rapid diagnosis directly from blood samples may provide faster results for timely and pathogen-directed antibiotic initiation. Multiplex real-time PCR assays show high specificity, while sensitivity may vary (20–90%) [61,62]. However, these PCR assays are limited by the primers’ availability. The BioFireFilmArray BCID2 assay identifies 33 organisms by multiplex PCR from positive blood cultures in one hour, and detects 10 resistance markers [63,64].
Next generation sequencing (NGS) approaches allow for nearly universal identification without a priori knowledge of pathogens, and have demonstrated improved pathogen detection in septic patients, surpassing blood cultures [65]. Furthermore, molecular tests have the potential to identify pathogens in patients with previous exposure to antibiotics, which may inhibit growth in blood cultures.
The major drawback is susceptibility testing. Molecular detection of resistance is possible for Gram-positive bacteria from positive blood cultures, such as mecA and vanA/vanB gene identification for methicillin-resistance in staphylococcal and vancomycin-resistance in enterococcal infections, respectively [66,67]. Identifying susceptibility patterns in Gram-negative bacteria, which may present with multiple mechanisms of resistance, are more complex, and phenotypic resistance is not as easily predicted by available assays. Commercial array-based kits for extended-spectrum beta-lactamase and carbapenemase detection from positive blood cultures demonstrated high accuracy, but sensitivity and specificity were lower when compared to conventional methods, and may miss less frequent, yet important resistance phenotypes [68]. Even with the development of NGS, phenotypic identification of resistance must be a priority [69].
Pathogen-directed antibiotic therapy may be achieved, anticipating de-escalation. Still, currently these tests only complement conventional culture-based methods.

4.6. Drug

Choosing the right antibiotic for the right patient requires multidisciplinary collaboration, and is based on multiple factors between the patient, the pathogen, and the drug.
The impact of inappropriate antibiotic therapy in critically ill patients is well established: higher mortality, increased duration of hospital stays, and risk of drug resistance [70,71,72,73]. Each hour delay in administration of effective antibiotic increases the risk of mortality [74,75,76,77,78]. Patients with febrile neutropenia require rapid administration of empiric antibiotics [78,79].
In patients with neutropenia, the choice of the appropriate antibiotic (i.e., an antibiotic with in vitro activity against the infective pathogen) is of major importance, considering that these patients may not have typical warning signs and symptoms of infection; thus, empirical therapy is mostly based on broad-spectrum antibiotics.
The ultimate choice is based on local factors (hospital susceptibility patterns) and patient factors (previous hospital admission, previous antibiotic exposure, infection/colonization past history). Facility-specific clinical practice guidelines based on local microbiological data establish clear recommendations for optimal antibiotic use at the hospital.
Monotherapy has an equivalent efficacy compared with combination therapy [80,81,82]. Initial empiric treatment should include monotherapy with antipseudomonal beta-lactams [41,83], such as cefepime (fourth generation cephalosporin), piperacillin/tazobactam, or a carbapenem (meropenem or imipenem/cilastatin).
Due to the emergence of multidrug-resistant Gram-negative bacilli, ESBL-E, CRE, or MDR Pseudomonas aeruginosa, broad-spectrum antibiotics are justified for most patients, particularly high-risk patients with neutropenia [84,85,86,87,88].
Empirical therapy (Table 3) on febrile neutropenia usually does not cover MRSA unless suspected, since a meta-analysis of randomized control trials did not show advantages with vancomycin [89,90]; Gram-positive coverage with vancomycin (or other equal spectrum antibiotics) is suggested in patients with neutropenia. MRSA coverage is also recommended in suspected catheter-related infection, skin, or soft tissue infection, pneumonia, or septic shock. An interesting retrospective study [91] with 1305 patients with febrile neutropenia concluded that the use of empiric vancomycin is unnecessary in the majority of cases and did not improve mortality. MRSA screening is an important tool in antimicrobial stewardship, reducing the spectrum of empirical therapy [92,93].
Preventing drug adverse effects is a priority in patient safety. Therapeutic reconciliation is an effective strategy in preventing adverse drug reactions [94]. Medication reconciliation allows for the creation of the best possible list of all patient medication, including dose, frequency, and route of administration, based on several sources of information. After a complete drug history, omissions, duplications, or inadequate doses are analysed to prevent related incidents. It must be implemented whenever therapeutic changes or transition of care occurs [94,95].
The absence of medication reconciliation is responsible for 46% of medication-related errors, and more than 20% of the adverse effects that occur in hospitals [94]. Polypharmacy in patients with malignancies makes antibiotic prescription a challenge; the implementation of therapeutic reconciliation must be part of a stewardship program.

4.7. Dose

Adequate dosing and administration based on pharmacokinetics/pharmacodynamics (PK/PD) should be a priority. PK/PD describes the relationship between drug dose and pharmacological effects, with changes in drug concentrations leading to different pharmacological effects.
Increased volume of distribution requires a loading dose on hydrophilic antibiotics to achieve therapeutic concentrations in the target site [75,96,97]. Antibiotic properties should be taken into account, such as hydrophilic vs. lipophilic, bacteriostatic vs. bactericidal, time-dependent vs. dose-dependent.
Standard dosage regimens are based on healthy volunteers and do not take into account patient/pathogen characteristics. Underdosing of antibiotics is frequent in patients with sepsis, and is associated with treatment failures and worse outcomes [35,97]. Patients with severe infections are at higher risk of suboptimal antibiotic dose. High-risk neutropenic patients have increased volume of distribution and/or capillary leak syndrome and/or a more rapid clearance of certain drugs, particularly if mechanically ventilated or requiring vasopressor [96,97]. Higher doses are needed to obtain adequate serum concentrations.
The optimal dose (Table 4) includes the minimum inhibitory concentration (MIC) of the pathogen, and the site and the severity of the infection; additionally, the patient volume of distribution, renal clearance, weight, organ support (e.g., renal replacement therapy or extracorporeal membrane oxygenation) should be considered [96].
Prompt antibiotic administration and administration of a loading dose when indicated are associated with better patient outcomes in patients with septic shock, particularly within the first hour [75,76,77,78,96,97,98]. EUCAST has recently updated criteria for susceptibility interpretation [99]; time-dependent antibiotic efficacy depends on maintaining drug concentration above the pathogen MIC for longer than 40% of the time. Continuous prolonged infusion of beta-lactams increases efficacy and is associated with better outcomes [97,100,101].
A recent review [102] suggests a three-step approach to optimized antibiotic dose: the first antibiotic dose is based on pharmacodynamics, suggesting a double dose in critically ill patients with beta-lactams, aminoglycosides, and glycopeptides. Patients under continuous renal replacement therapy may also need increased doses from linezolid or colistin [102].
Therapeutic drug monitoring (TDM) is necessary to achieve the efficacy dose and to avoid overdosing, and consequently, adverse effects. TDM of glycopeptides and aminoglycosides has already been used for a long time; Bayesian statistics utilize measured concentration and clinical covariates, and they are the most accurate estimation of individualized patient dosing [100,103,104].
Vancomycin is the standard of care for Gram-positive infections due to MRSA. Inappropriate dosing is associated with high failure rates, higher MICs, and toxicity. Adequate vancomycin dosing focused on AUC/MIC improves clinical response in infections due to MRSA [105,106]. Most patients with neutropenia have augmented vancomycin clearance, and may need increased vancomycin daily dosing; TDM in patients with neutropenia is crucial to achieve an adequate antibiotic dose and to limit vancomycin dose variation depending on patient conditions [105].
Aminoglycosides dose adjustment based on TDM did not correlate with better outcomes, but contributed to limiting toxicity; yet, optimal dosing and earlier clinical response was achieved [104].
Beta-lactams are by far the most common used antibiotics and are the cornerstone of therapy of the majority of infections. Patients with neutropenia are a special population where beta-lactam TDM could lead to better outcomes, though their use is still controversial and further studies are needed to demonstrate their benefit [107]. Their efficacy is unpredictable since TDM is currently not recommended by practice guidelines, and only a few hospitals use it on a routine basis. Beta-lactam TDM provides real antibiotic measurements so that optimal dosing can be achieved. TDM is particularly important in critically ill patients where the optimal dose defines the outcome of the patients; there is an urgent call for better beta-lactam TDM.

4.8. Duration/Early Discontinuation

Overuse of antibiotics includes unnecessarily long duration of antibiotics. Long exposure to antibiotics leads to higher side effects, renal or hepatic toxicities, and bacterial resistance due to selective antimicrobial pressure. The traditional 7 to 14-days antibiotic course for most bacterial infections is outdated, and several randomized controlled trials comparing short course vs. traditional course provide evidence of equal efficacy, and fewer adverse effects and diminished emergence of resistance with short courses [108,109,110,111].
Discontinuation of empirical antibiotics after 72 h in hemodynamically stable patients with neutropenia in the absence of fever for the past 48 h is recommended by European Conference on infections in Leukemia (ECIL-4) guidelines [3], but is rarely applied on daily clinical practice.
Discontinuation of antibiotics in patients with persistent neutropenia who fill these criteria seems to be safe, and several studies demonstrated no substantial rates of infection recurrence [111,112,113,114].
A recent open-label, multicenter, randomized trial of 281 patients with febrile neutropenia who received intensive chemotherapy or hematopoietic stem-cell transplantation between 2014 and 2019 compared short (72 h) vs. extended (≥9 days until being afebrile for 5 days, or neutrophil recovery) carbapenem treatment. Early discontinuation was noninferior (10% margin) to extended treatment regarding treatment failure, both in intention-to-treat analysis (19% vs. 15%, adjusted risk difference 4.0% (90% CI: 1.7–9.5%, p = 0.25)) and per-protocol analysis (n = 225; 23% vs. 16%, adjusted risk difference 7.3% (90% CI: 0.3–14.9%, p = 0.11)). However, serious adverse events (16% vs. 10%) and all-cause mortality (3% vs. 1%) were higher in the short treatment group, driven by patients who are persistently febrile [115]; this supports the ECIL-4 [3] recommendation of discontinuation in stable and afebrile patients.

4.9. De-Escalation

De-escalation means implementing a strategy to reduce the spectrum of the initial empirical antibiotic therapy, either by isolating one valuable causal agent, or by excluding others. De-escalation is only manageable for routine bacteriology diagnosis, and contributes to reducing antibiotic exposure to broad-spectrum antibiotics and, so, may have a relevant impact on the emergence of resistance. European ICU practitioners regarded antibiotic de-escalation and discontinuation as the most important intervention, with the potential to prevent MDR development [116].
Martire et al. [117] implemented an AMS intervention in high-risk patients with neutropenia with de-escalation and discontinuation strategies, and a significant reduction of carbapenems consumption was achieved; rates of infection relapse, increase of ICU referral, and bacteremia were unchanged.
De-escalation appears to be safe and does not have a detriment impact on outcomes [118,119]. This is a strategy that needs further studies, especially to evaluate the impact on resistance, which is still an important intervention in AMS. Early discontinuation is safe, even in patients that remain febrile and neutropenic, only after exclusion of bacterial infection; this intervention demonstrated reduced antibiotic use without worse outcomes [118,120].

5. Conclusions

AMS is essential to ensure effective and safe access to antibiotics, regardless of the clinical situation. The scarcity of studies regarding AMS programs on patients with neutropenia limits this narrative review. Antimicrobial stewardship is expanding; there are still many constraints to overcome, such as the resistance of clinicians to limit antibiotic use, and the scarcity of the literature on improving outcomes. These gaps and additional evidence focusing on outcomes represent a research opportunity. There may be a conflict between the recommendations for the empirical prescription of antibiotics and AMS programs. These approach strategies highlight the importance of patient stratification to decide broad-spectrum antibiotics, the need for nonculture diagnostics tests for early diagnosis, the critical first empirical antibiotic dose, and the safety on de-escalation/discontinuation.
In patients with neutropenia, right first time is crucial. Screening and molecular diagnostic tests have an important role in early diagnosis or antibiotic discontinuation. Fear of attending clinicians of suboptimal coverage in high-risk neutropenic patients is a major issue. Empowering health professionals with educational activities and a multidisciplinary team with trained and dedicated professionals for antimicrobial stewardship is a priority.

Author Contributions

Conceptualization, J.A. and J.R.; writing—original draft preparation, J.A., B.A. and P.P.; writing—review and editing, J.A., J.R., E.A. and T.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

J.R. served in advisory boards/speaker bureau for Pfizer, MSD, and ROCHE. Other authors disclose no conflict of interest regarding this manuscript.

References

  1. Shelton, B.; Stanik-Hutt, J.; Kane, J.; Jones, R. Implementing the Surviving Sepsis Campaign in an Ambulatory Clinic for Patients with Hematologic Malignancies. Clin. J. Oncol. Nurs. 2016, 20, 281–288. [Google Scholar] [CrossRef] [PubMed]
  2. Heinz, W.J.; Buchheidt, D.; Christopeit, M.; von Lilienfeld-Toal, M.; Cornely, O.A.; Einsele, H.; Karthaus, M.; Link, H.; Mahlberg, R.; Neumann, S.; et al. Diagnosis and empirical treatment of fever of unknown origin (FUO) in adult neutropenic patients: Guidelines of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Medical Oncology (DGHO). Ann. Hematol. 2017, 96, 1775–1792. [Google Scholar] [CrossRef] [PubMed]
  3. Averbuch, D.; Orasch, C.; Cordonnier, C.; Livermore, D.M.; Mikulska, M.; Viscoli, C.; Gyssens, I.C.; Kern, W.V.; Klyasova, G.; Marchetti, O.; et al. European guidelines for empirical antibacterial therapy for febrile neutropenic patients in the era of growing resistance: Summary of the 2011 4th European Conference on Infections in Leukemia. Haematologica 2013, 98, 1826–1835. [Google Scholar] [CrossRef] [PubMed]
  4. Stern, A.; Carrara, E.; Bitterman, R.; Yahav, D.; Leibovici, L.; Paul, M. Early discontinuation of antibiotics for febrile neutropenia versus continuation until neutropenia resolution in people with cancer. Cochrane Database Syst. Rev. 2019, 2019, CD012184. [Google Scholar] [CrossRef] [PubMed]
  5. McGowan, J.E.; Gerding, D.N. Does antibiotic restriction prevent resistance? New Horiz. 1996, 4, 370–376. [Google Scholar]
  6. Barlam, T.F.; Cosgrove, S.E.; Abbo, L.M.; MacDougall, C.; Schuetz, A.N.; Septimus, E.J.; Srinivasan, A.; Dellit, T.H.; Falck-Ytter, Y.T.; Fishman, N.O.; et al. Implementing an Antibiotic Stewardship Program: Guidelines by the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America. Clin. Infect. Dis. 2016, 62, e51–e77. [Google Scholar] [CrossRef]
  7. Schuts, E.C.; Hulscher, M.E.J.L.; Mouton, J.W.; Verduin, C.M.; Stuart, J.W.T.C.; Overdiek, H.W.P.M.; van der Linden, P.D.; Natsch, S.; Hertogh, C.M.P.M.; Wolfs, T.F.W.; et al. Current evidence on hospital antimicrobial stewardship objectives: A systematic review and meta-analysis. Lancet Infect. Dis. 2016, 16, 847–856. [Google Scholar] [CrossRef]
  8. Rice, L.B. Antimicrobial Stewardship and Antimicrobial Resistance. Med. Clin. N. Am. 2018, 102, 805–818. [Google Scholar] [CrossRef]
  9. Kollef, M.H.; Micek, S.T. Antimicrobial stewardship programs: Mandatory for all ICUs. Crit. Care 2012, 16, 179. [Google Scholar] [CrossRef]
  10. De Waele, J.J.; Dhaese, S. Antibiotic stewardship in sepsis management: Toward a balanced use of antibiotics for the severely ill patient. Expert Rev. Anti Infect. Ther. 2019, 17, 89–97. [Google Scholar] [CrossRef]
  11. Seidelman, J.L.; A Turner, N.; Wrenn, R.H.; Sarubbi, C.; Anderson, D.J.; Sexton, D.J.; Moehring, R.W. Impact of Antibiotic Stewardship Rounds in the Intensive Care Setting: A Prospective Cluster-Randomized Crossover Study. Clin. Infect. Dis. 2022, 74, 1986–1992. [Google Scholar] [CrossRef] [PubMed]
  12. Taur, Y.; Xavier, J.; Lipuma, L.; Ubeda, C.; Goldberg, J.; Gobourne, A.; Lee, Y.J.; Dubin, K.A.; Socci, N.D.; Viale, A.; et al. Intestinal Domination and the Risk of Bacteremia in Patients Undergoing Allogeneic Hematopoietic Stem Cell Transplantation. Clin. Infect. Dis. 2012, 55, 905–914. [Google Scholar] [CrossRef] [PubMed]
  13. Blijlevens, N.; Donnelly, J.; De Pauw, B. Mucosal barrier injury: Biology, pathology, clinical counterparts and consequences of intensive treatment for haematological malignancy: An overview. Bone Marrow Transplant. 2000, 25, 1269–1278. [Google Scholar] [CrossRef] [PubMed]
  14. Bow, E.J. There should be no ESKAPE for febrile neutropenic cancer patients: The dearth of effective antibacterial drugs threatens anticancer efficacy. J. Antimicrob. Chemother. 2013, 68, 492–495. [Google Scholar] [CrossRef]
  15. Cattaneo, C.; Zappasodi, P.; Mancini, V.; Annaloro, C.; Pavesi, F.; Skert, C.; Ferrario, A.; Todisco, E.; Saccà, V.; Verga, L.; et al. Emerging resistant bacteria strains in bloodstream infections of acute leukaemia patients: Results of a prospective study by the Rete Ematologica Lombarda (Rel). Ann. Hematol. 2016, 95, 1955–1963. [Google Scholar] [CrossRef]
  16. Scheich, S.; Weber, S.; Reinheimer, C.; Wichelhaus, T.A.; Hogardt, M.; Kempf, V.A.J.; Kessel, J.; Serve, H.; Steffen, B. Bloodstream infections with gram-negative organisms and the impact of multidrug resistance in patients with hematological malignancies. Ann. Hematol. 2018, 97, 2225–2234. [Google Scholar] [CrossRef]
  17. Ali, R.K.; Surme, S.; Balkan, I.I.; Salihoglu, A.; Ozdemir, M.S.; Ozdemir, Y.; Mete, B.; Can, G.; Ar, M.C.; Tabak, F.; et al. An eleven-year cohort of bloodstream infections in 552 febrile neutropenic patients: Resistance profiles of Gram-negative bacteria as a predictor of mortality. Ann. Hematol. 2020, 99, 1925–1932. [Google Scholar]
  18. Montassier, E.; Batard, E.; Gastinne, T.; Potel, G.; Cochetière, M.F. Recent changes in bacteremia in patients with cancer: A systematic review of epidemiology and antibiotic resistance. Eur. J. Clin. Microbiol. Infect. Dis. 2013, 32, 841–850. [Google Scholar] [CrossRef]
  19. Stoma, I.; Karpov, I.; Milanovich, N.; Uss, A.; Iskrov, I. Risk factors for mortality in patients with bloodstream infections during the pre-engraftment period after hematopoietic stem cell transplantation. Blood Res. 2016, 51, 102. [Google Scholar] [CrossRef]
  20. Charles, P.G.P.; Ward, P.B.; Johnson, P.D.R.; Howden, B.P.; Grayson, M.L. Clinical Features Associated with Bacteremia Due to Heterogeneous Vancomycin-Intermediate Staphylococcus aureus. Clin. Infect. Dis. 2004, 38, 448–451. [Google Scholar] [CrossRef]
  21. Vydra, J.; Shanley, R.M.; George, I.; Ustun, C.; Smith, A.R.; Weisdorf, D.J.; Young, J.-A. Enterococcal Bacteremia Is Associated with Increased Risk of Mortality in Recipients of Allogeneic Hematopoietic Stem Cell Transplantation. Clin. Infect. Dis. 2012, 55, 764–770. [Google Scholar] [CrossRef] [PubMed]
  22. Tleyjeh, I.M.; Tlaygeh, H.M.; Hejal, R.; Montori, V.M.; Baddour, L.M. The Impact of Penicillin Resistance on Short-Term Mortality in Hospitalized Adults with Pneumococcal Pneumonia: A Systematic Review and Meta-Analysis. Clin. Infect. Dis. 2006, 42, 788–797. [Google Scholar] [CrossRef] [PubMed]
  23. Huh, K.; Chung, D.R.; Ha, Y.E.; Ko, J.-H.; Kim, S.-H.; Kim, M.-J.; Huh, H.J.; Lee, N.Y.; Cho, S.Y.; Kang, C.-I.; et al. Impact of Difficult-to-Treat Resistance in Gram-negative Bacteremia on Mortality: Retrospective Analysis of Nationwide Surveillance Data. Clin. Infect. Dis. 2020, 71, e487–e496. [Google Scholar] [CrossRef]
  24. Weber, S.; Magh, A.; Hogardt, M.; Kempf, V.A.J.; Vehreschild, M.J.G.T.; Serve, H.; Scheich, S.; Steffen, B. Profiling of bacterial bloodstream infections in hematological and oncological patients based on a comparative survival analysis. Ann. Hematol. 2021, 100, 1593–1602. [Google Scholar] [CrossRef] [PubMed]
  25. Dellit, T.H.; Owens, R.C.; McGowan, J.E.; Gerding, D.N.; Weinstein, R.A.; Burke, J.P.; Huskins, W.C.; Paterson, D.L.; Fishman, N.O.; Carpenter, C.F.; et al. Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America Guidelines for Developing an Institutional Program to Enhance Antimicrobial Stewardship. Clin. Infect. Dis. 2007, 44, 159–177. [Google Scholar] [CrossRef] [PubMed]
  26. Dyar, O.J.; Huttner, B.; Schouten, J.; Pulcini, C. What is antimicrobial stewardship? Clin. Microbiol. Infect. 2017, 23, 793–798. [Google Scholar] [CrossRef]
  27. Tverdek, F.P.; Rolston, K.V.; Chemaly, R.F. Antimicrobial Stewardship in Patients with Cancer. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2012, 32, 722–734. [Google Scholar] [CrossRef]
  28. Gyssens, I.C.; Kern, W.V.; Livermore, D.M. The role of antibiotic stewardship in limiting antibacterial resistance among hematology patients. Haematologica 2013, 98, 1821–1825. [Google Scholar] [CrossRef]
  29. Abbo, L.M.; Ariza-Heredia, E.J. Antimicrobial Stewardship in Immunocompromised Hosts. Infect. Dis. Clin. N. Am. 2014, 28, 263–279. [Google Scholar] [CrossRef]
  30. Pillinger, K.E.; Bouchard, J.; Withers, S.T.; Mediwala, K.; McGee, E.U.; Gibson, G.M.; Bland, C.M.; Bookstaver, P.B. Inpatient Antibiotic Stewardship Interventions in the Adult Oncology and Hematopoietic Stem Cell Transplant Population: A Review of the Literature. Ann. Pharmacother. 2020, 54, 594–610. [Google Scholar] [CrossRef]
  31. Bielicki, J.A.; Manuel, O. Antimicrobial stewardship programs in solid-organ transplant recipients in Switzerland. Transpl. Infect. Dis. 2022, 24, e13902. [Google Scholar] [CrossRef]
  32. El-Sokkary, R.; Erdem, H.; Kullar, R.; Pekok, A.U.; Amer, F.; Grgić, S.; Carevic, B.; El-Kholy, A.; Liskova, A.; Özdemir, M.; et al. Self-reported antibiotic stewardship and infection control measures from 57 intensive care units: An international ID-IRI survey. J. Infect. Public Health 2022, 15, 950–954. [Google Scholar] [CrossRef] [PubMed]
  33. Plachouras, D.; Kärki, T.; Hansen, S.; Hopkins, S.; Lyytikäinen, O.; Moro, M.L.; Reilly, J.; Zarb, P.; Zingg, W.; Kinross, P.; et al. Antimicrobial use in European acute care hospitals: Results from the second point prevalence survey (PPS) of healthcare-associated infections and antimicrobial use, 2016 to 2017. Eurosurveillance 2018, 23, 1800393. [Google Scholar] [CrossRef] [PubMed]
  34. Rello, J.; Paiva, J.A. Antimicrobial stewardship at the emergency department: Dead bugs do not mutate! Eur. J. Intern. Med. 2023, 109, 30–32. [Google Scholar] [CrossRef] [PubMed]
  35. Vieceli, T.; Rello, J. Optimization of antimicrobial prescription in the hospital. Eur. J. Intern. Med. 2022, 106, 29–44. [Google Scholar] [CrossRef]
  36. Hecker, M.T.; Aron, D.C.; Patel, N.P.; Lehmann, M.K.; Donskey, C.J. Unnecessary Use of Antimicrobials in Hospitalized Patients. Arch. Intern. Med. 2003, 163, 972. [Google Scholar] [CrossRef]
  37. Thursky, K.A.; Worth, L.J. Can mortality of cancer patients with fever and neutropenia be improved? Curr. Opin. Infect. Dis. 2015, 28, 505–513. [Google Scholar] [CrossRef]
  38. Schnell, D.; Azoulay, E.; Benoit, D.; Clouzeau, B.; Demaret, P.; Ducassou, S.; Frange, P.; Lafaurie, M.; Legrand, M.; Meert, A.-P.; et al. Management of neutropenic patients in the intensive care unit (NEWBORNS EXCLUDED) recommendations from an expert panel from the French Intensive Care Society (SRLF) with the French Group for Pediatric Intensive Care Emergencies (GFRUP), the French Society of Anesthesia and Intensive Care (SFAR), the French Society of Hematology (SFH), the French Society for Hospital Hygiene (SF2H), and the French Infectious Diseases Society (SPILF). Ann. Intensive Care 2016, 6, 90. [Google Scholar]
  39. Kalil, A.C.; Sandkovsky, U.; Florescu, D.F. Severe infections in critically ill solid organ transplant recipients. Clin. Microbiol. Infect. 2018, 24, 1257–1263. [Google Scholar] [CrossRef]
  40. Rolston, K.V.I. New Trends in Patient Management: Risk-Based Therapy for Febrile Patients with Neutropenia. Clin. Infect. Dis. 1999, 29, 515–521. [Google Scholar] [CrossRef]
  41. Freifeld, A.G.; Bow, E.J.; Sepkowitz, K.A.; Boeckh, M.J.; Ito, J.I.; Mullen, C.A.; Raad, I.I.; Rolston, K.V.; Young, J.-A.H.; Wingard, J.R. Clinical Practice Guideline for the Use of Antimicrobial Agents in Neutropenic Patients with Cancer: 2010 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2011, 52, e56–e93. [Google Scholar] [CrossRef] [PubMed]
  42. Ballo, O.; Tarazzit, I.; Stratmann, J.; Reinheimer, C.; Hogardt, M.; Wichelhaus, T.A.; Kempf, V.; Serve, H.; Finkelmeier, F.; Brandts, C. Colonization with multidrug resistant organisms determines the clinical course of patients with acute myeloid leukemia undergoing intensive induction chemotherapy. PLoS ONE 2019, 14, e0210991. [Google Scholar] [CrossRef] [PubMed]
  43. Kömürcü, B.; Tükenmez Tigen, E.; Toptaş, T.; Fıratlı Tuğlular, T.; Korten, V. Rectal colonization with multidrug-resistant gram-negative bacteria in patients with hematological malignancies: A prospective study. Expert Rev. Hematol. 2020, 13, 923–927. [Google Scholar] [CrossRef]
  44. Torres, I.; Huntley, D.; Tormo, M.; Calabuig, M.; Hernández-Boluda, J.C.; Terol, M.J.; Carretero, C.; de Michelena, P.; Pérez, A.; Piñana, J.L.; et al. Multi-body-site colonization screening cultures for predicting multi-drug resistant Gram-negative and Gram-positive bacteremia in hematological patients. BMC Infect. Dis. 2022, 22, 172. [Google Scholar] [CrossRef] [PubMed]
  45. Micozzi, A.; Gentile, G.; Santilli, S.; Minotti, C.; Capria, S.; Moleti, M.L.; Barberi, W.; Cartoni, C.; Trisolini, S.M.; Testi, A.M.; et al. Reduced mortality from KPC-K.pneumoniae bloodstream infection in high-risk patients with hematological malignancies colonized by KPC-K.pneumoniae. BMC Infect. Dis. 2021, 21, 1079. [Google Scholar]
  46. Ambretti, S.; Bassetti, M.; Clerici, P.; Petrosillo, N.; Tumietto, F.; Viale, P.; Rossolini, G.M. Screening for carriage of carbapenem-resistant Enterobacteriaceae in settings of high endemicity: A position paper from an Italian working group on CRE infections. Antimicrob. Resist. Infect. Control 2019, 8, 136. [Google Scholar] [CrossRef] [PubMed]
  47. Gagliotti, C.; Ciccarese, V.; Sarti, M.; Giordani, S.; Barozzi, A.; Braglia, C.; Gallerani, C.; Gargiulo, R.; Lenzotti, G.; Manzi, O.; et al. Active surveillance for asymptomatic carriers of carbapenemase-producing Klebsiella pneumoniae in a hospital setting. J. Hosp. Infect. 2013, 83, 330–332. [Google Scholar] [CrossRef]
  48. Morency-Potvin, P.; Schwartz, D.N.; Weinstein, R.A. Antimicrobial Stewardship: How the Microbiology Laboratory Can Right the Ship. Clin. Microbiol. Rev. 2017, 30, 381–407. [Google Scholar] [CrossRef]
  49. Rowland, T.; Hilliard, H.; Barlow, G. Procalcitonin. In Advances in Clinical Chemistry; Elsevier: Amsterdam, The Netherlands, 2015; pp. 71–86. [Google Scholar]
  50. Layios, N.; Lambermont, B.; Canivet, J.-L.; Morimont, P.; Preiser, J.-C.; Garweg, C.; LeDoux, D.; Frippiat, F.; Piret, S.; Giot, J.-B.; et al. Procalcitonin usefulness for the initiation of antibiotic treatment in intensive care unit patients. Crit. Care Med. 2012, 40, 2304–2309. [Google Scholar] [CrossRef]
  51. Wirz, Y.; Meier, M.A.; Bouadma, L.; Luyt, C.E.; Wolff, M.; Chastre, J.; Tubach, F.; Schroeder, S.; Nobre, V.; Annane, D.; et al. Effect of procalcitonin-guided antibiotic treatment on clinical outcomes in intensive care unit patients with infection and sepsis patients: A patient-level meta-analysis of randomized trials. Crit. Care 2018, 22, 191. [Google Scholar] [CrossRef]
  52. Met, Ö.; Jensen, K.M.; Chamberlain, C.A.; Donia, M.; Svane, I.M. Principles of adoptive T cell therapy in cancer. Semin. Immunopathol. 2019, 41, 49–58. [Google Scholar] [CrossRef] [PubMed]
  53. Lagacé-Wiens, P.R.S.; Adam, H.J.; Karlowsky, J.A.; Nichol, K.A.; Pang, P.F.; Guenther, J.; Webb, A.A.; Miller, C.; Alfa, M.J. Identification of Blood Culture Isolates Directly from Positive Blood Cultures by Use of Matrix-Assisted Laser Desorption Ionization–Time of Flight Mass Spectrometry and a Commercial Extraction System: Analysis of Performance, Cost, and Turnaround Time. J. Clin. Microbiol. 2012, 50, 3324–3328. [Google Scholar] [CrossRef] [PubMed]
  54. Idelevich, E.A.; Sparbier, K.; Kostrzewa, M.; Becker, K. Rapid detection of antibiotic resistance by MALDI-TOF mass spectrometry using a novel direct-on-target microdroplet growth assay. Clin. Microbiol. Infect. 2018, 24, 738–743. [Google Scholar] [CrossRef]
  55. Livermore, D.M. Multicentre evaluation of the VITEK 2 Advanced Expert System for interpretive reading of antimicrobial resistance tests. J. Antimicrob. Chemother. 2002, 49, 289–300. [Google Scholar] [CrossRef]
  56. Marschal, M.; Bachmaier, J.; Autenrieth, I.; Oberhettinger, P.; Willmann, M.; Peter, S. Evaluation of the Accelerate Pheno System for Fast Identification and Antimicrobial Susceptibility Testing from Positive Blood Cultures in Bloodstream Infections Caused by Gram-Negative Pathogens. J. Clin. Microbiol. 2017, 55, 2116–2126. [Google Scholar] [CrossRef] [PubMed]
  57. Martin, E.C.; Colombier, M.A.; Limousin, L.; Daude, O.; Izarn, O.; Cahen, P.; Farfour, E.; Lesprit, P.; Vasse, M. Impact of EUCAST rapid antimicrobial susceptibility testing (RAST) on management of Gram-negative bloodstream infection. Infect. Dis. Now 2022, 52, 421–425. [Google Scholar] [CrossRef]
  58. Kim, J.-H.; Kim, T.S.; Chang, E.; Kang, C.K.; Choe, P.G.; Kim, N.J.; Oh, M.-D.; Park, W.B.; Kim, I. Effectiveness of antimicrobial stewardship programmes based on rapid antibiotic susceptibility testing of haematological patients having high-risk factors for bacteraemia-related mortality: A post-hoc analysis of a randomised controlled trial. Int. J. Antimicrob. Agents 2022, 60, 106604. [Google Scholar] [CrossRef]
  59. Demord, A.; Poirel, L.; D’Emidio, F.; Pomponio, S.; Nordmann, P. Rapid ESBL NP Test for Rapid Detection of Expanded-Spectrum β-Lactamase Producers in Enterobacterales. Microb. Drug Resist. 2021, 27, 1131–1135. [Google Scholar] [CrossRef]
  60. Feng, Y.; Palanisami, A.; Kuriakose, J.; Pigula, M.; Ashraf, S.; Hasan, T. Novel Rapid Test for Detecting Carbapenemase. Emerg. Infect. Dis. 2020, 26, 793–795. [Google Scholar] [CrossRef]
  61. Chang, S.-S.; Hsieh, W.-H.; Liu, T.-S.; Lee, S.-H.; Wang, C.-H.; Chou, H.-C.; Yeo, Y.H.; Tseng, C.-P.; Lee, C.-C. Multiplex PCR System for Rapid Detection of Pathogens in Patients with Presumed Sepsis—A Systemic Review and Meta-Analysis. PLoS ONE 2013, 8, e62323. [Google Scholar] [CrossRef]
  62. Trung, N.T.; Thau, N.S.; Bang, M.H.; Song, L.H. PCR-based Sepsis@Quick test is superior in comparison with blood culture for identification of sepsis-causative pathogens. Sci. Rep. 2019, 9, 13663. [Google Scholar] [CrossRef] [PubMed]
  63. El Sherif, H.M.; Elsayed, M.; El-Ansary, M.R.; Aboshanab, K.M.; El Borhamy, M.I.; Elsayed, K.M. BioFire FilmArray BCID2 versus VITEK-2 System in Determining Microbial Etiology and Antibiotic-Resistant Genes of Pathogens Recovered from Central Line-Associated Bloodstream Infections. Biology 2022, 11, 1573. [Google Scholar] [CrossRef] [PubMed]
  64. Holma, T.; Torvikoski, J.; Friberg, N.; Nevalainen, A.; Tarkka, E.; Antikainen, J.; Martelin, J.J. Rapid molecular detection of pathogenic microorganisms and antimicrobial resistance markers in blood cultures: Evaluation and utility of the next-generation FilmArray Blood Culture Identification 2 panel. Eur. J. Clin. Microbiol. Infect. Dis. 2022, 41, 363–371. [Google Scholar] [CrossRef] [PubMed]
  65. Fida, M.; Wolf, M.J.; Hamdi, A.; Vijayvargiya, P.; Garrigos, Z.E.; Khalil, S.; E Greenwood-Quaintance, K.; Thoendel, M.J.; Patel, R. Detection of Pathogenic Bacteria from Septic Patients Using 16S Ribosomal RNA Gene–Targeted Metagenomic Sequencing. Clin. Infect. Dis. 2021, 73, 1165–1172. [Google Scholar] [CrossRef]
  66. Mason, W.J.; Blevins, J.S.; Beenken, K.; Wibowo, N.; Ojha, N.; Smeltzer, M.S. Multiplex PCR Protocol for the Diagnosis of Staphylococcal Infection. J. Clin. Microbiol. 2001, 39, 3332–3338. [Google Scholar] [CrossRef] [PubMed]
  67. Both, A.; Berneking, L.; Berinson, B.; Lütgehetmann, M.; Christner, M.; Aepfelbacher, M.; Rohde, H. Rapid identification of the vanA/vanB resistance determinant in Enterococcus sp. from blood cultures using the Cepheid Xpert vanA/vanB cartridge system. Diagn. Microbiol. Infect. Dis. 2020, 96, 114977. [Google Scholar] [CrossRef]
  68. De Angelis, G.; Grossi, A.; Menchinelli, G.; Boccia, S.; Sanguinetti, M.; Posteraro, B. Rapid molecular tests for detection of antimicrobial resistance determinants in Gram-negative organisms from positive blood cultures: A systematic review and meta-analysis. Clin. Microbiol. Infect. 2020, 26, 271–280. [Google Scholar] [CrossRef]
  69. Rello, J.; Alonso-Tarrés, C. Emerging Technologies for Microbiologic Diagnosis of Sepsis: The Rapid Determination of Resistance to Antimicrobial Agents Should Be the Key. Clin. Infect. Dis. 2021, 73, 1173–1175. [Google Scholar] [CrossRef]
  70. Gaieski, D.F.; Mikkelsen, M.E.; Band, R.A.; Pines, J.M.; Massone, R.; Furia, F.F.; Shofer, F.S.; Goyal, M. Impact of time to antibiotics on survival in patients with severe sepsis or septic shock in whom early goal-directed therapy was initiated in the emergency department. Crit. Care Med. 2010, 38, 1045–1053. [Google Scholar] [CrossRef]
  71. Garnacho-Montero, J.; Gutiérrez-Pizarraya, A.; Escoresca-Ortega, A.; Fernández-Delgado, E.; López-Sánchez, J.M. Adequate antibiotic therapy prior to ICU admission in patients with severe sepsis and septic shock reduces hospital mortality. Crit. Care 2015, 19, 302. [Google Scholar] [CrossRef]
  72. Zhang, D.; Micek, S.T.; Kollef, M.H. Time to Appropriate Antibiotic Therapy Is an Independent Determinant of Postinfection ICU and Hospital Lengths of Stay in Patients with Sepsis. Crit. Care Med. 2015, 43, 2133–2140. [Google Scholar] [CrossRef] [PubMed]
  73. Flaherty, S.K.; Weber, R.L.; Chase, M.; Dugas, A.F.; Graver, A.M.; Salciccioli, J.D.; Cocchi, M.N.; Donnino, M.W. Septic Shock and Adequacy of Early Empiric Antibiotics in the Emergency Department. J. Emerg. Med. 2014, 47, 601–607. [Google Scholar] [CrossRef] [PubMed]
  74. Kollef, M.H. Optimizing antibiotic therapy in the intensive care unit setting. Crit. Care 2001, 5, 189–195. [Google Scholar] [CrossRef] [PubMed]
  75. Roberts, J.A.; Taccone, F.S.; Lipman, J. Understanding PK/PD. Intensive Care Med. 2016, 42, 1797–1800. [Google Scholar] [CrossRef]
  76. Simpson, N.; Lamontagne, F.; Shankar-Hari, M. Septic shock resuscitation in the first hour. Curr. Opin. Crit. Care 2017, 23, 561–566. [Google Scholar] [CrossRef]
  77. 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. Intensive Care Med. 2017, 43, 304–377. [Google Scholar] [CrossRef]
  78. Daniels, L.M.; Durani, U.; Barreto, J.N.; O’horo, J.C.; Siddiqui, M.A.; Park, J.G.; Tosh, P.K. Impact of time to antibiotic on hospital stay, intensive care unit admission, and mortality in febrile neutropenia. Support. Care Cancer 2019, 27, 4171–4177. [Google Scholar] [CrossRef]
  79. Bader, M.Z.; Obaid, A.T.; Al-Khateb, H.M.; Eldos, Y.T.; Elaya, M.M. Developing Adult Sepsis Protocol to Reduce the Time to Initial Antibiotic Dose and Improve Outcomes among Patients with Cancer in Emergency Department. Asia Pac. J. Oncol. Nurs. 2020, 7, 355–360. [Google Scholar] [CrossRef]
  80. Furno, P.; Bucaneve, G.; Del Favero, A. Monotherapy or aminoglycoside-containing combinations for empirical antibiotic treatment of febrile neutropenic patients: A meta-analysis. Lancet Infect. Dis. 2002, 2, 231–242. [Google Scholar] [CrossRef]
  81. Paul, M.; Yahav, D.; Fraser, A.; Leibovici, L. Empirical antibiotic monotherapy for febrile neutropenia: Systematic review and meta-analysis of randomized controlled trials. J. Antimicrob. Chemother. 2006, 57, 176–189. [Google Scholar] [CrossRef]
  82. Rolston, K.V.I. Comment on: Empirical antibiotic monotherapy for febrile neutropenia: Systematic review and meta-analysis of randomized controlled trials. J. Antimicrob. Chemother. 2006, 58, 478. [Google Scholar] [CrossRef]
  83. Klastersky, J.; de Naurois, J.; Rolston, K.; Rapoport, B.; Maschmeyer, G.; Aapro, M.; Herrstedt, J.; on behalf of the ESMO Guidelines Committee. Management of febrile neutropaenia: ESMO Clinical Practice Guidelines. Ann. Oncol. 2016, 27, v111–v118. [Google Scholar] [CrossRef]
  84. 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. [Google Scholar] [CrossRef] [PubMed]
  85. Tamma, P.D.; Aitken, S.L.; Bonomo, R.A.; Mathers, A.J.; van Duin, D.; Clancy, C.J. Infectious Diseases Society of America 2022 Guidance on the Treatment of Extended-Spectrum β-lactamase Producing Enterobacterales (ESBL-E), Carbapenem-Resistant Enterobacterales (CRE), and Pseudomonas aeruginosa with Difficult-to-Treat Resistance (DTR- P. aeruginosa). Clin. Infect. Dis. 2022, 75, 187–212. [Google Scholar] [PubMed]
  86. Rodríguez-Baño, J.; Gutiérrez-Gutiérrez, B.; Machuca, I.; Pascual, A. Treatment of Infections Caused by Extended-Spectrum-Beta-Lactamase-, AmpC-, and Carbapenemase-Producing Enterobacteriaceae. Clin. Microbiol. Rev. 2018, 31, e00079-17. [Google Scholar] [CrossRef] [PubMed]
  87. Hawkey, P.M.; E Warren, R.; Livermore, D.M.; McNulty, C.A.M.; A Enoch, D.; A Otter, J.; Wilson, A.P.R. Treatment of infections caused by multidrug-resistant Gram-negative bacteria: Report of the British Society for Antimicrobial Chemotherapy/Healthcare Infection Society/British Infection Association Joint Working Party. J. Antimicrob. Chemother. 2018, 73 (Suppl. S3), iii2–iii78. [Google Scholar] [CrossRef]
  88. Garcia-Vidal, C.; Stern, A.; Gudiol, C. Multidrug-resistant, gram-negative infections in high-risk haematologic patients: An update on epidemiology, diagnosis and treatment. Curr. Opin. Infect. Dis. 2021, 34, 314–322. [Google Scholar] [CrossRef]
  89. Paul, M.; Borok, S.; Fraser, A.; Vidal, L.; Leibovici, L. Empirical antibiotics against Gram-positive infections for febrile neutropenia: Systematic review and meta-analysis of randomized controlled trials. J. Antimicrob. Chemother. 2005, 55, 436–444. [Google Scholar] [CrossRef]
  90. Beyar-Katz, O.; Dickstein, Y.; Borok, S.; Vidal, L.; Leibovici, L.; Paul, M. Empirical antibiotics targeting gram-positive bacteria for the treatment of febrile neutropenic patients with cancer. Cochrane Database Syst. Rev. 2017, 2017, CD003914. [Google Scholar] [CrossRef]
  91. Guarana, M.; Nucci, M.; Nouér, S.A. Shock and Early Death in Hematologic Patients with Febrile Neutropenia. Antimicrob. Agents Chemother. 2019, 63, e01250-19. [Google Scholar] [CrossRef]
  92. Mergenhagen, K.A.; Starr, K.E.; Wattengel, B.A.; Lesse, A.J.; Sumon, Z.; Sellick, J.A. Determining the Utility of Methicillin-Resistant Staphylococcus aureus Nares Screening in Antimicrobial Stewardship. Clin. Infect. Dis. 2020, 71, 1142–1148. [Google Scholar] [CrossRef] [PubMed]
  93. Giancola, S.E.; Nguyen, A.T.; Le, B.; Ahmed, O.; Higgins, C.; Sizemore, J.A.; Orwig, K.W. Clinical utility of a nasal swab methicillin-resistant Staphylococcus aureus polymerase chain reaction test in intensive and intermediate care unit patients with pneumonia. Diagn. Microbiol. Infect. Dis. 2016, 86, 307–310. [Google Scholar] [CrossRef] [PubMed]
  94. Makiani, M.; Nasiripour, S.; Hosseini, M.; Mahbubi, A. Drug-drug interactions: The importance of medication reconciliation. J. Res. Pharm. Pract. 2017, 6, 61. [Google Scholar] [PubMed]
  95. Cadwallader, J.; Spry, K.; Morea, J.; Russ, A.L.; Duke, J.; Weiner, M. Design of a Medication Reconciliation Application. Appl. Clin. Inform. 2013, 4, 110–125. [Google Scholar]
  96. Ulldemolins, M.; Rello, J. The Relevance of Drug Volume of Distribution in Antibiotic Dosing. Curr. Pharm. Biotechnol. 2011, 12, 1996–2001. [Google Scholar] [CrossRef]
  97. Roberts, J.A.; Paul, S.K.; Akova, M.; Bassetti, M.; De Waele, J.J.; Dimopoulos, G.; Kaukonen, K.-M.; Koulenti, D.; Martin, C.; Montravers, P.; et al. DALI: Defining Antibiotic Levels in Intensive Care Unit Patients: Are Current β-Lactam Antibiotic Doses Sufficient for Critically Ill Patients? Clin. Infect. Dis. 2014, 58, 1072–1083. [Google Scholar] [CrossRef]
  98. Liu, V.X.; Fielding-Singh, V.; Greene, J.D.; Baker, J.M.; Iwashyna, T.J.; Bhattacharya, J.; Escobar, G.J. The Timing of Early Antibiotics and Hospital Mortality in Sepsis. Am. J. Respir. Crit. Care Med. 2017, 196, 856–863. [Google Scholar] [CrossRef]
  99. Giske, C.G.; Turnidge, J.; Cantón, R.; Kahlmeter, G. Update from the European Committee on Antimicrobial Susceptibility Testing (EUCAST). J. Clin. Microbiol. 2022, 60, e00276-21. [Google Scholar] [CrossRef]
  100. Sime, F.B.; Roberts, M.S.; Roberts, J.A. Optimization of dosing regimens and dosing in special populations. Clin. Microbiol. Infect. 2015, 21, 886–893. [Google Scholar] [CrossRef]
  101. Vardakas, K.Z.; Voulgaris, G.L.; Maliaros, A.; Samonis, G.; Falagas, M.E. Prolonged versus short-term intravenous infusion of antipseudomonal β-lactams for patients with sepsis: A systematic review and meta-analysis of randomised trials. Lancet Infect. Dis. 2018, 18, 108–120. [Google Scholar] [CrossRef]
  102. Timsit, J.-F.; Bassetti, M.; Cremer, O.; Daikos, G.; de Waele, J.; Kallil, A.; Kipnis, E.; Kollef, M.; Laupland, K.; Paiva, J.-A.; et al. Rationalizing antimicrobial therapy in the ICU: A narrative review. Intensive Care Med. 2019, 45, 172–189. [Google Scholar] [CrossRef]
  103. Tängdén, T.; Martín, V.R.; Felton, T.W.; Nielsen, E.I.; Marchand, S.; Brüggemann, R.J.; Bulitta, J.B.; Bassetti, M.; Theuretzbacher, U.; Tsuji, B.T.; et al. The role of infection models and PK/PD modelling for optimising care of critically ill patients with severe infections. Intensive Care Med. 2017, 43, 1021–1032. [Google Scholar] [CrossRef]
  104. Duszynska, W.; Taccone, F.; Hurkacz, M.; Kowalska-Krochmal, B.; Wiela-Hojeńska, A.; Kübler, A. Therapeutic drug monitoring of amikacin in septic patients. Crit. Care 2013, 17, R165. [Google Scholar] [CrossRef]
  105. Monteiro, J.F.; Hahn, S.R.; Gonçalves, J.; Fresco, P. Vancomycin therapeutic drug monitoring and population pharmacokinetic models in special patient subpopulations. Pharmacol. Res. Perspect. 2018, 6, e00420. [Google Scholar] [CrossRef] [PubMed]
  106. Prybylski, J.P. Vancomycin Trough Concentration as a Predictor of Clinical Outcomes in Patients with Staphylococcus aureus Bacteremia: A Meta-analysis of Observational Studies. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2015, 35, 889–898. [Google Scholar] [CrossRef] [PubMed]
  107. Huttner, A.; Harbarth, S.; Hope, W.W.; Lipman, J.; Roberts, J.A. Therapeutic drug monitoring of the β-lactam antibiotics: What is the evidence and which patients should we be using it for? J. Antimicrob. Chemother. 2015, 70, dkv201. [Google Scholar] [CrossRef] [PubMed]
  108. El Moussaoui, R.; Borgie, C.A.J.M.D.; Broek, P.V.D.; Hustinx, W.N.; Bresser, P.; Berk, G.E.L.V.D.; Poley, J.-W.; Berg, B.V.D.; Krouwels, F.H.; Bonten, M.J.M.; et al. Effectiveness of discontinuing antibiotic treatment after three days versus eight days in mild to moderate-severe community acquired pneumonia: Randomised, double blind study. BMJ 2006, 332, 1355. [Google Scholar] [CrossRef]
  109. Capellier, G.; Mockly, H.; Charpentier, C.; Annane, D.; Blasco, G.; Desmettre, T.; Roch, A.; Faisy, C.; Cousson, J.; Limat, S.; et al. Early-Onset Ventilator-Associated Pneumonia in Adults Randomized Clinical Trial: Comparison of 8 versus 15 Days of Antibiotic Treatment. PLoS ONE 2012, 7, e41290. [Google Scholar] [CrossRef] [PubMed]
  110. Yahav, D.; Franceschini, E.; Koppel, F.; Turjeman, A.; Babich, T.; Bitterman, R.; Neuberger, A.; Ghanem-Zoubi, N.; Santoro, A.; Eliakim-Raz, N.; et al. Seven Versus 14 Days of Antibiotic Therapy for Uncomplicated Gram-negative Bacteremia: A Noninferiority Randomized Controlled Trial. Clin. Infect. Dis. 2019, 69, 1091–1098. [Google Scholar] [CrossRef]
  111. Aguilar-Guisado, M.; Espigado, I.; Martín-Peña, A.; Gudiol, C.; Royo-Cebrecos, C.; Falantes, J.; Vázquez-López, L.; Montero, M.I.; Rosso-Fernández, C.; Martino, M.D.L.L.; et al. Optimisation of empirical antimicrobial therapy in patients with haematological malignancies and febrile neutropenia (How Long study): An open-label, randomised, controlled phase 4 trial. Lancet Haematol. 2017, 4, e573–e583. [Google Scholar] [CrossRef] [PubMed]
  112. Link, H.; Maschmeyer, G.; Meyer, P.; Hiddemann, W.; Stille, W.; Helmerking, M. Interventional antimicrobial therapy in febrile neutropenic patients. Ann. Hematol. 1994, 69, 231–243. [Google Scholar] [CrossRef]
  113. Cornelissen, J.J.; Rozenberg-Arska, M.; Dekker, A.W. Discontinuation of Intravenous Antibiotic Therapy During Persistent Neutropenia in Patients Receiving Prophylaxis with Oral Ciprofloxacin. Clin. Infect. Dis. 1995, 21, 1300–1302. [Google Scholar] [CrossRef]
  114. Slobbe, L.; van der Waal, L.; Jongman, L.R.; Lugtenburg, P.J.; Rijnders, B.J.A. Three-day treatment with imipenem for unexplained fever during prolonged neutropaenia in haematology patients receiving fluoroquinolone and fluconazole prophylaxis: A prospective observational safety study. Eur. J. Cancer 2009, 45, 2810–2817. [Google Scholar] [CrossRef] [PubMed]
  115. de Jonge, N.A.; Sikkens, J.J.; Zweegman, S.; Beeker, A.; Ypma, P.; Herbers, A.H.; Vasmel, W.; de Kreuk, A.; Coenen, J.L.L.M.; Lissenberg-Witte, B.; et al. Short versus extended treatment with a carbapenem in patients with high-risk fever of unknown origin during neutropenia: A non-inferiority, open-label, multicentre, randomised trial. Lancet Haematol. 2022, 9, e563–e572. [Google Scholar] [CrossRef] [PubMed]
  116. Rello, J.; on behalf of The Nine-I study Group; Sarda, C.; Mokart, D.; Arvaniti, K.; Akova, M.; Tabah, A.; Azoulay, E. Antimicrobial Stewardship in Hematological Patients at the intensive care unit: A global cross-sectional survey from the Nine-i Investigators Network. Eur. J. Clin. Microbiol. Infect. Dis. 2020, 39, 385–392. [Google Scholar] [CrossRef] [PubMed]
  117. La Martire, G.; Robin, C.; Oubaya, N.; Lepeule, R.; Beckerich, F.; Leclerc, M.; Barhoumi, W.; Toma, A.; Pautas, C.; Maury, S.; et al. De-escalation and discontinuation strategies in high-risk neutropenic patients: An interrupted time series analyses of antimicrobial consumption and impact on outcome. Eur. J. Clin. Microbiol. Infect. Dis. 2018, 37, 1931–1940. [Google Scholar] [CrossRef] [PubMed]
  118. Mathieu, C.; Pastene, B.; Cassir, N.; Martin-Loeches, I.; Leone, M. Efficacy and safety of antimicrobial de-escalation as a clinical strategy. Expert Rev. Anti Infect. Ther. 2019, 17, 79–88. [Google Scholar] [CrossRef]
  119. Schmidt-Hieber, M.; Teschner, D.; Maschmeyer, G.; Schalk, E. Management of febrile neutropenia in the perspective of antimicrobial de-escalation and discontinuation. Expert Rev. Anti Infect. Ther. 2019, 17, 983–995. [Google Scholar] [CrossRef] [PubMed]
  120. Le Clech, L.; Talarmin, J.-P.; Couturier, M.-A.; Ianotto, J.-C.; Nicol, C.; Le Calloch, R.; Dos Santos, S.; Hutin, P.; Tandé, D.; Cogulet, V.; et al. Early discontinuation of empirical antibacterial therapy in febrile neutropenia: The ANTIBIOSTOP study. Infect. Dis. 2018, 50, 539–549. [Google Scholar] [CrossRef] [PubMed]
Table 2. Five-point approach for optimizing antibiotic treatment in critically ill patients with neutropenia.
Table 2. Five-point approach for optimizing antibiotic treatment in critically ill patients with neutropenia.
Antimicrobial Stewardship StrategyRationaleEvidence Gaps
1. DiagnosisThe initiation of empirical antibiotic treatment should be prompted by fever and clinical signs, and not by C-reactive protein or other biomarkers.
Risk Assessment:
-
Patients with profound (<100 cell/mm3) and prolonged (>7 days) neutropenia have higher risk of infection and severity.
-
Screening for MDR pathogens (for ESBL-E, CRE, MRSA) may guide empirical antibiotics.
Appropriate screening methodology is unknown and dependent on local epidemiology.
Early and specific microbiology diagnosis is essential to directed therapy and minimizing the use of broad-spectrum antibiotics.Does not substitute conventional culture-based methods.
Cascade or selective antimicrobial susceptibility reports promote available narrow-spectrum antibiotics.No guidelines available.
Rapid antimicrobial susceptibility testing improves management of antibiotic therapy in patients with Gram-negative sepsis.Gram-positive infections.
2. DrugAppropriate empirical antibiotic is associated with better patient outcomes.
Facility-specific treatment guidelines standardize prescribing practices based on local epidemiology.
Monotherapy versus combination therapy has equivalent efficacy.
Therapeutic reconciliation is an effective strategy in preventing adverse drug reactions.
3. DoseUnderdosing of antibiotics is associated with treatment failures and worse outcomes.
Administer loading dose when indicated.
Prolonged infusion of beta-lactams is associated with better outcomes.
TDM achieves the efficacy dose and avoids overdosing and adverse effects.Further studies are needed to demonstrate the benefit of beta-lactam TDM.
4. Duration (and early discontinuation)Shorter antibiotic administration seems safe, does not have a detrimental impact on outcomes, and reduces the use of broad-spectrum antibiotics.Further studies are needed to reinforce the evidence and to evaluate the impact on resistance.
5. De-escalationDe-escalation based on microbiology diagnosis is safe, does not have a detrimental impact on outcomes, and reduces the use of broad-spectrum antibiotics.Further studies are needed to reinforce the evidence and to evaluate the impact on resistance.
PK/PD—pharmacokinetics/pharmacodynamics; TDM—therapeutic drug monitoring.
Table 3. Suggested empirical antibiotic treatment options for high-risk neutropenic patients *.
Table 3. Suggested empirical antibiotic treatment options for high-risk neutropenic patients *.
Local EpidemiologyPreferred Antibiotic TreatmentAlternative Antibiotic Treatment
Without risk for E-ESBL, CRE, MDR Pseudomonas aeruginosa, or MRSACefepime or

Piperacillin/tazobactam or

Meropenem or

Imipenem/cilastatin
Ceftazidime–avibactam or

Aztreonam (consider addition of vancomycin for Gram-positive coverage) or

Aminoglycoside (uncomplicated bloodstream infections with complete source control)
Presumed or confirmed extended-spectrum β-Lactamase-producing EnterobacteralesMeropenem or

Imipenem/cilastatin
Ceftazidime–avibactam or

Aztreonam (consider addition of vancomycin for Gram-positive coverage) or

Aminoglycoside (uncomplicated bloodstream infections with complete source control)
Presumed or confirmed carbapenem-resistant EnterobacteralesCeftazidime–avibactam or

Meropenem–vaborbactam or

Imipenem/cilastatin–relebactam
Cefiderocol or

Aminoglycoside (uncomplicated bloodstream infections with complete source control)
Presumed or confirmed difficult-to-treat Pseudomonas aeruginosaCeftolozane–tazobactam or

Ceftazidime–avibactam or

Imipenem/cilastatin–relebactam
Cefiderocol or

Aminoglycoside (uncomplicated bloodstream infections with complete source control)
Presumed or confirmed MRSAAdd vancomycinAdd Daptomycin or

Linezolide
* [2,3,41,83,84,85,86,87,88].
Table 4. Suggested dosing of antibiotics for the treatment of infections in high-risk patients with neutropenia.
Table 4. Suggested dosing of antibiotics for the treatment of infections in high-risk patients with neutropenia.
AntibioticAdult Dosage, Assuming Normal Renal and Liver Function
Amikacin20 mg/kg/dose IV (subsequent doses and dosing interval based on pharmacokinetic evaluation)
Aztreonam2 g IV every 8 h, infused over 3 h
Cefepime2 g IV every 8 h, infused over 3 h
Ceftazidime–avibactam2.5 g IV every 8 h, infused over 3 h
Ceftolozane–tazobactam3 g IV every 8 h, infused over 3 h
Gentamicin7 mg/kg/dose IV (subsequent doses and dosing interval based on pharmacokinetic evaluation)
Imipenem/cilastatin500 mg IV every 6 h, infused over 3 h
Imipenem/cilastatin–relebactam1.25 g IV every 6 h, infused over 30 min
Meropenem2 g IV every 8 h, infused over 3 h
Meropenem–vaborbactam4 g IV every 8 h, infused over 3 h
Piperacillin-tazobactam4.5 g IV every 6 h, infused over 3 h
Plazomicin15 mg/kg/dose IV (subsequent doses and dosing interval based on pharmacokinetic evaluation)
VancomycinLoading dose up to 35 mg/kg IV (maximum 3 g) and maintenance dose 15–20 mg/kg 8–12 h, adjusted to achieve target AUC24 of 400–600 (subsequent doses based on pharmacokinetic evaluation)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alves, J.; Abreu, B.; Palma, P.; Alp, E.; Vieceli, T.; Rello, J. Antimicrobial Stewardship on Patients with Neutropenia: A Narrative Review Commissioned by Microorganisms. Microorganisms 2023, 11, 1127. https://doi.org/10.3390/microorganisms11051127

AMA Style

Alves J, Abreu B, Palma P, Alp E, Vieceli T, Rello J. Antimicrobial Stewardship on Patients with Neutropenia: A Narrative Review Commissioned by Microorganisms. Microorganisms. 2023; 11(5):1127. https://doi.org/10.3390/microorganisms11051127

Chicago/Turabian Style

Alves, Joana, Betânia Abreu, Pedro Palma, Emine Alp, Tarsila Vieceli, and Jordi Rello. 2023. "Antimicrobial Stewardship on Patients with Neutropenia: A Narrative Review Commissioned by Microorganisms" Microorganisms 11, no. 5: 1127. https://doi.org/10.3390/microorganisms11051127

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

Article Metrics

Back to TopTop