Antibiotics in anesthesia and critical care
Review Article | Data-Driven Clinical Practice and Policy Making

Antibiotics in anesthesia and critical care

Antoine Premachandra1, Pierre Moine1,2,3

1Department of Intensive Care, Hôpital Raymond Poincaré, Groupe Hospitalo-Universitaire GHU AP-HP, University Versailles Saint Quentin-University Paris-Saclay, Garches, France; 2Laboratory of Infection & Inflammation - U1173, University of Versailles Saint-Quentin-en-Yvelines (UVSQ) - University Paris-Saclay - Institut National de la Santé et de la Recherche Médicale (INSERM), Garches, France; 3Fédération Hospitalo-Universitaire FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), Garches, France

Contributions: (I) Conception and design: Both authors; (II) Administrative support: P Moine; (III) Provision of study materials or patients: Both authors; (IV) Collection and assembly of data: Both authors; (V) Data analysis and interpretation: Both authors; (VI) Manuscript writing: Both authors; (VII) Final approval of manuscript: Both authors.

Correspondence to: Pierre Moine, MD, PhD. Department of Intensive Care, Hôpital Raymond Poincaré, Groupe Hospitalo-Universitaire GHU AP-HP, University Versailles Saint Quentin-University Paris-Saclay, 104 Bd Raymond Poincaré, 92380 Garches, France; Laboratory of Infection & Inflammation - U1173, University of Versailles Saint-Quentin-en-Yvelines (UVSQ) - University Paris-Saclay - Institut National de la Santé et de la Recherche Médicale (INSERM), Garches, France; Fédération Hospitalo-Universitaire FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), Garches, France. Email: pierre.moine@aphp.fr.

Abstract: Sepsis is life-threatening organ dysfunction due to a dysregulated host response to an underlying acute infection. Sepsis is a major worldwide healthcare problem. An annual estimated 48.9 million incident cases of sepsis is reported, with 11 million (20%) sepsis-related deaths. Administration of appropriate antimicrobials is one of the most effective therapeutic interventions to reduce mortality. The severity of illness informs the urgency of antimicrobial administration. Nevertheless, even used properly, they cause adverse effects and contribute to the development of antibiotic resistance. Both inadequate and unnecessarily broad empiric antibiotics are associated with higher mortality and also select for antibiotic-resistant germs. In this narrative review, we will first discuss important factors and potential confounders which may influence the occurrence of surgical site infection (SSI) and which should be considered in the provision of perioperative antibiotic prophylaxis (PAP). Then, we will summarize recent advances and perspectives to optimize antibiotic therapy in the intensive care unit (ICU). Finally, the major role of the microbiota and the impact of antimicrobials on it will be discussed. While expert recommendations help guide daily practice in the operating theatre and ICU, a thorough knowledge of pharmacokinetic/pharmacodynamic (PK/PD) rules is critical to optimize the management of complex patients and minimize the emergence of multidrug-resistant organisms.

Keywords: Antibiotics; surgical prophylaxis; microbiota; septic shock; pharmacokinetic/pharmacodynamic (PK/PD)


Submitted Nov 09, 2022. Accepted for publication Nov 06, 2023. Published online Dec 13, 2023.

doi: 10.21037/atm-22-5585


Introduction

Sepsis is life-threatening organ dysfunction due to a dysregulated host response to an underlying acute infection (1-3). Sepsis is a major worldwide healthcare problem (1,3). An annual estimated 48.9 million incident cases of sepsis is reported, with 11 million (20%) sepsis-related deaths (1).

Administration of appropriate antimicrobials is one of the most effective therapeutic interventions to reduce mortality (3). The severity of illness informs the urgency of antimicrobial administration (3). Nevertheless, even used properly, they cause side effects and contribute to the development of antibiotic resistance (4). Both inadequate and unnecessarily broad empiric antibiotics are associated with higher mortality and also select for antibiotic-resistant germs (4,5). In this narrative review, we will first discuss important factors and potential confounders which may influence the occurrence of surgical site infections (SSIs) and which should be considered in the provision of perioperative antibiotic prophylaxis (PAP). Then, we will summarize recent advances and perspectives to optimize antibiotic therapy in the intensive care unit (ICU) (6).


Surgical antibiotic prophylaxis (AP)

In Europe and the USA, SSI are one of the most common causes of hospital-acquired infections (7,8). They occur in up to 5% of patients who had surgery, and increase significantly hospital length of stay, morbidity and mortality, and costs of hospitalization (7-9). These infections are largely preventable and application of evidence-based strategies may prevent approximately 50% of SSIs (Table 1) (7-18).

Table 1

Evidence-based recommendations for the prevention of surgical site infections (7,8,10-18)—recommendations provided for adults

Antibiotic prophylaxis
   • Indications
    o Operative procedures with a high incidence of SSIs, or when foreign materials are implanted—Altemeier II (15)
    o High morbidity attributable to SSI
   • Optimal selection
    o Antibiotics effective against the pathogens most likely to be encountered during the surgery with a good diffusion in the targeted tissues (14)
    o Preferred β-lactams: first- and second-generation cephalosporins
    o Primarily agents not commonly used in curative antibiotic therapy
   • Route
    o Intravenous administration
   • Optimal timing
    o Before the surgical incision or within 60 minutes prior surgical incision
    o Vancomycin and fluoroquinolones within 60–120 minutes prior to incision
    o Timed such that a bactericidal concentration of the agent is established in the serum and tissues when the incision is made (10,12)
    o “At induction of anesthesia”
   • Dosing
    o Single dose of prophylactic antimicrobial agent (12)
   • Obese patients and weight-adjusted dosing
    o No recommendation/unresolved issue
   • Intraoperative redosing during prolonged procedures
    o Re-dosing should be based on the half-life of the antibiotic [redosing after two half-lives of the drug (16,17)] and excessive blood loss (>1,500 mL)§
   • Postoperative AP optimal duration
    o Surgical antibiotic prophylaxis should not be prolonged after completion of the operation or for less than 24 hours
    o “In clean and clean-contaminated procedures, do not administer additional prophylactic antimicrobial agent doses after surgical incision is closed in the operating room, even in presence of drain” (10)
Other evidence-based recommendations
   • Preoperative showering on at least the night before the operative day (soap or antiseptic agent)
   • Decolonization with mupirocin ointment in nasal carriers of S. aureus undergoing cardiothoracic and orthopedic surgery
   • Hair should either not be removed (use a clipper if absolutely necessary)
   • Alcohol-based skin preparation
   • Surgical hand preparation: scrubbing with antimicrobial soap and water or using alcohol-based hand rub
   • Enhanced nutritional support
   • Perioperative normothermia
   • Blood pressure control/adequate volume replacement—goal-directed fluid therapy suggested
   • Perioperative glucose control <200 mg/dL (between 110 and 150 mg/dL)
   • Increased intraoperatively and post-extubation 80% FiO2 (challenged) (13)

, each institution should develop guidelines/protocols for the proper surgical prophylaxis to standardize practices. , randomized controlled trials to evaluate the benefits and harms of weight-adjusted parenteral antimicrobial prophylaxis dosing and its effect on the risk of SSI were not identified in the literature (10). It is recommended to increase the single preoperative prophylactic antimicrobial agent dose for select prophylactic antimicrobial agents in obese and morbidly obese patients: for cefazolin 3.0 g if >120 kg; for aminoglycosides, dosing is calculated using the patient’s ideal body weight plus 40% of the difference between the actual and ideal body weight; vancomycin should be dosed at 15 to 30 mg/kg (10,17). §, randomized controlled trials to evaluate the benefits or harms of intraoperative redosing of parenteral prophylactic antimicrobial agents for the prevention of SSI were not identified in the literature (10). SSI, surgical site infection; AP, antibiotic prophylaxis; OR, operating room.

SSIs are therefore a quality indicator and a pay-for performance P4P metric in the United States (19). The Surgical Care Improvement Project (SCIP) performance has promoted three perioperative antibiotic recommendations: (I) appropriate timing: prophylactic antibiotic (PA) received within 1 hour prior to surgical incision; (II) drug selection: appropriate PA selection consistent with published guidelines; (III) duration: PA discontinued within 24 hours after surgery end time (48 hours for cardiac patients) (4-10,19-23). Wherefore, SSI prevention guidelines are published by major scientific and professional organizations (10-12,18), and are periodically revisited (12,23). PAP, among other evidence-based recommendations, is one of the most effective strategies for preventing SSIs, and considered a standard of care (7,8,10,11,13,18,19,24,25). Nevertheless, these SCIP recommendations failed to provide improved surgical outcomes (26). These best practice measures may be too general and other potential contributing factors, both patient-specific and non-specific, might require some considerations in clinical practice (Table 2). Nonetheless, these considerations would concern both surgery and critically ill patients.

Table 2

Factors which likely contribute to surgical site infections and are not targeted by SCIP measures

Selection of perioperative antibiotics according to effectiveness in specific types of surgery
Variability in antibiotic pharmacokinetics within various type of surgical patients/populations
Antibiotic concentrations at the “site of infection”
Antibiotic pharmacokinetic/pharmacodynamic characteristics
Appropriate antibiotic dosing and redosing
Obesity
Multidrug-resistant organisms
Local antibiotic resistance patterns
Allergies

SCIP, Surgical Care Improvement Project.

Effectiveness of selected antibiotic agents according to type of surgery

A fundamental question underlying the selection of antibiotics for PAP is whether the choice of specific intravenous antibiotics according to the type of surgery truly matters. The optimal choice of antibiotic for different types of surgery has not yet been definitively established (14). Current recommendations regarding choice of PAs are the result of consensus among multidisciplinary panels of experts and based on very limited evidence concerning relatively few agents, rather than based on high-quality evidence concerning true comparisons of various antibiotic options (14). However, evaluations of the association between appropriate antibiotic selection and SSI rates have identified trends approaching statistical significance; in addition, individual studies have shown significant differences in the occurrence of SSI between specific comparator antibiotics (27-30). For example, in orthopedic procedures, vancomycin as a sole agent was significantly and independently associated with higher SSI rates, compared to cefazolin, clindamycin, or vancomycin-cefazolin combination (26,31). Likewise, in a RCT comparing prophylactic ertapenem and cefotetan in colorectal surgery (CRS), significantly lower rates of SSIs were reported with ertapenem (32).

The above findings suggest that the agent selected for prophylaxis may indeed be important. Significant variations in effectiveness among the approved and accepted agents for SSI prophylaxis in colorectal and orthopedic surgeries have been reported (14,26,30,31). Such variability in reported efficacy is likely multifactorial and may be explained in part by the antibiotic spectrum of antimicrobial activity against bacteria most likely to contaminate the surgical site (SS), as well as by the potential development of antibiotic resistance over time. Furthermore, differences among antibiotics in terms of their pharmacokinetic (PK) and pharmacodynamic (PD) properties within different types of surgical patient populations may also explain observed variability in prophylactic effectiveness. Related to these PK/PD properties, differences in tissue penetration of antibiotics, the use of antibiotic loading doses, weight-based (or alternative) dosing strategies, and timely antibiotic redosing during prolonged surgical procedures may each play a role in antibiotic effectiveness; however, the interplay among all of these potential factors remains to be fully understood (33). PA effectiveness may also be explained by specific factors at the site of potential SSI, such as bacterial load, phase of bacterial growth, pH, or the presence or absence of oxygen at contaminated SSs (34). To compound the issue, other confounders such as surgical technique, specific patient factors such as age and obesity or comorbidities, and disease presentations likely influence the occurrence of SSIs (30). This issue is particularly relevant when patients report a β-lactam allergy. Several studies have suggested that the use of an alternative to β-lactam antibiotics in AP, such as clindamycin or vancomycin, is associated with an increased risk of SSIs and hospital length of stay (35-37). A β-lactam allergy is reported in up to 15% of the cases in some studies (38). However, up to 90% of patients who declared penicillin allergy could in fact receive penicillin after testing (39). This may be explained by misclassification (e.g., diarrhea), by a phenomena of desensitization over time, or even because penicillins used in therapy do not share the lateral chain causing the allergic reaction (40).

Antibiotic concentration at the “site of infection”

PAP is not intended to sterilize the tissues or prevent all possible contaminants, but rather provide protection against contamination with a high bacterial inoculum of dominant skin or site-specific flora (41,42). The risk of SSI has been shown to be markedly increased if the SS is contaminated with >105 microorganisms per gram of tissue (41,43). If foreign material is present, a ten-thousand-fold lower bacterial inoculum may result in infection (41,43). One unresolved critical variable is the minimal bacterial inoculum required to establish infection in different types of surgical procedures involving different tissues and procedural characteristics, as well as in individual patients according to comorbidities (44). Nevertheless, AP is used to reduce the number of viable bacteria resulting from intraoperative contamination to a level that cannot overwhelm host defenses (41). To accomplish this, antimicrobial agents must be administered with a correct timing so that a concentration of drug which is targeted to achieve a maximal antibacterial activity is established in serum and tissues by the time the skin is incised (41). These “therapeutic” concentrations of PAs should also be maintained in both serum and tissues at the SS throughout the operation and until, at most, a few hours after the incision is closed (41). The ability of an antimicrobial to reach the target tissues is clearly a key determinant of clinical outcome (45). Tissues at various SSs are not homogenous compartments, and the distribution of drug molecules in plasma and tissue depends on their physical-chemical properties (45). It is assumed that only the “free” unbound antibiotic at the target site is responsible for antimicrobial effects (45), and therefore the most relevant concentration for antibiotic efficacy may be the unbound concentration within the extracellular or interstitial space of target tissues where many SSIs occur. Traditionally only serum drug concentrations are monitored during clinical use or PK studies, leaving drug concentrations to be estimated at the tissue sites of interest (46). And for most antibiotics used for routine SP, with the possible exceptions of vancomycin and the aminoglycosides, drug concentrations are not routinely monitored.

Key physicochemical properties of antibiotics such as lipid solubility, protein binding (PB), molecular weight, and degree of ionization are known to influence antibiotic distribution (47,48). Hydrophilic agents have typically a smaller volume of distribution (Vd) and primarily distribute into the extracellular spaces with overall limited tissue distribution. An increased Vd has been demonstrated for hydrophilic antibiotics in many critically ill patients (47-49). Conversely, lipophilic agents (such as fluoroquinolones, macrolides, metronidazole, clindamycin) have a larger Vd because they partition intracellularly and into adipose tissue to a greater extent. Third-spacing of drugs due to pathophysiological changes may result in significantly increased Vd of hydrophilic antibiotics yet is comparatively insignificant for lipophilic agents (47-50). In addition, more extensive plasma PB of antibiotics may decrease their ability to penetrate into many tissues (45). Variations in PB may alter the PK of highly bound antibiotics, thus potentially compromising their efficacy in SP where achieving sufficient tissue concentrations of drug is considered key to success (51,52). Finally, the antibiotic dose, time since drug administration, types of tissue at the SS, and the patient’s comorbid conditions may also affect antibiotic distribution (53).

Variability in antibiotic pharmacokinetic within individual surgical patients and specific surgical populations

Large inter- and intra-individual variability is observed in the PK of intravenous antibiotics. A wide range of drug exposure is often observed when administering a fixed dose of drug to large numbers of patients within a target patient population (54-56). The variability in antibiotic PK within various surgical patients or populations remains to be adequately assessed. There is a lack of well-designed antibiotic PK studies within surgical populations, and population-PK models are almost non-existent. Their potential impact on appropriate drug dosing are therefore not well understood. Antibiotic PK/PD properties including the Vd, degree of PB, routes of drug metabolism, and rates of hepatic or renal drug clearance (CL) may be substantially altered by numerous patient-specific characteristics: underlying comorbidities such as obesity, renal or liver impairments; presence and degree of inflammation; need for cardiopulmonary bypass; complex and prolonged procedures such as liver transplant surgery; and critical illness resulting from trauma, sepsis, burn, or neurological insult (57). For example, both Vd and CL may be either increased or decreased in critically ill patients (58-60). Significant capillary leak syndromes, aggressive fluid resuscitation/loading, blood product transfusions, reduction in albumin serum concentration, advanced liver disease, presence of extra-corporal circuits, burns, trauma, surgical procedures, and positive pressure ventilation have all been shown to contribute to alterations in the PKs of many drugs (47,49,57). In addition, maldistribution of blood flow in the microcirculation may further decrease drug concentrations in certain tissues (49). The clinical importance of alterations in Vd and CL is particularly relevant for hydrophilic antimicrobials, such as β-lactams, vancomycin, linezolid, aminoglycosides or colistin (57). Although it is possible that some physiological alterations (e.g., renal impairment) may actually improve the PK/PD performance of perioperative antibiotics in some surgical patients, the potential for antibiotics to be adversely affected by such alterations with resultant increased risk of SSI is also highly likely and of greater concern. Furthermore, PK/PD alterations occurring within an individual patient may change according to the varying stages of illness, providing ample opportunity for significant intra-patient variability over time (57).

Again, the PK/PD properties of antibiotics in specific surgical populations or critically ill patients, and the corresponding clinical significance are not well characterized. The optimal dosing of antibiotics for surgical prophylaxis (SP) remains elusive. The duration of AP is generally limited to 24 hours postoperatively, and most antibiotics used prophylactically are generally well tolerated by patients. Therefore, the major concern with optimized dosing of perioperative antibiotics is related to insufficient dosing resulting in increased risk of SSI rather than excessive dosing resulting in drug accumulation and toxicity. Because significant and relatively unpredictable variability of PK parameters may exist within individual patients, the typical “one dose fits all” approach to AP which is currently used is likely flawed.

Antibiotic pharmacokinetic/pharmacodynamic PK/PD characteristics

Antibiotics are classified on the basis of their patterns of antibacterial activity as concentration dependent or time-dependent agents (Table 3) (3,6,55,61-67). The PK/PD indices which are thought to best represent the antibacterial activity of concentration-dependent antibiotics are the ratio of maximum serum antibiotic concentration (Cmax) to MIC (Cmax/MIC) and/or the ratio of the area under the concentration-time curve during a 24-hour dosing period (AUC0–24) to MIC (AUC0–24/MIC), whereas time-dependent antibiotics are best represented by the duration of time (expressed as a percentage of a given dosing interval or period) in which unbound antibiotic concentrations exceed the MIC (ƒT>MIC) (34,61).

Table 3

In vitro PK/PD parameters correlating with antimicrobial efficacy

Bactericidal pattern activity in vitro Antimicrobial agent Physical properties PK/PD index associated with bacterial killing Drug concentration target PK/PD optimization
Concentration-dependent Aminoglycosides Hydrophilic Cmax/MIC Cmax/MIC 8–10 30 min infusion and TDM
Fluoroquinolones Lipophilic AUC0–24/MIC or Cmax/MIC AUC0–24/MIC >125–250
Colistin Hydrophilic AUC0–24/MIC Unknown
Daptomycin Hydrophilic AUC0–24/MIC AUC0–24/MIC >200–600
Time-dependent Penicillins Hydrophilic T>MIC Cmin >MIC, ƒT >4–5, MIC 50–100% Extending the %ƒT>MIC (LD & EI or CI, and TDM)
Cephalosporins Hydrophilic T>MIC
Monobactams Hydrophilic T>MIC
Carbapenems Hydrophilic T>MIC
Vancomycin Hydrophilic AUC0–24/MIC AUC0–24/MIC 400 LD and CI & TDM
Linezolid Lipophilic AUC0–24/MIC ƒAUC0–24/MIC ≥80–120, Cmin 2–8 mg/L
Clindamycin Lipophilic AUC0–24/MIC %T>MIC 85%

PK/PD, pharmacokinetic/pharmacodynamic; MIC, minimum inhibitory concentration; Cmax/MIC, ratio of maximum serum antibiotic concentration to MIC; AUC0–24/MIC, ratio of the area under the concentration-time curve during a 24-hour dosing period (AUC0–24) to MIC; ƒT>MIC, duration of time (expressed as percentage of dosing interval or period) in which unbound antibiotic concentrations exceed the pathogen MIC; LD, loading dose; EI, extended infusion; CI, continuous infusion; TDM, therapeutic dosing monitoring; LD, loading dose.

Time-dependent antibiotics

β-lactams, the most commonly prescribed family of antibiotics for SP and ICU patients (12), are considered time-dependent antibiotics with ƒT>MIC being the best predictor of efficacy (34,54,61,66,68,69). However, extrapolations from neutropenic animal models describe differences among each β-lactam/organism combinations regarding the specific percentage of ƒT>MIC which is needed in order to achieve maximum efficacy (61). It has been proposed that bacteriostatic effects, rather than bactericidal, may be sufficient to achieve optimum antibacterial efficacy in non-neutropenic hosts (61); ƒT>MIC values required for bacteriostasis are approximately 29–34%, 35–55%, and 20–26% for penicillins, cephalosporins, and carbapenems, respectively (54,61,68). On the other hand, achievement of the maximal bactericidal effect requires ƒT>MIC of 50%, 60–70% and 40%, respectively for these β-lactam classes (54,61). In vitro, in vivo, and clinical studies have also suggested that larger drug exposures may be required in certain populations (70-75). In a febrile neutropenic population treated with meropenem, patients with a calculated median ƒT>MIC of 59% had a poor clinical response compared to those with ƒT>MIC >75% (76). This study also demonstrated a significant association between increasing %ƒT>MIC and increased probability of favorable clinical response (76). Other clinical data also suggest that higher and more prolonged exposure is associated with improved outcomes in critically ill patients (55,77-79). In an analysis of cefepime in gram-negative infections, clinical cure and microbiological eradication rates were improved when ƒT>MIC reached 100% (80). Similar associations of ƒT>MIC with drug efficacy for SP do not exist; however, based on consistency in other patient populations, it is reasonable to extrapolate that ƒT>MIC may also be applicable to surgical populations in the setting of PAP.

For the treatment of established infections in humans, the optimal %ƒT>MIC remains controversial and definitive values have yet to be established in specific patient populations. Moreover, whether time-dependent bactericidal action of β-lactams is achieved at any concentration above the MIC or whether some dose dependency is still operative is unclear (70,75,80). Data suggest that maximum bactericidal activity of β-lactams occurs at concentrations approximately ≥4–5 times the MIC for the entire dosing interval. Also, an association between AUC0–24/MIC and outcomes has been described for the cephalosporins (78,81-86), further suggesting an element of concentration dependency in the activity of these drugs since AUC0–24 reflects both the magnitude and duration of drug exposure. Moreover, there is a clear relationship between the AUC0–24/MIC parameter and the risk of development of bacterial resistance (86). This concentration-dependent effect may be observed with different pathogens, more resistant organisms or large inoculum sizes (69,87,88).

Based on available data from a variety of studies, it follows that not all β-lactams necessarily exhibit the same pharmacodynamic behavior in all organisms (77,78,80,87-90), or in patients with severe/life-threatening bacterial infections and/or infections caused by multidrug-resistant organisms (MDRO) (49). Evidence is lacking to clearly and definitively show that PD targets and response rates with β-lactams should be the same for different therapeutic endpoints, for different populations, among different pathogens, or even among strains of the same pathogen species with different antibiotic susceptibilities.

Given the previous discussion, optimal PD targets for PAP are even more unclear since all currently established PD targets concern treatment of infection rather than prevention of infection (established tissue infection rather than tissue contamination). Whether currently established PD targets for treatment of infection are applicable in determining the antibiotic dosages for SP has not been adequately studied (52,53,91,92). According to current guidelines, the goal of PAP is to achieve adequate free serum and tissue drug concentrations that exceed the MICs for pathogens most likely to contaminate the SS across the period of potential contamination (12,22,23). The importance of adequate antibiotic concentrations at closure in the prevention of SSI has been previously shown (22,93,94), and the SCIP project also recommends dosing to ensure adequate antimicrobial concentrations until wound closure (12,22,23). Because intra-operative contamination may occur at any time during the procedure, a β-lactam PD target of ƒT>MIC of 100% over the full duration of the surgical procedure is a reasonable goal for optimization of β-lactam prophylactic therapy (51-53), especially considering the many uncertainties of β-lactam PK/PD in various surgical populations and the lack of literature to the contrary. Nevertheless, it cannot be ruled out that Cmax/MIC and/or AUC0–24/MIC may also perhaps play a role in predicting optimal β-lactam activity in prophylactic therapy. Clearly, the optimal PK/PD parameters for β-lactams in SP remain to be established.

Concentration-dependent antibiotics

While the focus of this discussion has been on the β-lactams, similar uncertainties regarding optimal PK/PD targets for concentration-dependent antibiotics also exist. While Cmax/MIC and AUC0–24/MIC are the PK/PD parameters associated with concentration-dependent antibiotics, maintaining adequate concentrations at surgical wound closure have also been shown to be associated with reduced SSI during prophylactic aminoglycoside use (93). The optimal PD targets for concentration-dependent antibiotics in prophylaxis become equally uncertain when considering variables such as inoculum size and the potential timing of bacterial contamination (51,52). Unless the antibiotic has unusually high penetration and long persistence in tissues, achieving a high Cmax/MIC ratio prior to surgical incision should have no benefit if a high-inoculum bacterial contamination occurs near the end of a prolonged surgical procedure.

Most published antimicrobial PK/PD indices are based on antibiotic concentrations in the blood rather than in tissues. Tissue concentration of antibiotic at the wound site at the time of potential bacterial contamination is a critical factor in determining the efficacy of prophylaxis, but data related to tissue penetration of antibiotics are limited and highly variable among various studies (53). In heterogeneous patient populations, extrapolating optimal PK/PD indices to an outcome (SSI) which may be relatively infrequent in many types of procedures becomes somewhat problematic from the perspective of conducting meaningful clinical studies. Finally, as already underlined, the activity of the same antibiotic may be variably influenced by specific factors at the site of contamination (34,51).

Issues related to appropriate antibiotic dosing and redosing

Selection and dosing of antimicrobials based on PK/PD principles has become common in order to optimize therapeutic outcomes in the treatment of infections and to prevent the emergence of resistance. As discussed, use of these same PK/PD principles for evaluating antibiotics for SP is largely unsupported by clinical evidence and remains controversial (51,53,62). The actual guideline-recommended drugs and dosing regimens have been relatively unchanged over the past 20 years (12,23). Limited published data exist regarding appropriate antimicrobial selection and dosing for prophylaxis. To compound the problem, the antibiotic susceptibilities of clinically isolated pathogens have substantially changed over the past two decades while recommended drug doses have remained mostly unchanged (53). Thus, it is questionable whether appropriate doses and dosing frequencies of PAs are currently being used (53).

For example, standard dosing of ertapenem (1 g) was demonstrated to be more effective than cefotetan (2 g) in the prevention of SSI in patients undergoing elective CRSs (32). Whilst selection of a specific antimicrobial agent is only one of many considerations in reducing SSI rates, the primary reason for cefotetan failure in this study was most likely inferior achievement of optimal PK/PD targets against key organisms compared with ertapenem (33,53,95). Applying currently accepted PK/PD principles of antibiotic dosing and prediction models (i.e., Monte Carlo Simulation), cefotetan was shown to have extremely poor predicted PK/PD performance against Escherichia coli, Bacteroides fragilis and Staphylococcus aureus over the first 3–4 hours post-infusion (53). Cefotetan failed to meet any specified PK/PD target (e.g., ƒT>MIC of 100%) at any dose (1 and 2 g doses) or at any time point after dosing against any of the three targeted organisms, suggesting a cefotetan 2 g dose is entirely inadequate for routine use as a prophylactic agent in CRS (53). In comparison, ertapenem 1 g had virtually 100% probability of achieving 100% ƒT>MIC across the entire 4-hour period post-infusion and against all three organisms (53).

The optimal strategy for intraoperative redosing of PAs is still debated. According to published guidelines, redosing of a drug is needed to ensure adequate serum and tissue concentrations of the antibiotic if the duration of the procedure exceeds two half-lives of the drug or there is excessive blood loss (12,23). Increased duration of surgery is a well-known significant risk factor for SSIs (32,96,97). The relative risk of SSI significantly increases after roughly 2 hours in the operating room and increases further with every extra hour of operative time thereafter (97). However, there are no convincing data demonstrating significantly lower SSI rates with multiple-dose regimens versus single doses (12,91,92). It can be argued that additional dosing may increase the risk of resistant organisms and Clostridium difficile colitis (91). Nonetheless, it is recommended that additional intraoperative doses may be needed to optimize the potential effectiveness of the prophylaxis depending on the length of surgery, antibiotic PK/PD characteristics, and individual patient characteristics (51-53). Administration of β-lactams via extended or continuous infusions after an initial loading dose may optimize β-lactam PK/PD characteristics without increasing toxicity or cost (22). In addition, administering higher doses by extended infusion may also allow for more optimal drug exposures against strains of less-susceptible organisms with relatively high MICs (61). These alternative administration methods have already been suggested to optimize the PD profile of β-lactams for both therapeutic treatment of infection and SP (98,99).

In adults, the dosing of many antimicrobials is not typically based on body weight as is common in children (12,100). Administration of a “standard” dose instead of a weight-based dose may be insufficient to achieve adequate antibiotic concentrations in blood and tissues in many adults, particularly in patients with high body weight, increased body mass or abnormal mass composition (100). It was shown that up to 80% of patients undergoing elective total joint and spine surgery who receive vancomycin are under-dosed when given a standard 1-g dose preoperatively rather than 15 mg/kg (101). In that study, a 1 g dose would have been appropriate only for patients weighing <67 kg (101). Although weight-based dosing appears to offer clear PK/PD advantages in certain patient populations, data to support such recommendations are scarce and weight-based dosing is recommended for only three drugs in current prophylaxis guidelines (10,12).

Obesity

Differences in proportion of adipose tissue, lean muscle tissue and fluid status in obese patients can greatly affect antibiotic PK through alterations in drug distribution, metabolism and excretion (102-104). The Vd may be dramatically different compared to normal-weight patients (105). Beyond a greater percentage of adipose tissue compared to non-obese individuals, physiological changes such as tissue blood flow and changes in cardiac output may also alter drug distribution of both hydrophilic and lipophilic drugs in obesity (105). In addition, physiological changes in the liver and kidneys can alter metabolism and excretion (105). However, as already discussed, the dosing of many prophylactic antimicrobials in adults is not based on body weight or composition despite the known physiological differences and PK alterations known to occur in obese patients (12,100,105). Even if comprehensive reviews on antibacterial use in obese patients are available in the literature (65,106,107), limited published data exist regarding appropriate dosing of antimicrobials for prophylaxis in this population (56). Evidence-based antibiotic dose adjustments for obesity are lacking for many drugs, including antimicrobials for which optimal dosing in this population continues to present significant challenges (105,107,108). Again, the standardized “one dose fits all” approach to antibiotic dosing for prophylaxis cannot be assumed adequate for obese patients (107). In the study comparing ertapenem and cefotetan in CRS, nearly 30% of the enrolled patients were obese [body mass index (BMI) >30 kg/m2], yet no dose adjustments were made for either drug (32,102). Rates of SSI were significantly higher in patients with a BMI ≥30 kg/m2 compared to those with BMI <30 kg/m2 regardless of the antibiotic administered, perhaps because drug doses were inadequate for obesity (102). It is generally stated in published recommendations that PAs should be given in doses which are adequate based on patient weight, adjusted dosing weight, or BMI (12,23); however, specific doses for individuals with various BMI are not often provided. The 2005 National Surgical Infection Prevention project made weight-based dose recommendations for adults, but these doses were primarily derived from published pediatric recommendations (12,23). In 2013, although AP dosing was discussed in the context of obesity, weight-based dosing recommendations for adults were not addressed due to a lack of clinical data (12). If some authors argue that increasing the dose of hydrophilic antibiotic such as cephalosporins may be unable to increase tissue concentration, recent observational data suggest that higher dose of cefazolin would be more effective (109,110). Appropriate drug dosing for SP in obese patients is clearly an area in which well-designed clinical studies and evidence-based recommendations are sorely needed.

Multidrug resistant organisms (MDROs)

Emerging new pathogens and changing patterns of antibiotic resistance are potential factors further contributing to SSIs during use of traditional prophylactic drugs. Enterococcus species, methicillin-susceptible (MSSA) and methicillin-resistant S. aureus (MRSA) are playing an increasingly important role in SSIs (32,103,104,111,112). Approximately 50% of SSI complicating knee and hip primary arthroplasty are attributable to S. aureus and 40% of these infections are caused by MRSA (108). More challenging gram-negative bacteria such as extended-spectrum beta-lactamase (ESBL)-producing Enterobacteriaceae (ESBL-PE), inducible AmpC-cephalosporinase-producing Enterobacteriaceae, and multidrug-resistant P. aeruginosa are also more commonly found in SSIs (32,103,104,111,113-115). The prevalence of carriage of resistant GNB among patients about to undergo surgery in which endogenous GNB are the main cause of SSI varies from 1% to 46% and varies by geography and over time (116). The increasing proportions of SSIs caused by organisms intrinsically resistant to accepted β-lactams may seriously confound the optimal choice of antibiotic for SP (112,116). Patients colonized with ESBL prior to CRS have an increased risk of SSI from ESBL (113). The data establishing the efficacy of these accepted β-lactams for SP were generated before the appearance of these MRDOs (14). Exemplifying this point, the study comparing ertapenem and cefotetan for CRS found cefotetan resistance in 67% and 46% of isolated SSI pathogens in the cefotetan and ertapenem arms, respectively (32). Additionally, 16% of isolated pathogens in both treatment groups were also resistant to ertapenem (102). In a multicenter prospective nonrandomized, nonblinded, interventional study, presurgical screening for ESBL-PE carriage before CRS and personalizing prophylaxis for carriers is efficacious in reducing SSIs (117). In intention-to-treat analysis, any type of SSI rates [45/209 (21.5%) in the baseline phase as compared to 47/269 (17.5%) in the intervention phase] did not reach statistical difference, nor deep SSI rates (117). Thus the authors did not prove the effectiveness of the intervention (117). The as-treated analysis showed that ertapenem prophylaxis for ESBL-PE carriers significantly decreased the risk of SSI (a 33% reduction) and the risk of SSI caused by ESBL-PE (a 86% reduction), supporting the efficacy of the personalized prophylaxis strategy (117).

In the US, standard AP regimens (cefazolin or cefuroxime) used in coronary artery bypass graft and hip/knee arthroplasty procedures had inadequate activity against more than 50% of reported SSI pathogens (118). When vancomycin and/or an aminoglycoside were combined with cefazolin or cefuroxime, 14% to 19% of SSI pathogens were still resistant to these regimens (104). Of interest, the organism most commonly associated with antimicrobial failure in this study was MSSA, a pathogen which was susceptible in vitro to all administered regimens (104). This finding further highlights that the problem of SSI is not only one of appropriate drug selection, but also likely involves a constellation of aforementioned host-specific, pathogen-specific, PK/PD-related, and procedure-specific variables.

There is scant evidence to suggest that broad-spectrum antimicrobial agents result in lower rates of postoperative SSI compared with “older” antimicrobial agents with a narrower spectrum of activity, with notable exceptions being ertapenem in CRS, or prophylactic use of therapeutic antibiotics based on the results of preoperative bile cultures in hepato-pancreatico-biliary (HPB) surgery or broad-spectrum antibiotics in pancreatoduodenectomy (12,32,112,119). Some data have even demonstrated an association between development of SSI due to MSSA and use of vancomycin as a sole agent for AP (120), and that the addition of anti-MRSA agents (including vancomycin) does not reduce the incidence of MRSA infections (121). Likewise, there is little evidence and no consensus regarding AP in a patient with past infection/colonization with MDROs, nor in patients with risk factors for MDROs (12,116). The frequent use of broad-spectrum antimicrobials for prophylaxis may further contribute to the selection of drug-resistant pathogens, thereby decreasing prophylaxis efficacy and future options (104,111). Whether past infection or colonization with MDROs, or potential risk factors for MDRO, led to significant differences in effectiveness of AP and therefore require more broad-spectrum “customized patient-specific” regimens rather than the more traditional, narrow-spectrum “standardized population-based” regimens for PAP is still unclear. This topic was not addressed in recent guidelines for SSI prevention issued by the US Centers for Disease Control (10) and the World Health Organization (11). For other professional groups, the decision to alter PAP in known carriers of MDROs must take into consideration the pathogen, its susceptibility profile, the host, the procedure, and the distance between the carriage site and the operative site (12,116).

For cardiac and orthopedic procedures in patients known to be at high risk for MRSA infection or in hospitals with high rates of MRSA related SSIs, glycopeptides are recommended as part of the perioperative antimicrobial regimens (12,23). However, patients may still develop gram-positive and/or MRSA infections despite the use of glycopeptides as recommended (52,118,121,122). A previous study compared cefazolin to vancomycin for prophylaxis in patients undergoing cardiac surgery in an institution with a high prevalence of MRSA (123). Although not statistically different, patients who developed an SSI following cefazolin prophylaxis were more likely to be infected with MRSA, whereas those who received vancomycin were more likely to be infected with MSSA (123). Furthermore, while the choice of antimicrobial agent used for prophylaxis may have influenced the infecting organisms, it did not apparently alter the observed infection rates (118,122). These findings are consistent with those reported in the orthopedic literature (124) and have been confirmed by three systematic reviews (52,125-127).

Exceptions to the above findings regarding the tailoring of PAs to known risk factors for MDROs may include transplant recipients with history of colonization or infection with resistant pathogens, assuming that these pathogens are relevant to SSIs in the planned procedure (12,128). However, targeted or individualized perioperative antimicrobial regimens have not been clearly shown to result in reduction in SSI rates in such circumstances (103,129). No rigorous studies have formally addressed optimal prophylaxis for these patients (129). Some data provides evidence for screening for MDRO carriage before elective orthopedic, cardiac and colorectal surgeries and personalized AP for patients who screen positive (117).

Antibiotic-associated harm

Antibiotics-associated harm are more and more highlighted, such as drug toxicity, mitochondrial dysfunction, allergic reaction, microbiota alteration and C. difficile colitis that has been shown to be as frequent after AP than after antibiotic treatment (130,131). Moreover, PAs side effects are poorly reported (132). To ensure PA effectiveness and limit the risk of side effects, PAP must follow recommendations published by major organizations (10-12,23). Each institution should develop protocols for the proper surgical prophylaxis to standardize practices.


Antibiotics in critical care

Septic shock and sepsis are medical emergencies and, treatment and resuscitation should begin immediately (3). Early administration of appropriate antibiotics is one of the most effective interventions to decrease mortality in patients with sepsis (3). On a given day in ICU, 48% to 70% of the patients are receiving empirical or definite antimicrobial treatments (6,133). International guidelines for management of sepsis and septic shock are published, and periodically updated (3). Antimicrobial therapy recommendations are summarized in Table 4.

Table 4

Surviving Sepsis Campaign International Guidelines for Management of Sepsis and Septic shock in adult patients—antimicrobial administration recommendations (3)

Main considerations Recommandation
Indications • Septic shock and sepsis
• “For possible sepsis without shock, rapid assessment of the likelihood of infectious versus noninfectious causes of acute illness”
Optimal timing • Possible septic shock, or a high likelihood for sepsis: administer antimicrobials immediately, ideally within 1 h of recognition
• Possible sepsis without shock; it is suggested a time-limited course of rapid investigation and if concern for infection persists, the administration of antimicrobials within 3 h from the time when sepsis was first recognized
• Low likelihood of infection and without sepsis and/or shock: defer antimicrobials while continuing to closely monitor the patient
Optimal selection • The empiric antimicrobial regimen includes at least one effective agent active against the most common causative organisms. The optimal choice depends on the source of infection, potential causative agents, local/regional prevalence of resistant organisms, local antibiogram, patient characteristics and risk factors for resistant organisms, and the severity of illness
• According to the risk of MDR organisms:
o High risk: it is suggested using two GNB agents
o Low risk: it is suggested a single GNB agent for empiric treatment
• According to the risk of MRSA (sepsis or septic shock):
o High risk: empiric GNB broad-spectrum therapy with MRSA coverage
o Low risk: it is suggested against using empiric antimicrobials with MRSA coverage, as compared with using antimicrobials without MRSA coverage
• According to the risk of fungal infection§ (sepsis or septic shock):
o High risk: it is suggested using empiric antifungal therapy over no antifungal therapy
o Low risk: it is suggested against empiric use of antifungal therapy
Antimicrobial combination • According to the risk of MDR organisms:
o High risk: it is suggested using double GNB coverage for empiric treatment over one gram-negative agent
o Low risk: it is suggested against using two gram-negative agents for empiric treatment, compared with one gram-negative agent
Route • Intravenous administration
Dosing and delivery of antibiotics • For sepsis or septic shock, it is
o Suggested using prolonged infusion of beta-lactams for maintenance (after an initial bolus) over conventional bolus infusion
o Recommended optimizing dosing strategies of antimicrobials based on accepted PK/PD principles and specific drug properties
De-escalation • For sepsis or septic shock,
o Daily assessment for de-escalation of antibiotics over using fixed durations of therapy without daily reassessment for de-escalation
o It is suggested against using double gram-negative coverage once the causative pathogen(s) and the susceptibilities are known
o Once both the pathogen(s) and susceptibilities are known, it is encouraged stopping an antimicrobial that is no longer necessary or changing an antimicrobial to narrow the spectrum
Discontinuation-duration • “For suspected sepsis or septic shock but unconfirmed infection, it is recommended continuously re-evaluating and searching for alternative diagnoses and discontinuing empiric antimicrobials if an alternative cause of illness is demonstrated or strongly suspected”
• For an initial diagnosis of sepsis or septic shock and adequate source control:
o We suggest using shorter over longer duration of antimicrobial therapy (Table 5)
o Where optimal duration of therapy is unclear, we suggest using procalcitonin and clinical evaluation to decide when to discontinue antimicrobials over clinical evaluation alone

, risk factors for MDR pathogens (3,6): proven infection or colonization with antibiotic-resistant organisms within the preceding year (3), local prevalence of antibiotic-resistant organisms (3), hospital-acquired/healthcare-associated infection (vs. community-acquired infection) (3), broad-spectrum antibiotic use within the preceding 90 days (3), concurrent use selective digestive decontamination (3), travel to a highly endemic country within the preceding 90 days and hospitalization abroad within the preceding 90 days (3), advanced poor hygiene practices in hospital, co-morbid illnesses (6), immunosuppression (6), recent hospital stay (or ICU stay) (6), prior antifungals (6), broad-spectrum antibiotics or with potent activity against intestinal anaerobes are also risk factors for MDR pathogens (6). , MRSA risk factors (3): prior history of MRSA infection or colonization, recurrent skin infections or chronic wounds, recent IV antibiotics, presence of invasive devices, hemodialysis, recent hospitalization and severity of illness. §, risk factors for fungal infection (3): Candida colonization at multiple sites, neutropenia, surrogate markers such as serum beta-D-glucan, immunosuppression, severity of illness, longer ICU length of stay; central venous catheters, persons who inject drugs, total parenteral nutrition, broad spectrum antibiotics, gastrointestinal tract perforations and anastomotic leaks, emergency gastrointestinal or hepatobiliary surgery, acute renal failure and hemodialysis, severe thermal injury, prior surgery. MDR, multidrug resistant; GNB, gram-negative bacteria; MRSA, meticillin-resistant S. aureus; PK/PD, pharmacokinetic/pharmacodynamic; ICU, intensive care unit; IV, intravenous.

Table 5

Duration of antimicrobial therapy in ICU—shorter versus longer therapy according to clinical syndrome (3,134-136)

Diseases Short course therapy (days) Long course therapy (days)
Community-acquired pneumonia 3–5 7–10
Nosocomial pneumonia (including ventilator-associated pneumonia) 7–8 10–15
Bacteremia 5–7 10–14
Intra-abdominal infection 4–8 10–15
Skin infections (cellulitis, major abscesses, wound infections) 5–6 10
Urinary tract infections 5 10

ICU, intensive care unit.

The clinical diagnosis of sepsis (suspected or documented infection complicated with life-threatening organ dysfunction) is challenging and largely over-estimated in ICU (6,137). Up to 43% of patients treated for suspected sepsis are unlikely to have an infection (137). Aggressive empirical antibiotic use—antibiotics started as soon as infection is suspected before microbiological evidence of infection—might be harmful in ICU patients (138). Patients managed under an aggressive antibiotic therapy had a more rapid start of treatment, a lower chance of receiving initially appropriate treatment, a prolonged duration of antimicrobial treatment, and significantly lower survival (138). In the last Surviving Sepsis Campaign recommendations, “for adults with possible sepsis without shock, it is suggested a time-limited course of rapid investigation and if concern for infection persists, the administration of antimicrobials within 3 hours from the time when sepsis was first recognized” (3). “For adults with a low likelihood of infection without shock, it is even suggested to defer antimicrobials while continuing to closely monitor the patient” (3). Molecular diagnostics (PCR-based systems targeting selected pathogens and resistance markers), performed alongside conventional cultures, may accelerate the diagnosis process, identifying pathogens and their susceptibilities faster than conventional methods (6). Nevertheless, further studies are warranted to fully appraise their impact on patient outcome (6).

If the appropriateness of the initial empiric antibiotic treatment is directly correlated to the mortality (6,139-141), the dosing selection of the appropriate antibiotic treatment is challenging (Table 6) (6,55,142-145). Roberts and colleagues have shown large variability of β-lactam antibiotic plasma concentrations in critically ill patients and, thus, <50% of patients achieved PK/PD targets (defined as 50% ƒT>4×MIC) (55). Underdosing of antibiotics is thus frequent in ICU patients, and these patients are less likely to have positive clinical outcome (6,55). Variability in antibiotic pharmacokinetics within individual surgical or ICU patients is already developed above. Therefore, initial higher doses and routine therapeutic drug monitoring (TDM) appears necessary to avoid underdosing (maximizing drug efficacy through optimized PK), as well as overdosing (drug-related toxicity) (6). To properly achieve PK/PD targets and allow for a personalized antibiotic dosing, both the drug serum concentrations and the actual MICs of the causative pathogens need to be measured to treat sepsis and septic shock (55). Moreover, to increase the likelihood for PK/PD attainments and potential therapeutic success, maintaining exposure via continuous or extended infusion of β-lactams (which display a time-dependent mechanism of action) may be of great clinical benefits for critically-ill patients with sepsis and septic shock, particularly with higher MIC pathogens (146-148).

Table 6

Recommended doses of initial empirical antibiotics in ICU patients with sepsis and septic shock (6,142-145)

Antibiotics Timsit et al. 2019 IDSA guidelines (Kalil et al. 2016, Tamma et al. 2022)
Ceftaroline 600 mg q12h
Ceftazidime/avibactam 2.5 g q8h 2.5 g q8h, EI 3 h
Ceftolozane/tazobactam 1.5 g q8h; or 3 g q8h (VAP) 3 g q8 h, EI 3 h
ATM/AVI 6,500 mg ATM/2,167 mg AVI q24h on day 1 followed by 6,000 mg ATM/2,000 mg AVI q24h CAZ-AVI 2.5 g q8h EI 3 h + Aztreonam: 2 g q8h EI 3 h, infused during CAZ-AVI infusion
Meropenem/vaborbactam 2 g/2 g q8h 4 g q8h, EI 3 h
Cefiderocol 2 g q8h 2 g q8h, EI 3 h
Imipenem/relebactam 500 mg/250–125 mg q6h 1.25 g q6h, EI 30 min
Eravacycline 1 mg/kg q12h 1 mg/kg q12h
Plazomicin 15 mg/kg q24h 15 mg/kg × 1 dose
Tedizolid 200 mg q24h (IV or oral)
Piperacillin/tazobactam 4.5 g q6h CI 4.5 g q6h§
Ceftazidime 6 g q24h CI 2 g q8h§
Cefepime 2 g q8h or CI 2 g q8h, EI 3 h
Aztreonam 1 g (2 g) q8h 2 g q8h
Imipenem/cilastatin 500 mg (1 g) q6h 500 mg q6h§
ESBL/AmpC: EI 30 min
CRE & CRAB: EI 3 h
Meropenem 1 g (2 g) q8h or CI ESBL/AmpC: 1–2 g q8h EI 30 min
CRE & CRAB: 2 g q8h EI 3 h
Ertapenem 1 g q24h, EI 30 min
Tigecycline 100–200 mg loading dose, then 50–100 mg q12h 200 mg loading dose, then 100 mg q12h
Gentamicin 7 mg/kg/day q24h 7 mg/kg/day q24 h
Amikacin 25–30 mg/kg/day q24h 20 mg/kg q24h
Colistin 9 MU loading dose, then 4.5 MU q8–12 h 5 mg/kg IV loading dose, then 2.5 mg × (1.5 × CrCl + 30) daily, divided q12h
Polymixin B 2.5–3.0 mg/kg/d divided in 2 daily IV doses
Vancomycin 15–30 mg/kg LD, 30–60 mg/kg every 12 h, 6 h or CI 15 mg/kg IV q8–12h, consider a LD of 25–30 mg/kg × 1 for severe illness
Linezolid 600 mg q12h 600 mg q12h
Ciprofloxacin 400 mg q8h–12h
Levofloxacin 750 mg q24h
Trimethoprim-sulfamethoxazole 8–12 mg/kg/day trimethoprim IV/PO divided q8–12h (max 960 mg trimethoprim q24h)

, drug levels and adjustment of doses and/or intervals required (142,143). , vancomycin target trough level: 15–20 mg/L. §, extended infusions may be appropriate (143). , dosing is based on colistin-base activity CBA: one million IU of colistin is equivalent to about 30 mg of CBA which corresponds to about 80 mg of the prodrug colistimethate. Polymyxin B (1 mg = 10,000 units) (142-145). ICU, intensive care unit; IDSA, Infectious Diseases Society of America; EI, extended infusion; VAP, ventilator-associated pneumonia; ATM, aztreonam; AVI, avibactam; CAZ, ceftazidime; CI, continuous infusion; AmpC, ampicillin chromosomal cephalosporinase; CRE, carbapenem-resistant Enterobacteriaceae; CRAB, carbapenem-resistant A baumannii; IV, intravenous; LD, loading dose.

Also already underlined, emerging new pathogens, changing patterns of antibiotic resistance and continued development of new types of resistance are contributing to increased morbidity, mortality, length of stay in ICU and hospital, and costs (4,149,150). Recently, CDC reported that: “More than 2.8 million antibiotic-resistant infections occur in the United States each year, and more than 35,000 people die as a result. In addition, 223,900 people required hospital care for C. difficile and at least 12,800 people died in 2017” (4). Candida auris, Enterococcus faecium [vancomycin-resistant Enterococcus (VRE)], methicillin-resistant S. aureus (MRSA), ESBL-producing Enterobacteriaceae (essentially K. pneumoniae and E. coli), inducible AmpC-cephalosporinase-producing Enterobacteriaceae (Enterobacter species), and multidrug-resistant P. aeruginosa, Acinetobacter baumannii and Stenotrophomonas maltophilia are the most frequent antimicrobial-resistant germs playing an increasingly important role in healthcare-associated nosocomial infections and community-acquired infections, and represent a global threat to human health (4,149-152).

Concerning GNB, the most important emerging β-lactamases correspond to the production of ESBL, AmpC cephalosporinases, and carbapenem-hydrolizing β-lactamases, the latest conferring resistance to almost all β-lactams (Table 7) (5,151-159). The worldwide spread of these β-lactamase-producing GNBs is an important source of concern since these β-lactamase resistance mechanisms are frequently combined with other β-lactam-resistance mechanisms [drug uptake limitations (porin loss, biofilm), drug target alterations, and/or drug efflux pump] and non-β-lactam resistance mechanisms, therefore leading to multi-drug resistant (MDR/XDR) isolates (5,151,152). Infection rates due to MDROs are increasing steadily throughout the world and antimicrobial agents active against MDROs, especially carbapenem resistant (CR) GNBs, remain limited and are associated with high rates of mortality (6,149,151,160,161).

Table 7

Ambler classification and main concerning β-lactamases. Mechanisms of resistance among pathogens (GNB) with extended-drug resistance

β-lactamases Enzymes Species Effective inhibitors Susceptibility pattern
Class A serine β-lactamases/Penicillinases ESBL: SHV, TEM, CTX-M Enterobacteriaceae Clavulanate, tazobactam, sulbactam R: AMX, TZP, CTX, CAZ, ATM
I/R: ETP
S/I/R: IMP MER
Partially inhibited by Clav-A and TZB
Class A serine β-lactamases/Carbapenemases KPC, IMI, SME, GES, SHV Enterobacteriaceae, Pseudomonas spp, Acinetobacter spp Avibactam, relebactam, vaborbactam R: AMX, TZP, CTX, CAZ ATM
I/R: ETP
S/I/R: IMP MER
Partially inhibited by Clav-A
“S”: AVI, RE, VAB
Class B metallo-β-lactamases NDM, VIM, IMP, GIM, SPM, DIM, SIM Enterobacteriaceae, Pseudomonas spp, A. baumannii, S. maltophilia, Bacteroides fragilis R: AMX, AMC, CTX,
I/R: TZP, CAZ, ETP
S/I/R: IMP, MER
S: ATM
Class C serine β-lactamases AmpC, ACT, CMY, DHA, FOX, ADC Enterobacteriaceae, Pseudomonas spp, A. baumannii Avibactam, relebactam, vaborbactam R: AMX, AMC, CTX, TZP, CAZ
Class D serine β-lactamases-Oxacillinase OXA OXA-48, OXA-23, OXA-51, OXA-58, OXA-24/40, OXA-58… Enterobacteriaceae, A. baumannii Avibactam, relebactam (variable inhibition) R: AMX, AMC
S/I: CTX, IMP, ETP, MER
S: CAZ, ATM, CFP

, Serratia, Providencia, “indole-positive” Proteus species, Citrobacter, and Enterobacter species. GNB, gram-negative bacteria; SHV, sulfhydryl variant of the TEM enzyme; TEM, Temoneira class A extended-spectrum β-lactamase; CTX-M, cefotaxime-hydrolyzing β-lactamase-Munich; AMX, amoxicillin; TZP, piperacillin-tazobactam; CTX, cefotaxime; CAZ, ceftazidime; ATM, aztreonam; ETP, ertapenem; IMP, imipenemase metallo-β-lactamase; MER, meropenem; TZB, tazobactam; KPC, Klebsiella pneumoniae carbapenemase; IMI, imipenem hydrolyzing β-lactamase; SME, Serratia marcescens enzyme; GES, Guiana extended-spectrum β-lactamase; SHV, sulfhydryl variant of the TEM enzyme; NDM, New Delhi metallo-β-lactamase; VIM, Verona integron-encoded metallo-β-lactamase; GIM, German imipenemase; SPM, Sao Paulo metallo β-lactamase; DIM, Deutch imipinemase; SIM, Seoul imipenemase; AMC, amoxicillin-clavulanic acid; AmpC, ampicillin chromosomal cephalosporinase; ACT, AmpC type β-lactamase; CMY, cephamycin-hydrolyzing β-lactamase; FOX, plasmid-mediated class C β-lactamase; OXA, oxacillin carbapenemase/oxacillinase; CFP, cefoperazone.

Therefore, antimicrobial stewardship programs and carbapenems-sparing strategies must be implemented to preserve the effectiveness of carbapenem antibiotics and new β-lactams/β-lactamase-inhibitors (BLI) (3,134-136,142,149,153-155,162-177) (Tables 7,8). Early identification of colonized patients, appropriate and strict antibiotic control policies, care staff education and training, and contact/isolation precaution measures are of paramount importance to prevent transmission and spread of these CR and MDR/XDR-GNB (149).

Table 8

Newer intravenous antimicrobial agent with activity against MDR/XDR GNB (3,142,149,153-155,162-177)

Drugs Enterobacteriaceae Non-fermenting carbapenem-resistant GNB
Class A ESBL Class C AmpC Class A
Carbapenemase
KPC
Class B
Carbapenemase
NDM, IMP, VIM
Class D
Carbapenemase
OXA type
P. aeruginosa (MDR/XDR) A. baumannii (MDR/XDR) S. maltophilia
Ceftolozane-tazobactam Yes Yes/no No No No Yes§ No No
Ceftazidime-avibactam Yes Yes Yes No Yes Yes No No
Aztreonam-avibactam Yes Yes Yes Yes Yes Variable* No Yes
Ceftaroline-avibactam¥ Yes Yes Yes No Yes No No No
Cefepime-enmetazobactam Yes Yes No No No No No No
Cefepime-zidebactam Yes Yes Yes Yes Yes Yes Limited Yes
Cefepime-taniborbactam Yes Yes Yes Yes& Yes Yes No Yes
Meropenem-vaborbactam Yes Yes Yes No No NoÙ No No
Meropenem-nacubactam Yes Yes Yes Yes Yes£ Noδ Noδ No
Imipenem-cilastatin-relebactam Yes Yes Yes No No Yes No No
Sulbactam-durlobactam No No No No No No Yes No
Cefiderocol Yes Yes Yes Yes Yes Yes Yes Yes
Plazomicin Yes Yes Yes No Yes Variable# Variable# No
Eravacycline$ Yes Yes Yes Yes Yes No Yes Yes
Omadacycline$ Yes Yes Yes Yes Yes No Yes Yes

, Enterobacter cloacae, Klebsiella aerogenes, and Citrobacter freundii are at moderate to high risk (8% to 40% of infections caused by these pathogens) for clinically significant AmpC production (142). In contrast, Serratia marcescens, Morganella morganii and Providencia spp are unlikely (<5%) to overexpress AmpC (142). There is no significant investigation on Hafnia alvei, Citrobacter youngae and Yersinia enterolitica (142). , the role of ceftolozane-tazobactam in invasive infections due to ESBL-producing Enterobacteriaceae needs to be clarified (175). Carbapenems still remain the preferred treatment for any ESBL-producing Enterobacteriaceae infections (175). §, treatment option for mainly carbapenem-resistant non-carbapenemase producing P. aeruginosa (172). , the combination of ceftazidime-avibactam and aztreonam is suggested for moderate to severe S. maltophilia infections when neither TMP-SMX nor minocycline are considered viable treatment options (142). *, the combination of aztreonam and ceftazidime-avibactam may be a viable treatment option against serine-β-lactamase- and metallo-β-lactamase producing Enterobacteriaceae and P. aeruginosa (166). ¥, ceftaroline: “Novel” fifth generation cephalosporin with bactericidal against MRSA (and vancomycin-intermediate, heteroresistant vancomycin-intermediate, and vancomycin-resistant S. aureus) and MDR Streptococcus pneumoniae. &, not efficient against IMP class B metallo-β-lactamase (carbapenemase)—incompletely efficient (70–80%) against NDM (class B metallo-β-lactamase (carbapenemase) (167). Ù, similar activity as meropenem against P. aeruginosa (172). £, class D: only active against OXA-48/181 like (149). δ, similar activity to meropenem for Pseudomonas and Acinetobacter spp (172). #, Not superior to other aminoglycosides (amikacin, gentamicin, tobramycin) against P. aeruginosa and A. baumannii (169,172). $, inactive against Proteus spp, Providencia spp and P. aeruginosa. GNB, gram-negative bacteria; KPC, Klebsiella pneumoniae carbapenemase; NDM, New Delhi metallo-β-lactamase; IMP, imipenemase metallo-β-lactamase; VIM, Verona integron-encoded metallo-β-lactamase; OXA, oxacillin carbapenemase/oxacillinase; MDR, multidrug resistant; XDR, extensively drug-resistant.


Microbiota

Gut dysbiosis—reduction in the gastrointestinal tract in microbiota diversity and replacement of normal commensal microbial organisms [“health-promoting” colonizing commensal microbe population, such as Bacteroidetes (mainly Bacteroides spp, Prevotella spp) and Firmicutes (mainly Lactobacillus spp, Clostridium spp, but also small quantity of Enterococcus spp and Staphylococcus spp)] with pathogenic and virulent organisms (such as Proteobacteria, called “pathobionts”, mainly Enterobacterales and small quantity of P. aeruginosa)—is considered to play a major role in the pathogenesis for many acute or chronic diseases, and is associated in ICU patients with poor outcomes (associated with increased susceptibility to nosocomial infections, sepsis, and organ failures) (178-181). Therefore, even if the gut microbiota is unique to each individual and the “healthy microbiota” is not defined, the majority of the normal gut microbiota consists of obligate anaerobic bacteria, which inhibit the growth of pathogenic bacteria (essentially aerobic or facultative anaerobic bacteria such as E. coli and other Enterobacteriaceae) (179,182). Gut dysbiosis and secondary alteration of microbiota-derived metabolites interact with intestinal receptors, induce adverse local and systemic effects and affect different organ systems (178,179,182,183). During hospitalization, many factors, such as food, parenteral nutrition, discontinuation of the normal diet, general anesthesia, antibiotics, medications (antipsychotics, proton pump inhibitors, opioids, non-steroidal anti-inflammatory drugs, hypoglycemic agents, beta-blockers, amines, chemotherapeutic agents), surgery (stress, injury, hemorrhage and blood transfusion), critical illness, mechanical ventilation, sepsis, splanchnic hypoperfusion, inflammation, electrolytes disturbances, and/or decreased intestinal motility directly, reduce the diversity (qualitative alteration) and alter proportion of the bacterial species (quantitative alteration) in the gut microbiota within 1 or 2 days (178-185). Therefore, significant gut microbiome alterations occur in critically ill patients and are associated with disease severity and clinical outcome (182,186,187). Similarly, gut microbiome alterations occur also in the processes leading to endogenous SSIs and post-surgical complications (188-190).

Antibiotics, frequently used in ICUs, are probably the most common cause of dysbiosis (gut and upper respiratory tract microbiome dysregulation) (180,191). If antibiotic therapies target pathogenic bacteria, they are also targeting the commensal “health-promoting” bacteria making our microbiota (181,182,192). The use of antibiotics highly active against anaerobic strains increases the risk of MDROs compared with other antibiotic regimen (193,194). The microbiota is rapidly and sustainably affected by antibiotics, and is then the main reservoir for MDROs, which will be involved in further infections (182,188-204). The effects of antibiotics on microbiota are multifactorial and depend on the class of antibiotic used, the spectrum of activity, the activity against anaerobic bacteria and potential impact on the intestinal microbiota, the administration route, the pharmacokinetic and pharmacodynamics properties, the route of elimination, the bowel bioavailability and the comorbidities of the patient (179,182,205-207). Emergence of MDROs may occur from the very first day (202,203,208). A single day of imipenem therapy may increase the incidence of imipenem-resistant bacteria colonization in the ICU population (202). Even a single dose of surgical AP alters the bacterial epidemiology of early ventilator-associated pneumonia in brain-injured patients (208). It has been shown that a single dose of third generation cephalosporin is likely to modify the digestive and vaginal flora (209,210). Moreover, the impacts on the microbiota is specific of each antibiotic with different effects between antibiotics of the same class [ertapenem versus imipenem (211,212) or ceftriaxone versus cefotaxime (213)].

Gut microbiota disruption contributes to hospital-acquired infections, sepsis and influences the outcome of sepsis leading to multiple organ failures (180,190,192,214-216). Microbiota-targeted therapeutics or “dysbiosis therapy” aimed at restoring the balance between commensal “health-promoting” and pathogenic organisms may improve post-surgical patient and/or critically ill patient outcomes (180,190-192,217,218). Unaltered or restored microbiota is a key element for local mucosal gut and systemic immunities, and in the fight against MDROs (colonization, infection, and spread) (182,191). Probiotic/prebiotic/symbiotic nutrition or fecal microbiota transplantation strategies to modulate or modify the microbiome are potential interventions to prevent or cure infections (180,184,190,219-222). These different strategies may then contribute to reduce the use of antibiotics and multidrug resistance (222).


Antibiotic potential alternatives for GNB infections

Novel treatment approaches such as bacteriophage therapy (phagotherapy) targeting MDR/XDR strains of GNB (A. baumannii, P. aeruginosa or K. pneumonia), or lysins (bacteriophage-derived enzymes with bacteriolytic activity against Staphylococcus spp), lectin inhibitors (P. aeruginosa), antimicrobial peptides, quorum sensing inhibitors (P. aeruginosa), immunotherapy (therapeutic antibodies targeting S. aureus virulence factors, Acinetobacter baumannii capsule, P. aeruginosa lipopolysaccharides or other virulence factors, or antibiotic resistance mechanisms, and acting concomitantly or synergistically with antibiotics) and GNB vaccines are developed to decrease the pathogen virulence (antivirulence therapy), enhance the therapeutic arsenal against MDROs, and prevent MDRO infections (146,163,164,223-232). Unfortunately, their potential is yet to be explored to demonstrate consistent clinical efficacy in combination with antibiotic therapy.


Conclusions

Community-acquired and hospital-acquired infections caused by MDR bacteria are an ongoing therapeutic challenge. With the emergence and continuous spreading of ESBL- and carbapenemase-producing GNB, therapeutic options for clinicians are more and more limited. Despite the development of new antibiotics, most being combinations of a β-lactam and a β-lactamase inhibitor, MDROs cause difficult-to-treat infections with increased morbidity and mortality, and increased health care costs. Unnecessary antibiotic therapy—i.e., treatment duration longer than necessary, inappropriateness of treatment of noninfectious or nonbacterial syndromes, or inappropriateness of treatment of colonization/contamination—further contributes to the development of resistance and its dissemination, as well as to antibiotic-related harms to patients. In addition, most antibiotics cause collateral damage on commensal bacteria and gut microbiota. Antibiotic stewardship programs are critical to significantly decrease antibiotic inappropriate use, consumption and duration, and promote optimal use of newer antibiotics. Parallel provider education is also mandatory. Antimicrobial effectiveness is a precious and limited resource and should be preserved.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editors (Tobias Eckle and Benjamin Scott) for the series “Highlights in Anesthesia and Critical Care Medicine” published in Annals of Translational Medicine. The article has undergone external peer review.

Peer Review File: Available at https://atm.amegroups.com/article/view/10.21037/atm-22-5585/prf

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://atm.amegroups.com/article/view/10.21037/atm-22-5585/coif). The series “Highlights in Anesthesia and Critical Care Medicine” was commissioned by the editorial office without any funding or sponsorship. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Rudd KE, Johnson SC, Agesa KM, et al. Global, regional, and national sepsis incidence and mortality, 1990-2017: analysis for the Global Burden of Disease Study. Lancet 2020;395:200-11. [Crossref] [PubMed]
  2. Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016;315:801-10. [Crossref] [PubMed]
  3. Evans L, Rhodes A, Alhazzani W, et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock 2021. Crit Care Med 2021;49:e1063-143. [Crossref] [PubMed]
  4. Centers for Disease Control and Prevention (U.S.). Antibiotic resistance threats in the United States, 2019 . Centers for Disease Control and Prevention (U.S.); 2019 [cité 1 2022]. Available online: https://stacks.cdc.gov/view/cdc/82532
  5. Nordmann P, Naas T, Poirel L. Global spread of Carbapenemase-producing Enterobacteriaceae. Emerg Infect Dis 2011;17:1791-8. [Crossref] [PubMed]
  6. Timsit JF, Bassetti M, Cremer O, et al. Rationalizing antimicrobial therapy in the ICU: a narrative review. Intensive Care Med 2019;45:172-89. [Crossref] [PubMed]
  7. World Health Organization. Report on the burden of endemic health care-associated infection worldwide. Geneva: World Health Organization; 2011 [cité 1 2022]. 40 p. Available online: https://apps.who.int/iris/handle/10665/80135
  8. Allegranzi B, Bischoff P, de Jonge S, et al. New WHO recommendations on preoperative measures for surgical site infection prevention: an evidence-based global perspective. Lancet Infect Dis 2016;16:e276-87. [Crossref] [PubMed]
  9. Lamarsalle L, Hunt B, Schauf M, et al. Evaluating the clinical and economic burden of healthcare-associated infections during hospitalization for surgery in France. Epidemiol Infect 2013;141:2473-82. [Crossref] [PubMed]
  10. Berríos-Torres SI, Umscheid CA, Bratzler DW, et al. Centers for Disease Control and Prevention Guideline for the Prevention of Surgical Site Infection, 2017. JAMA Surg 2017;152:784-91. [Crossref] [PubMed]
  11. World Health Organization. Global Guidelines for the Prevention of Surgical Site Infection. Geneva: World Health Organization; 2016 [cité 21 2022]. Available online: https://apps.who.int/iris/handle/10665/250680
  12. Bratzler DW, Dellinger EP, Olsen KM, et al. Clinical practice guidelines for antimicrobial prophylaxis in surgery. Surg Infect (Larchmt) 2013;14:73-156. [Crossref] [PubMed]
  13. Bowater RJ, Stirling SA, Lilford RJ. Is antibiotic prophylaxis in surgery a generally effective intervention? Testing a generic hypothesis over a set of meta-analyses. Ann Surg 2009;249:551-6. [Crossref] [PubMed]
  14. Hendren S, Fritze D, Banerjee M, et al. Antibiotic choice is independently associated with risk of surgical site infection after colectomy: a population-based cohort study. Ann Surg 2013;257:469-75. [Crossref] [PubMed]
  15. Altemeier WA, American College of Surgeons, éditeurs. Manual on control of infection in surgical patients. Philadelphia: Lippincott; 1976. 280 p.
  16. Steinberg JP, Braun BI, Hellinger WC, et al. Timing of antimicrobial prophylaxis and the risk of surgical site infections: results from the Trial to Reduce Antimicrobial Prophylaxis Errors. Ann Surg 2009;250:10-6. [Crossref] [PubMed]
  17. Martin C, Auboyer C, Boisson M, et al. Antibioprophylaxie en chirurgie et médecine interventionnelle (patients adultes). Actualisation 2017. Anesthésie & Réanimation 2019;5:544-66. [Crossref] [PubMed]
  18. Ban KA, Minei JP, Laronga C, et al. American College of Surgeons and Surgical Infection Society: Surgical Site Infection Guidelines, 2016 Update. J Am Coll Surg 2017;224:59-74. [Crossref] [PubMed]
  19. Fields AC, Pradarelli JC, Itani KMF. Preventing Surgical Site Infections: Looking Beyond the Current Guidelines. JAMA 2020;323:1087-8. [Crossref] [PubMed]
  20. Umscheid CA, Mitchell MD, Doshi JA, et al. Estimating the proportion of healthcare-associated infections that are reasonably preventable and the related mortality and costs. Infect Control Hosp Epidemiol 2011;32:101-14. [Crossref] [PubMed]
  21. Rosenberger LH, Politano AD, Sawyer RG. The surgical care improvement project and prevention of post-operative infection, including surgical site infection. Surg Infect (Larchmt) 2011;12:163-8. [Crossref] [PubMed]
  22. Bratzler DW, Hunt DR. The surgical infection prevention and surgical care improvement projects: national initiatives to improve outcomes for patients having surgery. Clin Infect Dis 2006;43:322-30. [Crossref] [PubMed]
  23. Bratzler DW, Houck PM, et al. Antimicrobial prophylaxis for surgery: an advisory statement from the National Surgical Infection Prevention Project. Clin Infect Dis 2004;38:1706-15. [Crossref] [PubMed]
  24. Eykyn SJ, Jackson BT, Lockhart-Mummery HE, et al. Prophylactic peroperative intravenous metronidazole in elective colorectal surgery. Lancet 1979;2:761-4. [Crossref]
  25. Smaill FM, Grivell RM. Antibiotic prophylaxis versus no prophylaxis for preventing infection after cesarean section. Cochrane Database Syst Rev 2014;2014:CD007482. [Crossref] [PubMed]
  26. Hawn MT, Richman JS, Vick CC, et al. Timing of surgical antibiotic prophylaxis and the risk of surgical site infection. JAMA Surg 2013;148:649-57. [Crossref] [PubMed]
  27. Hawn MT, Vick CC, Richman J, et al. Surgical site infection prevention: time to move beyond the surgical care improvement program. Ann Surg 2011;254:494-9; discussion 499-501. [Crossref] [PubMed]
  28. Stulberg JJ, Delaney CP, Neuhauser DV, et al. Adherence to surgical care improvement project measures and the association with postoperative infections. JAMA 2010;303:2479-85. [Crossref] [PubMed]
  29. Hübner M, Diana M, Zanetti G, et al. Surgical site infections in colon surgery: the patient, the procedure, the hospital, and the surgeon. Arch Surg 2011;146:1240-5. [Crossref] [PubMed]
  30. Deierhoi RJ, Dawes LG, Vick C, et al. Choice of intravenous antibiotic prophylaxis for colorectal surgery does matter. J Am Coll Surg 2013;217:763-9. [Crossref] [PubMed]
  31. Ponce B, Raines BT, Reed RD, et al. Surgical Site Infection After Arthroplasty: Comparative Effectiveness of Prophylactic Antibiotics: Do Surgical Care Improvement Project Guidelines Need to Be Updated? J Bone Joint Surg Am 2014;96:970-7. [Crossref] [PubMed]
  32. Itani KM, Wilson SE, Awad SS, et al. Ertapenem versus cefotetan prophylaxis in elective colorectal surgery. N Engl J Med 2006;355:2640-51. [Crossref] [PubMed]
  33. Moine P, Asehnoune K. Antibiotic prophylaxis in colorectal surgery. N Engl J Med 2007;356:1684-author reply 1685. [Crossref] [PubMed]
  34. Gunderson BW, Ross GH, Ibrahim KH, et al. What do we really know about antibiotic pharmacodynamics? Pharmacotherapy 2001;21:302S-18S. [Crossref] [PubMed]
  35. Blumenthal KG, Ryan EE, Li Y, et al. The Impact of a Reported Penicillin Allergy on Surgical Site Infection Risk. Clin Infect Dis 2018;66:329-36. [Crossref] [PubMed]
  36. Kheir MM, Tan TL, Azboy I, et al. Vancomycin Prophylaxis for Total Joint Arthroplasty: Incorrectly Dosed and Has a Higher Rate of Periprosthetic Infection Than Cefazolin. Clin Orthop Relat Res 2017;475:1767-74. [Crossref] [PubMed]
  37. Shibamura-Fujiogi M, Ormsby J, Breibart M, et al. Risk factors for pediatric surgical site infection following neurosurgical procedures for hydrocephalus: a retrospective single-center cohort study. BMC Anesthesiol 2021;21:124. [Crossref] [PubMed]
  38. Shah NS, Ridgway JP, Pettit N, et al. Documenting Penicillin Allergy: The Impact of Inconsistency. PLoS One 2016;11:e0150514. [Crossref] [PubMed]
  39. Chen JR, Tarver SA, Alvarez KS, et al. A Proactive Approach to Penicillin Allergy Testing in Hospitalized Patients. J Allergy Clin Immunol Pract 2017;5:686-93. [Crossref] [PubMed]
  40. Solensky R, Earl HS, Gruchalla RS. Lack of penicillin resensitization in patients with a history of penicillin allergy after receiving repeated penicillin courses. Arch Intern Med 2002;162:822-6. [Crossref] [PubMed]
  41. Mangram AJ, Horan TC, Pearson ML, et al. Guideline for Prevention of Surgical Site Infection, 1999. Centers for Disease Control and Prevention (CDC) Hospital Infection Control Practices Advisory Committee. Am J Infect Control 1999;27:97-132; quiz 133-4; discussion 96.
  42. James M, Martinez EA. Antibiotics and perioperative infections. Best Pract Res Clin Anaesthesiol 2008;22:571-84. [Crossref] [PubMed]
  43. Krizek TJ, Robson MC. Evolution of quantitative bacteriology in wound management. Am J Surg 1975;130:579-84. [Crossref] [PubMed]
  44. Sinha B, van Assen S, Friedrich AW. Important issues for perioperative systemic antimicrobial prophylaxis in surgery. Curr Opin Anaesthesiol 2014;27:377-81. [Crossref] [PubMed]
  45. Liu P, Müller M, Derendorf H. Rational dosing of antibiotics: the use of plasma concentrations versus tissue concentrations. Int J Antimicrob Agents 2002;19:285-90. [Crossref] [PubMed]
  46. Barbour A, Scaglione F, Derendorf H. Class-dependent relevance of tissue distribution in the interpretation of anti-infective pharmacokinetic/pharmacodynamic indices. Int J Antimicrob Agents 2010;35:431-8. [Crossref] [PubMed]
  47. Udy AA, Roberts JA, Lipman J. Clinical implications of antibiotic pharmacokinetic principles in the critically ill. Intensive Care Med 2013;39:2070-82. [Crossref] [PubMed]
  48. Roberts JA, Lipman J. Pharmacokinetic issues for antibiotics in the critically ill patient. Crit Care Med 2009;37:840-51; quiz 859. [Crossref] [PubMed]
  49. Gonçalves-Pereira J, Póvoa P. Antibiotics in critically ill patients: a systematic review of the pharmacokinetics of β-lactams. Crit Care 2011;15:R206. [Crossref] [PubMed]
  50. McKenzie C. Antibiotic dosing in critical illness. J Antimicrob Chemother 2011;66:ii25-31. [Crossref] [PubMed]
  51. Asín-Prieto E, Soraluce A, Trocóniz IF, et al. Population pharmacokinetic models for cefuroxime and metronidazole used in combination as prophylactic agents in colorectal surgery: Model-based evaluation of standard dosing regimens. Int J Antimicrob Agents 2015;45:504-11. [Crossref] [PubMed]
  52. Lador A, Nasir H, Mansur N, et al. Antibiotic prophylaxis in cardiac surgery: systematic review and meta-analysis. J Antimicrob Chemother 2012;67:541-50. [Crossref] [PubMed]
  53. Moine P, Fish DN. Pharmacodynamic modelling of intravenous antibiotic prophylaxis in elective colorectal surgery. Int J Antimicrob Agents 2013;41:167-73. [Crossref] [PubMed]
  54. Drusano GL. Antimicrobial pharmacodynamics: critical interactions of 'bug and drug'. Nat Rev Microbiol 2004;2:289-300. [Crossref] [PubMed]
  55. Roberts JA, Paul SK, Akova M, 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;58:1072-83. [Crossref] [PubMed]
  56. Moine P, Mueller SW, Schoen JA, et al. Pharmacokinetic and Pharmacodynamic Evaluation of a Weight-Based Dosing Regimen of Cefoxitin for Perioperative Surgical Prophylaxis in Obese and Morbidly Obese Patients. Antimicrob Agents Chemother 2016;60:5885-93. [Crossref] [PubMed]
  57. Blot SI, Pea F, Lipman J. The effect of pathophysiology on pharmacokinetics in the critically ill patient--concepts appraised by the example of antimicrobial agents. Adv Drug Deliv Rev 2014;77:3-11. [Crossref] [PubMed]
  58. Wang Z, Chen F, Ward M, et al. Compliance with Surgical Care Improvement Project measures and hospital-associated infections following hip arthroplasty. J Bone Joint Surg Am 2012;94:1359-66. [Crossref] [PubMed]
  59. Udy AA, Roberts JA, Boots RJ, et al. Augmented renal clearance: implications for antibacterial dosing in the critically ill. Clin Pharmacokinet 2010;49:1-16. [Crossref] [PubMed]
  60. Udy AA, Putt MT, Shanmugathasan S, et al. Augmented renal clearance in the Intensive Care Unit: an illustrative case series. Int J Antimicrob Agents 2010;35:606-8. [Crossref] [PubMed]
  61. Craig WA. Pharmacokinetic/pharmacodynamic parameters: rationale for antibacterial dosing of mice and men. Clin Infect Dis 1998;26:1-10; quiz 11-2. [Crossref] [PubMed]
  62. Craig WA, Ebert SC. Killing and regrowth of bacteria in vitro: a review. Scand J Infect Dis Suppl 1990;74:63-70.
  63. Mouton JW, Vinks AA. Pharmacokinetic/pharmacodynamic modelling of antibacterials in vitro and in vivo using bacterial growth and kill kinetics: the minimum inhibitory concentration versus stationary concentration. Clin Pharmacokinet 2005;44:201-10. [Crossref] [PubMed]
  64. Nielsen EI, Friberg LE. Pharmacokinetic-pharmacodynamic modeling of antibacterial drugs. Pharmacol Rev 2013;65:1053-90. [Crossref] [PubMed]
  65. Alobaid AS, Wallis SC, Jarrett P, et al. Effect of Obesity on the Population Pharmacokinetics of Meropenem in Critically Ill Patients. Antimicrob Agents Chemother 2016;60:4577-84. [Crossref] [PubMed]
  66. Roberts JA, Abdul-Aziz MH, Lipman J, et al. Individualised antibiotic dosing for patients who are critically ill: challenges and potential solutions. Lancet Infect Dis 2014;14:498-509. [Crossref] [PubMed]
  67. Bandín-Vilar E, García-Quintanilla L, Castro-Balado A, et al. A Review of Population Pharmacokinetic Analyses of Linezolid. Clin Pharmacokinet 2022;61:789-817. [Crossref] [PubMed]
  68. Turnidge JD. The pharmacodynamics of beta-lactams. Clin Infect Dis 1998;27:10-22. [Crossref] [PubMed]
  69. Hyatt JM, McKinnon PS, Zimmer GS, et al. The importance of pharmacokinetic/pharmacodynamic surrogate markers to outcome. Focus on antibacterial agents. Clin Pharmacokinet 1995;28:143-60. [Crossref] [PubMed]
  70. Mouton JW, den Hollander JG. Killing of Pseudomonas aeruginosa during continuous and intermittent infusion of ceftazidime in an in vitro pharmacokinetic model. Antimicrob Agents Chemother 1994;38:931-6. [Crossref] [PubMed]
  71. Robaux MA, Dube L, Caillon J, et al. In vivo efficacy of continuous infusion versus intermittent dosing of ceftazidime alone or in combination with amikacin relative to human kinetic profiles in a Pseudomonas aeruginosa rabbit endocarditis model. J Antimicrob Chemother 2001;47:617-22. [Crossref] [PubMed]
  72. Cappelletty DM, Kang SL, Palmer SM, et al. Pharmacodynamics of ceftazidime administered as continuous infusion or intermittent bolus alone and in combination with single daily-dose amikacin against Pseudomonas aeruginosa in an in vitro infection model. Antimicrob Agents Chemother 1995;39:1797-801. [Crossref] [PubMed]
  73. Mouton JW, Horrevorts AM, Mulder PG, et al. Pharmacokinetics of ceftazidime in serum and suction blister fluid during continuous and intermittent infusions in healthy volunteers. Antimicrob Agents Chemother 1990;34:2307-11. [Crossref] [PubMed]
  74. Xiong YQ, Caillon J, Zhou XY, et al. Treatment of experimental rabbit infective endocarditis due to a multidrug-resistant Pseudomonas aeruginosa with high-dose ceftazidime alone and combined with amikacin or sulbactam or both. J Antimicrob Chemother 1995;35:697-706.
  75. Manduru M, Mihm LB, White RL, et al. In vitro pharmacodynamics of ceftazidime against Pseudomonas aeruginosa isolates from cystic fibrosis patients. Antimicrob Agents Chemother 1997;41:2053-6. [Crossref] [PubMed]
  76. Ariano RE, Nyhlén A, Donnelly JP, et al. Pharmacokinetics and pharmacodynamics of meropenem in febrile neutropenic patients with bacteremia. Ann Pharmacother 2005;39:32-8. [Crossref] [PubMed]
  77. Li C, Du X, Kuti JL, et al. Clinical pharmacodynamics of meropenem in patients with lower respiratory tract infections. Antimicrob Agents Chemother 2007;51:1725-30. [Crossref] [PubMed]
  78. McKinnon PS, Paladino JA, Schentag JJ. Evaluation of area under the inhibitory curve (AUIC) and time above the minimum inhibitory concentration (T>MIC) as predictors of outcome for cefepime and ceftazidime in serious bacterial infections. Int J Antimicrob Agents 2008;31:345-51. [Crossref] [PubMed]
  79. 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;44:357-63. [Crossref] [PubMed]
  80. Tam VH, McKinnon PS, Akins RL, et al. Pharmacodynamics of cefepime in patients with Gram-negative infections. J Antimicrob Chemother 2002;50:425-8. [Crossref] [PubMed]
  81. Mouton JW, Vinks AA. Is continuous infusion of beta-lactam antibiotics worthwhile?--efficacy and pharmacokinetic considerations. J Antimicrob Chemother 1996;38:5-15. [Crossref] [PubMed]
  82. Lee SY, Kuti JL, Nicolau DP. Cefepime pharmacodynamics in patients with extended spectrum beta-lactamase (ESBL) and non-ESBL infections. J Infect 2007;54:463-8. [Crossref] [PubMed]
  83. Schentag JJ, Nix DE, Adelman MH. Mathematical examination of dual individualization principles (I): Relationships between AUC above MIC and area under the inhibitory curve for cefmenoxime, ciprofloxacin, and tobramycin. DICP 1991;25:1050-7. [Crossref] [PubMed]
  84. Goss TF, Forrest A, Nix DE, et al. Mathematical examination of dual individualization principles (II): The rate of bacterial eradication at the same area under the inhibitory curve is more rapid for ciprofloxacin than for cefmenoxime. Ann Pharmacother 1994;28:863-8. [Crossref] [PubMed]
  85. Schentag JJ, Strenkoski-Nix LC, Nix DE, et al. Pharmacodynamic interactions of antibiotics alone and in combination. Clin Infect Dis 1998;27:40-6. [Crossref] [PubMed]
  86. Thomas JK, Forrest A, Bhavnani SM, et al. Pharmacodynamic evaluation of factors associated with the development of bacterial resistance in acutely ill patients during therapy. Antimicrob Agents Chemother 1998;42:521-7. [Crossref] [PubMed]
  87. Moine P, Mazoit JX. Streptococcus pneumoniae pneumonia in mice: optimal amoxicillin dosing predicted from a pharmacokinetic-pharmacodynamic model. J Pharmacol Exp Ther 1999;291:1086-92.
  88. Moine P, Mazoit JX, Bédos JP, et al. Correlation between in vitro and in vivo activity of amoxicillin against Streptococcus pneumoniae in a murine pneumonia model. J Pharmacol Exp Ther 1997;280:310-5.
  89. Lavoie GY, Bergeron MG. Influence of four modes of administration on penetration of aztreonam, cefuroxime, and ampicillin into interstitial fluid and fibrin clots and on in vivo efficacy against Haemophilus influenzae. Antimicrob Agents Chemother 1985;28:404-12. [Crossref] [PubMed]
  90. Onyeji CO, Nicolau DP, Nightingale CH, et al. Optimal times above MICs of ceftibuten and cefaclor in experimental intra-abdominal infections. Antimicrob Agents Chemother 1994;38:1112-7. [Crossref] [PubMed]
  91. Nelson RL, Gladman E, Barbateskovic M. Antimicrobial prophylaxis for colorectal surgery. Cochrane Database Syst Rev 2014;2014:CD001181. [Crossref] [PubMed]
  92. Song F, Glenny AM. Antimicrobial prophylaxis in colorectal surgery: a systematic review of randomized controlled trials. Br J Surg 1998;85:1232-41. [Crossref] [PubMed]
  93. Zelenitsky SA, Ariano RE, Harding GK, et al. Antibiotic pharmacodynamics in surgical prophylaxis: an association between intraoperative antibiotic concentrations and efficacy. Antimicrob Agents Chemother 2002;46:3026-30. [Crossref] [PubMed]
  94. Goldmann DA, Hopkins CC, Karchmer AW, et al. Cephalothin prophylaxis in cardiac valve surgery. A prospective, double-blind comparison of two-day and six-day regimens. J Thorac Cardiovasc Surg 1977;73:470-9.
  95. Sexton DJ. Carbapenems for surgical prophylaxis? N Engl J Med 2006;355:2693-5. [Crossref] [PubMed]
  96. Daley BJ, Cecil W, Clarke PC, et al. How slow is too slow? Correlation of operative time to complications: an analysis from the Tennessee Surgical Quality Collaborative. J Am Coll Surg 2015;220:550-8. [Crossref] [PubMed]
  97. Procter LD, Davenport DL, Bernard AC, et al. General surgical operative duration is associated with increased risk-adjusted infectious complication rates and length of hospital stay. J Am Coll Surg 2010;210:60-5.e1-2.
  98. Suffoletta TJ, Jennings HR, Oh JJ, et al. Continuous versus intermittent infusion of prophylactic cefoxitin after colorectal surgery: a pilot study. Pharmacotherapy 2008;28:1133-9. [Crossref] [PubMed]
  99. Ferraz ÁA, Siqueira LT, Campos JM, et al. Antibiotic prophylaxis in bariatric surgery: a continuous infusion of cefazolin versus ampicillin/sulbactam and ertapenem. Arq Gastroenterol 2015;52:83-7. [Crossref] [PubMed]
  100. Falagas ME, Karageorgopoulos DE. Adjustment of dosing of antimicrobial agents for bodyweight in adults. Lancet 2010;375:248-51. [Crossref] [PubMed]
  101. Catanzano A, Phillips M, Dubrovskaya Y, et al. The standard one gram dose of vancomycin is not adequate prophylaxis for MRSA. Iowa Orthop J 2014;34:111-7.
  102. Itani KM, Jensen EH, Finn TS, et al. Effect of body mass index and ertapenem versus cefotetan prophylaxis on surgical site infection in elective colorectal surgery. Surg Infect (Larchmt) 2008;9:131-7. [Crossref] [PubMed]
  103. Worth LJ, Bull AL, Spelman T, et al. Diminishing surgical site infections in Australia: time trends in infection rates, pathogens and antimicrobial resistance using a comprehensive Victorian surveillance program, 2002-2013. Infect Control Hosp Epidemiol 2015;36:409-16. [Crossref] [PubMed]
  104. Berríos-Torres SI, Yi SH, Bratzler DW, et al. Activity of commonly used antimicrobial prophylaxis regimens against pathogens causing coronary artery bypass graft and arthroplasty surgical site infections in the United States, 2006-2009. Infect Control Hosp Epidemiol 2014;35:231-9. [Crossref] [PubMed]
  105. Polso AK, Lassiter JL, Nagel JL. Impact of hospital guideline for weight-based antimicrobial dosing in morbidly obese adults and comprehensive literature review. J Clin Pharm Ther 2014;39:584-608. [Crossref] [PubMed]
  106. Meng L, Mui E, Holubar MK, et al. Comprehensive Guidance for Antibiotic Dosing in Obese Adults. Pharmacotherapy 2017;37:1415-31. [Crossref] [PubMed]
  107. Janson B, Thursky K. Dosing of antibiotics in obesity. Curr Opin Infect Dis 2012;25:634-49. [Crossref] [PubMed]
  108. Sirard S, Abou Chakra CN, Langlois MF, et al. Is Antimicrobial Dosing Adjustment Associated with Better Outcomes in Patients with Severe Obesity and Bloodstream Infections? An Exploratory Study. Antibiotics (Basel) 2020;9:707. [Crossref] [PubMed]
  109. Hussain Z, Curtain C, Mirkazemi C, et al. Prophylactic Cefazolin Dosing and Surgical Site Infections: Does the Dose Matter in Obese Patients? Obes Surg 2019;29:159-65. [Crossref] [PubMed]
  110. Karamian BA, Toci GR, Lambrechts MJ, et al. Cefazolin prophylaxis in spine surgery: patients are frequently underdosed and at increased risk for infection. Spine J 2022;22:1442-50. [Crossref] [PubMed]
  111. Si D, Rajmokan M, Lakhan P, et al. Surgical site infections following coronary artery bypass graft procedures: 10 years of surveillance data. BMC Infect Dis 2014;14:318. [Crossref] [PubMed]
  112. Okamura K, Tanaka K, Miura T, et al. Randomized controlled trial of perioperative antimicrobial therapy based on the results of preoperative bile cultures in patients undergoing biliary reconstruction. J Hepatobiliary Pancreat Sci 2017;24:382-93. [Crossref] [PubMed]
  113. Dubinsky-Pertzov B, Temkin E, Harbarth S, et al. Carriage of Extended-spectrum Beta-lactamase-producing Enterobacteriaceae and the Risk of Surgical Site Infection After Colorectal Surgery: A Prospective Cohort Study. Clin Infect Dis 2019;68:1699-704. [Crossref] [PubMed]
  114. Jolivet S, Lescure FX, Armand-Lefevre L, et al. Surgical site infection with extended-spectrum β-lactamase-producing Enterobacteriaceae after cardiac surgery: incidence and risk factors. Clin Microbiol Infect 2018;24:283-8. [Crossref] [PubMed]
  115. Aslan AT, Akova M. Extended spectrum β-lactamase producing enterobacteriaceae: carbapenem sparing options. Expert Rev Anti Infect Ther 2019;17:969-81. [Crossref] [PubMed]
  116. Temkin E, Margalit I, Nutman A, et al. Surgical antibiotic prophylaxis in patients colonized with multidrug-resistant Gram-negative bacteria: practical and conceptual aspects. J Antimicrob Chemother 2021;76:i40-6. [Crossref] [PubMed]
  117. Nutman A, Temkin E, Harbarth S, et al. Personalized Ertapenem Prophylaxis for Carriers of Extended-spectrum β-Lactamase-producing Enterobacteriaceae Undergoing Colorectal Surgery. Clin Infect Dis 2020;70:1891-7. [Crossref] [PubMed]
  118. Prokuski L. Prophylactic antibiotics in orthopaedic surgery. J Am Acad Orthop Surg 2008;16:283-93. [Crossref] [PubMed]
  119. Fromentin M, Mullaert J, Gille B, et al. Extended antibiotic prophylaxis after pancreatoduodenectomy reduces postoperative abdominal infection in high-risk patients: Results from a retrospective cohort study. Surgery 2022;172:205-11. [Crossref] [PubMed]
  120. Bull AL, Worth LJ, Richards MJ. Impact of vancomycin surgical antibiotic prophylaxis on the development of methicillin-sensitive staphylococcus aureus surgical site infections: report from Australian Surveillance Data (VICNISS). Ann Surg 2012;256:1089-92. [Crossref] [PubMed]
  121. Patrick S, James C, Ali A, et al. Vascular surgical antibiotic prophylaxis study (VSAPS). Vasc Endovascular Surg 2010;44:521-8. [Crossref] [PubMed]
  122. Lee B, Michaels MG. Prophylactic antimicrobials in solid organ transplant. Curr Opin Crit Care 2014;20:420-5. [Crossref] [PubMed]
  123. Finkelstein R, Rabino G, Mashiah T, et al. Vancomycin versus cefazolin prophylaxis for cardiac surgery in the setting of a high prevalence of methicillin-resistant staphylococcal infections. J Thorac Cardiovasc Surg 2002;123:326-32. [Crossref] [PubMed]
  124. Sewick A, Makani A, Wu C, et al. Does dual antibiotic prophylaxis better prevent surgical site infections in total joint arthroplasty? Clin Orthop Relat Res 2012;470:2702-7. [Crossref] [PubMed]
  125. Bolon MK, Morlote M, Weber SG, et al. Glycopeptides are no more effective than beta-lactam agents for prevention of surgical site infection after cardiac surgery: a meta-analysis. Clin Infect Dis 2004;38:1357-63. [Crossref] [PubMed]
  126. Chambers D, Worthy G, Myers L, et al. Glycopeptide vs. non-glycopeptide antibiotics for prophylaxis of surgical site infections: a systematic review. Surg Infect (Larchmt) 2010;11:455-62. [Crossref] [PubMed]
  127. Crawford T, Rodvold KA, Solomkin JS. Vancomycin for surgical prophylaxis? Clin Infect Dis 2012;54:1474-9. [Crossref] [PubMed]
  128. Primeggia J, Matsumoto CS, Fishbein TM, et al. Infection among adult small bowel and multivisceral transplant recipients in the 30-day postoperative period. Transpl Infect Dis 2013;15:441-8. [Crossref] [PubMed]
  129. Almeida RA, Hasimoto CN, Kim A, et al. Antibiotic prophylaxis for surgical site infection in people undergoing liver transplantation. Cochrane Database Syst Rev 2015;2015:CD010164. [Crossref] [PubMed]
  130. Arulkumaran N, Routledge M, Schlebusch S, et al. Antimicrobial-associated harm in critical care: a narrative review. Intensive Care Med 2020;46:225-35. [Crossref] [PubMed]
  131. Carignan A, Allard C, Pépin J, et al. Risk of Clostridium difficile infection after perioperative antibacterial prophylaxis before and during an outbreak of infection due to a hypervirulent strain. Clin Infect Dis 2008;46:1838-43. [Crossref] [PubMed]
  132. Fowler AJ, Dias P, Hui S, et al. Liberal or restrictive antimicrobial prophylaxis for surgical site infection: systematic review and meta-analysis of randomised trials. Br J Anaesth 2022;129:104-13. [Crossref] [PubMed]
  133. Versporten A, Zarb P, Caniaux I, et al. Antimicrobial consumption and resistance in adult hospital inpatients in 53 countries: results of an internet-based global point prevalence survey. Lancet Glob Health 2018;6:e619-29. [Crossref] [PubMed]
  134. Spellberg B. The New Antibiotic Mantra-"Shorter Is Better". JAMA Intern Med 2016;176:1254-5. [Crossref] [PubMed]
  135. Spellberg B. The Maturing Antibiotic Mantra: "Shorter Is Still Better". J Hosp Med 2018;13:361-2. [Crossref] [PubMed]
  136. Royer S, DeMerle KM, Dickson RP, et al. Shorter Versus Longer Courses of Antibiotics for Infection in Hospitalized Patients: A Systematic Review and Meta-Analysis. J Hosp Med 2018;13:336-42. [Crossref] [PubMed]
  137. Klein Klouwenberg PM, Cremer OL, van Vught LA, et al. Likelihood of infection in patients with presumed sepsis at the time of intensive care unit admission: a cohort study. Crit Care 2015;19:319. [Crossref] [PubMed]
  138. Hranjec T, Rosenberger LH, Swenson B, et al. Aggressive versus conservative initiation of antimicrobial treatment in critically ill surgical patients with suspected intensive-care-unit-acquired infection: a quasi-experimental, before and after observational cohort study. Lancet Infect Dis 2012;12:774-80. [Crossref] [PubMed]
  139. Harbarth S, Garbino J, Pugin J, et al. Inappropriate initial antimicrobial therapy and its effect on survival in a clinical trial of immunomodulating therapy for severe sepsis. Am J Med 2003;115:529-35. [Crossref] [PubMed]
  140. MacArthur RD, Miller M, Albertson T, et al. Adequacy of early empiric antibiotic treatment and survival in severe sepsis: experience from the MONARCS trial. Clin Infect Dis 2004;38:284-8. [Crossref] [PubMed]
  141. Kumar A, Roberts D, Wood KE, et al. Duration of hypotension before initiation of effective antimicrobial therapy is the critical determinant of survival in human septic shock. Crit Care Med 2006;34:1589-96. [Crossref] [PubMed]
  142. Tamma PD, Aitken SL, Bonomo RA, et al. Infectious Diseases Society of America Guidance on the Treatment of AmpC β-Lactamase-Producing Enterobacterales, Carbapenem-Resistant Acinetobacter baumannii, and Stenotrophomonas maltophilia Infections. Clin Infect Dis 2022;74:2089-114. [Crossref] [PubMed]
  143. Kalil AC, Metersky ML, Klompas M, et al. Management of Adults With Hospital-acquired and Ventilator-associated Pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis 2016;63:e61-e111. [Crossref] [PubMed]
  144. Nation RL, Li J, Cars O, et al. Framework for optimisation of the clinical use of colistin and polymyxin B: the Prato polymyxin consensus. Lancet Infect Dis 2015;15:225-34. [Crossref] [PubMed]
  145. Garonzik SM, Li J, Thamlikitkul V, et al. Population pharmacokinetics of colistin methanesulfonate and formed colistin in critically ill patients from a multicenter study provide dosing suggestions for various categories of patients. Antimicrob Agents Chemother 2011;55:3284-94. [Crossref] [PubMed]
  146. Koulenti D, Song A, Ellingboe A, et al. Infections by multidrug-resistant Gram-negative Bacteria: What's new in our arsenal and what's in the pipeline?. Int J Antimicrob Agents 2019;53:211-24. [Crossref] [PubMed]
  147. Dulhunty JM, Roberts JA, Davis JS, et al. Continuous infusion of beta-lactam antibiotics in severe sepsis: a multicenter double-blind, randomized controlled trial. Clin Infect Dis 2013;56:236-44. [Crossref] [PubMed]
  148. Drusano GL, Lodise TP. Saving lives with optimal antimicrobial chemotherapy. Clin Infect Dis 2013;56:245-7. [Crossref] [PubMed]
  149. Jean SS, Harnod D, Hsueh PR. Global Threat of Carbapenem-Resistant Gram-Negative Bacteria. Front Cell Infect Microbiol 2022;12:823684. [Crossref] [PubMed]
  150. De Oliveira DMP, Forde BM, Kidd TJ, et al. Antimicrobial Resistance in ESKAPE Pathogens. Clin Microbiol Rev 2020;33:e00181-19. [Crossref] [PubMed]
  151. Nordmann P, Gniadkowski M, Giske CG, et al. Identification and screening of carbapenemase-producing Enterobacteriaceae. Clin Microbiol Infect 2012;18:432-8. [Crossref] [PubMed]
  152. Nordmann P, Poirel L. Strategies for identification of carbapenemase-producing Enterobacteriaceae. J Antimicrob Chemother 2013;68:487-9. [Crossref] [PubMed]
  153. Karaiskos I, Lagou S, Pontikis K, et al. The "Old" and the "New" Antibiotics for MDR Gram-Negative Pathogens: For Whom, When, and How. Front Public Health 2019;7:151. [Crossref] [PubMed]
  154. Boyd SE, Livermore DM, Hooper DC, et al. Metallo-β-Lactamases: Structure, Function, Epidemiology, Treatment Options, and the Development Pipeline. Antimicrob Agents Chemother 2020;64:e00397-20. [Crossref] [PubMed]
  155. Mo Y, Lorenzo M, Farghaly S, et al. What's new in the treatment of multidrug-resistant gram-negative infections?. Diagn Microbiol Infect Dis 2019;93:171-81. [Crossref] [PubMed]
  156. Nordmann P, Poirel L. Epidemiology and Diagnostics of Carbapenem Resistance in Gram-negative Bacteria. Clin Infect Dis 2019;69:S521-8. [Crossref] [PubMed]
  157. Bush K, Jacoby GA. Updated functional classification of beta-lactamases. Antimicrob Agents Chemother 2010;54:969-76. [Crossref] [PubMed]
  158. Bush K, Bradford PA. Epidemiology of β-Lactamase-Producing Pathogens. Clin Microbiol Rev 2020;33:e00047-19. [Crossref] [PubMed]
  159. Peri AM, Doi Y, Potoski BA, et al. Antimicrobial treatment challenges in the era of carbapenem resistance. Diagn Microbiol Infect Dis 2019;94:413-25. [Crossref] [PubMed]
  160. Sy CL, Chen PY, Cheng CW, et al. Recommendations and guidelines for the treatment of infections due to multidrug resistant organisms. J Microbiol Immunol Infect 2022;55:359-86. [Crossref] [PubMed]
  161. Falagas ME, Tansarli GS, Karageorgopoulos DE, et al. Deaths attributable to carbapenem-resistant Enterobacteriaceae infections. Emerg Infect Dis 2014;20:1170-5. [Crossref] [PubMed]
  162. Bou Zerdan M, Al Hassan S, Shaker W, et al. Carbapenemase Inhibitors: Updates on Developments in 2021. J Clin Med Res 2022;14:251-9. [Crossref] [PubMed]
  163. Assefa M. Multi-drug resistant gram-negative bacterial pneumonia: etiology, risk factors, and drug resistance patterns. Pneumonia (Nathan) 2022;14:4. [Crossref] [PubMed]
  164. Kunz Coyne AJ, El Ghali A, Holger D, et al. Therapeutic Strategies for Emerging Multidrug-Resistant Pseudomonas aeruginosa. Infect Dis Ther 2022;11:661-82. [Crossref] [PubMed]
  165. Doi Y. Treatment Options for Carbapenem-resistant Gram-negative Bacterial Infections. Clin Infect Dis 2019;69:S565-75. [Crossref] [PubMed]
  166. Lee M, Abbey T, Biagi M, et al. Activity of aztreonam in combination with ceftazidime-avibactam against serine- and metallo-β-lactamase-producing Pseudomonas aeruginosa. Diagn Microbiol Infect Dis 2021;99:115227. [Crossref] [PubMed]
  167. Mushtaq S, Vickers A, Doumith M, et al. Activity of β-lactam/taniborbactam (VNRX-5133) combinations against carbapenem-resistant Gram-negative bacteria. J Antimicrob Chemother 2021;76:160-70. [Crossref] [PubMed]
  168. Bassetti M, Garau J. Current and future perspectives in the treatment of multidrug-resistant Gram-negative infections. J Antimicrob Chemother 2021;76:iv23-37. [Crossref] [PubMed]
  169. Losito AR, Raffaelli F, Del Giacomo P, et al. New Drugs for the Treatment of Pseudomonas aeruginosa Infections with Limited Treatment Options: A Narrative Review. Antibiotics (Basel) 2022;11:579. [Crossref] [PubMed]
  170. Shapiro S. Cefepime/Enmetazobactam Is a Clinically Effective Combination Targeting Extended-Spectrum β-Lactamase-Producing Enterobacterales. Antimicrob Agents Chemother 2022;66:e0029822. [Crossref] [PubMed]
  171. Principe L, Lupia T, Andriani L, et al. Microbiological, Clinical, and PK/PD Features of the New Anti-Gram-Negative Antibiotics: β-Lactam/β-Lactamase Inhibitors in Combination and Cefiderocol-An All-Inclusive Guide for Clinicians. Pharmaceuticals (Basel) 2022;15:463. [Crossref] [PubMed]
  172. Yahav D, Giske CG, Grāmatniece A, et al. New β-Lactam-β-Lactamase Inhibitor Combinations. Clin Microbiol Rev 2020;34:e00115-20. [Crossref] [PubMed]
  173. Khan Z, Iregui A, Landman D, et al. Activity of cefepime/zidebactam (WCK 5222) against Enterobacteriaceae, Pseudomonas aeruginosa and Acinetobacter baumannii endemic to New York City medical centres. J Antimicrob Chemother 2019;74:2938-42. [Crossref] [PubMed]
  174. Zhanel GG, Esquivel J, Zelenitsky S, et al. Omadacycline: A Novel Oral and Intravenous Aminomethylcycline Antibiotic Agent. Drugs 2020;80:285-313. [Crossref] [PubMed]
  175. Monogue ML, Heil EL, Aitken SL, et al. The role of tazobactam-based combinations for the management of infections due to extended-spectrum β-lactamase-producing Enterobacterales: Insights from the Society of Infectious Diseases Pharmacists. Pharmacotherapy 2021;41:864-80. [Crossref] [PubMed]
  176. Kanj SS, Bassetti M, Kiratisin P, et al. Clinical data from studies involving novel antibiotics to treat multidrug-resistant Gram-negative bacterial infections. Int J Antimicrob Agents 2022;60:106633. [Crossref] [PubMed]
  177. Wright H, Bonomo RA, Paterson DL. New agents for the treatment of infections with Gram-negative bacteria: restoring the miracle or false dawn? Clin Microbiol Infect 2017;23:704-12. [Crossref] [PubMed]
  178. Ho KM, Kalgudi S, Corbett JM, et al. Gut microbiota in surgical and critically ill patients. Anaesth Intensive Care 2020;48:179-95. [Crossref] [PubMed]
  179. Wozniak H, Beckmann TS, Fröhlich L, et al. The central and biodynamic role of gut microbiota in critically ill patients. Crit Care 2022;26:250. [Crossref] [PubMed]
  180. Bassetti M, Bandera A, Gori A. Therapeutic Potential of the Gut Microbiota in the Management of Sepsis. Crit Care 2020;24:105. [Crossref] [PubMed]
  181. Blaser MJ. Antibiotic use and its consequences for the normal microbiome. Science 2016;352:544-5. [Crossref] [PubMed]
  182. Szychowiak P, Villageois-Tran K, Patrier J, et al. The role of the microbiota in the management of intensive care patients. Ann Intensive Care 2022;12:3. [Crossref] [PubMed]
  183. Nalluri H, Kizy S, Ewing K, et al. Peri-operative antibiotics acutely and significantly impact intestinal microbiota following bariatric surgery. Sci Rep 2020;10:20340. [Crossref] [PubMed]
  184. Haak BW, Prescott HC, Wiersinga WJ. Therapeutic Potential of the Gut Microbiota in the Prevention and Treatment of Sepsis. Front Immunol 2018;9:2042. [Crossref] [PubMed]
  185. Dickson RP. The microbiome and critical illness. Lancet Respir Med 2016;4:59-72. [Crossref] [PubMed]
  186. Wei R, Chen X, Hu L, et al. Dysbiosis of intestinal microbiota in critically ill patients and risk of in-hospital mortality. Am J Transl Res 2021;13:1548-57.
  187. Minerbi A, Shen S. Gut Microbiome in Anesthesiology and Pain Medicine. Anesthesiology 2022;137:93-108. [Crossref] [PubMed]
  188. Long DR, Alverdy JC, Vavilala MS. Emerging Paradigms in the Prevention of Surgical Site Infection: The Patient Microbiome and Antimicrobial Resistance. Anesthesiology 2022;137:252-62. [Crossref] [PubMed]
  189. Lauka L, Sobhani I, Brunetti F, et al. Human Colonic Microbiota and Short-Term Postoperative Outcomes in Colorectal Cancer Patients: A Pilot Study. Microorganisms 2021;10:41. [Crossref] [PubMed]
  190. Mustansir Dawoodbhoy F, Patel BK, Patel K, et al. Gut Microbiota Dysbiosis as a Target for Improved Post-Surgical Outcomes and Improved Patient Care: A Review of Current Literature. Shock 2021;55:441-54. [Crossref] [PubMed]
  191. Pettigrew MM, Johnson JK, Harris AD. The human microbiota: novel targets for hospital-acquired infections and antibiotic resistance. Ann Epidemiol 2016;26:342-7. [Crossref] [PubMed]
  192. Wischmeyer PE, McDonald D, Knight R. Role of the microbiome, probiotics, and 'dysbiosis therapy' in critical illness. Curr Opin Crit Care 2016;22:347-53. [Crossref] [PubMed]
  193. Donskey CJ, Chowdhry TK, Hecker MT, et al. Effect of antibiotic therapy on the density of vancomycin-resistant enterococci in the stool of colonized patients. N Engl J Med 2000;343:1925-32. [Crossref] [PubMed]
  194. Perez F, Pultz MJ, Endimiani A, et al. Effect of antibiotic treatment on establishment and elimination of intestinal colonization by KPC-producing Klebsiella pneumoniae in mice. Antimicrob Agents Chemother 2011;55:2585-9. [Crossref] [PubMed]
  195. Bell BG, Schellevis F, Stobberingh E, et al. A systematic review and meta-analysis of the effects of antibiotic consumption on antibiotic resistance. BMC Infect Dis 2014;14:13. [Crossref] [PubMed]
  196. Ruppé E, Lixandru B, Cojocaru R, et al. Relative fecal abundance of extended-spectrum-β-lactamase-producing Escherichia coli strains and their occurrence in urinary tract infections in women. Antimicrob Agents Chemother 2013;57:4512-7. [Crossref] [PubMed]
  197. Andremont O, Armand-Lefevre L, Dupuis C, et al. Semi-quantitative cultures of throat and rectal swabs are efficient tests to predict ESBL-Enterobacterales ventilator-associated pneumonia in mechanically ventilated ESBL carriers. Intensive Care Med 2020;46:1232-42. [Crossref] [PubMed]
  198. Quale JM, Landman D, Bradford PA, et al. Molecular epidemiology of a citywide outbreak of extended-spectrum beta-lactamase-producing Klebsiella pneumoniae infection. Clin Infect Dis 2002;35:834-41. [Crossref] [PubMed]
  199. Neuhauser MM, Weinstein RA, Rydman R, et al. Antibiotic resistance among gram-negative bacilli in US intensive care units: implications for fluoroquinolone use. JAMA 2003;289:885-8. [Crossref] [PubMed]
  200. Lepper PM, Grusa E, Reichl H, et al. Consumption of imipenem correlates with beta-lactam resistance in Pseudomonas aeruginosa. Antimicrob Agents Chemother 2002;46:2920-5. [Crossref] [PubMed]
  201. Ling ML, Tee YM, Tan SG, et al. Risk factors for acquisition of carbapenem resistant Enterobacteriaceae in an acute tertiary care hospital in Singapore. Antimicrob Resist Infect Control 2015;4:26. [Crossref] [PubMed]
  202. Armand-Lefèvre L, Angebault C, Barbier F, et al. Emergence of imipenem-resistant gram-negative bacilli in intestinal flora of intensive care patients. Antimicrob Agents Chemother 2013;57:1488-95. [Crossref] [PubMed]
  203. Tacconelli E, Górska A, De Angelis G, et al. Estimating the association between antibiotic exposure and colonization with extended-spectrum β-lactamase-producing Gram-negative bacteria using machine learning methods: a multicentre, prospective cohort study. Clin Microbiol Infect 2020;26:87-94. [Crossref] [PubMed]
  204. Sommer MO, Dantas G. Antibiotics and the resistant microbiome. Curr Opin Microbiol 2011;14:556-63. [Crossref] [PubMed]
  205. Woerther PL, Lepeule R, Burdet C, et al. Carbapenems and alternative β-lactams for the treatment of infections due to extended-spectrum β-lactamase-producing Enterobacteriaceae: What impact on intestinal colonisation resistance? Int J Antimicrob Agents 2018;52:762-70. [Crossref] [PubMed]
  206. Hilliquin D, Le Guern R, Thepot Seegers V, et al. Risk factors for acquisition of OXA-48-producing Klebsiella pneumonia among contact patients: a multicentre study. J Hosp Infect 2018;98:253-9. [Crossref] [PubMed]
  207. Razazi K, Derde LP, Verachten M, et al. Clinical impact and risk factors for colonization with extended-spectrum β-lactamase-producing bacteria in the intensive care unit. Intensive Care Med 2012;38:1769-78. [Crossref] [PubMed]
  208. Premachandra A, Mazeraud A, Schimpf C, et al. Is amoxicillin/clavulanic acid the best option to treat early-onset ventilator-acquired pneumonia in brain-injured patients. J Glob Antimicrob Resist 2021;27:247-9. [Crossref] [PubMed]
  209. Bräutigam HH, Knothe H, Rangoonwala R. Impact of cefotaxime and ceftriaxone on the bowel and vaginal flora after single-dose prophylaxis in vaginal hysterectomy. Drugs 1988;35:163-8. [Crossref] [PubMed]
  210. Michéa-Hamzehpour M, Auckenthaler R, Kunz J, et al. Effect of a single dose of cefotaxime or ceftriaxone on human faecal flora. A double-blind study. Drugs 1988;35:6-11. [Crossref] [PubMed]
  211. Grall N, Lazarevic V, Gaïa N, et al. Unexpected persistence of extended-spectrum β-lactamase-producing Enterobacteriaceae in the faecal microbiota of hospitalised patients treated with imipenem. Int J Antimicrob Agents 2017;50:81-7. [Crossref] [PubMed]
  212. Samonis G, Maraki S, Leventakos K, et al. Comparative effects of ertapenem, imipenem, and meropenem on the colonization of the gastrointestinal tract of mice by Candida albicans. Med Mycol 2006;44:233-5. [Crossref] [PubMed]
  213. Tan BK, Vivier E, Bouziad KA, et al. A hospital-wide intervention replacing ceftriaxone with cefotaxime to reduce rate of healthcare-associated infections caused by extended-spectrum β-lactamase-producing Enterobacteriaceae in the intensive care unit. Intensive Care Med 2018;44:672-3. [Crossref] [PubMed]
  214. Klingensmith NJ, Coopersmith CM. The Gut as the Motor of Multiple Organ Dysfunction in Critical Illness. Crit Care Clin 2016;32:203-12. [Crossref] [PubMed]
  215. Freedberg DE, Zhou MJ, Cohen ME, et al. Pathogen colonization of the gastrointestinal microbiome at intensive care unit admission and risk for subsequent death or infection. Intensive Care Med 2018;44:1203-11. [Crossref] [PubMed]
  216. Lukovic E, Moitra VK, Freedberg DE. The microbiome: implications for perioperative and critical care. Curr Opin Anaesthesiol 2019;32:412-20. [Crossref] [PubMed]
  217. Liu Z, Qin H, Yang Z, et al. Randomised clinical trial: the effects of perioperative probiotic treatment on barrier function and post-operative infectious complications in colorectal cancer surgery - a double-blind study. Aliment Pharmacol Ther 2011;33:50-63. [Crossref] [PubMed]
  218. Haak BW, Levi M, Wiersinga WJ. Microbiota-targeted therapies on the intensive care unit. Curr Opin Crit Care 2017;23:167-74. [Crossref] [PubMed]
  219. Kinross JM, Markar S, Karthikesalingam A, et al. A meta-analysis of probiotic and synbiotic use in elective surgery: does nutrition modulation of the gut microbiome improve clinical outcome? JPEN J Parenter Enteral Nutr 2013;37:243-53. [Crossref] [PubMed]
  220. Chen Y, Qi A, Teng D, et al. Probiotics and synbiotics for preventing postoperative infectious complications in colorectal cancer patients: a systematic review and meta-analysis. Tech Coloproctol 2022;26:425-36. [Crossref] [PubMed]
  221. Veziant J, Bonnet M, Occean BV, et al. Probiotics/Synbiotics to Reduce Infectious Complications after Colorectal Surgery: A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Nutrients 2022;14:3066. [Crossref] [PubMed]
  222. Kasatpibal N, Whitney JD, Saokaew S, et al. Effectiveness of Probiotic, Prebiotic, and Synbiotic Therapies in Reducing Postoperative Complications: A Systematic Review and Network Meta-analysis. Clin Infect Dis 2017;64:S153-60. [Crossref] [PubMed]
  223. Proctor LL, Ward WL, Roggy CS, et al. Potential Therapeutic Targets for Combination Antibody Therapy against Pseudomonas aeruginosa Infections. Antibiotics (Basel) 2021;10:1530. [Crossref] [PubMed]
  224. Liao C, Huang X, Wang Q, et al. Virulence Factors of Pseudomonas Aeruginosa and Antivirulence Strategies to Combat Its Drug Resistance. Front Cell Infect Microbiol 2022;12:926758. [Crossref] [PubMed]
  225. McCarthy MW. Exebacase: A Novel Approach to the Treatment of Staphylococcal Infections. Drugs R D 2022;22:113-7. [Crossref] [PubMed]
  226. Nielsen TB, Yan J, Slarve M, et al. Monoclonal Antibody Therapy against Acinetobacter baumannii. Infect Immun 2021;89:e0016221. [Crossref] [PubMed]
  227. Hauser AR, Cobb E, Bodi M, et al. Type III protein secretion is associated with poor clinical outcomes in patients with ventilator-associated pneumonia caused by Pseudomonas aeruginosa. Crit Care Med 2002;30:521-8. [Crossref] [PubMed]
  228. François B, Luyt CE, Dugard A, et al. Safety and pharmacokinetics of an anti-PcrV PEGylated monoclonal antibody fragment in mechanically ventilated patients colonized with Pseudomonas aeruginosa: a randomized, double-blind, placebo-controlled trial. Crit Care Med 2012;40:2320-6. [Crossref] [PubMed]
  229. Skurnik D, Kropec A, Roux D, et al. Natural antibodies in normal human serum inhibit Staphylococcus aureus capsular polysaccharide vaccine efficacy. Clin Infect Dis 2012;55:1188-97. [Crossref] [PubMed]
  230. Skurnik M. Can Bacteriophages Replace Antibiotics? Antibiotics (Basel) 2022;11:575. [Crossref] [PubMed]
  231. Skurnik D, Cywes-Bentley C, Pier GB. The exceptionally broad-based potential of active and passive vaccination targeting the conserved microbial surface polysaccharide PNAG. Expert Rev Vaccines 2016;15:1041-53. [Crossref] [PubMed]
  232. Cywes-Bentley C, Skurnik D, Zaidi T, et al. Antibody to a conserved antigenic target is protective against diverse prokaryotic and eukaryotic pathogens. Proc Natl Acad Sci U S A 2013;110:E2209-18. [Crossref] [PubMed]
Cite this article as: Premachandra A, Moine P. Antibiotics in anesthesia and critical care. Ann Transl Med 2024;12(1):6. doi: 10.21037/atm-22-5585

Download Citation