SciELO - Scientific Electronic Library Online

 
vol.46 número3Patient-reported outcomes y aplicaciones móviles. Revisión de su impacto en los resultados en salud de los pacientesAnálisis de la certificación en el marco de especialización farmacéutica a nivel internacional índice de autoresíndice de assuntospesquisa de artigos
Home Pagelista alfabética de periódicos  

Serviços Personalizados

Journal

Artigo

Indicadores

Links relacionados

  • Em processo de indexaçãoCitado por Google
  • Não possue artigos similaresSimilares em SciELO
  • Em processo de indexaçãoSimilares em Google

Compartilhar


Farmacia Hospitalaria

versão On-line ISSN 2171-8695versão impressa ISSN 1130-6343

Farm Hosp. vol.46 no.3 Toledo Mai./Jun. 2022  Epub 25-Jul-2022

https://dx.doi.org/10.7399/fh.13170 

REVIEWS

Pharmacokinetics and pharmacodynamics of beta-lactam antibiotics in critically ill patients

Helmi Sulaiman1  , Jason A Roberts2  3  4  5  , Mohd H Abdul–Aziz2 

1Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.

2University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane, Australia

3Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia

4Department of Pharmacy, Royal Brisbane and Women's Hospital, Brisbane, Australia

5Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France

Abstract

Optimal antibiotic therapy for critically ill patients can be complicated by the altered physiology associated with critical illness. Antibiotic pharmacokinetics and exposures can be altered driven by the underlying critical illness and medical interventions that critically ill patients receive in the intensive care unit. Furthermore, pathogens that are usually isolated in the intensive care unit are commonly less susceptible and “resistant” to common antibiotics. Indeed, antibiotic dosing that does not consider these unique differences will likely fail leading to poor clinical outcomes and the emergence of antibiotic resistance in the intensive care unit. The aims of this narrative review were to describe the pharmacokinetics of beta-lactam antibiotics in critically ill patients, to highlight pharmacokinetic/pharmacodynamic targets for both non-critically ill and critically ill patients, and to discuss important strategies that can be undertaken to optimize beta-lactam antibiotic dosing for critically ill patients in the intensive care unit.

KEYWORDS Antibiotics; Beta-lactamics; Criticaly ill patient; Clinical pharmacokinetics

Overview

Sepsis is defined as a life-threatening organ dysfunction due to dysregulated physiological response following infection1. In a large audit involving 10,069 critically ill patients managed in intensive care units (ICU) worldwide, 13.6% to 39.3% of ICU patients were diagnosed with sepsis2. Mortality rates due to sepsis may range between 15% to 20%3-5, with in-hospital mortality rates reported as high as 50.9% in Germany and 58.6% in Italy when septic shock is present6,7. The Surviving Sepsis Campaign International Guidelines for the Management of Sepsis and Septic Shock recommends aggressive resuscitation, source identification and source control, optimization of glycemic control, as well as timely administration of empiric antibiotic therapy during the early hours of sepsis8. Timely and optimal antibiotic therapy (including both spectrum of antibiotic activity and therapeutic concentration) is an important intervention for critically ill patients with sepsis or septic shock. In a recent study by Seymour et al.9 , the odds for in-hospital mortality increased by 1.04 for every hour that an appropriate antibiotic is delayed in such a patient population. However, optimal antibiotic therapy, which includes prompt delivery of antibiotics in sufficient concentrations, can be influenced and complicated by the altered physiology in critically ill patients. Antibiotic pharmacokinetic (PK) disposition, namely absorption, distribution, metabolism, and elimination can be altered in such patients, driven by the underlying critical illness and medical interventions (i.e., mechanical and pharmacological) that the patient receives. Additionally, pathogens that are usually isolated in the ICU are commonly less susceptible and “resistant” to common antibiotics10. Antibiotic dosing that does not consider these unique differences will likely fail leading to poor clinical outcomes and the emergence of antibiotic resistance in the ICU.

The aims of this narrative review were to describe the PK of beta-lactam antibiotics in critically ill patients, to highlight pharmacokinetic/pharmacodynamic (PK/PD) targets for both non-critically ill and critically ill patients, and to discuss important strategies that can be undertaken to optimize betalactam antibiotic dosing for critically ill patients in the ICU.

Pharmacokinetic changes

Absorption

The amount of drug absorbed from the site of administration (e.g., enteral, subcutaneous, and intramuscular) to the systemic circulation is influenced by physicochemical properties of beta-lactam antibiotics (e.g., solubility and molecular size), as well as the properties of the organ/tissue through which the beta-lactam antibiotics are absorbed11. In sepsis and septic shock, reduced gut motility, diminished regional blood flow, and delayed gastric emptying have been suggested to reduce drug absorption12. Intravenous (IV) administration of antibiotics is preferred to account for impaired drug absorption in critically ill patients in the ICU.

Distribution

Abnormal fluid balance following aggressive fluid resuscitation and capillary leak syndrome can result in the “third spacing” phenomenon and cumulative fluid accumulation13. This can lead to an increase in the volume of distribution, especially for hydrophilic antibiotics, including beta-lactam antibiotics, glycopeptides, aminoglycosides, linezolid, and lipopeptides14-18. The impact is more pronounced for hydrophilic antibiotics compared to lipophilic antibiotics, as the latter already has a larger volume of distribution compared to the former17. A systematic review of clinical studies that evaluated the PK of beta-lactam antibiotics in critically ill patients reported that large volume of distribution differences were commonly observed and most studies reported a 2-fold variation in this PK parameter when compared with the non-critically ill population19. This phenomenon is likely to decrease concentrations of beta-lactam antibiotics, particularly in the earlier phase of the disease. Therefore, higher initial loading doses should be applied in critically ill patients with sepsis or septic shock to compensate for the enlarged volume of distribution. Numerous studies have shown that higher initial loading doses of beta-lactam antibiotics and other antibiotics (e.g., amikacin, colistin, gentamicin, teicoplanin, and vancomycin) are required to rapidly attain effective concentrations in patients with sepsis or septic shock20-25.

Hypoalbuminemia (serum albumin < 25 g/L) is also common in critically ill patients, and this can lead to increased antibiotic distribution and clearance especially for moderately to highly-protein bound beta-lactam antibiotics (e.g., flucloxacillin, ceftriaxone, and ertapenem)26-29. The volume of distribution for highly-protein bound beta-lactam antibiotics, such as ceftriaxone and flucloxacillin, are found to be increased (as much as 90%) in critically ill patients with hypoalbuminemia. However, tissue concentrations remain low due to the “third spacing” phenomenon and cumulative fluid accumulation associated with this patient population. Furthermore, as these beta-lactam antibiotics are also cleared renally, the increase in the free fraction of drugs will also result in rapid drug clearance. The altered volume of distribution and clearance for beta-lactam antibiotics may lead to low antibiotic concentrations particularly at the end of the dosing interval. Maintenance doses for these antibiotics should be increased to compensate for this phenomenon and this is particularly relevant for time-dependent antibiotics.

Several medical interventions in the ICU, such as aggressive fluid resuscitation30, mechanical ventilation31, extracorporeal circuits32, the presence of post-surgical drains33, and total parenteral nutrition34, have also been reported to be associated with enlarged volume of distribution and consequently decreased concentrations of hydrophilic antimicrobials.

Clearance

Enhanced renal clearance of antibiotics due to elevated glomerular filtration and tubular secretion/reabsorption has been increasingly described in critically ill patients, leading to subtherapeutic concentrations of antibiotics and treatment failure35. This phenomenon is known as augmented renal clearance (ARC), which is defined as glomerular filtration rate (GFR) of above 130 mL/min/1.73 m2 (preferably based on urinary creatinine clearance)36. The phenomenon is likely observed in polytrauma, postoperative and head trauma patients, as well as younger patients, especially in those with lower disease severity37,38. Augmented renal clearance has been strongly associated with suboptimal beta-lactam antibiotic39,40 and vancomycin41-43 exposures, which may partly explain the poor clinical outcomes associated with critically ill patients receiving these antibiotics. Therefore, for these antibiotics, which display time-dependent properties and predominantly cleared by the kidneys, applying altered dosing strategies, such as extended or continuous infusion, may likely maintain effective drug concentrations for a longer duration in critically ill patients with ARC.

On the other hand, reduced antibiotic metabolism and clearance might occur following organ hypoperfusion leading to renal and/or hepatic dysfunction. As the disease progresses in a critically ill patient, myocardial depression may occur leading to decreased organ perfusion and microcirculatory failure. These could cause end-organ damage or in extreme cases, multi-organ dysfunction syndrome32. This syndrome often includes renal and/or hepatic dysfunction that consequently results in decreased antibiotic clearance. Renal dysfunction or acute kidney injury (AKI) leads to reduced GFR and clearance of renally eliminated antibiotics. Nonetheless, dose reduction of antibiotics is not straight forward in patients with AKI, as the decision should consider patient's residual kidney function and fluid status, use of renal replacement therapy (RRT), and the antibiotic PK/PD target44. Beta-lactam antibiotic dosing requirements for critically ill patients are highly dynamic, and regular dosing reviews and modifications are likely required to prevent both suboptimal dosing and development of adverse events.

Mechanical intervention

Mechanical intervention for organ support has been shown to alter antibiotic PK45-49. Mechanical ventilation can alter antibiotic PK by increasing the intrathoracic pressure, leading to a decreased venous return to the heart50. This can lead to an increase in the antibiotic volume of distribution, as well as reduced clearance due to decreased GFR47,51,52. In a study performed by Medellín-Garibay et al., mechanical ventilation was shown to reduce the clearance of vancomycin by 20%46. The decrease in drug clearance was linked to hemodynamic changes in mechanically ventilated patients, which reduced the renal blood flow leading to a reduction in the glomerular function and urine output53. Renal replacement therapy may alter the PK of beta-lactam antibiotics by augmenting antibiotic clearance and volume of distribution54. This is dependent on a few factors including dialysate flow rate, mode of dialysis, type of dialysis membrane, dialysis duration, as well as antibiotic physiochemical properties, and the degree of protein binding55. Patients with AKI receive various forms of RRT, but continuous renal replacement therapy (CRRT) remains the common mode of RRT for critically ill patients in the ICU56,57. CRRT is commonly not applied in a uniform way and therefore, antibiotic clearance may greatly vary and be lower than what has been initially prescribed48,57. No conclusive dosing recommendations can be made currently for critically ill patients receiving CRRT but as a general rule, antibiotics with a high volume of distribution (1 L/kg or greater) and/or that are highly protein bound (80% or greater) are generally poorly eliminated by CRRT58. Current data suggest that a significant proportion of CRRT patients are at an increased risk for either antibiotic underexposure or overexposure48.

Another form of mechanical intervention in the ICU is extracorporeal membrane oxygenation (ECMO). It is an artificial and temporary respiratory and/or cardiac support that is carried out extracorporeally (i.e., cardio-pulmonary bypass) in patients with cardiorespiratory failure refractory to conventional medical therapies59. Its use has increased steadily, especially during the current COVID-19 pandemic, whereby the World Health Organization (WHO) recommends its use in COVID-19 patients with profound hypoxemia (with or without hypercapnia) not amenable to the lung-protective ventilation60. Earlier neonatal and paediatric data suggest that ECMO has the potential to alter the PK of many important antibiotics including beta-lactam antibiotics61. ECMO extracorporeal circuits consisting of conduit tubing provide an additional “compartment” through which beta-lactam antibiotics can distribute62. The circuit tubes, as well as the oxygenator membrane, introduce additional surface areas that the beta-lactam antibiotics can adhere and sequester onto. This is particularly problematic to antibiotics that are lipophilic63, highly-protein bound (e.g., ceftriaxone64), or chemically unstable (e.g., meropenem65). The priming of the ECMO circuit can also dilute and sequester the drug further66. All the above are theorized to lead to an increase in the beta-lactams volume of distribution and possibly treatment failure due to subtherapeutic concentrations65. In addition, ECMO patients have lower drug clearance when compared to patients not undergoing ECMO67. However, based on current clinical evidence: (1) modern ECMO circuits have minimal impact on the PK of most antibiotics, including beta-lactam antibiotics; (2) PK changes in patients receiving ECMO are more reflective of critical illness rather than ECMO therapy itself; and (3) apart from lipophilic and highlyprotein bound antibiotics, the impact of ECMO on the PK and dosing requirements is likely to be minimal68-73.

In conclusion, profound alteration of beta-lactam antibiotic PK is common in critically ill patients, especially in those receiving mechanical organ support. Nonetheless, contemporary antibiotic dosing is largely based on dose-finding studies which mostly included healthy participants and patients who are not critically ill. Such a dosing approach has been increasingly shown to increase risks of suboptimal beta-lactam antibiotic exposure in a large proportion of critically ill patients74,75.

Beta-lactam antibiotic pharmacodynamics

Antibiotics can be broadly categorized into three pharmacokinetics/ pharmacodynamics (PK/PD) groups based on their modes of bacterial killing76: concentration-dependent, time-dependent, and both concentration- and time-dependent agents. For concentration-dependent antibiotics, a direct relationship exists between antibiotic concentration and efficacy where increasing concentrations enhance bacterial killing. For these antibiotics (e.g., aminoglycosides and fluoroquinolones), the maximum concentration (Cmax) relative to the minimum inhibitory concentration (Cmax/MIC) best describes their activity. For time-dependent antibiotics (e.g., beta-lactam antibiotics), prolonging the duration of exposure enhances bacterial killing and it is the percentage of the dosing interval that the free drug concentrations remain above the MIC (%ƒT>MIC) that drives their efficacy. For antibiotics that display both concentration- and time-dependent killing characteristics, the ratio of area under the concentration-time curve (AUC) to MIC (AUC/MIC) best describes their activity. For antibiotics, achieving these PK/PD indices may increase the likelihood of microbiological and clinical response.

The PK/PD index associated with optimal beta-lactam antibiotic activity is the % ƒT>MIC (40 – 70%)77. Beta-lactam antibiotics demonstrate superior bacterial killing the longer that drug concentrations remain above the MIC of a pathogen. Clinical data from critically ill patients suggest that these patients may benefit from longer (e.g., 100%ƒT>MIC) and higher (e.g., 2 – 5 x MIC) beta-lactam exposures than those previously described in in vitro and in vivo animal model studies77.

Beta-lactam antibiotic optimization in critically ill patients

Pharmacokinetic changes of beta-lactam antibiotics in critically ill patients

Poor beta-lactam antibiotic PK/PD target attainment in critically ill patients has been illustrated in two recently published multicentre clinical studies48,74. In a large point prevalence PK study of beta-lactam antibiotics (i.e., the DALI study) involving 384 patients, up to 500-fold variations were seen in the unbound concentrations of the studied beta-lactam antibiotics74. Of these, 248 (64.5%) patients were treated for infection and 40 (16.0%) of them did not achieve the predefined PK/PD target (50%ƒT>MIC) and they were 32.0% less likely to have a positive clinical outcome. Similarly, poor target attainment was seen in the SMARRT study, a large prospective, multinational PK study, involving 381 patients on RRT receiving either meropenem, piperacillin-tazobactam or vancomycin48. Up to 55% of the concentrations failed to achieve the low target trough concentrations, with higher failure rates (up to 72.0%) seen with the high target trough concentrations. The trough concentrations were inversely associated with the estimated total renal clearance of the prescribed RRT and residual renal clearance. In addition, highly variable trough concentrations (up to 8-fold) were seen in this study.

Strategies to improve PK/PD target attainment

Altered dosing strategy via prolonged infusion

Numerous studies have shown better PK/PD target attainment with prolonged (PI) or continuous infusion (CI) of beta-lactam antibiotic compared to intermittent bolus (IB) dosing78,79. The altered dosing strategy increases the percentage of time that free beta-lactams concentrations remains above the target MIC for a given dosing interval, allowing enhanced bacterial killing80. Roberts et al., demonstrated median steady-state concentrations of 16.6 mg/L with CI of piperacillin when compared to median Cmin concentrations of 4.9 mg/L following IB dosing, despite a lower total CI daily dose (25% lower than the IB regimens)79. Noteworthy, systemic review and meta-analyses comparing between CI/PI and IB dosing of beta-lactam antibiotics in terms of survival benefit have shown mixed results81-87. However, when the studies are limited to patients with severe sepsis who received equivalent doses of antibiotics in both arms (IB vs. PI/CI), lower in-hospital mortality was shown in patients receiving CI of beta-lactam antibiotics81.

Prior to the rollout of CI or PI dosing of beta-lactam antibiotics in the ICU, physicians should consider the following: 1) the use of loading dose; 2) antibiotic stability; 3) residual volume or dead space; 4) compatibility with other drugs; 5) drug accumulation in patients with renal dysfunction; and 6) target population and knowledge of susceptibility data for antibiotics aimed for PI program. Application of loading dose would shorten the time to therapeutic exposure88,89. A loading dose is a short-term dose given as an intermittent bolus (30-60 minutes) followed by the total recommended daily dose given as PI or CI90. Infusion duration, beta-lactam antibiotic concentrations (with lower final concentrations post reconstitution being more stable compared to higher concentrations), types of diluents, and container used for reconstitution can affect the beta-lactam stability, and therefore, these need to be considered prior to the beta-lactam antibiotic CI/PI dosing rollout in the ICU. Of the beta-lactam antibiotics that have been studied for PI, both meropenem and imipenem show the shortest duration of stability (4 to 9 hours after reconstitution with water for injection)91,92. Other beta-lactam antibiotics including amoxycillin, benzylpenicillin, and ceftazidime have also been reported to be stable less than 24 hours93-95. Therefore, multiple infusions need to be given over 24 hours when these agents are used. Next, residual volume following beta-lactams infusion can reduce the total amount of beta-lactams given to patients. Bolla et al., showed that more than 10% of antimicrobial would be lost for 26 of 39 studied antibiotics if the residual volume is not infused back to patients at the end of the infusion96. Noteworthy, rapid flushing of the intravenous (IV) line to deliver this “infusion line dead space” to patients is contra to the principle of PI as the residual volume will be delivered in a bolus form rather than prolonged infusion97. Therefore, the residual volume should instead be infused at an appropriate rate with clear administration instructions given to the nurses. Another strategy that can be used to overcome this is by giving higher doses to compensate for the loss from the residual volume98. To the best of our knowledge, there is yet a published PK study assessing the effectiveness of these strategies in improving beta-lactam antibiotic exposures in patients with the infusion line dead space. Another consideration is the beta-lactam antibiotics compatibility with other IV drugs when the beta-lactams cannot be given through a dedicated line. The issue can be handled through three possible ways: 1) co-administration of a drug/drugs that is proven to be compatible with the beta-lactam antibiotics; 2) the placement of additional IV line; and 3) shortening the infusion time of the beta-lactams (i.e., less than 24 hours) with the adjustment of medication administration time to allow the incompatible drug(s) to be administered at different occasions99.

TDM-based strategy

Dose personalization for beta-lactam antibiotics guided by therapeutic drug monitoring (TDM) is still under investigation as beta-lactam antibiotics have a wide therapeutic range with a favorable safety profile, unlike aminoglycosides and vancomycin. However, given the high PK variability seen with beta-lactam antibiotics in critically ill patients19, TDM might be useful in optimizing drug exposure as the variability might lead to subtherapeutic or supratherapeutic concentrations. It might also be useful in a certain group of critically ill patients, such as those who are obese, immunocompromised, infected by resistant bacterial strains, and in those receiving RRT or having augmented renal clearance100-103. These patients are at high risk of underdosing. Nonetheless, there are only two published randomized controlled trials (RCT) on beta-lactam antibiotics TDM, with both using PK/PD target attainment rather than clinical endpoints (e.g., survival benefit) as their outcome measures104,105. Currently, there are two ongoing randomized controlled trials (RCTs) investigating this question: does beta-lactam antibiotics TDM improve clinical outcomes106,107?

Several observational studies on beta-lactam antibiotics TDM have been published (Table 1)108-112. In these studies, samples for TDM were performed at steady-state prior to the next dosing (i.e., trough or Cmin concentration), except in one study, whereby mid-dose and trough samplings were performed in patients receiving IB dosing of beta-lactams (in the same study, the sampling was performed at steady state in the CI arm)108. Variable PK/PD targets were used, with dosing adjustments performed in patients who did not achieve the predefined targets. For example, Wong et al., increased the dosing frequency of the beta-lactams by 25% to 50% or changed the infusion strategy from IB to PI/CI dosing when the beta-lactams concentrations were below the PK/PD target (i.e., 100%ƒT>MIC)108. Conversely, the dose was either decreased by 50% or the frequency was reduced by 25% to 50% of the same daily dose when the PK/PD exposures were higher than 100%ƒT>MIC. The rates of PK/PD target attainment for 100%ƒT>MIC and 100%ƒT>4 X MIC were 66.9% and 36.6%, respectively for the seven studied beta-lactam antibiotics. Collectively, these studies showed that a significant number of critically ill patients did not achieve the desired PK/PD targets for patient benefits. Poor target attainment, even with alternative dosing strategy (e.g., PI dosing)108 in some studies, underlines the difficulty of getting the dose right in critically ill patients. Therefore, applying aggressive initial dosing regimen (e.g., high-dose regimens via CI or PI during the initial empiric therapy) coupled with TDM may currently be the best strategy to optimize of beta-lactam antibiotic exposures in critically ill patients.

Table 1.  Observational TDM studies of beta lactams 

CI: continuous infusion; IB: intermittent bolus; ICU: intensive care unit.

Table 1 (cont.).  Observational TDM studies of beta lactams 

CI: continuous infusion; IB: intermittent bolus; ICU: intensive care unit.

Conclusion

Appropriate beta-lactam antibiotic dosing remains a challenge in critically ill patients, who are at a high risk of mortality due to sepsis and septic shock. These patients are usually managed in the ICU, where infections by resistant pathogens, especially Gram-negative microorganisms are common. This makes the attainment of PK/PD target in this group even more difficult. Therefore, alternative dosing strategies for beta-lactam antibiotics should be considered for these patients, which may include the use of PI or CI dosing. Personalized dosing guided by TDM can also improve PK/PD target attainment within an individual patient, and data from ongoing RCTs are needed to support a global practice of performing TDM for beta-lactam antibiotics in critically ill patients.

Bibliography

1. Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, et al. The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. 2016;315:801-10. DOI: 10.1001/jama.2016.0287 [ Links ]

2. Sakr Y, Jaschinski U, Wittebole X, Szakmany T, Lipman J, Ñamendys-Silva SA, et al. Sepsis in intensive care unit patients: worldwide data from the intensive care over nations audit. Open Forum Infect Dis. 2018;5:313. DOI: 10.1093./ofid/ofy313 [ Links ]

3. Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of sepsis in the United States from 1979 through 2000. N Eng J Med. 2003;348:1546-54. DOI: 10.1056/NEJMoa022139 [ Links ]

4. Kaukonen KM, Bailey M, Suzuki S, Pilcher D, Bellomo R. Mortality related to severe sepsis and septic shock among critically ill patients in Australia and New Zealand, 2000-2012. JAMA. 2014;311:1308-16. DOI: 10.1001/jama.2014.2637 [ Links ]

5. Rhee C, Dantes R, Epstein L, Murphy D, Seymour CW, Iwashyna TJ, et al. Incidence and Trends of Sepsis in US Hospitals Using Clinical vs Claims Data, 2009-2014. JAMA. 2017;318:1241-9. DOI: 10.1001/jama.2017.13836 [ Links ]

6. SepNet Critical Care Trial Group. Incidence of severe sepsis and septic shock in German intensive care units: the prospective, multicentre INSEP study. Intensive Care Med. 2016;42:1980-9. DOI: 10.1007/s00134-016-4504-3 [ Links ]

7. Sakr Y, Elia C, Mascia L, Barberis B, Cardellino S, Livigni L, et al. Epidemiology and outcome of sepsis syndromes in Italian ICUs: a muticentre, observational cohort study in the region of Piedmont. Minerva Anestesiol. 2013;79:993-1002. [ Links ]

8. Rhodes A, Evans LE, Alhazzani W, Levy MM, Antonelli M, Ferrer R, et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Crit Care Med. 2017;45:486-552. DOI: 10.1097/CCM.0000000000002255 [ Links ]

9. Seymour CW, Gesten F, Prescott HC, Friedrich ME, Iwashyna TJ, Pjillips GS, et al. Time to treatment and mortality during mandated emergency care for sepsis. N Eng J Med. 2017;376:2235-44. DOI: 10.1056/NEJMoa1703058 [ Links ]

10. Sievert DM, Ricks P, Edwards JR, Schneider A, Patel J, Srinivasan A, et al. Antimicrobial-resistant pathogens associated with healthcare-associated infections: summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2009-2010. Infect Control Hosp Epidemiol. 2013;34:1-14. DOI: 10.1086/668770 [ Links ]

11. 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. DOI: 10.1016/j.addr.2014.07.006 [ Links ]

12. Jochberger S, Mayr V, Luckner G, Fries DR, Mayr AJ, Friesenecker BE, et al. Antifactor Xa activity in critically ill patients receiving antithrombotic prophylaxis with standard dosages of certoparin: a prospective, clinical study. Crit Care. 2005;9:1-8. DOI: 10.1186/cc37923 [ Links ]

13. Mehta RL. Fluid balance issues in the critically ill patient. Fluid Overload. 2010;164:69-78. DOI: 10.1159/000313722 [ Links ]

14. Conil J, Georges B, Brenden A, Segonds C, Lavit M, Seguin T, et al. Increased amikacin dosage requirements in burn patients receiving a once-daily regimen. Int J Antimicrob Agents. 2006;28:226-30. DOI: 10.1016/j.ijantimicag [ Links ]

15. Marik P. Aminoglycoside volume of distribution and illness severity in critically ill septic patients. Anaesth Intensive Care. 1993;21:172-3. DOI: 10.1177/0310057X9302100206 [ Links ]

16. Buerger C, Plock N, Dehghanyar P, Joukhadar C, Kloft C. Pharmacokinetics of unbound linezolid in plasma and tissue interstitium of critically ill patients after multiple dosing using microdialysis. Antimicrob Agents Chemother. 2006;50:2455-63. DOI: 10.1128/AAC.01468-05 [ Links ]

17. Roberts JA, Abdul-Aziz MH, Lipman J, Mouton JW, Vinks AA, Felton TW, et al. Individualised antibiotic dosing for patients who are critically ill: challenges and potential solutions. Lancet Infect Dis. 2014;14:498-509. DOI: 10.1016/S1473.3099(14)70036.-2 [ Links ]

18. De Winter S, Van Hest R, Dreesen E, Annaert P, Wauters J, Meersseman W, et al. Quantification and Explanation of the Variability of First-Dose Amikacin Concentrations in Critically Ill Patients Admitted to the Emergency Department: A Population Pharmacokinetic Analysis. Eur J Drug Metabol Pharmacokin. 2011;1:11. DOI: 10.1007/s13318-021-00698-w [ Links ]

19. Goncalves-Pereira J, Povoa P. Antibiotics in critically ill patients: a systematic review of the pharmacokinetics of beta-lactams. Crit Care. 2011;15:R206. DOI: 10.1186/cc10441 [ Links ]

20. Taccone FS, Laterre PF, Spapen H, Dugernier T, Delattre I, Layeux B, et al. Revisiting the loading dose of amikacin for patients with severe sepsis and septic shock. Crit Care. 2010;14:1-10. DOI: 101186/cc8945 [ Links ]

21. Delattre IK, Hites M, Laterre PF, Dugernier T, Spapen H, Wallemacq PE, et al. What is the optimal loading dose of broad-spectrum β-lactam antibiotics in septic patients? Results from pharmacokinetic simulation modelling. Int J Antimicrob Agents. 2020;56:106-13. DOI: 10.1016/j.ijantimicag.2020.106113 [ Links ]

22. Tsuji BT, Pogue JM, Zavascki AP, Paul M, Daikos GL, Forrest A, et al. International consensus guidelines for the optimal use of the polymyxins. Pharmacotherapy. 2019;39;10-39. DOI: 10.1002/phar.2209 [ Links ]

23. Goncalves-Pereira J, Martins A, Povoa P. Pharmacokinetics of gentamicin in critically ill patients: pilot study evaluating the first dose. Clin Microbiol Infect. 2010;16:1258-63. DOI: 10.11/j.1469-0691.2009.03-074.x [ Links ]

24. Sato M, Chida K, Suda T, Gemma H, Nakumura H, Muramatsu H, et al. Recommended initial loading dose of teicoplanin, established by therapeutic drug monitoring, and outcome in terms of optimal trough level. J Infect Chemother. 2006;12:185-9. DOI: 10.1007/s10156-006-0446-Y [ Links ]

25. Álvarez O, Plaza-Plaza JC, Ramírez M, Peralta A, Amador CA, Amador R, et al. Pharmacokinetic assessment of vancomycin loading dose in critically ill patients. Antimicrob Agents Chemother. 2017;61:e00280-00217. DOI: 10.1128/AAC.00280-17 [ Links ]

26. Ulldemolins M, Roberts JA, Wallis SC, Rello J, Lipman J. Flucloxacillin dosing in critically ill patients with hypoalbuminaemia: special emphasis on unbound pharmacokinetics. J Antimicrob Chemother. 2010;65:1771-8. DOI: 10.1093/jac/dkq184 [ Links ]

27. Schleibinger M, Steinbach C, Töpper C, Kratzer A, Liebchen U, Kees F, et al. Protein binding characteristics and pharmacokinetics of ceftriaxone in intensive care unit patients. Br J Clin Pharmacol. 2015;80:525-33. DOI: 10.1111/bcp.12636 [ Links ]

28. Brink A, Richards G, Schillack V, Kiem S, Schentag J. Pharmacokinetics of oncedaily dosing of ertapenem in critically ill patients with severe sepsis. Int J Antimicrob Agents. 2009;33:432-6. DOI: 10.1016/j.ijantimicag.2008.10.005 [ Links ]

29. Gregoire N, Chauzy A, Buyck J, Rammaert B, Couet W, Marchand S, et al. Clinical Pharmacokinetics of Daptomycin. Clin Pharmacokinet. 2021;60:271-81. DOI: 10.1007/s40262-020-00968-x [ Links ]

30. Ocampos-Martinez E, Penaccini L, Scolleta S, Abdelhadii A, Devigli A, Cianferoni S, et al. Determinants of early inadequate vancomycin concentrations during continuous infusion in septic patients. Int J Antimicrob Agents. 2012;39:332-7. DOI: 10.1016/j.ijantimicag.2011.12.008 [ Links ]

31. Conil JM, Georges B, Lavit M, Laguerre J, Samii K, Houin G, et al. A population pharmacokinetic approach to ceftazidime use in burn patients: influence of glomerular filtration, gender and mechanical ventilation. Br J Clin Pharmacol. 2007;64:27-35. DOI: 10.1111/j.1365-2125.2007.02857.x [ Links ]

32. Hites M, Dell'Anna AM, Scolletta S, Taccone FS. The challenges of multiple organ dysfunction syndrome and extra-corporeal circuits for drug delivery in critically ill patients. Adv Drug Deliv Rev. 2014;77:12-21. DOI: 10.1016/j.addr.2014.05.007 [ Links ]

33. Adnan S, Xuanhui J, Wallis S, Rudd M, Jarret P, Paterson D, et al. Pharmacokinetics of meropenem and piperacillin in critically ill patients with indwelling surgical drains. Int J Antimicrob Agents. 2013;42:90-3. DOI: 10.1016/j.ijantimicag.2013.02.023 [ Links ]

34. Ronchera-Oms CL, Tormo C, Ordovás JP, Abad J, Jiménez NV. Expanded gentamicin volume of distribution in critically ill adult patients receiving total parenteral nutrition. J Clin Pharm Ther. 1995;20:253-8. DOI: 10.1111/j.1365-2710.1995.tb00659.x [ Links ]

35. Claus B, Colpaert K, Hoste E, Decruyenaere J, De Waele J. Increased glomerular filtration in the critically ill patient receiving anti-infective treatment. Crit Care. 2010;14:1-2. [ Links ]

36. Bilbao-Meseguer I, Rodríguez-Gascón A, Barrasa H, Isla A, Solinís MÁ. Augmented renal clearance in critically ill patients: a systematic review. Clin Pharmacokinet. 2018;57:1107-21. DOI: 10.1007/s40262-018-0636-7 [ Links ]

37. Fuster-Lluch O, Gerónimo-Pardo M, Peyró-García R, Lizán-García M. Glomerular hyperfiltration and albuminuria in critically ill patients. Anaesth Intensive Care. 2008,36:674-80. DOI: 10.1177/0310057X0803600507 [ Links ]

38. Udy AA, Jarret P, Lassing-Smith M, Stuart J, Starr T, Dunlop R, et al. Augmented Renal Clearance in Traumatic Brain Injury: A Single-Center Observational Study of Atrial Natriuretic Peptide, Cardiac Output, and Creatinine Clearance. J Neurotraum. 2017;34:137-44. DOI: 10.1089/neu.2015.4328 [ Links ]

39. Carrie C, Petit L, D´Houdain N, Sauvage N, Cottenceau V, Laffite M, et al. Association between augmented renal clearance, antibiotic exposure and clinical outcome in critically ill septic patients receiving high doses of beta-lactams administered by continuous infusion: a prospective observational study. Int J Antimicrob Agents. 2018;51:443-9. DOI: 10.1016/j.ijantimicag.2017.11.013 [ Links ]

40. Huttner A, Von Dach E, Renzoni A, Huttner BD, Affaticati M, Pagani L, et al. Augmented renal clearance, low beta-lactam concentrations and clinical outcomes in the critically ill: an observational prospective cohort study. Int J Antimicrob Agents. 2015;45:385-92. DOI: 10.1016/j.ijantimicag.2014.12.017 [ Links ]

41. Bakke V, Sporsem H, Von der Lippe E, Nordoy I, lao Y, Nyrerod HC, et al. Vancomycin levels are frequently subtherapeutic in critically ill patients: a prospective observational study. Acta Anaesthesiol Scand. 2017;61:627-35. DOI: 10.1111/aas.12897 [ Links ]

42. Hirai K, Ishii H, Shimosshikiryo T, Shimomura T, Tsuji D, Inoue K, et al. Augmented Renal Clearance in Patients With Febrile Neutropenia is Associated With Increased Risk for Subtherapeutic Concentrations of Vancomycin. Ther Drug Monit. 2016;38:706-10. DOI: 10.1097/FTD.0000000000000346 [ Links ]

43. Baptista JP, Sousa E, Martins PJ, Pimentel JM. Augmented renal clearance in septic patients and implications for vancomycin optimisation. Int J Antimicrob Agents. 2012;39:420-3. DOI: 10.1016/j.ijantimicag.2011.12.011 [ Links ]

44. Eyler RF, Mueller BA. Antibiotic dosing in critically ill patients with acute kidney injury. Nat Rev Nephrol. 2011;7:226-35. DOI: 10.1038/nrneph.2011.12 [ Links ]

45. Jamal JA, Udy AA, Lipman J, Roberts JA. The impact of variation in renal replacement therapy settings on piperacillin, meropenem, and vancomycin drug clearance in the critically ill: an analysis of published literature and dosing regimens. Crit Care Med. 2014;42:1640-50. DOI: 10.1097/CCM.0000000000000317 [ Links ]

46. Medellín-Garibay SE, Romano-Moreno S, Tejedor-Prado P, Rubio-Álvaro N, Rueda-Naharro A, Blasco-Navalpotro MA, et al. Influence of mechanical ventilation on the pharmacokinetics of vancomycin administered by continuous infusion in critically ill patients. Antimicrob Agents Chemother. 2017;61:e01249-01217. DOI: 10.1128/AAC.01249-17 [ Links ]

47. Conil JM, Georges B, Labit M, Laguerre J, Samii K, Houin G, et al. A population pharmacokinetic approach to ceftazidime use in burn patients: influence of glomerular filtration, gender and mechanical ventilation. Br J Clin Pharmacol. 2007;64:27-35. DOI: 10.1111/j.1365-2125.2007.02857.x [ Links ]

48. Roberts JA, Goynt GM, Lee A, Choi G, Bellomo R, Kanji S, et al. The effect of renal replacement therapy and antibiotic dose on antibiotic concentrations in critically ill patients: data from the multinational sampling antibiotics in renal replacement therapy study. Clin Infect Dis. 2021;72:1369-78. DOI: 10.1093/cid/ciaa224 [ Links ]

49. Burdet C, Pajot O, Couffignal C, Armand-Lefèvre L, Foucrier A, Laouènan C, et al. Population pharmacokinetics of single-dose amikacin in critically ill patients with suspected ventilator-associated pneumonia. Eur J Clin Pharmacol. 2015;71:75-83. DOI: 10.1007/s00228-014-1766-y [ Links ]

50. Der Merwe F, Wallis S, Udy A. Understanding the impact of critical illness on drug pharmacokinetics-scientifically robust study design. J Clinic Toxicol. 2012;S4:2161-0495. [ Links ]

51. Georges B, Conil JM, Seguin T, Ruiz S, Minville V, Cougot P, et al. Population pharmacokinetics of ceftazidime in intensive care unit patients: influence of glomerular filtration rate, mechanical ventilation, and reason for admission. Antimicrob Agents Chemother. 2009;53:4483-9. DOI: 10.1128/AAC.00430-09 [ Links ]

52. Martin C, Lambert D, Bruguerolle B, Saux P, Freney J, Fleurette J, et al. Ofloxacin pharmacokinetics in mechanically ventilated patients. Antimicrob Agents Chemother. 1991;35:1582-5. DOI: 10.1128/AAC.35.8.1582 [ Links ]

53. Perkins MW, Dasta JF, Dehaven B. Physiologic implications of mechanical ventilation on pharmacokinetics. DICP. 1989;23:316-23. DOI: 10.1177/106002808902300408 [ Links ]

54. Roberts JA, Joynt GM, Lee A, Choi G, Bellomo R, Kanji S, et al. The Effect of Renal Replacement Therapy and Antibiotic Dose on Antibiotic Concentrations in Critically Ill Patients: Data From the Multinational Sampling Antibiotics in Renal Replacement Therapy Study. Clin Infect Dis. 2021;72:1369-78. DOI: 10.1093/cid/ciaa224 [ Links ]

55. Pistolesi V, Morabito S, Di Mario F, Regolisti G, Cantarelli C, Fiaccadori E. A guide to understanding antimicrobial drug dosing in critically ill patients on renal replacement therapy. Antimicrob Agents Chemother. 2019;63:e00583-00519. DOI: 10.1128/AAC.00583-19 [ Links ]

56. Legrand M, Darmon M, Joannidis M, Payen D. Management of renal replacement therapy in ICU patients: an international survey. Intensive Care Med. 2013;39:101-8. DOI: 10.1007/s00134-012-2706-x [ Links ]

57. Bellomo R, Cass A, Cole L, Finfer S, Gallaher M, Goldsmith D, et al. Renal replacement therapy for acute kidney injury in Australian and New Zealand intensive care units: a practice survey. Crit Care Resusc. 2008;10:225-30. [ Links ]

58. Sime FB, Roberts JA. Antibiotic dosing in critically ill patients receiving renal replacement therapy. Expert Rev Clin Pharmacol. 2016;9:497-9. DOI: 10.1586/17512433.2016.1133290 [ Links ]

59. Gattinoni L, Carlesso E, Langer T. Clinical review: Extracorporeal membrane oxygenation. Crit Care. 2011;15:1-6. DOI: 10.1186/cc10490 [ Links ]

60. World Health Organization. Clinical management of severe acute respiratory infection when novel coronavirus (2019-nCoV) infection is suspected: interim guidance. in Clinical management of severe acute respiratory infection when novel coronavirus (2019-nCoV) infection is suspected: Interim guidance 21-21 (2020). [ Links ]

61. Sherwin J, Heath T, Watt K. Pharmacokinetics and Dosing of Anti-infective Drugs in Patients on Extracorporeal Membrane Oxygenation: A Review of the Current Literature. Clin Therap. 2016;38:1976-94. DOI: 10.1016/j.clinthera.2016.07.169 [ Links ]

62. Cheng V, Abdul-Aziz MH, Roberts JA, Shekar K. Optimising drug dosing in patients receiving extracorporeal membrane oxygenation. J Thorac Dis. 2018;10:S629. DOI: 10.21037/jtd.2017.09.154 [ Links ]

63. Wildschut E, Ahsman M, Allegaert K, Mathot R, Tibboel D. Determinants of drug absorption in different ECMO circuits. Intensive Care Med. 2010;36:2109-16. DOI: 10.1007/s00134-010-2041-z [ Links ]

64. Shekar K, Roberts JA, Barnett AG, Diab S, Wallis SC, Fung YL, et al. Can physicochemical properties of antimicrobials be used to predict their pharmacokinetics during extracorporeal membrane oxygenation? Illustrative data from ovine models. Crit Care. 2015;19:1-11. DOI: 10.1186/s13054-015-1151-y [ Links ]

65. Shekar K, Roberts JA, McDonald CI, Fisquet S, Barnett AG, Mullany DV, et al. Sequestration of drugs in the circuit may lead to therapeutic failure during extracorporeal membrane oxygenation. Crit Care. 2012;16:1-7. DOI: 10.1186/cc11679 [ Links ]

66. Buck ML. Pharmacokinetic changes during extracorporeal membrane oxygenation. Clin Pharmacokinet. 2003;42:403-17. DOI: 10.2165/00003088-200342050-00001 [ Links ]

67. Shekar K, Fraser JF, Smith MT, Roberts JA. Pharmacokinetic changes in patients receiving extracorporeal membrane oxygenation. J Crit Care. 2012;27:741-9. DOI: 10.1016/j.jcrc.2012.02.013 [ Links ]

68. Cheng V, Abdul-Aziz MH, Burrows F, Buscher H, Cho YJ, Corley A, et al. Population Pharmacokinetics of Piperacillin and Tazobactam in Critically Ill Patients Receiving Extracorporeal Membrane Oxygenation: an ASAP ECMO Study. Antimicrob Agents Chemother. 2021;65:e0143821. DOI: 10.1128/AAC.01438-21 [ Links ]

69. Cheng V, Abdul-Aziz MH, Burrows F, Buscher H, Cho YJ, Corley A, et al. Population pharmacokinetics of vancomycin in critically ill adult patients receiving extracorporeal membrane oxygenation (an ASAP ECMO study). Antimicrob Agents Chemother. 2021;AAC0137721. DOI: 10.1128/AAC.01377-21 [ Links ]

70. Cheng V, Abdul-Aziz MH, Burrows F, Buscher H, Corley A, Dielh A, et al. Population pharmacokinetics of cefepime in critically ill patients receiving extracorporeal membrane oxygenation (an ASAP ECMO study). Int J Antimicrob Agents. 2021;58:106466. DOI: 10.1016/j.ijantimicag.2021.106466 [ Links ]

71. Dhanani JA, Lipman J, Pincus J, Townsend S, Livermore A, Wallis SC, et al. Pharmacokinetics of fluconazole and ganciclovir as combination antimicrobial chemotherapy on ECMO: a case report. Int J Antimicrob Agents. 2021;106431. DOI: 10.1016/j.ijantimicag.2021.106431 [ Links ]

72. Dhanani JA, Lipman J, Pincus J, Townsend S, Livermore A, Wallis SC, et al. Pharmacokinetics of Sulfamethoxazole and Trimethoprim During Venovenous Extracorporeal Membrane Oxygenation: A Case Report. Pharmacotherapy. 2020;40(7):713-7. DOI: 10.1002/phar.2413 [ Links ]

73. Dhanani JA, Lipman J, Pincus J, Townsend S, Livermore A, Wallis SC, et al. Pharmacokinetics of Total and Unbound Cefazolin during Veno-Arterial Extracorporeal Membrane Oxygenation: A Case Report. Chemotherapy. 2019;64:115-8. DOI: 10.1159/000502474 [ Links ]

74. Roberts JA, Paul SK, Akova M, Bassetti M, De Waele JJ, Dimopoulos G, 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. DOI: 10.1093/cid/ciu027 [ Links ]

75. Blot S, Koulenti D, Akova M, Bassetti M, De Waele JJ, Dimopoulos G, et al. Does contemporary vancomycin dosing achieve therapeutic targets in a heterogeneous clinical cohort of critically ill patients? Data from the multinational DALI study. Crit Care. 2014;18:1-12. DOI: 10.1186/cc13874 [ Links ]

76. Craig WA. Pharmacokinetic/pharmacodynamic parameters: rationale for antibacterial dosing of mice and men. Clin Infect Dis. 1998;26:1-10. DOI: 10.1086/516284 [ Links ]

77. Abdul-Aziz MH, Alffenaar JWC, Bassetti M, Bracht H, Dimopoulos G, Marriot D, et al. Antimicrobial therapeutic drug monitoring in critically ill adult patients: a Position Paper. Intensive Care Med. 2020;46:1127-53. DOI: 10.1007/s00134-020-06050-1 [ Links ]

78. Abdul-Aziz MH, Sulaiman H, Mat-Nor MB, Rai V, Wong KK, Hasan MS, et al. Beta-Lactam Infusion in Severe Sepsis (BLISS): a prospective, two-centre, open-labelled randomised controlled trial of continuous versus intermittent beta-lactam infusion in critically ill patients with severe sepsis. Intensive Care Med. 2016;42:1535-45. DOI: 10.1007/s00134-015-4188-0 [ Links ]

79. Roberts JA, Roberts MS, Robertson TA, Dalley AJ, Lipman J. Piperacillin penetration into tissue of critically ill patients with sepsis—bolus versus continuous administration? Crit Care Med. 2009;37:926-33. DOI: 10.1097/CCM.0b013e3181968e44 [ Links ]

80. Eagle H, Fleischman R, Musselman AD. Effect of Schedule of Administration on the Therapeutic Efficacy of Penicillin. Importance of the Aggregate Time Penicillin remains at Effectively Bactericidal Levels. Am J Med. 1950;9:280-99. DOI: 10.1016/0002-9343(50)90425-6 [ Links ]

81. Roberts JA, Abdul-Aziz MH, Davis JS, Dulhunty JM, O Cotta M, Myburgh J, et al. Continuous versus intermittent β-lactam infusion in severe sepsis. A meta-analysis of individual patient data from randomized trials. Am J Respir Crit Care Med. 2016;194(6):681-91. DOI: 10.1164/rccm.201601-0024OC [ Links ]

82. Roberts JA, Webb S, Paterson D, Ho KM, Lipman J. A systematic review on clinical benefits of continuous administration of β-lactam antibiotics. Crit Care Med. 2009;37:2071-8. DOI: 10.1097/CCM.0b013e3181a0054d [ Links ]

83. Kasiakou SK, Sermaides GJ, Michalopoulos A, Soteriades ES, Falagas ME. Continuous versus intermittent intravenous administration of antibiotics: a meta-analysis of randomised controlled trials. Lancet Infect Dis. 2005;5:581-9. DOI: 10.1016/S1473-3099(05)70218-8 [ Links ]

84. Falagas ME, Tansarli GS, Ikawa K, Vardakas KZ. Clinical outcomes with extended or continuous versus short-term intravenous infusion of carbapenems and piperacillin/tazobactam: a systematic review and meta-analysis. Clin Infect Dis. 2013;56:272-82. DOI: 10.1093/cid/cis857 [ Links ]

85. Teo J, Liew Y, Lee W, Kwa ALH. Prolonged infusion versus intermittent boluses of β-lactam antibiotics for treatment of acute infections: a meta-analysis. Int J Antimicrob Agents. 2014;43:403-11. DOI: 10.1016/j.ijantimicag.2014.01.027 [ Links ]

86. Yusuf E, Spapen H, Piérard D. Prolonged vs intermittent infusion of piperacillin/ tazobactam in critically ill patients: a narrative and systematic review. J Crit Care. 2014;29:1089-95. DOI: 10.1016/j.jcrc.2014.07.033 [ Links ]

87. Shiu JR, Wang E, Tejani AM, Wasdell M. Continuous versus intermittent infusions of antibiotics for the treatment of severe acute infections. Cochrane Database Syst Rev. 2013(3):CD008481. DOI: 10.1002/14651858.CD008481.pub2 [ Links ]

88. Rhodes NJ, MacVane SH, Kuti JL, Scheetz MH. Impact of loading doses on the time to adequate predicted beta-lactam concentrations in prolonged and continuous infusion dosing schemes. Clin Infect Dis. 2014;59:905-7. DOI: 10.1093/cid/ciu402 [ Links ]

89. Roberts JA, Paul SK, Akova M, Bassetti M, De Waele JJ, Dimopoulos G, et al. Reply to Rhodes et al. Clin Infect Dis. 2014,59:907-8. DOI: 10.1093/cid/ciu403 [ Links ]

90. Paul M, Theuretzbacher U. β-lactam prolonged infusion: it's time to implement! Lancet Infect Dis. 2017;18:13-4. DOI: 10.1016/S1473-3099(17)30614-X [ Links ]

91. Bigley FP, Forsyth RJ, Henley MW. Compatibility of imipenem-cilastatin sodium with commonly used intravenous solutions. Am J Hosp Pharm. 1986;43:2803-9. [ Links ]

92. Patel PR, Cook SE. Stability of meropenem in intravenous solutions. Am J Health Syst Pharm. 1997;54:412-21. DOI: 10.1093/ajhp/54.4.412 [ Links ]

93. Cook B, Hill S, Lynn B. The stability of amoxycillin sodium in intravenous infusion luids. J Clin Pharm Ther. 1982;7:245-50. DOI: 10.1111/j.1365-2710.1982. tb01029.x [ Links ]

94. Stiles ML, Allen LV. Stability of nafcillin sodium, oxacillin sodium, penicillin G potassium, penicillin G sodium, and tobramycin sulfate in polyvinyl chloride drug reservoirs. Am J Health Syst Pharm. 1997;54:1068-70. DOI: 10.1093/ajhp/54.9.1068 [ Links ]

95. Stewart JT, Warren FW, Johnson SM, Fox JL, Mullaney J. Stability of ceftazidime in plastic syringes and glass vials under various storage conditions. Am J Hosp Pharm. 1992;49:2765-8. [ Links ]

96. Bolla B, Buxani Y, Wong R, Jones L, Dube M. Understanding IV antimicrobial drug losses: the importance of flushing infusion administration sets. JAC-Antimicrobial Resistance. 2020,2:dlaa061. DOI: 10.1093/jacamr/dlaa061 [ Links ]

97. Peyko V. An Unrecognized Problem in Optimizing Antimicrobial Therapy: Significant Residual Volume Remaining in Intravenous Tubing With Extended-Infusion Piperacillin–Tazobactam. J Pharm Pract. 2021:08971900211033462. DOI: 10.1177/08971900211033462 [ Links ]

98. Lam WJ, Bhowmick T, Gross A, Vanschooneveld TC, Weinstein MP. Using higher doses to compensate for tubing residuals in extended-infusion piperacillin-tazobactam. Ann Pharmacother. 2013;47:886-91. DOI: 10.1345/aph.1R721 [ Links ]

99. Hermsen ED, Fehrenbacher L. Antibiotic Stewardship and Applications of Pharmacodynamics. Antibiotic Pharmacodynamics. 2016;633-47. [ Links ]

100. Udy AA, Roberts JA, Lipman J, Blot S. The effects of major burn related pathophysiological changes on the pharmacokinetics and pharmacodynamics of drug use: An appraisal utilizing antibiotics. Adv Drug Deliv Rev. 2018;123:65-74. DOI: 10.1016/j.addr.2017.09.019 [ Links ]

101. Alobaid AS, Wallis SC, Jarrett P, Starr T, Stuart J, Lassig-Smith M, et al. Population pharmacokinetics of piperacillin in nonobese, obese, and morbidly obese critically ill patients. Antimicrob Agents Chemother. 2017;61:e01276-01216. DOI: 10.1128/AAC.01276-16 [ Links ]

102. Roberts JA, Udy AA, Jarrett P, Wallis SC, Hope WW, Sharma R, et al. Plasma and target-site subcutaneous tissue population pharmacokinetics and dosing simulations of cefazolin in post-trauma critically ill patients. J Antimicrob Chemother. 2015;70:1495-502. DOI: 10.1093/jac/dku564 [ Links ]

103. Weber N, Jackson K, McWhinney B, Ungerer J, Kennedy G, Lipman J, et al. Evaluation of pharmacokinetic/pharmacodynamic and clinical outcomes with 6-hourly empiric piperacillin-tazobactam dosing in hematological malignancy patients with febrile neutropenia. J Infect Chemother. 2019;25:503-8. DOI: 10.1016/j.jiac.2019.02.014 [ Links ]

104. Sime FB, Roberts MS, Tiong IS, Gardner JH, Lehman S, Peake SL, et al. Can therapeutic drug monitoring optimize exposure to piperacillin in febrile neutropenic patients with haematological malignancies? A randomized controlled trial. J Antimicrob Chemother. 2015;70:2369-75. DOI: 10.1093/jac/dkv123 [ Links ]

105. De Waele JJ, Carrette S, Carlier M, Stove V, Boelens J, Claeys G, et al. Therapeutic drug monitoring-based dose optimisation of piperacillin and meropenem: a randomised controlled trial. Intensive Care Med. 2014:40:380-7. DOI: 10.1007/s00134-013-3187-2 [ Links ]

106. Hagel S, Fiedler S, Hohn A, Brinkmann A, Frey OR, Hoyer H, et al. Therapeutic drug monitoring-based dose optimisation of piperacillin/tazobactam to improve outcome in patients with sepsis (TARGET): a prospective, multi-centre, randomised controlled trial. Trials. 2019;20:330. DOI: 10.1186/s13063-019-3437-x [ Links ]

107. Abdulla A, Ewoldt TM, Hunfeld NGM, Muller AE, Rietdijk WJR, Polinder S, et al. The effect of therapeutic drug monitoring of beta-lactam and fluoroquinolones on clinical outcome in critically ill patients: the DOLPHIN trial protocol of a multicentre randomised controlled trial. BMC Infect Dis. 2020;20:57. DOI: 10.1186/s12879-020-4781-x [ Links ]

108. Wong G, Briscoe S, McWhinney B, Ally M, Ungerer J, Lipman J, et al. Therapeutic drug monitoring of β-lactam antibiotics in the critically ill: direct measurement of unbound drug concentrations to achieve appropriate drug exposures. J Antimicrob Chemother. 2018;73:3087-94. DOI: 10.1093/jac/dky314 [ Links ]

109. Economou CJP, Wong G, McWhinney B, Ungerer JPJ, Lipman J, Roberts JA, et al. Impact of β-lactam antibiotic therapeutic drug monitoring on dose adjustments in critically ill patients undergoing continuous renal replacement therapy. Int J Antimicrob Agents. 2017;49:589-94. DOI: 10.1016/j.ijantimicag.2017.01.009 [ Links ]

110. Fournier A, Eggimann P, Pagani JL, Revelly JP, Decosterd LA, Marchetti O, et al. Impact of the introduction of real-time therapeutic drug monitoring on empirical doses of carbapenems in critically ill burn patients. Burns. 2015;41:956-68. DOI: 10.1016/j.burns.2015.01.001 [ Links ]

111. Patel BM, Paratz J, See NC, Muller MJ, Rudd M, Paterson D, et al. Therapeutic drug monitoring of beta-lactam antibiotics in burns patients—a oneyear prospective study. Ther Drug Monit. 2012;34:160-4. DOI: 10.1097/FTD.0b013e31824981a6 [ Links ]

112. Roberts JA, Ulldemolins M, Roberts M, Roberts MS, McWhinney B, Ungerer J, et al. Therapeutic drug monitoring of β-lactams in critically ill patients: proof of concept. Int J Antimicrob Agents. 2010;36:332-9. DOI: 10.1016/j.ijantimicag.2010.06.008 [ Links ]

How to cite this paperSulaiman H, Roberts JA, Abdul-Aziz MH. Pharmacokinetics and pharmacodynamics of beta-lactam antibiotics in critically ill patients. Farm Hosp. 2022;46(3):182-90.

FundingNo funding.

Received: December 04, 2021; Accepted: February 06, 2022; pub: March 25, 2022

Conflict of interest

No conflict of interests.

Author of correspondence: Jason A. Roberts. University of Queensland Centre for Clinical Research (UQCCR). Faculty of Medicine. The University of Queensland, Brisbane 4029 QLD. Australia. Email: j.roberts2@uq.edu.au

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License