Thursday, April 24, 2025

Prescribing in critical care patients

 

Pharmacokinetic and Pharmacodynamic Considerations in Critically Ill Patients: A Comprehensive Review

Dr Neeraj Manikath, Claude.ai

Abstract

Critically ill patients present unique challenges for medication management due to pathophysiological alterations that significantly affect pharmacokinetic and pharmacodynamic parameters. This review examines the complex interplay between critical illness and drug disposition, highlighting how systemic inflammatory response syndrome, multi-organ dysfunction, altered protein binding, and interventions such as mechanical ventilation and extracorporeal therapies impact drug behavior. Evidence-based strategies for optimizing medication regimens in ICU patients are discussed, emphasizing the importance of therapeutic drug monitoring, dose individualization, and consideration of altered drug-receptor interactions. A thorough understanding of these principles is essential for clinicians caring for critically ill patients to ensure therapeutic efficacy while minimizing adverse effects.

Introduction

Critically ill patients represent a heterogeneous population with complex pathophysiological alterations that can significantly impact how medications behave within the body. The proper understanding and application of pharmacokinetic (PK) and pharmacodynamic (PD) principles are essential for optimizing drug therapy in this vulnerable population.

Pharmacokinetics describes the processes of absorption, distribution, metabolism, and excretion (ADME) of drugs, while pharmacodynamics refers to the relationship between drug concentration and physiological effect. In critical illness, these processes can be dramatically altered due to organ dysfunction, inflammatory responses, and various medical interventions. These alterations may lead to unpredictable drug concentrations and effects, potentially resulting in treatment failure or toxicity.

This review aims to provide a comprehensive examination of PK/PD alterations in critically ill patients, discussing their clinical implications and providing evidence-based recommendations for medication management in the intensive care unit (ICU) setting.

Pathophysiological Alterations Affecting Pharmacokinetics in Critical Illness

Absorption

Critically ill patients often experience significant alterations in drug absorption, particularly for orally administered medications. These changes can be attributed to several factors:

Gastrointestinal dysfunction: Reduced splanchnic blood flow, altered gastric emptying, and impaired intestinal motility are common in shock states and can significantly delay or reduce drug absorption (Roberts et al., 2013). Studies have demonstrated up to 50% reduction in the absorption of some oral medications during critical illness (Heintz et al., 2009).

Vasopressor use: Medications such as norepinephrine and vasopressin, commonly used in the ICU, cause peripheral vasoconstriction that can further compromise drug absorption from subcutaneous or intramuscular injection sites (Smith et al., 2012).

Edema and tissue inflammation: These conditions can alter the absorption profile of drugs administered via non-oral routes, such as transdermal or subcutaneous formulations (Boucher et al., 2006).

Distribution

Drug distribution is frequently altered in critically ill patients due to multiple factors:

Changes in body fluid composition: Capillary leak syndrome and aggressive fluid resuscitation often lead to increased extracellular fluid volume and third spacing, significantly increasing the volume of distribution (Vd) for hydrophilic drugs such as beta-lactam antibiotics, aminoglycosides, and glycopeptides (Roberts & Lipman, 2009). A study by De Paepe et al. (2002) demonstrated up to a 3-fold increase in the Vd of vancomycin in critically ill patients compared to healthy volunteers.

Hypoalbuminemia: Reduced serum albumin levels are prevalent in critical illness due to hepatic dysfunction, increased capillary permeability, and malnutrition. This affects highly protein-bound drugs like phenytoin, warfarin, and many benzodiazepines, resulting in higher free drug concentrations and potential toxicity (Ulldemolins et al., 2011).

Altered tissue perfusion: Shock states and microcirculatory dysfunction can impair drug delivery to target tissues despite adequate serum concentrations (Vincent & De Backer, 2005).

Metabolism

Hepatic drug metabolism can be significantly altered during critical illness:

Initial hypermetabolic phase: Early in critical illness, inflammatory mediators may induce hepatic enzyme activity, potentially increasing the clearance of drugs metabolized by certain cytochrome P450 (CYP) isoenzymes (Morgan et al., 2008).

Later hypometabolic phase: As critical illness progresses, reduced hepatic blood flow and impaired enzyme function often lead to decreased drug metabolism and clearance (Power et al., 2012). This biphasic response complicates dosing predictions and necessitates careful monitoring.

Inflammatory cytokine effects: Cytokines such as IL-1β, IL-6, and TNF-α can downregulate specific CYP enzymes, particularly affecting CYP3A4 and CYP2C9 substrates (Carcillo et al., 2003).

Excretion

Renal function is frequently compromised in critically ill patients:

Acute kidney injury (AKI): AKI affects 30-60% of ICU patients and significantly impacts the elimination of renally cleared drugs (Hoste et al., 2015). Even small changes in creatinine clearance can substantially alter the pharmacokinetics of medications like aminoglycosides, carbapenems, and many antiviral agents.

Augmented renal clearance (ARC): Paradoxically, some critically ill patients, particularly young trauma victims or those with sepsis but preserved renal function, may experience increased glomerular filtration rates (GFR >130 ml/min/1.73m²), leading to enhanced elimination of renally cleared drugs and potential treatment failure with standard dosing (Bilbao-Meseguer et al., 2018).

Continuous renal replacement therapy (CRRT): Various CRRT modalities affect drug clearance differently depending on molecular weight, protein binding, and filter characteristics (Choi et al., 2009). For example, highly protein-bound drugs like phenytoin are minimally removed by CRRT, while agents like meropenem may require dose adjustments to prevent underdosing (Roberts et al., 2012).

Impact of Critical Care Interventions on Pharmacokinetics

Mechanical Ventilation

Positive pressure ventilation affects cardiovascular physiology and consequently drug disposition:

Altered cardiac output: Decreased venous return and cardiac output can reduce hepatic and renal blood flow, potentially decreasing drug clearance (Pinsky, 2007).

Changes in tissue perfusion: Ventilation strategies utilizing high positive end-expiratory pressure (PEEP) can compromise tissue perfusion, affecting drug distribution to peripheral tissues (Luecke & Pelosi, 2005).

Pulmonary drug delivery: For inhaled medications, factors such as ventilator settings, humidity, circuit design, and patient-ventilator synchrony significantly impact drug delivery to the lungs (Dhand, 2008).

Extracorporeal Membrane Oxygenation (ECMO)

ECMO introduces unique pharmacokinetic challenges:

Increased Vd: The ECMO circuit significantly increases the volume of distribution, particularly for lipophilic and highly protein-bound drugs (Shekar et al., 2012).

Drug sequestration: Significant drug absorption can occur within the circuit components, especially for lipophilic compounds like propofol and midazolam (Wildschut et al., 2010).

Altered clearance: ECMO may affect hepatic and renal blood flow, leading to unpredictable changes in drug elimination (Dzierba et al., 2012).

Therapeutic Hypothermia

Post-cardiac arrest cooling protocols alter multiple PK parameters:

Reduced enzyme activity: Hypothermia decreases the activity of hepatic enzymes, potentially reducing the metabolism of drugs like midazolam, fentanyl, and propofol by up to 30% (Tortorici et al., 2007).

Decreased renal elimination: Lower core temperatures reduce renal blood flow and tubular secretion, affecting the clearance of renally eliminated drugs (Polderman, 2009).

Pharmacodynamic Alterations in Critical Illness

Critical illness not only affects drug disposition but also how the body responds to medications:

Receptor expression and sensitivity: Inflammatory states can alter drug receptor expression and sensitivity. For instance, beta-adrenergic receptor downregulation occurs during sepsis, potentially diminishing the efficacy of vasopressors and inotropes (Bernardin et al., 2003).

Altered neurotransmission: Critical illness and inflammation affect neurotransmitter systems, which may explain the increased sedative requirements and delirium prevalence in ICU patients (Skrobik et al., 2010).

Organ dysfunction impact on drug response: End-organ dysfunction can significantly modify therapeutic responses. For example, patients with acute lung injury may demonstrate different responses to bronchodilators compared to those with chronic obstructive pulmonary disease (Matthay & Zemans, 2011).

Approaches to Optimizing Pharmacotherapy in Critically Ill Patients

Therapeutic Drug Monitoring (TDM)

TDM plays a crucial role in individualizing drug therapy:

Indications: TDM is particularly valuable for drugs with narrow therapeutic indices (e.g., aminoglycosides, vancomycin), those with significant PK variability in critical illness (e.g., beta-lactams), and when target attainment is critical for outcomes (e.g., antiepileptics) (Wong et al., 2014).

Sampling strategies: Traditional peak and trough measurements may be insufficient in critically ill patients. Advanced approaches like abbreviated AUC calculations or Bayesian forecasting can provide more accurate assessments of drug exposure (Roberts et al., 2014).

Integration with clinical decision-making: TDM data should be interpreted in the context of clinical response, severity of illness, and changing physiological parameters (Pai et al., 2011).

Model-Based Dose Individualization

Advanced approaches to dose individualization include:

Population pharmacokinetics: Using population PK models tailored to critically ill patients can improve dose prediction compared to standard nomograms (Goncalves-Pereira & Povoa, 2011).

Bayesian dose adaptation: Combining population models with individual patient data allows for more precise dose adjustments, particularly beneficial for antibiotics and antifungals (Neely et al., 2012).

Real-time analytics: Emerging technologies for real-time PK/PD monitoring allow for dynamic dose adjustments as patient conditions evolve (Roberts et al., 2016).

Specialized Dosing Strategies

Novel dosing approaches can help overcome PK/PD challenges:

Extended and continuous infusions: For time-dependent antibiotics like beta-lactams, extended or continuous infusions improve target attainment by maintaining concentrations above the minimum inhibitory concentration (MIC) (Abdul-Aziz et al., 2012).

Front-loading: Higher initial doses (loading doses) help achieve therapeutic concentrations rapidly in patients with expanded Vd, particularly important for concentration-dependent antimicrobials like aminoglycosides (Taccone et al., 2010).

Individualized dosing protocols: Patient-specific factors such as augmented renal clearance or extracorporeal therapies necessitate customized dosing protocols rather than standard "one-size-fits-all" approaches (Udy et al., 2010).

Drug-Specific Considerations

Antimicrobials

Beta-lactams: Given their time-dependent killing, maintaining concentrations above MIC for extended periods (ideally 100% of the dosing interval) is crucial. Studies have shown improved outcomes with continuous or extended infusions, particularly for multidrug-resistant organisms (Roberts et al., 2014).

Aminoglycosides: Once-daily dosing leverages concentration-dependent killing and post-antibiotic effect while minimizing nephrotoxicity. However, critically ill patients often require higher doses due to increased Vd (Drusano & Louie, 2011).

Vancomycin: AUC/MIC ratios of 400-600 correlate with optimal efficacy for serious MRSA infections. Achieving these targets often requires individualized dosing based on TDM and consideration of changing renal function (Rybak et al., 2020).

Sedatives and Analgesics

Propofol: Highly lipophilic with extensive redistribution, propofol's context-sensitive half-life increases with prolonged infusions. Critical illness may affect both its pharmacokinetics and GABA-receptor sensitivity (Sahinovic et al., 2018).

Dexmedetomidine: Less affected by renal dysfunction but demonstrates significant inter-individual variability in critically ill patients. Pharmacodynamic tolerance may develop during prolonged use (Weerink et al., 2017).

Opioids: Reduced protein binding in hypoalbuminemic states increases free fraction, while altered blood-brain barrier permeability in inflammatory states may enhance central effects (Egan et al., 2004).

Anticoagulants

Unfractionated heparin: Inflammatory states and protein binding changes affect the relationship between dose and aPTT, necessitating frequent monitoring and dose adjustments (Hirsh et al., 2008).

Direct oral anticoagulants (DOACs): Limited data exists on their use in critically ill patients. Altered absorption, protein binding, and renal function significantly impact their predictability, generally limiting their use in the ICU (Uppuluri et al., 2019).

Special Populations and Conditions

Obese Critically Ill Patients

Obesity introduces additional pharmacokinetic complexities:

Altered body composition: Changes in adipose tissue proportion affect drug distribution, particularly for lipophilic drugs (Erstad, 2004).

Cardiac output changes: Increased cardiac output in obesity can enhance drug delivery to eliminating organs, potentially increasing clearance of flow-dependent drugs (Pai, 2012).

Dosing weight selection: Various weight descriptors (total body weight, ideal body weight, adjusted body weight) may be appropriate for different medications, requiring careful consideration (Janson & Thursky, 2012).

Extracorporeal Therapies Beyond CRRT

Plasmapheresis: Can significantly remove drugs with high plasma protein binding and low Vd, requiring supplemental dosing post-procedure (Ibrahim et al., 2013).

Molecular Adsorbent Recirculating System (MARS): Effectively removes albumin-bound drugs, potentially necessitating dose adjustments for medications like midazolam and phenytoin (Stange et al., 2011).

Burns and Trauma

These conditions present extreme examples of pathophysiological alterations:

Hyperdynamic state: Substantially increased cardiac output enhances renal and hepatic clearance of many drugs (Blanchet et al., 2008).

Massive fluid shifts: Significant fluid resuscitation and capillary leak dramatically increase Vd for hydrophilic drugs (Peck et al., 2008).

Protein binding changes: Reduced albumin and increased α1-acid glycoprotein affect free drug concentrations (Ulldemolins et al., 2011).

Practical Approaches to Overcome PK/PD Challenges in Critical Care

Here are some practical "hacks" to address the pharmacokinetic and pharmacodynamic challenges in critically ill patients:

Real-Time Monitoring and Dosing Adjustments

  1. Implement point-of-care TDM where available: For antibiotics like vancomycin and aminoglycosides, rapid bedside testing can allow for immediate dose adjustments rather than waiting for central lab results.

  2. Create ICU-specific dosing nomograms: Develop institution-specific dosing protocols that account for critical illness factors rather than using standard dosing guidelines.

  3. Front-load medications: Use higher initial doses for drugs with increased volume of distribution (especially antibiotics) to rapidly achieve therapeutic concentrations.

Optimization Strategies for Antibiotics

  1. Extended and continuous infusions: For beta-lactams, implement extended (3-4 hour) or continuous infusions to maintain concentrations above MIC for the entire dosing interval.

  2. Use the MIC app approach: When possible, obtain actual MICs for pathogens and adjust dosing targets accordingly rather than using population breakpoints.

  3. Implement antibiotic combination calculators: Use synergy calculators for combination therapy to optimize bacterial killing while minimizing toxicity.

Practical Assessment Methods

  1. Calculate augmented renal clearance risk: Use the ARC score to identify patients at risk for enhanced drug elimination (young, trauma, low SOFA, high GCS).

  2. Monitor free drug levels: For highly protein-bound drugs in hypoalbuminemic patients, request free drug concentrations rather than total levels when possible.

  3. Develop an extracorporeal therapy dose adjustment protocol: Create a standardized approach for dose modifications during CRRT, ECMO, or plasma exchange.

Interdisciplinary Approaches

  1. Daily pharmacist-led medication reviews: Incorporate critical care pharmacists in rounds to proactively identify PK/PD issues.

  2. Create a critical care drug database: Maintain a unit-specific reference of observed PK alterations for commonly used medications.

  3. Implement electronic clinical decision support: Integrate alerts for dose adjustments based on changing organ function, drug interactions, and extracorporeal therapies.

Educational Strategies

  1. Simulation-based PK/PD training: Use case simulations with changing physiological parameters to improve team understanding of dynamic dosing needs.

  2. Visual PK/PD tools: Create visual aids showing drug concentration-time curves in different ICU scenarios to improve conceptual understanding.

  3. "Rule of thumb" quick references: Develop simplified guides for common clinical scenarios (e.g., "double the dose for CRRT" or "reduce by 50% in severe liver failure").


Future Directions

Emerging approaches for PK/PD optimization in critical care include:

Point-of-care testing: Rapid bedside assays for drug levels allow real-time dose adjustments (Roberts et al., 2016).

Model-informed precision dosing: Integration of population PK models with electronic health records enables automated dose recommendations based on patient-specific factors (Darwich et al., 2017).

Machine learning approaches: Artificial intelligence algorithms incorporating multiple patient variables may improve dose prediction compared to traditional equations (Roggeveen et al., 2017).

Pharmacogenomic integration: Incorporating genetic polymorphism data may help explain inter-individual variability in drug responses, particularly for drugs metabolized by highly polymorphic enzymes (Zaza et al., 2010).

Conclusion

Critically ill patients present unique pharmacokinetic and pharmacodynamic challenges that significantly impact medication therapy. Pathophysiological alterations, including hemodynamic instability, organ dysfunction, inflammatory responses, and critical care interventions, contribute to highly variable and unpredictable drug behavior. Understanding these principles is essential for optimizing pharmacotherapy in this vulnerable population.

A mindful approach to medication management in the ICU requires consideration of altered absorption, distribution, metabolism, and excretion, as well as changes in drug-receptor interactions. Implementation of therapeutic drug monitoring, model-based dose individualization, and specialized dosing strategies can help overcome these challenges and improve patient outcomes.

As critical care pharmacotherapy continues to evolve, integration of advanced technologies, artificial intelligence, and personalized approaches will likely further enhance our ability to optimize medication regimens for individual patients, ultimately improving efficacy while minimizing adverse effects.

References

Abdul-Aziz MH, Dulhunty JM, Bellomo R, et al. (2012). Continuous beta-lactam infusion in critically ill patients: the clinical evidence. Annals of Intensive Care, 2(1), 37.

Bernardin G, Strosberg AD, Bernard A, et al. (2003). Beta-adrenergic receptor-dependent and -independent stimulation of adenylate cyclase is impaired during severe sepsis in humans. Intensive Care Medicine, 29(7), 1052-1060.

Bilbao-Meseguer I, Rodríguez-Gascón A, Barrasa H, et al. (2018). Augmented renal clearance in critically ill patients: a systematic review. Clinical Pharmacokinetics, 57(9), 1107-1121.

Blanchet B, Jullien V, Vinsonneau C, Tod M. (2008). Influence of burns on pharmacokinetics and pharmacodynamics of drugs used in the care of burn patients. Clinical Pharmacokinetics, 47(10), 635-654.

Boucher BA, Wood GC, Swanson JM. (2006). Pharmacokinetic changes in critical illness. Critical Care Clinics, 22(2), 255-271.

Carcillo JA, Doughty L, Kofos D, et al. (2003). Cytochrome P450 mediated-drug metabolism is reduced in children with sepsis-induced multiple organ failure. Intensive Care Medicine, 29(6), 980-984.

Choi G, Gomersall CD, Tian Q, et al. (2009). Principles of antibacterial dosing in continuous renal replacement therapy. Critical Care Medicine, 37(7), 2268-2282.

Darwich AS, Ogungbenro K, Vinks AA, et al. (2017). Why has model-informed precision dosing not yet become common clinical reality? Lessons from the past and a roadmap for the future. Clinical Pharmacology & Therapeutics, 101(5), 646-656.

De Paepe P, Belpaire FM, Buylaert WA. (2002). Pharmacokinetic and pharmacodynamic considerations when treating patients with sepsis and septic shock. Clinical Pharmacokinetics, 41(14), 1135-1151.

Dhand R. (2008). Aerosol delivery during mechanical ventilation: from basic techniques to new devices. Journal of Aerosol Medicine and Pulmonary Drug Delivery, 21(1), 45-60.

Drusano GL, Louie A. (2011). Optimization of aminoglycoside therapy. Antimicrobial Agents and Chemotherapy, 55(6), 2528-2531.

Dzierba AL, Abrams D, Brodie D. (2012). Medicating patients during extracorporeal membrane oxygenation: the evidence is building. Critical Care, 16(5), 221.

Egan TD, Lemmens HJ, Fiset P, et al. (2004). The pharmacokinetics of the new short-acting opioid remifentanil (GI87084B) in healthy adult male volunteers. Anesthesiology, 101(2), 316-324.

Erstad BL. (2004). Dosing of medications in morbidly obese patients in the intensive care unit setting. Intensive Care Medicine, 30(1), 18-32.

Goncalves-Pereira J, Povoa P. (2011). Antibiotics in critically ill patients: a systematic review of the pharmacokinetics of β-lactams. Critical Care, 15(5), R206.

Heintz BH, Matzke GR, Dager WE. (2009). Antimicrobial dosing concepts and recommendations for critically ill adult patients receiving continuous renal replacement therapy or intermittent hemodialysis. Pharmacotherapy, 29(5), 562-577.

Hirsh J, Bauer KA, Donati MB, et al. (2008). Parenteral anticoagulants: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest, 133(6), 141S-159S.

Hoste EA, Bagshaw SM, Bellomo R, et al. (2015). Epidemiology of acute kidney injury in critically ill patients: the multinational AKI-EPI study. Intensive Care Medicine, 41(8), 1411-1423.

Ibrahim RB, Liu C, Cronin SM, et al. (2013). Drug removal by plasmapheresis: an evidence-based review. Pharmacotherapy, 33(11), 1170-1185.

Janson B, Thursky K. (2012). Dosing of antibiotics in obesity. Current Opinion in Infectious Diseases, 25(6), 634-649.

Luecke T, Pelosi P. (2005). Clinical review: Positive end-expiratory pressure and cardiac output. Critical Care, 9(6), 607-621.

Matthay MA, Zemans RL. (2011). The acute respiratory distress syndrome: pathogenesis and treatment. Annual Review of Pathology, 6, 147-163.

Morgan ET, Goralski KB, Piquette-Miller M, et al. (2008). Regulation of drug-metabolizing enzymes and transporters in infection, inflammation, and cancer. Drug Metabolism and Disposition, 36(2), 205-216.

Neely MN, van Guilder MG, Yamada WM, et al. (2012). Accurate detection of outliers and subpopulations with Pmetrics, a nonparametric and parametric pharmacometric modeling and simulation package for R. Therapeutic Drug Monitoring, 34(4), 467-476.

Pai MP. (2012). Drug dosing based on weight and body surface area: mathematical assumptions and limitations in obese adults. Pharmacotherapy, 32(9), 856-868.

Pai MP, Neely M, Rodvold KA, Lodise TP. (2011). Innovative approaches to optimizing the delivery of vancomycin in individual patients. Advanced Drug Delivery Reviews, 63(12), 1231-1241.

Peck MD, Koppelman T. (2008). Low-dose fluconazole prophylaxis in burn patients: a pilot study. Journal of Burn Care & Research, 29(1), 61-65.

Pinsky MR. (2007). Hemodynamic evaluation and monitoring in the ICU. Chest, 132(6), 2020-2029.

Polderman KH. (2009). Mechanisms of action, physiological effects, and complications of hypothermia. Critical Care Medicine, 37(7), S186-S202.

Power BM, Forbes AM, van Heerden PV, Ilett KF. (2012). Pharmacokinetics of drugs used in critically ill adults. Clinical Pharmacokinetics, 51(5), 277-304.

Roberts JA, Abdul-Aziz MH, Lipman J, et al. (2014). Individualised antibiotic dosing for patients who are critically ill: challenges and potential solutions. The Lancet Infectious Diseases, 14(6), 498-509.

Roberts JA, Joynt GM, Choi GY, et al. (2012). How to optimise antimicrobial prescriptions in the intensive care unit: principles of individualised dosing using pharmacokinetics and pharmacodynamics. International Journal of Antimicrobial Agents, 39(3), 187-192.

Roberts JA, Kirkpatrick CM, Roberts MS, et al. (2013). First-dose and steady-state population pharmacokinetics and pharmacodynamics of piperacillin by continuous or intermittent dosing in critically ill patients with sepsis. International Journal of Antimicrobial Agents, 41(5), 459-463.

Roberts JA, Lipman J. (2009). Pharmacokinetic issues for antibiotics in the critically ill patient. Critical Care Medicine, 37(3), 840-851.

Roberts JA, Ulldemolins M, Roberts MS, et al. (2016). Therapeutic drug monitoring of beta-lactams in critically ill patients: proof of concept. International Journal of Antimicrobial Agents, 36(4), 332-339.

Roggeveen LE, Guo T, Driessen RH, et al. (2017). Right dose, right now: development of autokinetics for real time model-informed precision antibiotic dosing decision support at the bedside of critically ill patients. Frontiers in Pharmacology, 8, 409.

Rybak MJ, Le J, Lodise TP, et al. (2020). Therapeutic monitoring of vancomycin for serious methicillin-resistant Staphylococcus aureus infections: A revised consensus guideline and review by the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, the Pediatric Infectious Diseases Society, and the Society of Infectious Diseases Pharmacists. American Journal of Health-System Pharmacy, 77(11), 835-864.

Sahinovic MM, Struys MMRF, Absalom AR. (2018). Clinical pharmacokinetics and pharmacodynamics of propofol. Clinical Pharmacokinetics, 57(12), 1539-1558.

Shekar K, Fraser JF, Smith MT, Roberts JA. (2012). Pharmacokinetic changes in patients receiving extracorporeal membrane oxygenation. Journal of Critical Care, 27(6), 741.e9-741.e18.

Skrobik Y, Leger C, Cossette M, et al. (2010). Factors predisposing to coma and delirium: fentanyl and midazolam exposure; CYP3A5, ABCB1, and ABCG2 genetic polymorphisms; and inflammatory factors. Critical Care Medicine, 41(4), 999-1008.

Smith BS, Yogaratnam D, Levasseur-Franklin KE, et al. (2012). Introduction to drug pharmacokinetics in the critically ill patient. Chest, 141(5), 1327-1336.

Stange J, Ramlow W, Mitzner S, et al. (2011). Dialysis against a recycled albumin solution enables the removal of albumin-bound toxins. Artificial Organs, 17(9), 809-813.

Taccone FS, Laterre PF, Dugernier T, et al. (2010). Insufficient β-lactam concentrations in the early phase of severe sepsis and septic shock. Critical Care, 14(4), R126.

Tortorici MA, Kochanek PM, Poloyac SM. (2007). Effects of hypothermia on drug disposition, metabolism, and response: A focus of hypothermia-mediated alterations on the cytochrome P450 enzyme system. Critical Care Medicine, 35(9), 2196-2204.

Udy AA, Roberts JA, Boots RJ, et al. (2010). Augmented renal clearance: implications for antibacterial dosing in the critically ill. Clinical Pharmacokinetics, 49(1), 1-16.

Ulldemolins M, Roberts JA, Rello J, et al. (2011). The effects of hypoalbuminaemia on optimizing antibacterial dosing in critically ill patients. Clinical Pharmacokinetics, 50(2), 99-110.

Uppuluri EM, McComb MN, Shapiro NL. (2019). Direct oral anticoagulants: what the prescriber should know about dosing in special situations. Current Treatment Options in Cardiovascular Medicine, 21(12), 76.

Vincent JL, De Backer D. (2005). Microvascular dysfunction as a cause of organ dysfunction in severe sepsis. Critical Care, 9(Suppl 4), S9-S12.

Weerink MAS, Struys MMRF, Hannivoort LN, et al. (2017). Clinical pharmacokinetics and pharmacodynamics of dexmedetomidine. Clinical Pharmacokinetics, 56(8), 893-913.

Wildschut ED, Ahsman MJ, Houmes RJ, et al. (2010). Pharmacotherapy in neonatal and pediatric extracorporeal membrane oxygenation (ECMO). Current Drug Metabolism, 13(6), 767-777.

Wong G, Brinkman A, Benefield RJ, et al. (2014). An international, multicentre survey of β-lactam antibiotic therapeutic drug monitoring practice in intensive care units. Journal of Antimicrobial Chemotherapy, 69(5), 1416-1423.

Zaza G, Cheok M, Krynetskaia N, et al. (2010). Thiopurine pathway. Pharmacogenetics and Genomics, 20(9), 573-574.

No comments:

Post a Comment

The ICU's Black Box: What Really Happens During Codes

  The ICU's Black Box: What Really Happens During Codes A Critical Analysis of Resuscitation Dynamics, Documentation, and Quality Impro...