Practical Drug Dosing in Conservative Management of CKD Patients in ICU: A Comprehensive Review
Abstract
The management of critically ill patients with chronic kidney disease (CKD) presents unique challenges in drug dosing and pharmacokinetics. This comprehensive review addresses the complexities of drug dosing in CKD patients in intensive care settings who are not receiving renal replacement therapy (RRT) but are managed conservatively. We examine the physiological alterations in CKD that affect drug disposition, provide evidence-based dosing recommendations for commonly used medications in critical care, and propose practical frameworks for clinical decision-making. Special emphasis is placed on antimicrobials, analgesics, sedatives, cardiovascular agents, and anticoagulants, with consideration of altered pharmacokinetics, pharmacodynamics, and the dynamic nature of renal function in critical illness. This review aims to optimize therapeutic outcomes while minimizing adverse drug events in this vulnerable patient population.
Keywords: chronic kidney disease, critical care, drug dosing, pharmacokinetics, conservative management, intensive care unit
Introduction
Chronic kidney disease (CKD) affects approximately 10-15% of the adult population worldwide and is associated with significant morbidity and mortality in critical care settings.^1,2^ The prevalence of CKD among intensive care unit (ICU) patients ranges from 20-30%, with up to 60% experiencing acute-on-chronic kidney injury during their ICU stay.^3^
Drug dosing in CKD patients who are critically ill presents a significant clinical challenge due to several factors:
- Altered pharmacokinetics (PK) and pharmacodynamics (PD) associated with impaired renal function
- Dynamic changes in renal function during critical illness
- The effects of critical illness itself on drug disposition
- Complex polypharmacy and drug-drug interactions
- Paucity of clinical trials specifically addressing drug dosing in this population^4,5^
While renal replacement therapy (RRT) offers one approach to managing these patients, many CKD patients in the ICU are managed conservatively without RRT due to clinical considerations, resource constraints, or in adherence to advanced directives. This review focuses specifically on drug dosing strategies for this important subgroup.
The stakes for appropriate drug dosing are particularly high in the ICU setting. Underdosing may lead to treatment failure and increased mortality, particularly with antimicrobials, while overdosing may result in toxicity, prolonged ICU stays, and increased healthcare costs.^6^ The challenge lies in achieving therapeutic efficacy while minimizing adverse drug events in patients with already compromised renal function.
This review aims to provide evidence-based recommendations and practical frameworks for drug dosing in critically ill CKD patients under conservative management. We will discuss the physiological basis for altered drug handling in CKD, examine specific drug classes commonly used in critical care, and provide practical approaches to individualized dosing strategies.
Physiological Considerations in CKD Affecting Drug Disposition
Alterations in Pharmacokinetics
Absorption
While absorption is generally less affected by CKD than other pharmacokinetic parameters, several factors in critically ill CKD patients can alter drug absorption:
- Delayed gastric emptying and altered gastrointestinal pH, particularly common in uremia^7^
- Reduced splanchnic blood flow in critically ill patients^8^
- Edema of the intestinal wall affecting absorption of certain drugs
- Concomitant use of phosphate binders, antacids, and other medications that may chelate drugs and impair absorption^9^
These factors should be considered when administering oral medications, with potential consideration for parenteral routes when rapid and reliable drug delivery is essential.
Distribution
Drug distribution is significantly altered in CKD patients due to:
- Changes in body fluid composition, with increased total body water and extracellular fluid volume^10^
- Hypoalbuminemia, which affects the binding of highly protein-bound drugs^11^
- Altered tissue binding due to uremic toxins^12^
- Changes in acid-base balance affecting ionization of drugs^13^
These changes affect the volume of distribution (Vd) of many drugs, with water-soluble drugs typically showing decreased Vd and lipophilic drugs often showing minimal changes or increased Vd. This has important implications for loading doses, which are determined primarily by Vd rather than elimination.^14^
Metabolism
Hepatic drug metabolism may be altered in CKD through several mechanisms:
- Reduced activity of certain cytochrome P450 enzymes, particularly CYP3A4 and CYP2C19^15^
- Decreased phase II conjugation reactions, including glucuronidation^16^
- Reduced first-pass metabolism leading to increased bioavailability of certain drugs^17^
- Altered expression of drug transporters^18^
- Accumulation of uremic toxins affecting enzyme function^19^
These alterations can lead to unpredictable changes in drug metabolism, potentially affecting both parent drugs and active metabolites.
Elimination
Renal elimination is most significantly affected in CKD, with alterations including:
- Decreased glomerular filtration rate (GFR) affecting filtration of drugs and metabolites^20^
- Reduced tubular secretion affecting organic anions and cations^21^
- Diminished renal blood flow affecting drug delivery to elimination sites^22^
- Potential competition between drugs and uremic toxins for tubular secretion pathways^23^
For drugs with significant renal elimination, dosage adjustments based on estimated GFR (eGFR) are often necessary, though critical illness introduces additional complexity to this assessment.
Pharmacodynamic Changes
CKD not only affects pharmacokinetics but can also alter pharmacodynamic responses:
- Increased sensitivity to certain drugs, particularly those acting on the central nervous system^24^
- Altered receptor sensitivity due to uremic toxins^25^
- Electrolyte and acid-base disturbances affecting drug responses^26^
These alterations may necessitate dose adjustments beyond what would be predicted by pharmacokinetic changes alone.
Assessment of Renal Function in Critically Ill CKD Patients
Limitations of Conventional eGFR Equations
Accurate assessment of renal function is fundamental to appropriate drug dosing. However, conventional eGFR equations have significant limitations in critically ill patients:
- Cockroft-Gault, MDRD, and CKD-EPI equations were developed in stable, non-critically ill populations^27^
- These equations assume steady-state serum creatinine, which is rarely the case in dynamic ICU settings^28^
- Altered creatinine production due to critical illness, malnutrition, and reduced muscle mass leads to overestimation of GFR^29^
- Volume overload may dilute serum creatinine, further overestimating GFR^30^
Approaches to Estimating GFR in Critical Illness
Several approaches may improve GFR estimation in critically ill CKD patients:
-
Measured creatinine clearance: Collection of timed urine samples (e.g., 8-hour collections) may provide more accurate assessment than equations, though practical difficulties exist in ICU settings^31^
-
Kinetic eGFR: Incorporating the rate of change of serum creatinine into GFR estimation may better account for non-steady-state conditions^32^
-
Cystatin C-based equations: Less affected by muscle mass and nutritional status, though influenced by inflammation and corticosteroid use common in ICU settings^33^
-
Iohexol clearance: Gold standard for GFR measurement but rarely practical in routine ICU care^34^
For practical purposes, clinicians should recognize the limitations of standard eGFR equations and consider using the more conservative estimate when discrepancies exist, particularly for high-risk medications with narrow therapeutic windows.
Principles of Drug Dosing in CKD Patients in ICU
Assessment of Drug Properties
When dosing medications in CKD patients in the ICU, the following drug properties should be considered:
-
Fraction eliminated unchanged by the kidney (fe): Drugs with fe >0.3 (30%) generally require dosage adjustment in CKD^35^
-
Therapeutic index: Drugs with narrow therapeutic indices require more careful adjustment and potentially therapeutic drug monitoring (TDM)^36^
-
Protein binding: Hypoalbuminemia in critically ill CKD patients increases the free fraction of highly protein-bound drugs, potentially increasing toxicity^37^
-
Active metabolites: Some drugs have active metabolites that accumulate in CKD, contributing to efficacy or toxicity^38^
Dosing Adjustment Strategies
Three main approaches to dosage adjustment in CKD include:
-
Dose reduction: Maintaining the standard dosing interval but reducing the individual dose amount
-
Interval extension: Maintaining the standard dose amount but extending the time between doses
-
Combined approach: Reducing both the dose and extending the interval
The appropriate strategy depends on the drug's characteristics:
- Concentration-dependent agents (e.g., aminoglycosides) often benefit from maintaining higher peak concentrations with extended intervals^39^
- Time-dependent agents (e.g., beta-lactams) may benefit from dose reduction while maintaining standard intervals to ensure time above MIC^40^
Loading Doses
Loading doses are primarily determined by the volume of distribution (Vd) rather than elimination. For critically ill CKD patients:
- Full loading doses are generally recommended for most drugs, especially antimicrobials, to rapidly achieve therapeutic concentrations^41^
- Exceptions include drugs with very narrow therapeutic indices or those where even brief exposure to high concentrations may cause toxicity^42^
Augmented Renal Clearance (ARC)
A frequently overlooked phenomenon in critically ill patients is augmented renal clearance (ARC), characterized by increased glomerular filtration beyond normal physiological levels (typically defined as creatinine clearance >130 mL/min/1.73m²).^43^
While seemingly counterintuitive in CKD patients, those with mild-to-moderate CKD (CKD stages 2-3) may experience relative ARC during critical illness due to:
- Increased cardiac output and renal blood flow from systemic inflammatory response
- Aggressive fluid resuscitation
- Vasopressor use
- Improved cardiac function with treatment
This phenomenon may lead to unexpected subtherapeutic drug levels, particularly for antimicrobials with predominant renal elimination.^44,45^ Clinicians should consider this possibility in patients showing poor clinical response despite seemingly appropriate dosing, particularly in the early phases of critical illness.
Specific Drug Classes in Critical Care
Antimicrobials
Beta-lactam Antibiotics
Beta-lactams exhibit time-dependent killing, with efficacy determined by the time the free drug concentration remains above the minimum inhibitory concentration (MIC).^46^ In CKD patients:
- Penicillins require dose adjustment with moderate-severe CKD (eGFR <30 mL/min)
- Cephalosporins vary in the degree of renal elimination; third and fourth-generation cephalosporins typically require more significant adjustments
- Carbapenems require dose adjustment even with mild renal impairment
Practical Recommendations:
- Consider extended or continuous infusions to optimize pharmacodynamics, particularly for difficult infections^47^
- For severe infections, use standard loading doses followed by adjusted maintenance doses
- Cefepime requires careful monitoring in CKD due to risk of neurotoxicity with accumulation^48^
TABLE 1: Recommended Dosing of Common Beta-lactams in CKD
Antibiotic | Normal Dose | CKD Stage 3 (eGFR 30-59 mL/min) | CKD Stage 4-5 (eGFR <30 mL/min) |
---|---|---|---|
Piperacillin-tazobactam | 4.5g q6h | 4.5g q8h | 2.25g q8h |
Ceftriaxone | 2g q24h | 2g q24h | 2g q24h* |
Cefepime | 2g q8h | 2g q12h | 1g q24h |
Meropenem | 1g q8h | 1g q12h | 0.5g q24h |
*Ceftriaxone has significant hepatic elimination and requires minimal adjustment in CKD.
Aminoglycosides
Aminoglycosides exhibit concentration-dependent killing and post-antibiotic effect, but have significant nephrotoxicity potential.^49^ In CKD patients:
- Extended-interval dosing (once-daily) is preferred when feasible to minimize toxicity
- Therapeutic drug monitoring is essential
- Consider alternative agents when possible in advanced CKD
Practical Recommendations:
- Use ideal body weight for dosing calculations
- Monitor trough levels for traditional dosing and 12-18h post-dose levels for extended-interval dosing
- Consider loading dose of 5-7 mg/kg (gentamicin/tobramycin) regardless of renal function, followed by adjusted maintenance dosing^50^
Vancomycin
Vancomycin efficacy correlates with AUC/MIC ratio, with target AUC/MIC ≥400 for most serious infections.^51^ In CKD patients:
- Loading doses of 20-25 mg/kg actual body weight remain appropriate regardless of renal function
- Maintenance doses and intervals require significant adjustment based on eGFR
- AUC-guided dosing using Bayesian software is preferred when available^52^
Practical Recommendations:
- For empiric dosing without Bayesian software, target trough concentrations of 15-20 mg/L for serious infections
- Monitor for AKI, particularly with concomitant nephrotoxic agents
- Consider alternative agents (e.g., linezolid, daptomycin) in severe CKD where appropriate
Fluoroquinolones
Fluoroquinolones exhibit concentration-dependent killing with moderate post-antibiotic effect. In CKD patients:
- Ciprofloxacin requires significant dose adjustment, particularly for systemically administered doses
- Levofloxacin clearance is predominantly renal, requiring substantial adjustment in CKD
- Moxifloxacin has minimal renal clearance and generally does not require adjustment^53^
Practical Recommendations:
- For serious infections in CKD, consider alternative agents when appropriate
- Monitor for QT prolongation, particularly with other QT-prolonging drugs commonly used in ICU
- Consider targeting higher end of dosing range for ciprofloxacin due to reduced peak concentrations in critically ill patients^54^
Antifungals
Echinocandins (caspofungin, micafungin, anidulafungin):
- Undergo minimal renal elimination and generally do not require dose adjustment in CKD^55^
- Preferred antifungals for most ICU patients with CKD
Azoles:
- Fluconazole is predominantly renally eliminated and requires significant adjustment in CKD
- Voriconazole and posaconazole undergo minimal renal elimination but require careful TDM due to variable pharmacokinetics in critically ill patients^56^
Amphotericin B formulations:
- All formulations are nephrotoxic and require careful consideration in CKD patients
- Lipid formulations may have less nephrotoxicity but still pose significant risk in advanced CKD^57^
Antivirals
Acyclovir/Valacyclovir:
- Require significant dose reduction in CKD
- Risk of neurotoxicity with accumulation, particularly in elderly CKD patients^58^
Neuraminidase inhibitors:
- Oseltamivir requires dose reduction in CKD; preferred over other agents for influenza treatment in renal impairment^59^
HIV antivirals:
- Complex adjustments based on specific agent; consultation with clinical pharmacist recommended
- Tenofovir requires particular caution due to potential for additional renal injury^60^
Analgesics and Sedatives
Opioid Analgesics
Opioid pharmacokinetics are variably affected by CKD:
- Morphine produces active metabolites (M6G) that accumulate in CKD, potentially causing prolonged sedation and respiratory depression^61^
- Hydromorphone produces fewer active metabolites but still requires dose adjustment
- Fentanyl and sufentanil undergo primarily hepatic metabolism and are generally preferred in CKD patients^62^
Practical Recommendations:
- Avoid morphine when possible in moderate-severe CKD; if used, reduce dose by 50-75% and extend interval
- Reduce hydromorphone dose by 50% in severe CKD
- Monitor closely for oversedation and respiratory depression
- Consider shorter half-life agents (fentanyl) for ease of titration
Benzodiazepines
- Midazolam and its active metabolite (1-hydroxymidazolam) accumulate in CKD, potentially prolonging sedation^63^
- Lorazepam undergoes glucuronidation with inactive metabolites but still requires dose adjustment in CKD
- Diazepam has multiple active metabolites with prolonged half-lives in CKD^64^
Practical Recommendations:
- Reduce benzodiazepine doses by 25-50% in moderate CKD and 50-75% in severe CKD
- Prefer shorter-acting agents for procedures
- Consider non-benzodiazepine alternatives when appropriate
Non-benzodiazepine Sedatives
- Propofol undergoes primarily extrahepatic metabolism with minimal impact from CKD, making it preferable for short-term sedation^65^
- Dexmedetomidine undergoes almost complete hepatic metabolism and requires minimal adjustment in CKD^66^
Practical Recommendations:
- Propofol and dexmedetomidine are generally preferred sedatives for CKD patients in ICU
- Monitor propofol infusion syndrome risk factors in patients requiring high doses or prolonged therapy
- Be aware of potential for relative bradycardia with dexmedetomidine, particularly in patients susceptible to hemodynamic instability
Non-opioid Analgesics
- Acetaminophen undergoes predominantly hepatic metabolism with minimal renal adjustment needed; limit dose to 3g/day in advanced CKD^67^
- NSAIDs should generally be avoided due to risk of worsening renal function
- Gabapentin and pregabalin require substantial dose reduction in CKD due to renal elimination^68^
Practical Recommendations:
- Acetaminophen can be used as an opioid-sparing agent
- Schedule gabapentinoids once daily or every other day in severe CKD, with close monitoring for CNS side effects
- Consider topical agents where appropriate for localized pain
Cardiovascular Medications
Vasopressors and Inotropes
- Norepinephrine, epinephrine, and phenylephrine undergo complex metabolism with minimal impact from renal dysfunction^69^
- Vasopressin clearance is affected by renal function, but clinical significance is unclear in the dosing ranges used for shock^70^
- Dobutamine and milrinone clearances are affected by renal function, with milrinone requiring significant dose adjustment in CKD^71^
Practical Recommendations:
- Titrate vasopressors to target MAP based on clinical context rather than specific dose adjustments
- For milrinone, reduce maintenance infusion rate by 50-70% in severe CKD; loading doses are typically avoided in critical care settings
- Monitor for increased sensitivity to vasoactive medications due to altered receptor responses in uremia
Antiarrhythmics
- Amiodarone undergoes primarily hepatic metabolism and requires minimal adjustment in CKD^72^
- Digoxin requires significant dose reduction and careful monitoring in CKD
- Lidocaine clearance may be reduced and requires careful titration^73^
Practical Recommendations:
- Reduce digoxin maintenance dose by 25-75% based on degree of renal impairment
- Consider measuring digoxin levels 7-14 days after initiation or dose changes
- For lidocaine infusions, start at lower doses (1 mg/min) and titrate cautiously
Anticoagulants
- Unfractionated heparin (UFH) is minimally affected by renal function and preferred over LMWH for therapeutic anticoagulation in severe CKD^74^
- Low molecular weight heparins (LMWH) accumulate in CKD, requiring dose adjustment or anti-Xa monitoring
- Direct oral anticoagulants (DOACs) have variable renal clearance and specific restrictions in advanced CKD^75^
Practical Recommendations:
- For prophylactic LMWH in severe CKD, consider 30-50% dose reduction or use UFH
- For therapeutic anticoagulation, UFH with aPTT monitoring is preferred in severe CKD
- If LMWH is used therapeutically, consider anti-Xa monitoring
- Avoid DOACs in severe CKD (CrCl <15-30 mL/min depending on agent) except in specific circumstances with specialist consultation
Miscellaneous Critical Care Medications
Stress Ulcer Prophylaxis
- H2-receptor antagonists (famotidine, ranitidine) require significant dose reduction in CKD^76^
- Proton pump inhibitors (PPIs) generally require minimal adjustment, with omeprazole and pantoprazole preferred^77^
Practical Recommendations:
- Reduce famotidine to 20mg daily in severe CKD
- Standard PPI dosing is generally appropriate, though twice daily dosing may be unnecessary
Neuromuscular Blocking Agents
- Atracurium and cisatracurium undergo Hofmann elimination and ester hydrolysis, independent of renal function^78^
- Rocuronium and vecuronium may have prolonged effect in CKD due to accumulation^79^
Practical Recommendations:
- Prefer cisatracurium for prolonged neuromuscular blockade in CKD patients
- If rocuronium is used, monitor for prolonged blockade and consider dose reduction (20-30%) for maintenance doses
Electrolyte Replacements
- Phosphate: Risk of hyperphosphatemia in CKD; replacement should be cautious with frequent monitoring
- Potassium: Reduced renal excretion increases risk of hyperkalemia; modify replacement protocols
- Magnesium: Accumulates in CKD; reduce replacement doses by 50-75% in severe CKD^80^
Practical Recommendations:
- Develop ICU-specific electrolyte replacement protocols for CKD patients
- Consider lower thresholds for replacement and smaller incremental doses
- Monitor more frequently during replacement therapy
Special Considerations
Drug-Drug Interactions in CKD
Polypharmacy is common in critically ill CKD patients, increasing the risk for drug-drug interactions:
- Competitive inhibition of renal tubular secretion may increase drug levels (e.g., trimethoprim inhibiting creatinine secretion)^81^
- Altered protein binding may increase free fraction of highly protein-bound drugs
- Inhibition or induction of metabolic enzymes may have exaggerated effects in CKD^82^
A systematic approach to medication reconciliation and careful consideration of the potential for drug-drug interactions should be part of the daily ICU workflow for CKD patients.
Dosing in Obesity and CKD
Obesity introduces additional complexity to drug dosing in CKD patients:
- Creatinine-based eGFR equations may be less accurate in obesity
- Some drugs require adjustment based on actual body weight, others on ideal or adjusted body weight^83^
- Volume of distribution changes may be more pronounced, affecting loading doses
Practical Recommendations:
- Consider measured creatinine clearance when feasible
- For antimicrobials, use total body weight for loading doses of hydrophilic drugs
- Consider clinical pharmacist consultation for complex cases
Extracorporeal Membrane Oxygenation (ECMO) and CKD
The combination of ECMO and CKD introduces unique considerations:
- ECMO circuits may sequester drugs, particularly lipophilic and highly protein-bound medications^84^
- The impact of ECMO on drug clearance is often unpredictable and may compound alterations from CKD
- Limited clinical data exists for drug dosing in these patients^85^
Practical Recommendations:
- Consider increased doses for sedatives and analgesics that may be sequestered by the circuit
- TDM is essential when available
- Empiric dose increases of 30-50% may be needed for certain highly sequestered medications
Transition from Conservative Management to RRT
Patients may transition from conservative management to RRT during their ICU stay, requiring reassessment of drug regimens:
- The initiation of RRT significantly alters drug clearance, particularly for hydrophilic, low-protein-bound medications
- Different RRT modalities (CRRT, IHD, PIRRT) have varying impacts on drug clearance^86^
- Drug properties determining RRT clearance include molecular weight, protein binding, and volume of distribution^87^
Practical Recommendations:
- Reassess all medication dosing upon initiation of RRT
- Consider specific RRT modality, intensity, and duration when adjusting doses
- Consult clinical pharmacy resources specific to the RRT modality being used
Practical Approach to Drug Dosing in ICU Patients with CKD
Stepwise Approach to Individualized Dosing
We propose a practical, stepwise approach to drug dosing for ICU patients with CKD:
-
Assess renal function:
- Recognize limitations of eGFR equations in critical illness
- Consider measured creatinine clearance when feasible
- Account for dynamic changes in renal function
-
Evaluate drug characteristics:
- Determine extent of renal elimination (fe)
- Identify active metabolites eliminated renally
- Consider therapeutic index and consequences of under/overdosing
-
Determine appropriate dosing strategy:
- Decide between dose reduction, interval extension, or combined approach
- Consider loading doses for concentration-dependent agents
- Evaluate need for therapeutic drug monitoring
-
Monitor response and adjust:
- Clinical response (efficacy, toxicity)
- TDM where available and appropriate
- Reassess with changes in clinical status or renal function
Role of Clinical Pharmacists
Clinical pharmacists play a vital role in optimizing medication therapy for CKD patients in the ICU:
- Providing specialized knowledge of altered pharmacokinetics
- Assisting with complex dosing calculations
- Developing institutional protocols and guidelines
- Recommending appropriate TDM strategies
- Facilitating medication reconciliation and transitions of care^88^
A collaborative approach between intensivists, nephrologists, and clinical pharmacists is essential for optimal medication management in this complex patient population.
Emerging Technologies and Approaches
Several emerging approaches may improve drug dosing in the future:
-
Bayesian dose optimization software:
- Incorporates population PK models with individual patient factors
- Updates dosing recommendations based on measured drug levels
- Adaptable to changing physiological parameters^89^
-
Real-time GFR monitoring:
- Novel technologies to provide continuous assessment of renal function
- Potential for more responsive drug dosing in dynamic ICU settings^90^
-
Model-informed precision dosing (MIPD):
- Integration of physiologically-based PK modeling with electronic health records
- Patient-specific dosing recommendations based on comprehensive physiological and pharmacological models^91^
Conclusion
Drug dosing in critically ill CKD patients under conservative management requires a nuanced understanding of altered pharmacokinetics and pharmacodynamics. The dynamic nature of critical illness, combined with the baseline alterations of CKD, creates a complex environment where standard dosing approaches may lead to either therapeutic failure or toxicity.
A systematic approach considering drug characteristics, accurate assessment of renal function, appropriate dosing strategies, and vigilant monitoring provides the foundation for optimized pharmacotherapy. While general principles and recommendations can guide practice, individualization remains paramount.
Future research should focus on validating drug dosing strategies specifically in critically ill CKD populations, developing more accurate methods of assessing renal function in dynamic ICU settings, and implementing technological solutions to facilitate precision dosing in this vulnerable patient population.
References
-
Webster AC, Nagler EV, Morton RL, Masson P. Chronic kidney disease. Lancet. 2017;389(10075):1238-1252.
-
Hoste EA, Bagshaw SM, Bellomo R, et al. Epidemiology of acute kidney injury in critically ill patients: the multinational AKI-EPI study. Intensive Care Med. 2015;41(8):1411-1423.
-
Chawla LS, Eggers PW, Star RA, Kimmel PL. Acute kidney injury and chronic kidney disease as interconnected syndromes. N Engl J Med. 2014;371(1):58-66.
-
Matzke GR, Aronoff GR, Atkinson AJ Jr, et al. Drug dosing consideration in patients with acute and chronic kidney disease-a clinical update from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int. 2011;80(11):1122-1137.
-
Bouchard J, Macedo E, Mehta RL. Dosing of drugs in patients with renal failure in the intensive care unit. UpToDate. 2023.
-
Roberts JA, Lipman J. Pharmacokinetic issues for antibiotics in the critically ill patient. Crit Care Med. 2009;37(3):840-851.
-
Ashley C, Dunleavy A, eds. The Renal Drug Handbook. 5th ed. CRC Press; 2018.
-
Boucher BA, Wood GC, Swanson JM. Pharmacokinetic changes in critical illness. Crit Care Clin. 2006;22(2):255-271.
-
Sica DA. Drug absorption in the management of congestive heart failure: loop diuretics. Congest Heart Fail. 2003;9(5):287-292.
-
Vilay AM, Churchwell MD, Mueller BA. Drug metabolism and nonrenal clearance in acute kidney injury. Crit Care. 2008;12(6):235.
-
Verbeeck RK, Musuamba FT. Pharmacokinetics and dosage adjustment in patients with renal dysfunction. Eur J Clin Pharmacol. 2009;65(8):757-773.
-
Lam YW, Banerji S, Hatfield C, Talbert RL. Principles of drug administration in renal insufficiency. Clin Pharmacokinet. 1997;32(1):30-57.
-
Joy MS, Frye RF, Nolin TD, et al. In vivo alterations in drug metabolism and transport pathways in patients with chronic kidney diseases. Pharmacotherapy. 2014;34(2):114-122.
-
Roberts JA, Lipman J. Antibacterial dosing in intensive care: pharmacokinetics, degree of disease and pharmacodynamics of sepsis. Clin Pharmacokinet. 2006;45(8):755-773.
-
Nolin TD, Naud J, Leblond FA, Pichette V. Emerging evidence of the impact of kidney disease on drug metabolism and transport. Clin Pharmacol Ther. 2008;83(6):898-903.
-
Sun H, Frassetto L, Benet LZ. Effects of renal failure on drug transport and metabolism. Pharmacol Ther. 2006;109(1-2):1-11.
-
Yeung CK, Shen DD, Thummel KE, Himmelfarb J. Effects of chronic kidney disease and uremia on hepatic drug metabolism and transport. Kidney Int. 2014;85(3):522-528.
-
Naud J, Nolin TD, Leblond FA, Pichette V. Current understanding of drug disposition in kidney disease. J Clin Pharmacol. 2012;52(S1):10S-22S.
-
Dreisbach AW, Lertora JJ. The effect of chronic renal failure on drug metabolism and transport. Expert Opin Drug Metab Toxicol. 2008;4(8):1065-1074.
-
Matzke GR, Frye RF. Drug administration in patients with renal insufficiency. Minimising renal and extrarenal toxicity. Drug Saf. 1997;16(3):205-231.
-
Brater DC. Drug dosing in patients with impaired renal function. Clin Pharmacol Ther. 2009;86(5):483-489.
-
Nolin TD. Altered nonrenal drug clearance in ESRD. Curr Opin Nephrol Hypertens. 2008;17(6):555-559.
-
Verbeeck RK, Musuamba FT. Pharmacokinetics and dosage adjustment in patients with renal dysfunction. Eur J Clin Pharmacol. 2009;65(8):757-773.
-
Gabardi S, Abramson S. Drug dosing in chronic kidney disease. Med Clin North Am. 2005;89(3):649-687.
-
Churchwell MD, Mueller BA. Drug dosing during kidney disease. Adv Chronic Kidney Dis. 2010;17(5):e48-e59.
-
Matzke GR, Frye RF. Drug administration in patients with renal insufficiency. Minimising renal and extrarenal toxicity. Drug Saf. 1997;16(3):205-231.
-
Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604-612.
-
Udy AA, Roberts JA, Shorr AF, Boots RJ, Lipman J. Augmented renal clearance in septic and traumatized patients with normal plasma creatinine concentrations: identifying at-risk patients. Crit Care. 2013;17(1):R35.
-
Bragadottir G, Redfors B, Ricksten SE. Assessing glomerular filtration rate (GFR) in critically ill patients with acute kidney injury - true GFR versus urinary creatinine clearance and estimating equations. Crit Care. 2013;17(3):R108.
-
Macedo E, Bouchard J, Soroko SH, et al. Fluid accumulation, recognition and staging of acute kidney injury in critically-ill patients. Crit Care. 2010;14(3):R82.
-
Bouchard J, Macedo E, Soroko S, et al. Comparison of methods for estimating glomerular filtration rate in critically ill patients with acute kidney injury. Nephrol Dial Transplant. 2010;25(1):102-107.
-
Chen S. Retooling the creatinine clearance equation to estimate kinetic GFR when the plasma creatinine is changing acutely. J Am Soc Nephrol. 2013;24(6):877-888.
-
Herget-Rosenthal S, Bökenkamp A, Hofmann W. How to estimate GFR-serum creatinine, serum cystatin C or equations? Clin Biochem. 2007;40(3-4):153-161.
-
Hoste EAJ, Damen J, Vanholder RC, et al. Assessment of renal function in recently admitted critically ill patients with normal serum creatinine. Nephrol Dial Transplant. 2005;20(4):747-753.
-
Lea-Henry TN, Carland JE, Stocker SL, Sevastos J, Roberts DM. Clinical pharmacokinetics in kidney disease: fundamental principles. Clin J Am Soc Nephrol. 2018;13(7):1085-1095.
-
Olyaei AJ, Bennett WM. Drug dosing in the elderly patients with chronic kidney disease. Clin Geriatr Med. 2009;25(3):459-527.
-
Verbeeck RK. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 2008;64(12):1147-1161.
-
Churchwell MD, Mueller BA. Drug dosing during continuous renal replacement therapy. Semin Dial. 2009;22(2):185-188.
-
Drusano GL. Antimicrobial pharmacodynamics: critical interactions of 'bug and drug'. Nat Rev Microbiol. 2004;2(4):289-300.
-
Craig WA. Basic pharmacodynamics of antibacterials with clinical applications to the use of beta-lactams, glycopeptides, and linezolid. Infect Dis Clin North Am. 2003;17(3):479-501.
-
Taccone FS, Laterre PF, Dugernier T, et al. Insufficient β-lactam concentrations in the early phase of severe sepsis and septic shock. Crit Care. 2010;14(4):R126.
-
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(6):498-509.
-
Udy AA, Baptista JP, Lim NL, et al. Augmented renal clearance in the ICU: results of a multicenter observational study of renal function in critically ill patients with normal plasma creatinine concentrations. Crit Care Med. 2014;42(3):520-527.
-
Claus BO, Hoste EA, Colpaert K, Robays H, Decruyenaere J, De Waele JJ. Augmented renal clearance is a common finding with worse clinical outcome in critically ill patients receiving antimicrobial therapy. J Crit Care. 2013;28(5):695-700.
-
Udy AA, Roberts JA, Boots RJ, Paterson DL, Lipman J. Augmented renal clearance: implications for antibacterial dosing in the critically ill. Clin Pharmacokinet. 2010;49(1):1-16.
-
Roberts JA, Lipman J. Pharmacokinetic issues for antibiotics in the critically ill patient. Crit Care Med. 2009;37(3):840-851.
-
Abdul-Aziz MH, Lipman J, Mouton JW, Hope WW, Roberts JA. Applying pharmacokinetic/pharmacodynamic principles in critically ill patients: optimizing efficacy and reducing resistance development. Semin Respir Crit Care Med. 2015;36(1):136-153.
-
Fugate JE, Kalimullah EA, Hocker SE, Clark SL, Wijdicks EF, Rabinstein AA. Cefepime neurotoxicity in the intensive care unit: a cause of severe, underappreciated encephalopathy. Crit Care. 2013;17(6):R264.
-
Drusano GL, Ambrose PG, Bhavnani SM, Bertino JS, Nafziger AN, Louie A. Back to the future: using aminoglycosides again and how to dose them optimally. Clin Infect Dis. 2007;45(6):753-760.
-
Nicolau DP, Freeman CD, Belliveau PP, Nightingale CH, Ross JW, Quintiliani R. Experience with a once-daily aminoglycoside program administered to 2,184 adult patients. Antimicrob Agents Chemother. 1995;39(3):650-655.
-
Rybak M, Lomaestro B, Rotschafer JC, et al. Therapeutic monitoring of vancomycin in adult patients: a consensus review of the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, and the Society of Infectious Diseases Pharmacists. Am J Health Syst Pharm. 2009;66(1):82-98.
-
Neely MN, Youn G, Jones B, et al. Are vancomycin trough concentrations adequate for optimal dosing? Antimicrob Agents Chemother. 2014;58(1):309-316.
-
Pea F, Viale P, Pavan F, Furlanut M. Pharmacokinetic considerations for antimicrobial therapy in patients receiving renal replacement therapy. Clin Pharmacokinet. 2007;46(12):997-1038.
-
Zelenitsky SA, Ariano RE. Support for higher ciprofloxacin AUC 24/MIC targets in treating Enterobacteriaceae bloodstream infection. J Antimicrob Chemother. 2010;65(8):1725-1732.
-
Eschenauer GA, Carver PL, Lin SW, et al. Fluconazole versus an echinocandin for Candida glabrata fungaemia: a retrospective cohort study. J Antimicrob Chemother. 2013;68(4):922-926.
-
Ashbee HR, Barnes RA, Johnson EM, Richardson MD, Gorton R, Hope WW. Therapeutic drug monitoring (TDM) of antifungal agents: guidelines from the British Society for Medical Mycology. J Antimicrob Chemother. 2014;69(5):1162-1176.
-
Lestner JM, Smith PB, Cohen-Wolkowiez M, Benjamin DK Jr, Hope WW. Antifungal agents and therapy for infants and children with invasive fungal infections: a pharmacological perspective. Br J Clin Pharmacol. 2013;75(6):1381-1395.
-
Hellden A, Odar-Cederlof I, Diener P, et al. High serum concentrations of the acyclovir main metabolite 9-carboxymethoxymethylguanine in renal failure patients with acyclovir-related neuropsychiatric side effects: an observational study. Nephrol Dial Transplant. 2003;18(6):1135-1141.
-
Robson R, Buttimore A, Lynn K, Brewster M, Ward P. The pharmacokinetics and tolerability of oseltamivir suspension in patients on haemodialysis and continuous ambulatory peritoneal dialysis. Nephrol Dial Transplant. 2006;21(9):2556-2562.
-
Yao X, Panichpisal K, Kurtzman N, Nugent K. Cisplatin nephrotoxicity: a review. Am J Med Sci. 2007;334(2):115-124.
-
Dean M. Opioids in renal failure and dialysis patients. J Pain Symptom Manage. 2004;28(5):497-504.
-
Mercadante S, Arcuri E. Opioids and renal function. J Pain. 2004;5(1):2-19.
-
Bauer TM, Ritz R, Haberthur C, et al. Prolonged sedation due to accumulation of conjugated metabolites of midazolam. Lancet. 1995;346(8968):145-147.
-
Höjer J, Baehrendtz S, Gustafsson L. Benzodiazepine poisoning: experience of 702 admissions to an intensive care unit during a 14-year period. J Intern Med. 1989;226(2):117-122.
-
Marik PE. Propofol: therapeutic indications and side-effects. Curr Pharm Des. 2004;10(29):3639-3649.
-
Devlin JW, Lau AK, Tanios MA. Propofol-associated hypertriglyceridemia and pancreatitis in the intensive care unit: an analysis of frequency and risk factors. Pharmacotherapy. 2005;25(10):1348-1352.
-
Vree TB, van Dongen RT, Koopman-Kimenai PM. Codeine analgesia is due to codeine-6-glucuronide, not morphine. Int J Clin Pract. 2000;54(6):395-398.
-
Randinitis EJ, Posvar EL, Alvey CW, Sedman AJ, Cook JA, Bockbrader HN. Pharmacokinetics of pregabalin in subjects with various degrees of renal function. J Clin Pharmacol. 2003;43(3):277-283.
-
Jentzer JC, Coons JC, Link CB, Schmidhofer M. Pharmacotherapy update on the use of vasopressors and inotropes in the intensive care unit. J Cardiovasc Pharmacol Ther. 2015;20(3):249-260.
-
Russell JA. Vasopressin in vasodilatory and septic shock. Curr Opin Crit Care. 2007;13(4):383-391.
-
Zangrillo A, Putzu A, Monaco F, et al. Levosimendan reduces mortality in patients with severe sepsis and septic shock: a meta-analysis of randomized trials. J Crit Care. 2015;30(5):908-913.
-
Lafuente-Lafuente C, Alvarez JC, Leenhardt A, et al. Amiodarone concentrations in plasma and fat tissue during chronic treatment and related toxicity. Br J Clin Pharmacol. 2009;67(5):511-519.
-
Collinsworth KA, Kalman SM, Harrison DC. The clinical pharmacology of lidocaine as an antiarrhythmic drug. Circulation. 1974;50(6):1217-1230.
-
Hirsh J, Bauer KA, Donati MB, et al. Parenteral anticoagulants: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest. 2008;133(6 Suppl):141S-159S.
-
Steffel J, Verhamme P, Potpara TS, et al. The 2018 European Heart Rhythm Association Practical Guide on the use of non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation. Eur Heart J. 2018;39(16):1330-1393.
-
Schentag JJ, Cerra FB, Calleri G, DeGlopper E, Rose JQ, Bernhard H. Pharmacokinetic and clinical studies in patients with cimetidine-associated mental confusion. Lancet. 1979;1(8109):177-181.
-
Tytgat GN. Pharmacological aspects of therapy in inflammatory bowel diseases: anti-inflammatory drugs and corticosteroids. Dig Dis. 2009;27(1):22-26.
-
Mertes PM, Laxenaire MC, Alla F. Anaphylactic and anaphylactoid reactions occurring during anesthesia in France in 1999-2000. Anesthesiology. 2003;99(3):536-545.
-
Baumüller E, Lierz P, Nadstawek J, Mertzlufft F, Hering R. Incidence of neuromuscular blockade and requirements of vecuronium during propofol-alfentanil and thiopental-isoflurane anesthesia. Anaesthesist. 1998;47(1):29-33.
-
Kraft MD, Btaiche IF, Sacks GS, Kudsk KA. Treatment of electrolyte disorders in adult patients in the intensive care unit. Am J Health Syst Pharm. 2005;62(16):1663-1682.
-
Matzke GR, Frye RF. Drug administration in patients with renal insufficiency. Minimising renal and extrarenal toxicity. Drug Saf. 1997;16(3):205-231.
-
Bogard KN, Peterson NT, Plumb TJ, Erwin MW, Fuller PD, Olsen KM. Antibiotic dosing during sustained low-efficiency dialysis: special considerations in adult critically ill patients. Crit Care Med. 2011;39(3):560-570.
-
Erstad BL. Dosing of medications in morbidly obese patients in the intensive care unit setting. Intensive Care Med. 2004;30(1):18-32.
-
Shekar K, Fraser JF, Smith MT, Roberts JA. Pharmacokinetic changes in patients receiving extracorporeal membrane oxygenation. J Crit Care. 2012;27(6):741.e9-741.e18.
-
Shekar K, Roberts JA, McDonald CI, et al. Sequestration of drugs in the circuit may lead to therapeutic failure during extracorporeal membrane oxygenation. Crit Care. 2012;16(5):R194.
-
Mueller BA, Smoyer WE. Challenges in developing evidence-based drug dosing guidelines for adults and children receiving renal replacement therapy. Clin Pharmacol Ther. 2009;86(5):479-482.
-
Li AM, Gomersall CD, Choi G, Tian Q, Joynt GM, Lipman J. A systematic review of antibiotic dosing regimens for septic patients receiving continuous renal replacement therapy: do current studies supply sufficient data? J Antimicrob Chemother. 2009;64(5):929-937.
-
Kane-Gill SL, Dasta JF, Buckley MS, Devabhakthuni S, Liu M, Cohen H. Clinical practice guideline: safe medication use in the ICU. Crit Care Med. 2017;45(9):e877-e915.
-
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(6):498-509.
-
Bagshaw SM, Gibney RT. Conventional markers of kidney function. Crit Care Med. 2008;36(4 Suppl):S152-S158.
-
Darwich AS, Ogungbenro K, Vinks AA, et al. Why has model-informed precision dosing not yet become common clinical reality? Lessons from the past and a roadmap for the future. Clin Pharmacol Ther. 2017;101(5):646-656.
-
Spruill WJ, Wade WE, Cobb HH 3rd. Continuing the use of the Cockcroft-Gault equation for drug dosing in patients with impaired renal function. Clin Pharmacol Ther. 2009;86(5):468-470.
-
Lewis SJ, Mueller BA. Antibiotic dosing in patients with acute kidney injury: "enough but not too much". J Intensive Care Med. 2016;31(3):164-176.
-
Lewis SJ, Mueller BA. Antibiotic dosing in critically ill patients with acute kidney injury and high-dose continuous venovenous hemofiltration. Pharmacotherapy. 2014;34(12):1334-1343.
-
Pai MP, Nafziger AN, Bertino JS Jr. Simplified estimation of aminoglycoside pharmacokinetics in underweight and obese adult patients. Antimicrob Agents Chemother. 2011;55(9):4006-4011.
-
Beumier M, Roberts JA, Kabtouri H, et al. A new regimen for continuous infusion of vancomycin during continuous renal replacement therapy. J Antimicrob Chemother. 2013;68(12):2859-2865.
-
Heintz BH, Matzke GR, Dager WE. Antimicrobial dosing concepts and recommendations for critically ill adult patients receiving continuous renal replacement therapy or intermittent hemodialysis. Pharmacotherapy. 2009;29(5):562-577.
-
Pea F, Viale P, Furlanut M. Antimicrobial therapy in critically ill patients: a review of pathophysiological conditions responsible for altered disposition and pharmacokinetic variability. Clin Pharmacokinet. 2005;44(10):1009-1034.
-
Ulldemolins M, Roberts JA, Lipman J, Rello J. Antibiotic dosing in multiple organ dysfunction syndrome. Chest. 2011;139(5):1210-1220.
-
Wong G, Brinkman A, Benefield RJ, et al. An international, multicentre survey of β-lactam antibiotic therapeutic drug monitoring practice in intensive care units. J Antimicrob Chemother. 2014;69(5):1416-1423.