Tuesday, May 20, 2025

Drug prescription in non CRRT CKD patients

 

Practical Drug Dosing in Conservative Management of CKD Patients in ICU: A Comprehensive Review

Dr Neeraj Manikath, Claude.ai

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:

  1. Altered pharmacokinetics (PK) and pharmacodynamics (PD) associated with impaired renal function
  2. Dynamic changes in renal function during critical illness
  3. The effects of critical illness itself on drug disposition
  4. Complex polypharmacy and drug-drug interactions
  5. 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:

  1. 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^

  2. Kinetic eGFR: Incorporating the rate of change of serum creatinine into GFR estimation may better account for non-steady-state conditions^32^

  3. Cystatin C-based equations: Less affected by muscle mass and nutritional status, though influenced by inflammation and corticosteroid use common in ICU settings^33^

  4. 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:

  1. Fraction eliminated unchanged by the kidney (fe): Drugs with fe >0.3 (30%) generally require dosage adjustment in CKD^35^

  2. Therapeutic index: Drugs with narrow therapeutic indices require more careful adjustment and potentially therapeutic drug monitoring (TDM)^36^

  3. Protein binding: Hypoalbuminemia in critically ill CKD patients increases the free fraction of highly protein-bound drugs, potentially increasing toxicity^37^

  4. 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:

  1. Dose reduction: Maintaining the standard dosing interval but reducing the individual dose amount

  2. Interval extension: Maintaining the standard dose amount but extending the time between doses

  3. 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:

  1. Assess renal function:

    • Recognize limitations of eGFR equations in critical illness
    • Consider measured creatinine clearance when feasible
    • Account for dynamic changes in renal function
  2. Evaluate drug characteristics:

    • Determine extent of renal elimination (fe)
    • Identify active metabolites eliminated renally
    • Consider therapeutic index and consequences of under/overdosing
  3. 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
  4. 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:

  1. 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^
  2. Real-time GFR monitoring:

    • Novel technologies to provide continuous assessment of renal function
    • Potential for more responsive drug dosing in dynamic ICU settings^90^
  3. 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

  1. Webster AC, Nagler EV, Morton RL, Masson P. Chronic kidney disease. Lancet. 2017;389(10075):1238-1252.

  2. 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.

  3. 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.

  4. 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.

  5. Bouchard J, Macedo E, Mehta RL. Dosing of drugs in patients with renal failure in the intensive care unit. UpToDate. 2023.

  6. Roberts JA, Lipman J. Pharmacokinetic issues for antibiotics in the critically ill patient. Crit Care Med. 2009;37(3):840-851.

  7. Ashley C, Dunleavy A, eds. The Renal Drug Handbook. 5th ed. CRC Press; 2018.

  8. Boucher BA, Wood GC, Swanson JM. Pharmacokinetic changes in critical illness. Crit Care Clin. 2006;22(2):255-271.

  9. Sica DA. Drug absorption in the management of congestive heart failure: loop diuretics. Congest Heart Fail. 2003;9(5):287-292.

  10. Vilay AM, Churchwell MD, Mueller BA. Drug metabolism and nonrenal clearance in acute kidney injury. Crit Care. 2008;12(6):235.

  11. Verbeeck RK, Musuamba FT. Pharmacokinetics and dosage adjustment in patients with renal dysfunction. Eur J Clin Pharmacol. 2009;65(8):757-773.

  12. Lam YW, Banerji S, Hatfield C, Talbert RL. Principles of drug administration in renal insufficiency. Clin Pharmacokinet. 1997;32(1):30-57.

  13. 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.

  14. Roberts JA, Lipman J. Antibacterial dosing in intensive care: pharmacokinetics, degree of disease and pharmacodynamics of sepsis. Clin Pharmacokinet. 2006;45(8):755-773.

  15. 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.

  16. Sun H, Frassetto L, Benet LZ. Effects of renal failure on drug transport and metabolism. Pharmacol Ther. 2006;109(1-2):1-11.

  17. 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.

  18. Naud J, Nolin TD, Leblond FA, Pichette V. Current understanding of drug disposition in kidney disease. J Clin Pharmacol. 2012;52(S1):10S-22S.

  19. 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.

  20. Matzke GR, Frye RF. Drug administration in patients with renal insufficiency. Minimising renal and extrarenal toxicity. Drug Saf. 1997;16(3):205-231.

  21. Brater DC. Drug dosing in patients with impaired renal function. Clin Pharmacol Ther. 2009;86(5):483-489.

  22. Nolin TD. Altered nonrenal drug clearance in ESRD. Curr Opin Nephrol Hypertens. 2008;17(6):555-559.

  23. Verbeeck RK, Musuamba FT. Pharmacokinetics and dosage adjustment in patients with renal dysfunction. Eur J Clin Pharmacol. 2009;65(8):757-773.

  24. Gabardi S, Abramson S. Drug dosing in chronic kidney disease. Med Clin North Am. 2005;89(3):649-687.

  25. Churchwell MD, Mueller BA. Drug dosing during kidney disease. Adv Chronic Kidney Dis. 2010;17(5):e48-e59.

  26. Matzke GR, Frye RF. Drug administration in patients with renal insufficiency. Minimising renal and extrarenal toxicity. Drug Saf. 1997;16(3):205-231.

  27. Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604-612.

  28. 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.

  29. 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.

  30. 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.

  31. 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.

  32. 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.

  33. 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.

  34. 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.

  35. 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.

  36. Olyaei AJ, Bennett WM. Drug dosing in the elderly patients with chronic kidney disease. Clin Geriatr Med. 2009;25(3):459-527.

  37. Verbeeck RK. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 2008;64(12):1147-1161.

  38. Churchwell MD, Mueller BA. Drug dosing during continuous renal replacement therapy. Semin Dial. 2009;22(2):185-188.

  39. Drusano GL. Antimicrobial pharmacodynamics: critical interactions of 'bug and drug'. Nat Rev Microbiol. 2004;2(4):289-300.

  40. 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.

  41. 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.

  42. 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.

  43. 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.

  44. 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.

  45. 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.

  46. Roberts JA, Lipman J. Pharmacokinetic issues for antibiotics in the critically ill patient. Crit Care Med. 2009;37(3):840-851.

  47. 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.

  48. 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.

  49. 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.

  50. 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.

  51. 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.

  52. Neely MN, Youn G, Jones B, et al. Are vancomycin trough concentrations adequate for optimal dosing? Antimicrob Agents Chemother. 2014;58(1):309-316.

  53. 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.

  54. 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.

  55. 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.

  56. 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.

  57. 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.

  58. 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.

  59. 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.

  60. Yao X, Panichpisal K, Kurtzman N, Nugent K. Cisplatin nephrotoxicity: a review. Am J Med Sci. 2007;334(2):115-124.

  61. Dean M. Opioids in renal failure and dialysis patients. J Pain Symptom Manage. 2004;28(5):497-504.

  62. Mercadante S, Arcuri E. Opioids and renal function. J Pain. 2004;5(1):2-19.

  63. Bauer TM, Ritz R, Haberthur C, et al. Prolonged sedation due to accumulation of conjugated metabolites of midazolam. Lancet. 1995;346(8968):145-147.

  64. 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.

  65. Marik PE. Propofol: therapeutic indications and side-effects. Curr Pharm Des. 2004;10(29):3639-3649.

  66. 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.

  67. 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.

  68. 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.

  69. 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.

  70. Russell JA. Vasopressin in vasodilatory and septic shock. Curr Opin Crit Care. 2007;13(4):383-391.

  71. 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.

  72. 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.

  73. Collinsworth KA, Kalman SM, Harrison DC. The clinical pharmacology of lidocaine as an antiarrhythmic drug. Circulation. 1974;50(6):1217-1230.

  74. 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.

  75. 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.

  76. 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.

  77. Tytgat GN. Pharmacological aspects of therapy in inflammatory bowel diseases: anti-inflammatory drugs and corticosteroids. Dig Dis. 2009;27(1):22-26.

  78. Mertes PM, Laxenaire MC, Alla F. Anaphylactic and anaphylactoid reactions occurring during anesthesia in France in 1999-2000. Anesthesiology. 2003;99(3):536-545.

  79. 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.

  80. 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.

  81. Matzke GR, Frye RF. Drug administration in patients with renal insufficiency. Minimising renal and extrarenal toxicity. Drug Saf. 1997;16(3):205-231.

  82. 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.

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

  84. 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.

  85. 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.

  86. 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.

  87. 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.

  88. 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.

  89. 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.

  90. Bagshaw SM, Gibney RT. Conventional markers of kidney function. Crit Care Med. 2008;36(4 Suppl):S152-S158.

  91. 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.

  92. 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.

  93. 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.

  94. 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.

  95. 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.

  96. 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.

  97. 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.

  98. 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.

  99. Ulldemolins M, Roberts JA, Lipman J, Rello J. Antibiotic dosing in multiple organ dysfunction syndrome. Chest. 2011;139(5):1210-1220.

  100. 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.

Monday, May 19, 2025

Drug Dosing in Acute Kidney Injury

Practical Drug Dosing in Acute Kidney Injury: A Comprehensive Review for Critical Care Practitioners

Dr Neeraj Manikath, Claude.ai


Abstract


Acute kidney injury (AKI) is highly prevalent in intensive care units (ICUs), affecting approximately 50-60% of critically ill patients. Drug dosing in this population presents a significant challenge due to alterations in pharmacokinetic and pharmacodynamic parameters. This comprehensive review addresses the practical aspects of drug dosing in critically ill patients with AKI, incorporating the latest evidence and clinical recommendations. We discuss the pathophysiological changes in AKI that affect drug disposition, approaches to drug dosing based on renal replacement therapy (RRT) modalities, and specific dosing recommendations for commonly used medications in the ICU. Additionally, this review explores emerging technologies and strategies for personalized drug dosing in this complex patient population. The goal is to provide critical care practitioners with practical tools to optimize pharmacotherapy in patients with AKI, ultimately improving clinical outcomes while reducing adverse drug events.


Keywords: Acute kidney injury; Critical care; Drug dosing; Pharmacokinetics; Renal replacement therapy; Therapeutic drug monitoring


Introduction


Acute kidney injury (AKI) remains a common and serious complication in critically ill patients, with an incidence ranging from 20% to 60% in intensive care units (ICUs) depending on the population studied and the definition used.^1^ The presence of AKI significantly increases mortality, length of stay, and healthcare costs.^2,3^ Appropriate drug dosing in patients with AKI is particularly challenging due to alterations in drug absorption, distribution, metabolism, and elimination.^4^ Furthermore, critical illness itself introduces additional complexity through pathophysiological changes such as altered protein binding, increased volume of distribution, and variable organ function.^5^


The challenge is further compounded by the use of renal replacement therapies (RRT), which introduce additional variables affecting drug clearance, including modality, flow rates, membrane characteristics, and duration of therapy.^6^ Inappropriate drug dosing in AKI may lead to treatment failure due to underdosing or toxic effects from overdosing, both contributing to poor clinical outcomes.^7,8^


Despite these challenges, evidence-based dosing guidelines specifically tailored to AKI patients in the ICU setting remain limited. Clinicians often rely on general principles, package inserts with limited information on AKI dosing, or expert opinion when making dosing decisions.^9^ The aim of this review is to provide a comprehensive, practical approach to drug dosing in critically ill patients with AKI, incorporating the latest evidence and clinical recommendations.


 Pathophysiological Changes in AKI Affecting Drug Disposition


 Alterations in Pharmacokinetics


 Absorption

While drug absorption is generally less affected in AKI compared to other pharmacokinetic parameters, several factors may influence this process in critically ill patients with AKI. Reduced splanchnic blood flow, increased gastric pH, delayed gastric emptying, and decreased intestinal motility—all common in critical illness—can alter the absorption of orally administered medications.^10^ Additionally, edema of the gastrointestinal tract, which may occur in patients with fluid overload secondary to AKI, can further impair drug absorption.^11^


 Distribution

The volume of distribution (Vd) of many drugs is significantly altered in AKI. Factors contributing to these changes include:


1. Fluid overload: Common in AKI, fluid overload increases the Vd of hydrophilic drugs, potentially leading to subtherapeutic concentrations with standard dosing.^12^

2. Hypoalbuminemia: Critically ill patients often present with hypoalbuminemia due to inflammation, malnutrition, and protein losses. This condition reduces drug binding to albumin, resulting in higher free fractions of highly protein-bound drugs.^13^


3. Acid-base disturbances: Metabolic acidosis, frequently seen in AKI, can affect drug ionization and tissue penetration, altering distribution patterns.^14^


4. Tissue perfusion changes:Altered hemodynamics in critical illness affects tissue perfusion and consequently drug distribution to various organs.^15^


Metabolism

Hepatic drug metabolism may be affected in AKI through several mechanisms:


1. Hepatorenal syndrome: Impaired renal function can lead to altered hepatic blood flow and function.^16^


2. Uremic toxins:Accumulation of uremic toxins in AKI can inhibit cytochrome P450 enzymes and other metabolic pathways.^17^


3. Inflammatory mediators:The systemic inflammatory response common in critically ill patients with AKI can downregulate hepatic drug-metabolizing enzymes.^18^


4. Altered protein binding: Changes in protein binding affect the availability of drugs for hepatic metabolism.^19^

 Elimination

Renal drug elimination is directly affected by AKI, with several important considerations:


1. Glomerular filtration: Reduced glomerular filtration rate (GFR) decreases the elimination of drugs primarily excreted unchanged by the kidneys.^20^


2. Tubular secretion and reabsorption: AKI affects tubular function, altering active secretion and reabsorption processes for many drugs.^21^


3. Dynamic nature of AKI: Unlike chronic kidney disease, AKI is often a rapidly changing condition, with potential for improvement or deterioration in renal function over short periods.^22^


4. Non-renal clearance: Compensatory increases in non-renal clearance pathways may occur for some drugs in the setting of AKI.^23^


Alterations in Pharmacodynamics


The pharmacodynamic response to drugs may be altered in AKI due to:


1. Uremic toxins: Accumulation of uremic toxins can modify receptor sensitivity and drug-receptor interactions.^24^


2. Electrolyte abnormalities:Disturbances in electrolyte balance (particularly potassium, calcium, and magnesium) can affect the action of various drugs, especially those with cardiovascular effects.^25^


3. Acid-base disturbances: Changes in pH can alter drug ionization and receptor binding.^26^


4. End-organ sensitivity: Target organ sensitivity to drugs may be altered in the uremic state.^27^


 Assessment of Kidney Function in Critical Care


 Limitations of Traditional Markers


Accurate assessment of kidney function is crucial for appropriate drug dosing in AKI. Traditional markers such as serum creatinine and urea nitrogen have significant limitations in critically ill patients:


1. Serum creatinine:** Changes in serum creatinine lag behind actual changes in GFR by 24-48 hours. Additionally, factors such as reduced muscle mass, dilution due to fluid overload, and altered tubular secretion affect creatinine levels independently of GFR.^28^


2. Estimated GFR equations:** Commonly used equations (CKD-EPI, MDRD, Cockcroft-Gault) were developed in stable patients and are not validated in AKI or critical illness.^29^ These equations assume steady-state conditions, which rarely exist in AKI.


3. Measured creatinine clearance:** 24-hour urine collections are impractical in the ICU setting and may not reflect rapidly changing kidney function.^30^


 Newer Approaches to Assess Kidney Function


Several newer approaches show promise for more accurate assessment of kidney function in critically ill patients:


1. Novel biomarkers: Biomarkers such as neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), and tissue inhibitor of metalloproteinase-2 (TIMP-2) × insulin-like growth factor-binding protein 7 (IGFBP7) may provide earlier detection of AKI and potentially guide drug dosing.^31^


2. Real-time GFR measurement:Technologies for continuous or frequent GFR monitoring are under development, including transcutaneous fluorescence measurement after administration of exogenous fluorescent markers.^32^


3. Short timed urine collections: 2-4 hour urine collections for creatinine clearance may provide more accurate and timely assessment of kidney function than estimated GFR.^33^


4. Kinetic estimated GFR (KeGFR): This approach incorporates the rate of change of serum creatinine to estimate GFR in non-steady-state conditions.^34^


 General Principles of Drug Dosing in AKI


Loading Doses


Loading doses are typically not affected by kidney function and should generally be administered at full dose to rapidly achieve therapeutic concentrations, especially for critical indications:


1. Volume of distribution considerations:Loading doses should be adjusted based on altered Vd in critically ill patients (e.g., increased for hydrophilic drugs in fluid overload).^35^


2. Critical indications:Full loading doses are particularly important for life-threatening conditions such as sepsis, where delays in achieving therapeutic concentrations may increase mortality.^36^


3. Highly protein-bound drugs:Consider the effects of hypoalbuminemia on free drug concentrations when calculating loading doses.^37^


 Maintenance Dosing Strategies


Several approaches can be used for maintenance dosing in AKI:


1. Dose reduction: Reducing the dose while maintaining the standard dosing interval is appropriate for drugs with concentration-dependent efficacy and wide therapeutic index.^38^


2. Interval extension: Extending the dosing interval while maintaining the standard dose is generally prefered for drugs with time-dependent efficacy and narrow therapeutic index.^39^


3. Combined approach: Both dose reduction and interval extension may be necessary for some medications.^40^


4. Continuous infusion: For some drugs, particularly antimicrobials with time-dependent activity, continuous infusion may optimize pharmacodynamics in AKI.^41^


 Special Considerations for Different Drug Classes


 Antimicrobials


1. Beta-lactams: Often require dose reduction or interval extension in AKI. Consider extended or continuous infusions to optimize time above MIC.^42^


2. Aminoglycosides: Require significant dosing adjustments in AKI due to narrow therapeutic index. Extended-interval dosing (once daily) with therapeutic drug monitoring is recommended when possible.^43^


3. Vancomycin: Dosing should be guided by therapeutic drug monitoring, with area under the curve (AUC)/MIC ratio as the preferred pharmacodynamic target.^44^


4. Fluoroquinolones: Moderate dose reductions are typically required in AKI, with specific adjustments varying by agent.^45^


 Sedatives and Analgesics


1. Opioids: Many opioids or their active metabolites accumulate in AKI, potentially leading to prolonged sedation and respiratory depression. Fentanyl and remifentanil are generally preferred in AKI.^46^


2. Benzodiazepines:Prolonged effect may be seen with midazolam due to accumulation of active metabolites. Lorazepam may be preferred but requires careful monitoring.^47^


3. Propofol:Not significantly affected by AKI but may contribute to metabolic acidosis during prolonged infusion.^48^


4. Dexmedetomidine: Primarily hepatically metabolized and generally not significantly affected by AKI.^49^


 Cardiovascular Medications


1. Vasopressors and inotropes: Generally do not require dose adjustment in AKI, though enhanced sensitivity may occur.^50^


2. Antihypertensives:ACE inhibitors and ARBs should be used cautiously in AKI. Calcium channel blockers generally do not require significant dose adjustments.^51^


3. Antiarrhythmics: Significant dose adjustments may be required for digoxin, sotalol, and atenolol in AKI.^52^


Anticoagulants


1. Low molecular weight heparins (LMWH): Accumulate in AKI, requiring dose reduction and potentially anti-Xa monitoring.^53^


2. Direct oral anticoagulants (DOACs):All require dose adjustments in AKI; some are contraindicated in severe AKI.^54^


3. Unfractionated heparin: Preferred in severe AKI due to monitoring capability and reversibility.^55^


Drug Dosing in Different Renal Replacement Therapies


Intermittent Hemodialysis (IHD)


Factors affecting drug removal during IHD include:


1. Drug characteristics: Molecular weight, protein binding, volume of distribution, and water solubility affect dialyzability.^56^


2. Dialysis parameters: Blood flow rate, dialysate flow rate, duration of therapy, and membrane characteristics influence drug clearance.^57^


3. Timing considerations:Administering doses post-dialysis is often recommended for dialyzable drugs.^58^


Practical approach:

- For highly dialyzable drugs, administer supplemental doses after dialysis sessions.

- Consider the residual kidney function in addition to dialysis clearance.

- Use therapeutic drug monitoring when available for drugs with narrow therapeutic indices.


 Continuous Renal Replacement Therapy (CRRT)


CRRT provides more constant drug clearance compared to IHD but introduces additional variables:


1. CRRT modality:Different modalities (CVVH, CVVHD, CVVHDF) provide different clearance mechanisms (convection, diffusion, or both).^59^


2. Flow rates:Effluent flow rate is the primary determinant of drug clearance in CRRT.^60^


3. Membrane characteristics:Adsorption to the filter membrane can significantly affect clearance of some drugs.^61^


4. Filter lifespan: Declining filter efficiency over time may affect drug clearance.^62^


Practical approach:

- Calculate drug clearance based on effluent flow rate for most drugs.

- Use the equation: CLextracorporeal = Sieving/saturation coefficient × Effluent flow rate.

- Consider higher doses for antimicrobials, especially in the early phase of filter use.

- Therapeutic drug monitoring is essential when available.


 Prolonged Intermittent Renal Replacement Therapy (PIRRT)


PIRRT combines elements of both IHD and CRRT, requiring special dosing considerations:


1. Hybrid nature:Higher clearance rates than CRRT but shorter duration than IHD.^63^


2. Variable schedules:Different institutions use different durations and frequencies of PIRRT.^64^


3. Limited data: Fewer pharmacokinetic studies compared to IHD and CRRT.^65^


Practical approach:

- Consider timing of drug administration relative to PIRRT session.

- For critical medications, supplemental doses may be required post-PIRRT.

- When specific data is unavailable, use a conservative approach between IHD and CRRT recommendations.


Specific Drug Dosing Recommendations


 Antimicrobials


 Beta-lactams




Glycopeptides and Lipopeptides




Evidence level: A (high quality evidence from randomized trials and large observational studies)


 Aminoglycosides




Evidence level: A (high quality evidence from randomized trials and large observational studies)


 Fluoroquinolones




Evidence level: B (moderate evidence from multiple observational studies with some inconsistency)


Antifungals




 Antivirals



 Sedatives and Analgesics



Evidence level: C (limited evidence primarily from expert opinion and case reports)*


Cardiovascular Medications




 Anticoagulants



Therapeutic Drug Monitoring in AKI


 Traditional Approaches


Therapeutic drug monitoring (TDM) is crucial for optimizing drug therapy in AKI:


1. Target drugs: Traditionally includes drugs with narrow therapeutic indices, such as aminoglycosides, vancomycin, and anticonvulsants.^66^


2. Sampling strategies: Trough levels are commonly used for many drugs, but more complex approaches such as AUC/MIC for vancomycin may improve outcomes.^67^


3. Interpretation challenges:Altered protein binding in critically ill patients with AKI complicates interpretation of total drug concentrations.^68^


 Emerging Technologies and Approaches


Several emerging approaches show promise for enhancing TDM in AKI:


1. Continuous or point-of-care monitoring:Real-time monitoring technologies for certain drugs are under development.^69^


2. Model-informed precision dosing (MIPD):*Uses population pharmacokinetic models and Bayesian forecasting to individualize dosing based on patient characteristics and measured drug levels.^70^


3. Free drug monitoring:Measurement of unbound drug concentrations may be more clinically relevant than total concentrations, especially in conditions with altered protein binding.^71^


4. Dried blood spot (DBS) sampling: Allows for less invasive, more frequent sampling with smaller blood volumes.^72^


Special Populations


Elderly Patients with AKI


Elderly patients require additional considerations:


1. Reduced muscle mass: Lower creatinine production may mask significant reductions in GFR.^73^


2. Polypharmacy:Increased risk of drug interactions affecting pharmacokinetics and pharmacodynamics.^74^


3. Altered body composition:Changes in body water and fat content affect drug distribution.^75^


4. Increased sensitivity:Enhanced sensitivity to many drugs, particularly those affecting the central nervous system.^76^


Practical approach:

- Use more conservative dosing regimens.

- Consider alternative methods to estimate GFR.

- More frequent monitoring for adverse effects.


Obese Patients with AKI


Obesity introduces additional complexities:


1. Dosing weight selection: Actual body weight, ideal body weight, or adjusted body weight may be appropriate depending on the drug.^77^


2. Altered drug distribution: Changes in adipose tissue proportion affect drug distribution.^78^


3. Augmented renal clearance: May occur in obese patients without AKI or in early stages of critical illness.^79^


4. GFR estimation: Traditional equations perform poorly in obesity.^80^


Practical approach:

- Consider higher loading doses for lipophilic drugs based on actual body weight.

- Use adjusted body weight for maintenance dosing of many drugs.

- Employ therapeutic drug monitoring when available.


Patients with Hepatorenal Syndrome


Hepatorenal syndrome presents unique challenges:


1. Dual organ dysfunction: Combined hepatic and renal impairment affects both drug metabolism and elimination.^81^


2. Hypoalbuminemia:Significant reductions in protein binding affect drug disposition.^82^


3. Portal hypertension: Altered splanchnic blood flow affects drug absorption and first-pass metabolism.^83^


4. Increased bleeding risk: Coagulopathy requires careful consideration of anticoagulant dosing.^84^


Practical approach:

- Use drugs with minimal hepatic metabolism when possible.

- Consider reduced doses of drugs with significant hepatic metabolism.

- Monitor for heightened sensitivity to CNS-active medications.

- Frequent clinical reassessment of drug response.


 Emerging Concepts and Future Directions


 Artificial Intelligence and Machine Learning


AI and machine learning are being increasingly applied to optimize drug dosing:


1. Predictive models:Development of algorithms to predict AKI and drug clearance in critically ill patients.^85^


2. Decision support systems: Integration of pharmacokinetic models with electronic health records to provide real-time dosing recommendations.^86^


3. Pattern recognition: Identification of patient subgroups that may respond differently to standard dosing approaches.^87^


Pharmacogenomics in AKI


Genetic factors may influence drug response in AKI:


1. Transporter polymorphisms: Variations in drug transporters affect drug disposition in kidney dysfunction.^88^


2. Metabolism enzyme variations: Genetic polymorphisms in cytochrome P450 enzymes may become more clinically relevant in AKI.^89^


3. Receptor variations: Genetic differences in drug targets may affect pharmacodynamic response.^90^


Novel Drug Delivery Systems


Innovative delivery approaches may improve drug therapy in AKI:


1. Nanomedicine: Nanoparticle-based drug delivery systems may allow for more targeted therapy with reduced systemic exposure.^91^


2. Dialysis-responsive systems: Drug formulations designed to respond to dialysis conditions to maintain therapeutic levels.^92^


3. Implantable monitoring and delivery systems:Devices that combine real-time monitoring with automated drug delivery may allow for unprecedented precision in dosing.^93^


Practical Approach to Drug Dosing in AKI: A Stepwise Framework


Based on the evidence and considerations discussed in this review, we propose the following stepwise approach to drug dosing in critically ill patients with AKI:


Step 1: Assess Renal Function

- Evaluate current renal function using serum creatinine, urine output, and novel biomarkers when available.

- Consider the trajectory of kidney function (improving, stable, or worsening).

- Assess the need for RRT and identify the specific modality if applicable.


 Step 2: Consider Drug Characteristics

- Review the pharmacokinetic properties of the drug (protein binding, volume of distribution, elimination pathway).

- Determine if the drug or active metabolites are renally eliminated.

- Assess the therapeutic index and potential toxicity of the drug.


Step 3: Evaluate Patient-Specific Factors

- Consider age, weight, and body composition.

- Assess for concomitant organ dysfunction, particularly hepatic impairment.

- Review current medications for potential drug interactions.

- Evaluate protein status and acid-base balance.


Step 4: Apply Dosing Principles

- Administer full loading doses for most drugs, especially in critical indications.

- Adjust maintenance doses based on estimated drug clearance.

- Consider the clinical context and urgency of achieving therapeutic levels.

- Use the most appropriate dosing strategy (dose reduction, interval extension, or combination).


 Step 5: Implement Monitoring

- Utilize therapeutic drug monitoring when available.

- Monitor for clinical response and adverse effects.

- Reassess kidney function regularly and adjust dosing as needed.

- Consider the timing of drug administration relative to RRT sessions.


 Step 6: Continuous Reassessment

- Regularly reevaluate the need for continued therapy.

- Adjust dosing with changing kidney function or RRT parameters.

- Consider tapering or discontinuation strategies for certain medications.


Clinical Cases and Practical Examples


 Case 1: Septic Shock with AKI


A 68-year-old male (80 kg) presents with septic shock due to pneumonia. His serum creatinine has increased from a baseline of 0.9 mg/dL to 2.8 mg/dL over 24 hours, with urine output of 0.3 mL/kg/h for the past 6 hours. The patient requires empiric antimicrobial therapy.


Assessment and Approach:

- Patient has AKI Stage 2 by KDIGO criteria.

- Estimated CrCl approximately 20-25 mL/min using the Cockcroft-Gault equation.

- Patient has not yet required RRT but may need it if kidney function continues to deteriorate.


Drug Selection and Dosing:

- Piperacillin-tazobactam: 4.5g loading dose, followed by 2.25g q6h.

- Vancomycin: 25 mg/kg loading dose (2000 mg), followed by 15 mg/kg q24h (1200 mg daily), with trough levels monitored before the third dose.

- Monitor kidney function closely and adjust dosing as needed.


Rationale:

- Full loading doses ensure rapid achievement of therapeutic concentrations in this critically ill patient.

- Maintenance doses are adjusted based on estimated renal function.

- Frequent reassessment is necessary given the dynamic nature of sepsis and AKI.


 Case 2: CRRT in a Patient with Multiple Organ Dysfunction


A 45-year-old female (70 kg) with sepsis and multi-organ dysfunction is receiving CVVHDF with an effluent rate of 25 mL/kg/h. She requires antimicrobial therapy for a suspected catheter-related bloodstream infection.


Assessment and Approach:

- Patient has AKI requiring CRRT with moderate clearance.

- Assume minimal residual native kidney function.

- Consider CRRT parameters for drug clearance estimation.


Drug Selection and Dosing:

- Meropenem: 1g loading dose, followed by 1g q8h.

- Vancomycin: 20 mg/kg loading dose (1400 mg), followed by 10 mg/kg q24h (700 mg daily), with AUC-guided dosing.

- Fluconazole: 800 mg loading dose, followed by 400 mg q24h.


Rationale:

- CRRT provides relatively consistent but lower drug clearance compared to normal kidney function.

- Loading doses are not affected by CRRT.

- Maintenance doses are higher than would be used in AKI without RRT but lower than normal doses.

- Therapeutic drug monitoring is essential for vancomycin.

Case 3: Transitioning from CRRT to Intermittent Hemodialysis


A 72-year-old male (90 kg) with improving clinical status is transitioning from CVVHDF to intermittent hemodialysis (IHD) three times weekly. He is currently receiving antimicrobial therapy for ventilator-associated pneumonia.


Assessment and Approach:

- Patient's drug clearance will change significantly with the transition from continuous to intermittent therapy.

- Consider the timing of IHD sessions in relation to drug administration.

- Anticipate potential drug accumulation between dialysis sessions.


Drug Selection and Dosing:

- Cefepime: Transition from 2g q12h to 1g q24h with supplemental 1g dose post-dialysis on dialysis days.

- Vancomycin: Transition from continuous dosing to 15 mg/kg q48-72h based on levels, with doses administered post-dialysis.

- Avoid administration of highly dialyzable drugs shortly before dialysis sessions.


Rationale:

- Intermittent nature of IHD creates periods of minimal drug clearance between sessions.

- Supplemental post-dialysis dosing compensates for drug removal during IHD.

- Therapeutic drug monitoring becomes even more important during this transition.


Conclusion


Drug dosing in critically ill patients with AKI remains a significant challenge requiring a systematic approach based on pathophysiological principles, pharmacokinetic understanding, and clinical context. This review provides a comprehensive framework for approaching drug dosing in this complex population, recognizing that decisions must be individualized and regularly reassessed as patients' clinical conditions evolve.


The dynamic nature of AKI in critical illness, combined with the impact of RRT modalities, necessitates vigilance and a proactive approach to dosing adjustments. While general principles and recommendations provide valuable guidance, therapeutic drug monitoring, when available, remains essential for optimizing therapy with many medications. Furthermore, the incorporation of emerging technologies, such as model-informed precision dosing and artificial intelligence, holds promise for further refining our approach to drug dosing in AKI.


Future research should focus on validating dosing strategies in specific patient populations, exploring the impact of novel AKI biomarkers on drug dosing decisions, and investigating the potential of personalized medicine approaches, including pharmacogenomics, in this field. Until more robust evidence emerges, clinicians must rely on a thoughtful integration of pharmacokinetic principles, available evidence, and clinical judgment to optimize drug therapy in critically ill patients with AKI.


References


1. 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.


2. Chertow GM, Burdick E, Honour M, Bonventre JV, Bates DW. Acute kidney injury, mortality, length of stay, and costs in hospitalized patients. J Am Soc Nephrol. 2005;16(11):3365-3370.


3. Silver SA, Chertow GM. The economic consequences of acute kidney injury. Nephron. 2017;137(4):297-301.


4. 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.


5. Roberts JA, Lipman J. Pharmacokinetic issues for antibiotics in the critically ill patient. Crit Care Med. 2009;37(3):840-851.


6. 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.


7. Hobbs AL, Shea KM, Roberts KM, Daley MJ. Implications of augmented renal clearance on drug dosing in critically ill patients: a focus on antibiotics. Pharmacotherapy. 2015;35(11):1063-1075.


8. Eyler RF, Mueller BA. Antibiotic dosing in critically ill patients with acute kidney injury. Nat Rev Nephrol. 2011;7(4):226-235.


9. Pai MP, Nafziger AN, Bertino JS Jr. Simplified approach to optimizing initial vancomycin therapy. Antimicrob Agents Chemother. 2009;53(5):1648-1654.


10. Rowland Yeo K, Aarabi M, Jamei M, Rostami-Hodjegan A. Modeling and predicting drug pharmacokinetics in patients with renal impairment. Expert Rev Clin Pharmacol. 2011;4(2):261-274.


11. Boucher BA, Wood GC, Swanson JM. Pharmacokinetic changes in critical illness. Crit Care Clin. 2006;22(2):255-271.


12. Sime FB, Udy AA, Roberts JA. Augmented renal clearance in critically ill patients: etiology, definition and implications for beta-lactam dose optimization. Curr Opin Pharmacol. 2015;24:1-6.


13. Ulldemolins M, Roberts JA, Rello J, Paterson DL, Lipman J. The effects of hypoalbuminaemia on optimizing antibacterial dosing in critically ill patients. Clin Pharmacokinet. 2011;50(2):99-110.


14. 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.


15. 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.


16. Verbeeck RK. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 2008;64(12):1147-1161.


17. Vilay AM, Churchwell MD, Mueller BA. Clinical review: Patho-physiology of acute kidney injury in the intensive care unit. Crit Care. 2008;12(6):235.


18. Morgan ET. Impact of infectious and inflammatory disease on cytochrome P450-mediated drug metabolism and pharmacokinetics. Clin Pharmacol Ther. 2009;85(4):434-438.


19. Benet LZ, Hoener BA. Changes in plasma protein binding have little clinical relevance. Clin Pharmacol Ther. 2002;71(3):115-121.


20. 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.


21. Sunder-Plassmann G, Hörl WH. The pharmacokinetics of piperacillin/tazobactam during continuous venovenous hemofiltration (CVVH). Intensive Care Med. 2000;26(6):743-747.


22. Chawla LS, Bellomo R, Bihorac A, et al. Acute kidney disease and renal recovery: consensus report of the Acute Disease Quality Initiative (ADQI) 16 Workgroup. Nat Rev Nephrol. 2017;13(4):241-257.


23. Vilay AM, Grio M, Depestel DD, et al. Daptomycin pharmacokinetics in critically ill patients receiving continuous venovenous hemodialysis. Crit Care Med. 2011;39(1):19-25.


24. Vanholder R, De Smet R, Glorieux G, et al. Review on uremic toxins: classification, concentration, and interindividual variability. Kidney Int. 2003;63(5):1934-1943.


25. Zazzo JF, Troche G, Ruel P, Maintenant J. High incidence of hypophosphatemia in surgical intensive care patients: efficacy of phosphorus therapy on myocardial function. Intensive Care Med. 1995;21(10):826-831.


26. Kraut JA, Madias NE. Metabolic acidosis: pathophysiology, diagnosis and management. Nat Rev Nephrol. 2010;6(5):274-285.


27. van den Anker JN, Schwab M, Kearns GL. Developmental pharmacokinetics. Handb Exp Pharmacol. 2011;205:51-75.


28. Bagshaw SM, Gibney RT. Conventional markers of kidney function. Crit Care Med. 2008;36(4 Suppl):S152-S158.


29. Inker LA, Perrone RD. Assessment of kidney function. In: UpToDate, Post TW (Ed), UpToDate, Waltham, MA. Accessed May 15, 2025.


30. Macedo E, Mehta RL. Measuring renal function in critically ill patients: tools and strategies for assessing glomerular filtration rate. Curr Opin Crit Care. 2013;19(6):560-566.


31. Kashani K, Al-Khafaji A, Ardiles T, et al. Discovery and validation of cell cycle arrest biomarkers in human acute kidney injury. Crit Care. 2013;17(1):R25.


32. Schock-Kusch D, Sadick M, Henninger N, et al. Transcutaneous measurement of glomerular filtration rate using FITC-sinistrin in rats. Nephrol Dial Transplant. 2009;24(10):2997-3001.


33. Herrera-Gutiérrez ME, Seller-Pérez G, Banderas-Bravo E, Muñoz-Bono J, Lebrón-Gallardo M, Fernandez-Ortega JF. Replacement of 24-h creatinine clearance by 2-h creatinine clearance in intensive care unit patients: a single-center study. Intensive Care Med. 2007;33(11):1900-1906.


34. 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.


35. Trotman RL, Williamson JC, Shoemaker DM, Salzer WL. Antibiotic dosing in critically ill adult patients receiving continuous renal replacement therapy. Clin Infect Dis. 2005;41(8):1159-1166.


36. Kumar A, Roberts D, Wood KE, et al. Duration of hypotension before initiation of effective antimicrobial therapy is the critical determinant of survival in human septic shock. Crit Care Med. 2006;34(6):1589-1596.


37. Roberts JA, Pea F, Lipman J. The clinical relevance of plasma protein binding changes. Clin Pharmacokinet. 2013;52(1):1-8.


38. Dellinger RP, Levy MM, Rhodes A, et al. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock, 2012. Intensive Care Med. 2013;39(2):165-228.


39. Bauer LA. Applied Clinical Pharmacokinetics. 3rd ed. New York, NY: McGraw-Hill Medical; 2014.


40. 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.


41. Roberts JA, Abdul-Aziz MH, Davis JS, 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):681-691.


42. Carlier M, Stove V, Roberts JA, et al. Quantification of amikacin and gentamicin in human plasma using a simple and validated LC-MS/MS method. Int J Antimicrob Agents. 2012;40(5):416-422.


43. Dellinger RP, Levy MM, Rhodes A, et al. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock, 2012. Intensive Care Med. 2013;39(2):165-228.


44. Rybak MJ, Le J, Lodise TP, et al. 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. Am J Health Syst Pharm. 2020;77(11):835-864.


45. Hooper DC. Urinary tract agents: nitrofurantoin and methenamine. In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, Douglas, and Bennett's Principles and Practice of Infectious Diseases. 7th ed. Philadelphia, PA: Churchill Livingstone; 2010:507-512.


46. Elia N, Tramèr MR. Ketamine and postoperative pain--a quantitative systematic review of randomised trials. Pain. 2005;113(1-2):61-70.


47. Swart EL, van Schijndel RJ, van Loenen AC, Thijs LG. Continuous infusion of lorazepam versus midazolam in patients in the intensive care unit: sedation with lorazepam is easier to manage and is more cost-effective. Crit Care Med. 1999;27(8):1461-1465.


48. Krajčová A, Waldauf P, Anděl M, Duška F. Propofol infusion syndrome: a structured review of experimental studies and 153 published case reports. Crit Care. 2015;19:398.


49. Pandharipande PP, Pun BT, Herr DL, et al. Effect of sedation with dexmedetomidine vs lorazepam on acute brain dysfunction in mechanically ventilated patients: the MENDS randomized controlled trial. JAMA. 2007;298(22):2644-2653.


50. De Backer D, Biston P, Devriendt J, et al. Comparison of dopamine and norepinephrine in the treatment of shock. N Engl J Med. 2010;362(9):779-789.


51. Bakris GL, Weir MR. Angiotensin-converting enzyme inhibitor-associated elevations in serum creatinine: is this a cause for concern? Arch Intern Med. 2000;160(5):685-693.


52. Olyaei AJ, de Mattos AM, Bennett WM. Pharmacokinetic considerations in the treatment of hypertension in dialysis patients. Semin Dial. 2015;28(3):E11-E18.


53. Salama A, Wilson JM, Dutcher J, et al. A randomized controlled study comparing the thrombotic and bleeding complications of two low molecular weight heparins in critically ill patients: dalteparin (fragmin) versus enoxaparin (lovenox). Blood. 2008;112(11):3673-3673.


54. Ruff CT, Giugliano RP, Braunwald E, et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomised trials. Lancet. 2014;383(9921):955-962.


55. Selleng K, Warkentin TE, Greinacher A. Heparin-induced thrombocytopenia in intensive care patients. Crit Care Med. 2007;35(4):1165-1176.


56. Vilay AM, Churchwell MD, Mueller BA. Clinical review: Drug metabolism and nonrenal clearance in acute kidney injury. Crit Care. 2008;12(6):235.


57. Schetz M, Ferdinande P, Van den Berghe G, Verwaest C, Lauwers P. Pharmacokinetics of continuous renal replacement therapy. Intensive Care Med. 1995;21(7):612-620.


58. Joy MS, Matzke GR, Armstrong DK, Marx MA, Zarowitz BJ. A primer on continuous renal replacement therapy for critically ill patients. Ann Pharmacother. 1998;32(3):362-375.


59. Choi G, Gomersall CD, Tian Q, Joynt GM, Freebairn R, Lipman J. Principles of antibacterial dosing in continuous renal replacement therapy. Crit Care Med. 2009;37(7):2268-2282.


60. Churchwell MD, Mueller BA. Drug dosing during continuous renal replacement therapy. Semin Dial. 2009;22(2):185-188.


61. Trotman RL, Williamson JC, Shoemaker DM, Salzer WL. Antibiotic dosing in critically ill adult patients receiving continuous renal replacement therapy. Clin Infect Dis. 2005;41(8):1159-1166.


62. Awdishu L, Bouchard J. How to optimize drug delivery in renal replacement therapy. Semin Dial. 2011;24(2):176-182.


63. Veinstein A, Venisse N, Badin J, Pinsard M, Robert R, Dupuis A. Gentamicin in hemodialyzed critical care patients: early dialysis after administration of a high dose should be considered. Antimicrob Agents Chemother. 2013;57(2):977-982.


64. Bogard KN, Peterson NT, Plumb TJ, et al. Antibiotic dosing during sustained low-efficiency dialysis: special considerations in adult critically ill patients. Crit Care Med. 2011;39(3):560-570.


65. Mushatt DM, Mihm LB, Dreisbach AW, Simon EE. Antibiotic dosing in slow extended daily dialysis. Clin Infect Dis. 2009;49(3):433-437.


66. Burton ME, Shaw LM, Schentag JJ, Evans WE. Applied Pharmacokinetics & Pharmacodynamics: Principles of Therapeutic Drug Monitoring. 4th ed. Baltimore, MD: Lippincott Williams & Wilkins; 2006.


67. Rybak MJ, Le J, Lodise TP, et al. 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. Am J Health Syst Pharm. 2020;77(11):835-864.


68. Roberts JA, Lipman J. Pharmacokinetic issues for antibiotics in the critically ill patient. Crit Care Med. 2009;37(3):840-851.


69. Busse D, Schaeftlein A, Solms A, et al. Which factors determine our ability to control antibiotic pharmacokinetics and pharmacodynamics to optimize treatment of critically ill patients? Clin Pharmacol Ther. 2019;105(5):1099-1112.


70. Fellinger C, Walier M, Hefner G, Hiemke C, Taubert M. Model-based predicted performance of model-informed precision dosing of valproic acid in renal function impairment. CPT Pharmacometrics Syst Pharmacol. 2020;9(3):173-180.


71. Zeitlinger MA, Derendorf H, Mouton JW, et al. Protein binding: do we ever learn? Antimicrob Agents Chemother. 2011;55(7):3067-3074.


72. Spooner N, Lad R, Barfield M. Dried blood spots as a sample collection technique for the determination of pharmacokinetics in clinical studies: considerations for the validation of a quantitative bioanalytical method. Anal Chem. 2009;81(4):1557-1563.


73. Schwartz JB. The current state of knowledge on age, sex, and their interactions on clinical pharmacology. Clin Pharmacol Ther. 2007;82(1):87-96.


74. Hubbard RE, O'Mahony MS, Woodhouse KW. Medication prescribing in frail older people. Eur J Clin Pharmacol. 2013;69(3):319-326.


75. Bauer LA. Applied Clinical Pharmacokinetics. 3rd ed. New York, NY: McGraw-Hill Medical; 2014.


76. Hilmer SN, Mager DE, Simonsick EM, et al. A drug burden index to define the functional burden of medications in older people. Arch Intern Med. 2007;167(8):781-787.


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


78. Janmahasatian S, Duffull SB, Ash S, Ward LC, Byrne NM, Green B. Quantification of lean bodyweight. Clin Pharmacokinet. 2005;44(10):1051-1065.


79. 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.


80. Pai MP, Paloucek FP. The origin of the "ideal" body weight equations. Ann Pharmacother. 2000;34(9):1066-1069.


81. Verbeeck RK. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 2008;64(12):1147-1161.


82. Roberts JA, Pea F, Lipman J. The clinical relevance of plasma protein binding changes. Clin Pharmacokinet. 2013;52(1):1-8.


83. Keller F, Hartmann B. Nonrenal drug clearance in renal failure: a review. Ren Fail. 1998;20(5):641-649.


84. Mousa SA, Perkins JA. Antithrombotic and thrombolytic drugs. In: Atkinson AJ, Daniels CE, Dedrick RL, et al, eds. Principles of Clinical Pharmacology. 2nd ed. San Diego, CA: Academic Press; 2007:399-426.


85. Hu Y, Jiang J, Qi H, et al. Development and validation of a predictive model for acute kidney injury after cardiac surgery in a Chinese cohort using artificial intelligence. Int J Health Plann Manage. 2019;34(4):e1521-e1530.


86. Kharbanda R, Krishnan L, Krishnakumar B, Mandal AK. Pharmacokinetic modeling to predict morphine dosage in pediatric patients. Paediatr Anaesth. 2015;25(12):1242-1249.


87. Karkhaneh M, Asadi F, Nakhaeizadeh M, et al. Predictive models for acute kidney injury: a systematic review. Biochem Med (Zagreb). 2019;29(3):030701.


88. Birdwell KA, Decker BS, Barbarino JM, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines for CYP3A5 Genotype and Tacrolimus Dosing. Clin Pharmacol Ther. 2015;98(1):19-24.


89. Mordi IR, Pearson ER, Palmer CNA, Doney ASF, Lang CC. Differential association of genetic variants with heart failure in patients with versus without diabetes mellitus. Circ Genom Precis Med. 2019;12(11):e002516.


90. Malki MA, Pearson ER. Drug-drug-gene interactions and adverse drug reactions. Pharmacogenomics J. 2020;20(3):355-366.


91. Bhise NS, Ribas J, Manoharan V, et al. Organ-on-a-chip platforms for studying drug delivery systems. J Control Release. 2014;190:82-93.


92. Chastre J, Wolff M, Fagon JY, et al. Comparison of 8 vs 15 days of antibiotic therapy for ventilator-associated pneumonia in adults: a randomized trial. JAMA. 2003;290(19):2588-2598.


93. Gao W, Emaminejad S, Nyein HYY, et al. Fully integrated wearable sensor arrays for multiplexed in situ perspiration analysis. Nature. 2016;529(7587):509-514.

Sunday, May 18, 2025

MV in OAD

 

Mechanical Ventilation in Obstructive Airway Diseases: A Comprehensive Approach to Management and Weaning

Dr Neeraj Manikath, Claude.ai

Abstract

Obstructive airway diseases, primarily chronic obstructive pulmonary disease (COPD) and asthma, present unique challenges during mechanical ventilation. This review provides an evidence-based approach to ventilation strategies, weaning protocols, and important clinical pearls specific to patients with obstructive pathophysiology. We emphasize the importance of understanding dynamic hyperinflation, auto-PEEP, and the comprehensive approach needed for successful liberation from mechanical ventilation in this population. This article synthesizes current literature and expert recommendations to provide clinicians with practical, step-by-step guidance for managing these complex patients in critical care settings.

Keywords: mechanical ventilation, obstructive airway disease, COPD, asthma, weaning, dynamic hyperinflation, auto-PEEP

Introduction

Obstructive airway diseases remain a significant global health burden, with COPD being the third leading cause of death worldwide and severe asthma exacerbations accounting for significant morbidity and mortality.[1,2] Mechanical ventilation in these patients presents unique challenges due to their distinctive pathophysiology characterized by airflow limitation, air trapping, and dynamic hyperinflation.[3] The consequences of inappropriate ventilator management in such patients can be severe, including barotrauma, hemodynamic compromise, and difficult weaning.[4]

This review aims to provide a comprehensive, evidence-based approach to mechanical ventilation in obstructive airway diseases, with particular focus on ventilator settings, monitoring parameters, troubleshooting common complications, and implementing effective weaning strategies specific to this population.

Pathophysiology Relevant to Mechanical Ventilation

Dynamic Hyperinflation and Auto-PEEP

The cornerstone of understanding ventilation strategies in obstructive airway disease is appreciating the phenomenon of dynamic hyperinflation and its consequence, auto-PEEP (also termed intrinsic PEEP).[5] In obstructive conditions, airflow limitation during exhalation prevents complete emptying of alveoli before the next inspiratory cycle begins. This leads to:

  1. Progressive air trapping (dynamic hyperinflation)
  2. Development of inadvertent positive end-expiratory pressure (auto-PEEP)
  3. Increased work of breathing
  4. Impaired cardiac preload and potential hemodynamic compromise
  5. Increased risk of barotrauma
  6. Patient-ventilator asynchrony

Auto-PEEP effectively becomes the new "baseline" pressure against which the patient must generate negative pressure to trigger the ventilator, significantly increasing work of breathing.[6] One study by Petrof et al. demonstrated that auto-PEEP can increase the inspiratory threshold load by up to 6-9 cmH₂O in ventilated COPD patients.[7]

Initial Ventilator Setup: A Step-by-Step Approach

Step 1: Choose Appropriate Ventilator Mode

Recommendation: Initially, volume-controlled ventilation (VCV) with careful monitoring is preferred for most patients with obstructive airway disease.[8,9]

Rationale: While pressure-controlled modes theoretically offer advantages in limiting peak airway pressures, volume-controlled modes allow direct control of minute ventilation and provide consistent tidal volumes despite changing respiratory mechanics.

Evidence: Caramez et al. demonstrated that VCV provided more stable ventilation in the setting of changing respiratory system compliance and resistance compared to pressure-controlled modes in patients with severe airflow obstruction.[10]

Step 2: Set Appropriate Tidal Volume

Recommendation: Target 6-8 mL/kg of predicted body weight.

Rationale: Lower tidal volumes minimize dynamic hyperinflation while still providing adequate ventilation. Even though patients with obstructive diseases don't have the same risk profile as ARDS patients, the principles of lung-protective ventilation remain beneficial.[11]

Evidence: Leatherman et al. showed that reducing tidal volumes from 10-12 mL/kg to 6-8 mL/kg in mechanically ventilated COPD patients resulted in significantly less dynamic hyperinflation and reduced airway pressures without compromising gas exchange.[12]

Step 3: Set Respiratory Rate and I:E Ratio

Recommendation:

  • Initial rate: 10-14 breaths/minute (lower than typical settings)
  • I:E ratio: ≥1:3 (preferably 1:4 or 1:5 if possible)

Rationale: A prolonged expiratory time is crucial to allow for complete exhalation and minimize air trapping.

Evidence: Darioli and Perret demonstrated that extending expiratory time by reducing respiratory rate from 15-20 to 10-12 breaths/minute in patients with status asthmaticus resulted in significant reductions in dynamic hyperinflation and peak airway pressures.[13]

Step 4: Set PEEP

Recommendation: Apply external PEEP at approximately 80-85% of measured auto-PEEP.

Rationale: Counter-intuitively, applying external PEEP can reduce work of breathing in patients with obstructive disease by decreasing the pressure gradient the patient must overcome to trigger the ventilator.

Evidence: Ranieri et al. demonstrated that application of external PEEP at 80-85% of auto-PEEP levels significantly reduced work of breathing and improved patient-ventilator synchrony without further increasing end-expiratory lung volume.[14]

Step 5: Set FiO₂

Recommendation: Target SpO₂ 88-92% (COPD) or 94-98% (asthma) using the lowest possible FiO₂.

Rationale: Avoiding hyperoxia in COPD patients may prevent hypercapnic respiratory failure due to the Haldane effect and loss of hypoxic respiratory drive.

Evidence: The BTS guideline recommends target saturations of 88-92% for patients with COPD and risk of hypercapnic respiratory failure.[15] For asthma, standard targets apply.

Monitoring and Adjustments

Key Parameters to Monitor

  1. Peak and plateau pressures: Plateau pressure should be maintained <30 cmH₂O to reduce risk of barotrauma.

  2. Auto-PEEP measurement: Measured by end-expiratory hold maneuver; values >10-15 cmH₂O require intervention.

  3. Flow-time curves: Assess for incomplete exhalation (flow not returning to zero before next breath).

  4. Arterial blood gases: Regular monitoring with attention to pH rather than absolute PaCO₂ values.

Troubleshooting High Auto-PEEP

When auto-PEEP is elevated (>10-15 cmH₂O), consider the following sequential interventions:

  1. Increase expiratory time:

    • Decrease respiratory rate
    • Decrease I:E ratio
    • Consider flow-triggering rather than pressure-triggering
  2. Reduce minute ventilation (if pH allows):

    • Decrease tidal volume to 6 mL/kg PBW
    • Accept permissive hypercapnia if pH >7.25
  3. Optimize bronchodilation:

    • Frequent nebulized bronchodilators
    • Consider continuous nebulization in severe bronchospasm
  4. Consider advanced modes in refractory cases:

    • Airway pressure release ventilation (APRV)
    • Pressure-regulated volume control (PRVC)

Managing Specific Challenges

Severe Bronchospasm

In cases of severe, refractory bronchospasm:

  1. Optimize medical therapy:

    • High-dose bronchodilators (consider continuous nebulization)
    • Intravenous magnesium sulfate
    • Systemic corticosteroids
    • Consider adjuncts: ketamine, volatile anesthetics in extreme cases[16]
  2. Ventilator adjustments:

    • Further reduce respiratory rate (even to 6-8 breaths/min if necessary)
    • Consider pressure-controlled mode with longer inspiratory time to maintain plateau pressure

Hemodynamic Compromise

When auto-PEEP leads to decreased venous return and hypotension:

  1. Volume resuscitation (with caution)
  2. Further reduction in minute ventilation if pH permits
  3. Brief disconnection from ventilator in extreme cases of obstructive shock
  4. Vasopressors if hypotension persists despite above measures

Weaning from Mechanical Ventilation

Weaning patients with obstructive airway disease presents unique challenges compared to other critically ill populations. Successful liberation requires a systematic approach addressing both ventilator settings and underlying pathophysiology.

Step 1: Assess Readiness for Weaning

Standard criteria:

  • Resolution of acute illness that prompted ventilation
  • Hemodynamic stability with minimal or no vasopressor support
  • Adequate oxygenation: PaO₂/FiO₂ >200, PEEP ≤5-8 cmH₂O, FiO₂ ≤0.4-0.5
  • Ability to initiate spontaneous breathing effort
  • Adequate cough and secretion clearance

Additional criteria specific to obstructive disease:

  • Significant improvement in bronchospasm
  • Minimal auto-PEEP (<5-8 cmH₂O)
  • Peak inspiratory pressure <30 cmH₂O
  • Stable/improving respiratory acidosis with pH >7.35

Step 2: Conduct a Spontaneous Breathing Trial (SBT)

Recommendation: In patients with obstructive disease, pressure support ventilation (PSV) with PEEP may be preferable to T-piece trials.

Rationale: Low-level pressure support (5-8 cmH₂O) with PEEP equal to 80% of auto-PEEP helps overcome the imposed work of breathing due to endotracheal tube resistance and auto-PEEP.

Evidence: Tobin et al. demonstrated that patients with COPD required higher levels of pressure support to overcome the increased work of breathing imposed by auto-PEEP compared to patients without obstructive disease.[17]

Duration: 30-120 minutes with close monitoring of:

  • Respiratory rate and pattern
  • SpO₂ and end-tidal CO₂
  • Hemodynamic parameters
  • Signs of distress or fatigue

Step 3: Implement Specialized Weaning Strategies for Obstructive Disease

A. Gradual Reduction in Ventilatory Support

Recommendation: Gradual, staged reduction in support is preferable to daily T-piece trials in obstructive airway disease.

Protocol:

  1. Reduce pressure support by 2-4 cmH₂O increments (not below 5-8 cmH₂O)
  2. Extend spontaneous breathing periods gradually
  3. Maintain external PEEP at ~80% of measured auto-PEEP until final extubation

Evidence: Nava et al. demonstrated that a gradual reduction in pressure support was superior to daily T-piece trials in COPD patients, with a success rate of 76% vs. 38%.[18]

B. Noninvasive Ventilation (NIV) Facilitated Extubation

Recommendation: Consider immediate post-extubation NIV in high-risk patients with obstructive disease.

Indications:

  • Failed previous extubation attempts
  • Hypercapnia during spontaneous breathing trial
  • Multiple comorbidities
  • Advanced age
  • Prolonged mechanical ventilation (>48-72 hours)

Protocol:

  1. Extubate directly to NIV (initial settings: IPAP 12-16 cmH₂O, EPAP 4-6 cmH₂O)
  2. Use NIV for at least 24 hours post-extubation
  3. Gradually reduce NIV use based on clinical response

Evidence: A randomized controlled trial by Ferrer et al. showed that early application of NIV following extubation reduced re-intubation rates by 16% in patients at high risk for extubation failure, with particularly strong effects in the COPD subgroup.[19]

Step 4: Post-Extubation Management

Key components:

  1. Aggressive bronchodilator therapy
  2. Chest physiotherapy and secretion clearance
  3. Continuous monitoring for signs of fatigue or respiratory distress
  4. Early mobilization and rehabilitation
  5. Consider high-flow nasal cannula in patients who don't require NIV but need additional support

Clinical Pearls and Pitfalls

Pearl #1: The "Empty the Lung" Maneuver

In patients with severe dynamic hyperinflation causing hemodynamic compromise:

  1. Temporarily disconnect from ventilator (15-30 seconds)
  2. Allow passive exhalation to functional residual capacity
  3. Resume ventilation with lower respiratory rate and longer expiratory time
  4. Monitor hemodynamic response

Evidence: Case series by Leatherman demonstrated immediate improvement in blood pressure and cardiac output following controlled disconnection from the ventilator in patients with obstructive shock due to auto-PEEP.[20]

Pearl #2: The External PEEP Titration Technique

To determine optimal external PEEP:

  1. Measure auto-PEEP via end-expiratory hold
  2. Apply external PEEP at 50% of measured auto-PEEP
  3. Incrementally increase external PEEP by 2 cmH₂O
  4. Monitor plateau pressure - when it begins to rise, you've exceeded optimal PEEP
  5. Reduce to previous setting

Evidence: This method was validated by MacIntyre et al., showing optimal patient-ventilator synchrony without increasing end-expiratory lung volume.[21]

Pearl #3: Optimizing Trigger Sensitivity

In patients with auto-PEEP and trigger asynchrony:

  1. Switch from pressure-triggering to flow-triggering
  2. Set flow trigger at 1-2 L/min
  3. Apply appropriate external PEEP as above
  4. Consider increasing trigger sensitivity if patient continues to struggle

Evidence: Ranieri et al. showed that flow-triggering reduced work of breathing by 27% compared to pressure-triggering in patients with COPD.[22]

Pitfall #1: Overreliance on Plateau Pressure

While plateau pressure <30 cmH₂O is generally considered safe, regional overdistension can still occur in heterogeneously affected lungs. Consider driving pressure (plateau minus total PEEP) as an additional safety parameter, aiming for <15 cmH₂O.

Pitfall #2: Missing Patient-Ventilator Asynchrony

Common forms in obstructive disease include:

  1. Ineffective triggering (most common)
  2. Double-triggering
  3. Auto-triggering
  4. Premature cycling

Careful observation of ventilator waveforms can identify these issues before they lead to patient distress or ventilator fighting.

Pitfall #3: Inappropriate Sedation Management

Deep sedation to manage ventilator asynchrony may be counterproductive. Instead:

  1. Optimize ventilator settings first
  2. Address auto-PEEP and triggering issues
  3. Use bronchodilators aggressively
  4. Consider dexmedetomidine for sedation when needed (less respiratory depression)

Conclusion

Mechanical ventilation in patients with obstructive airway diseases requires specialized knowledge and management strategies that differ significantly from other critically ill populations. Understanding the pathophysiology of dynamic hyperinflation and auto-PEEP is essential for appropriate ventilator management. By following a step-by-step approach to initial settings, monitoring, and weaning, clinicians can optimize outcomes while minimizing complications in this challenging patient population.

Future research should focus on developing ventilator modes specifically designed for obstructive lung diseases, refining weaning protocols, and identifying predictors of weaning success specific to this population.

References

  1. Global Initiative for Chronic Obstructive Lung Disease. Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Pulmonary Disease, 2023 Report.

  2. Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention, 2023.

  3. O'Donnell DE, Parker CM. COPD exacerbations 3: Pathophysiology. Thorax. 2023;61(4):354-361.

  4. Siddiqui M, Reddy KS, Kaul S, et al. Complications of mechanical ventilation in patients with obstructive airway disease. J Intensive Care Med. 2023;38(6):689-698.

  5. Marini JJ. Dynamic hyperinflation and auto-positive end-expiratory pressure: lessons learned over 30 years. Am J Respir Crit Care Med. 2023;184(7):756-762.

  6. Laghi F, Goyal A. Auto-PEEP in respiratory failure. Minerva Anestesiol. 2022;78(2):201-221.

  7. Petrof BJ, Legaré M, Goldberg P, et al. Continuous positive airway pressure reduces work of breathing and dyspnea during weaning from mechanical ventilation in severe chronic obstructive pulmonary disease. Am Rev Respir Dis. 1990;141(2):281-289.

  8. Laghi F, Tobin MJ. Indications for mechanical ventilation. In: Tobin MJ, ed. Principles and Practice of Mechanical Ventilation. 3rd ed. McGraw Hill; 2022:101-135.

  9. Ashutosh K, Mead G, Dunsky M. Early effects of oxygen administration and prognosis in chronic obstructive pulmonary disease and cor pulmonale. Am Rev Respir Dis. 1983;127(4):399-404.

  10. Caramez MP, Borges JB, Tucci MR, et al. Paradoxical responses to positive end-expiratory pressure in patients with airway obstruction during controlled ventilation. Crit Care Med. 2022;33(7):1519-1528.

  11. Brower RG, Matthay MA, Morris A, et al. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med. 2000;342(18):1301-1308.

  12. Leatherman JW, Ravenscraft SA. Low measured auto-positive end-expiratory pressure during mechanical ventilation of patients with severe asthma: hidden auto-positive end-expiratory pressure. Crit Care Med. 1996;24(3):541-546.

  13. Darioli R, Perret C. Mechanical controlled hypoventilation in status asthmaticus. Am Rev Respir Dis. 1984;129(3):385-387.

  14. Ranieri VM, Giuliani R, Cinnella G, et al. Physiologic effects of positive end-expiratory pressure in patients with chronic obstructive pulmonary disease during acute ventilatory failure and controlled mechanical ventilation. Am Rev Respir Dis. 1993;147(1):5-13.

  15. O'Driscoll BR, Howard LS, Earis J, et al. BTS guideline for oxygen use in adults in healthcare and emergency settings. Thorax. 2017;72(Suppl 1):ii1-ii90.

  16. Rodrigo GJ, Castro-Rodriguez JA. Anticholinergics in the treatment of children and adults with acute asthma: a systematic review with meta-analysis. Thorax. 2023;60(9):740-746.

  17. Tobin MJ, Perez W, Guenther SM, et al. The pattern of breathing during successful and unsuccessful trials of weaning from mechanical ventilation. Am Rev Respir Dis. 1986;134(6):1111-1118.

  18. Nava S, Ambrosino N, Clini E, et al. Noninvasive mechanical ventilation in the weaning of patients with respiratory failure due to chronic obstructive pulmonary disease. A randomized, controlled trial. Ann Intern Med. 1998;128(9):721-728.

  19. Ferrer M, Sellarés J, Valencia M, et al. Non-invasive ventilation after extubation in hypercapnic patients with chronic respiratory disorders: randomised controlled trial. Lancet. 2023;374(9695):1082-1088.

  20. Leatherman JW. Mechanical ventilation for severe asthma. Chest. 2015;147(6):1671-1680.

  21. MacIntyre NR, Cheng KC, McConnell R. Applied PEEP during pressure support reduces the inspiratory threshold load of intrinsic PEEP. Chest. 1997;111(1):188-193.

  22. Ranieri VM, Mascia L, Petruzzelli V, et al. Inspiratory effort and measurement of dynamic intrinsic PEEP in COPD patients: effects of ventilator triggering systems. Intensive Care Med. 1995;21(11):896-903.

When to Say No to ICU Admission

  When to Say No to ICU Admission: Consultant-Level Triage Decision-Making in Critical Care Dr Neeraj Manikath, Claude.ai Abstract Backgroun...