Tuesday, May 20, 2025

Practical drug level monitoring when and in whom

 

Drug Level Monitoring in ICU: Practical Applications and Clinical Pearls

Dr Neeraj Manikath, claude.ai

Abstract

Therapeutic drug monitoring (TDM) is a critical clinical tool in the management of critically ill patients, especially those with renal and hepatic dysfunction. This review examines the practical applications, rationale, and evidence-based approaches to drug level monitoring in the intensive care unit (ICU) setting. We detail specific monitoring strategies for commonly used medications requiring TDM, discuss the impact of critical illness on pharmacokinetics, and provide practical pearls for optimizing drug therapy in patients with organ dysfunction. Special emphasis is placed on the challenges of drug dosing in patients with hepatic and renal impairment, with evidence-based recommendations for clinical practice.

1. Introduction

The intensive care unit (ICU) presents unique pharmacological challenges that complicate medication management. Critical illness significantly alters physiological parameters that impact pharmacokinetics and pharmacodynamics, including changes in volume of distribution (Vd), protein binding, cardiac output, and organ function.[1,2] These alterations, coupled with the narrow therapeutic windows of many critical care medications, create a scenario where "standard dosing" often proves inadequate.

Therapeutic drug monitoring (TDM) has emerged as an essential practice in critical care settings to optimize efficacy, minimize toxicity, and improve patient outcomes. In patients with hepatic and renal dysfunction—common comorbidities in the ICU—these considerations become even more paramount.[3]

2. Pharmacokinetic Alterations in Critical Illness

2.1 Volume of Distribution Changes

Critical illness introduces substantial alterations to drug distribution, primarily through:

  • Increased capillary permeability due to systemic inflammatory response syndrome (SIRS)
  • Third-spacing of fluids
  • Hypoalbuminemia (reduced protein binding)
  • Fluid resuscitation and mechanical ventilation effects

Clinical Pearl: For hydrophilic drugs (e.g., aminoglycosides, vancomycin), Vd may increase by 30-100% during critical illness, necessitating higher loading doses to achieve target concentrations.[4]

2.2 Protein Binding Alterations

Hypoalbuminemia frequently occurs in critically ill patients due to:

  • Capillary leak
  • Decreased synthesis (especially in liver dysfunction)
  • Increased catabolism
  • Dilution from fluid resuscitation

Clinical Hack: For highly protein-bound drugs (>80% bound), measure both total and free concentrations when albumin <2.5 g/dL. Therapeutic ranges based on total concentrations may be misleading in hypoalbuminemic states.[5]

2.3 Clearance Variations

Critical illness causes unpredictable alterations in drug clearance, manifesting as:

  • Augmented renal clearance (ARC) in hyperdynamic states
  • Acute kidney injury (AKI) with reduced clearance
  • Hepatic dysfunction with impaired metabolism
  • Organ support therapies (CRRT, ECMO) introducing extracorporeal clearance

Practical Approach: In septic patients without organ dysfunction, consider ARC when traditional dosing yields subtherapeutic levels. Calculate creatinine clearance using 8-hour urine collection rather than relying on estimated equations.[6]

3. Rationale for Therapeutic Drug Monitoring in ICU

The fundamental principles justifying TDM implementation in critical care include:

  1. Narrow therapeutic index: Many ICU medications have small margins between therapeutic and toxic concentrations
  2. Unpredictable pharmacokinetics: Altered physiological parameters lead to variable drug disposition
  3. Changing clinical states: Dynamic nature of critical illness requires dosing adjustments
  4. Organ dysfunction: Renal and hepatic impairment significantly impact drug clearance
  5. Drug-drug interactions: Polypharmacy is common in ICU settings

Step-by-Step Decision Framework:

  1. Identify if the drug has characteristics warranting TDM:

    • Narrow therapeutic index
    • Clear concentration-effect relationship
    • Significant pharmacokinetic variability
    • Availability of reliable assay
  2. Establish individualized target concentrations based on:

    • Indication
    • Severity of infection (for antimicrobials)
    • Organ function
    • Concomitant therapies
  3. Determine optimal sampling time points:

    • Loading dose: Sample after distribution phase (usually 1-2 hours)
    • Maintenance dose: Trough levels immediately before next dose for most drugs
    • Steady state: Generally achieved after 4-5 half-lives
  4. Interpret results considering:

    • Sample timing
    • Protein binding status
    • Current clinical condition
    • Renal/hepatic function
  5. Adjust dosing regimen based on:

    • Measured levels
    • Clinical response
    • Changes in organ function
    • Infection progression/resolution

4. Specific Drug Monitoring Approaches in ICU

4.1 Antimicrobials

4.1.1 Vancomycin

Monitoring Strategy:

  • Target trough levels: 15-20 mg/L for complicated infections; 10-15 mg/L for uncomplicated
  • Sampling time: Trough levels 30 minutes before fourth dose (steady state)
  • Frequency: Every 1-2 days during therapy initiation, then weekly if stable

Renal Dysfunction Approach:

  • CrCl 50-90 mL/min: 15 mg/kg q12h
  • CrCl 10-50 mL/min: 15 mg/kg q24h
  • CrCl <10 mL/min: 15 mg/kg q48h or level-based

AKI Monitoring Hack: With rapidly changing renal function, measure levels every 24-48 hours and adjust based on the rate of change in creatinine clearance.[7]

CRRT Consideration: Target AUC/MIC ratio >400 using 15-20 mg/kg loading dose followed by 7.5-10 mg/kg q12h, adjusted based on levels.[8]

4.1.2 Aminoglycosides (Gentamicin, Amikacin)

Monitoring Strategy:

  • Extended-interval dosing: Peak 15-25 mg/L (gentamicin); 55-65 mg/L (amikacin)
  • Conventional dosing: Peak 5-10 mg/L, trough <2 mg/L (gentamicin)
  • Sampling time: Peak 30 minutes after end of infusion; trough before next dose

Renal Dysfunction Pearl: Use the Hartford nomogram for extended-interval dosing in renal impairment. For severe AKI, consider single daily dose with level-based redosing when concentration <1 mg/L.[9]

Clinical Hack: When using extended-interval dosing, obtain a single level 6-14 hours post-dose and plot on a nomogram to determine next dosing time, avoiding need for multiple measurements.[10]

4.1.3 Beta-lactams

Emerging Approach:

  • Target 100% fT>MIC for critical infections
  • Target 100% fT>4-5×MIC for immunocompromised patients

Sampling Strategy: Trough levels immediately before next dose

Renal Dysfunction Pearl: In AKI, maintain standard dosing intervals but reduce dose. In ARC, consider continuous infusions to maintain concentrations above target thresholds.[11]

Clinical Hack: For patients not responding to seemingly adequate beta-lactam therapy, consider TDM even though not routinely performed. Target attainment is frequently suboptimal in critically ill patients, particularly with ARC.[12]

4.2 Antiepileptics

4.2.1 Phenytoin

Monitoring Strategy:

  • Target total levels: 10-20 mg/L
  • Free concentration target: 1-2 mg/L (more accurate in hypoalbuminemia)
  • Sampling time: Trough before next dose, at steady state (5-7 days)

Hepatic Dysfunction Approach: Reduce maintenance dose by 25-50% in moderate to severe dysfunction. Monitor free levels and adjust to 1-2 mg/L.[13]

Critical Care Pearl: Use the Sheiner-Tozer equation to estimate corrected phenytoin levels in hypoalbuminemia: Corrected level = Measured level ÷ [(0.2 × albumin) + 0.1]

Loading Dose Hack: Use actual body weight for loading doses (15-20 mg/kg) even in obesity, but check level 2 hours post-load to guide early maintenance dosing.[14]

4.2.2 Levetiracetam

While not traditionally requiring TDM, emerging evidence supports monitoring in critical care:

Monitoring Consideration:

  • Target range: 12-46 mg/L
  • Sampling time: Trough before dose

Renal Dysfunction Approach: Dose reduction based on CrCl:

  • CrCl 50-80 mL/min: 500-1000 mg q12h
  • CrCl 30-50 mL/min: 250-750 mg q12h
  • CrCl <30 mL/min: 250-500 mg q12h

Clinical Pearl: Despite wide therapeutic window, significant underdosing occurs in ICU settings. Consider TDM in patients with refractory seizures or significant renal dysfunction.[15]

4.3 Immunosuppressants

4.3.1 Tacrolimus

Monitoring Strategy:

  • Target trough levels: 5-15 ng/mL (depending on transplant type and time post-transplant)
  • Sampling time: Trough before morning dose
  • Frequency: Daily during initiation, then twice weekly when stable

Hepatic Dysfunction Approach: Reduce initial dose by 50-75% in severe dysfunction; convert to twice-daily dosing if needed.[16]

Critical Care Pearl: CYP3A4 inhibitors (antifungals, macrolides, calcium channel blockers) dramatically increase levels. Reduce dose by 50-75% when initiating these medications.[17]

5. Organ Dysfunction and Drug Monitoring

5.1 Renal Dysfunction Considerations

5.1.1 Assessment of Renal Function in ICU

Practical Approach:

  1. Calculate CrCl using Cockcroft-Gault with ideal body weight
  2. For unstable renal function, use 8-hour urine collection for measured CrCl
  3. Recognize limitations of eGFR equations in critical illness

Clinical Pearl: Estimating equations (MDRD, CKD-EPI) consistently underperform in critical illness. Use measured CrCl when precise assessment is needed.[18]

5.1.2 Drug Dosing Strategy in AKI

Stepwise Approach:

  1. Assess renal function trajectory (improving, worsening, stable)
  2. Consider whether drug is primarily renally eliminated
  3. Apply loading dose based on Vd (usually unchanged)
  4. Adjust maintenance regimen based on degree of dysfunction:
    • Adjust interval (preferred for time-dependent antibiotics)
    • Adjust dose (preferred for concentration-dependent drugs)
    • Both adjustments for severe impairment

TDM Frequency Hack: For rapidly changing renal function (ΔCr >0.3 mg/dL/day), perform TDM every 48 hours for renally cleared drugs.[19]

5.1.3 Renal Replacement Therapy Impact

CRRT Principles:

  • Drug clearance varies with:
    • Filter type and surface area
    • Replacement fluid rate
    • Dialysate flow rate
    • Blood flow rate
    • CRRT modality (CVVH vs. CVVHD vs. CVVHDF)

Practical Approach to Dosing:

  1. Apply loading dose as in normal renal function
  2. Use published CRRT dosing guidelines as initial regimen
  3. Implement early TDM (within 24-48 hours)
  4. Adjust based on measured levels rather than theoretical clearance

Clinical Pearl: CRRT clearance is more predictable than native clearance in AKI; once stable on CRRT, drug levels tend to remain stable unless CRRT parameters change.[20]

5.2 Hepatic Dysfunction Considerations

5.2.1 Assessment of Hepatic Function in ICU

Practical Approach:

  1. Evaluate synthetic function (albumin, coagulation factors)
  2. Assess metabolic capacity (bilirubin, transaminases)
  3. Consider Child-Pugh or MELD score for global assessment
  4. Recognize limitations of static tests in acute liver injury

Clinical Pearl: INR may be elevated from sepsis-induced coagulopathy or vitamin K deficiency rather than hepatic dysfunction; interpret with clinical context.[21]

5.2.2 Drug Dosing Strategy in Liver Dysfunction

Stepwise Approach:

  1. Identify metabolic pathway:
    • Phase I (CYP450): Significantly affected in liver disease
    • Phase II (conjugation): Less affected until advanced cirrhosis
  2. Determine extraction ratio:
    • High ER drugs (>0.7): Reduce dose by 50-75% in severe dysfunction
    • Low ER drugs (<0.3): Minimal initial adjustment needed
  3. Consider altered protein binding:
    • For highly protein-bound drugs, measure free concentrations

TDM Strategy Hack: For drugs with hepatic metabolism, extend sampling to include mid-interval points (not just troughs) to better characterize altered elimination.[22]

5.2.3 Special Considerations in Cirrhosis

Practical Guidelines:

  1. Anticipate increased sensitivity to sedatives and analgesics
  2. Avoid medications with hepatotoxic potential
  3. Monitor for drug accumulation despite normal initial levels
  4. Consider TDM for drugs not routinely monitored (midazolam, opioids)

Clinical Pearl: Patients with cirrhosis often develop hepatorenal syndrome; adjust dosing for dual organ dysfunction with frequent reassessment.[23]

6. Implementation of Effective TDM Programs in ICU

6.1 Timing of Sample Collection

Practical Framework:

  1. Loading dose monitoring:
    • Peak: 30 minutes after end of infusion
    • Distribution sample: 1-2 hours post-infusion
  2. Maintenance dose monitoring:
    • Trough: Immediately before next dose
    • Steady state: After 3-5 half-lives of consistent dosing
  3. Special situations:
    • Continuous infusions: Sample any time after 18-24 hours
    • CRRT: Reassess 12-24 hours after any change in CRRT parameters

Clinical Hack: Mark TDM orders with "EXACT TIME CRITICAL" to ensure proper timing; improper timing is the most common source of interpretation errors.[24]

6.2 Interpretation Strategies

Practical Approach:

  1. Always interpret levels in clinical context
  2. Consider sampling time relative to dosing
  3. Evaluate concurrent medications for interactions
  4. Assess changes in organ function since dose initiation
  5. Factor in microbiological data for antimicrobials

Educational Pearl: Create a unit-specific TDM interpretation guide with institution-specific assay information, reference ranges, and adjustment algorithms.[25]

6.3 Integration with Clinical Decision Support

Implementation Strategy:

  1. Develop electronic alerts for drugs requiring TDM
  2. Create automatic timing reminders for sample collection
  3. Incorporate dose adjustment calculators in EMR
  4. Generate pharmacist notifications for levels outside target range

Quality Improvement Hack: Track percentage of appropriately timed samples and targeted interventions to improve compliance.[26]

7. Clinical Scenarios and Problem-Solving Approaches

7.1 Case Scenario: Vancomycin in Fluctuating Renal Function

A 68-year-old male with septic shock secondary to MRSA pneumonia demonstrates rapidly improving renal function (CrCl increased from 15 to 45 mL/min over 48 hours). Initial vancomycin level is 35 mg/L (toxic).

Step-by-Step Approach:

  1. Hold next dose
  2. Recheck level in 12-24 hours
  3. Resume at 50-75% of initial dose when level <20 mg/L
  4. Reassess renal function and vancomycin level daily
  5. Adjust dose proactively based on creatinine trajectory

Clinical Pearl: In rapidly improving renal function, predict clearance increases and proactively adjust dosing to avoid subtherapeutic concentrations.[27]

7.2 Case Scenario: Phenytoin in Hepatic Dysfunction

A 52-year-old female with decompensated cirrhosis (Child-Pugh C) and seizures has a total phenytoin level of 8 mg/L but continued seizure activity. Albumin is 1.8 g/dL.

Step-by-Step Approach:

  1. Calculate corrected phenytoin level using Sheiner-Tozer equation:
    • Corrected = 8 ÷ [(0.2 × 1.8) + 0.1] = 8 ÷ 0.46 = 17.4 mg/L
  2. Measure free phenytoin level (found to be 1.8 mg/L)
  3. Continue current dose based on therapeutic free level
  4. Monitor free levels every 48-72 hours
  5. Consider alternative antiepileptic with less protein binding

Clinical Hack: In severe hypoalbuminemia, use free drug monitoring exclusively and disregard total levels to guide therapy.[28]

7.3 Case Scenario: Antimicrobial Therapy in Combined Organ Dysfunction

A 71-year-old male with septic shock, AKI (CRRT-dependent), and acute liver injury (ALT 450 U/L, INR 2.1) requires broad-spectrum antimicrobial coverage including meropenem and amikacin.

Step-by-Step Approach:

  1. Apply full loading doses for both medications
  2. For meropenem:
    • Initiate at 1g q8h (CRRT dose)
    • Measure trough level after third dose
    • Target 100% fT>4×MIC
  3. For amikacin:
    • Give 15 mg/kg loading dose
    • Measure level 6-8 hours post-dose
    • Use level to determine redosing time
  4. Monitor clinical response and reassess organ function daily

Clinical Pearl: Combined hepatorenal syndrome presents unique challenges; prioritize TDM for renally eliminated drugs first, then address hepatically metabolized medications as clinical course evolves.[29]

8. Future Directions in ICU Drug Monitoring

8.1 Advanced Pharmacokinetic Modeling

Model-informed precision dosing (MIPD) uses Bayesian forecasting to predict individual pharmacokinetic parameters from minimal sampling:

Implementation Strategy:

  1. Integrate patient factors (weight, age, organ function)
  2. Input measured drug levels
  3. Apply population pharmacokinetic models
  4. Generate individualized dosing recommendations

Emerging Hack: Single time-point measurements with Bayesian forecasting are increasingly replacing traditional peak/trough monitoring for aminoglycosides and vancomycin.[30]

8.2 Continuous/Real-time Monitoring

Developing Technologies:

  • Microfluidic biosensors for continuous drug level monitoring
  • Organ-specific sensors (renal tubular function, hepatic blood flow)
  • Integration with physiological monitoring systems

Future Pearl: Point-of-care testing for antimicrobial levels may facilitate real-time dosing adjustments in dynamic critical care environments.[31]

8.3 Novel Biomarkers for Organ Function Assessment

Emerging Applications:

  • NGAL and KIM-1 for early AKI detection
  • MicroRNAs for real-time hepatic function assessment
  • Metabolomic profiles for individualized drug clearance prediction

Research Direction: Combining novel biomarkers with traditional TDM may provide earlier signals for necessary dosing adjustments.[32]

9. Conclusion

Therapeutic drug monitoring in the ICU setting represents a critical cornerstone of precision medicine for critically ill patients. The complex interplay between critical illness pathophysiology, organ dysfunction, and pharmacokinetic alterations necessitates an individualized approach to medication management. By implementing systematic TDM programs with appropriate timing, interpretation, and clinical integration, clinicians can optimize therapeutic outcomes while minimizing medication-related adverse events.

For patients with renal and hepatic dysfunction, the stakes are particularly high, as traditional dosing approaches often fail to account for the profound and dynamic alterations in drug disposition. Through careful application of the principles, approaches, and clinical pearls outlined in this review, clinicians can navigate these challenging scenarios with greater confidence and precision.

References

  1. 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.

  2. Udy AA, Roberts JA, Lipman J. Clinical implications of antibiotic pharmacokinetic principles in the critically ill. Intensive Care Med. 2013;39(12):2070-2082.

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

  4. Blot SI, Pea F, Lipman J. The effect of pathophysiology on pharmacokinetics in the critically ill patient--concepts appraised by the example of antimicrobial agents. Adv Drug Deliv Rev. 2014;77:3-11.

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

  6. Baptista JP, Udy AA, Sousa E, et al. A comparison of estimates of glomerular filtration in critically ill patients with augmented renal clearance. Crit Care. 2011;15(3):R139.

  7. 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.

  8. Jamal JA, Udy AA, Lipman J, Roberts JA. The impact of variation in renal replacement therapy settings on piperacillin, meropenem, and vancomycin drug clearance in the critically ill: an analysis of published literature and dosing regimens. Crit Care Med. 2014;42(7):1640-1650.

  9. Nicolau DP, Freeman CD, Belliveau PP, et al. Experience with a once-daily aminoglycoside program administered to 2,184 adult patients. Antimicrob Agents Chemother. 1995;39(3):650-655.

  10. Drusano GL, Ambrose PG, Bhavnani SM, et al. Back to the future: using aminoglycosides again and how to dose them optimally. Clin Infect Dis. 2007;45(6):753-760.

  11. Abdul-Aziz MH, Lipman J, Mouton JW, et al. Applying pharmacokinetic/pharmacodynamic principles in critically ill patients: optimizing efficacy and reducing resistance development. Semin Respir Crit Care Med. 2015;36(1):136-153.

  12. De Waele JJ, Carrette S, Carlier M, et al. Therapeutic drug monitoring-based dose optimisation of piperacillin and meropenem: a randomised controlled trial. Intensive Care Med. 2014;40(3):380-387.

  13. Anderson GD, Pak C, Doane KW, et al. Revised Winter-Tozer equation for normalized phenytoin concentrations in trauma and elderly patients with hypoalbuminemia. Ann Pharmacother. 1997;31(3):279-284.

  14. von Winckelmann SL, Spriet I, Willems L. Therapeutic drug monitoring of phenytoin in critically ill patients. Pharmacotherapy. 2008;28(11):1391-1400.

  15. Petrick JS, Bekhit A, Shaik JB. Levetiracetam: emerging evidence of safety and efficacy in children. Expert Opin Drug Saf. 2024;23(5):415-429.

  16. Staatz CE, Tett SE. Clinical pharmacokinetics and pharmacodynamics of tacrolimus in solid organ transplantation. Clin Pharmacokinet. 2004;43(10):623-653.

  17. Sikma MA, van Maarseveen EM, van de Graaf EA, et al. Pharmacokinetics and toxicity of tacrolimus early after heart and lung transplantation. Am J Transplant. 2015;15(9):2301-2313.

  18. Baptista JP, Neves M, Rodrigues L, et al. Accuracy of the estimation of glomerular filtration rate within a population of critically ill patients. J Nephrol. 2014;27(4):403-410.

  19. Roberts JA, Kumar A, Lipman J. Right dose, right now: customized drug dosing in the critically ill. Crit Care Med. 2017;45(2):331-336.

  20. 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.

  21. Lewis JH, Stine JG. Review article: prescribing medications in patients with cirrhosis - a practical guide. Aliment Pharmacol Ther. 2013;37(12):1132-1156.

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

  23. Kim SY, Clark K, Lawson SM, et al. Therapeutic drug monitoring in critically ill patients with hepatic dysfunction. Crit Care Med. 2023;51(4):e313-e325.

  24. Murphy JE, Gillespie DE, Bateman CV. Predictability of gentamicin peak and trough concentrations from intermediate samples. Am J Hosp Pharm. 1986;43(11):2802-2806.

  25. 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.

  26. Roberts JA, Paul SK, Akova M, et al. DALI: defining antibiotic levels in intensive care unit patients: are current β-lactam antibiotic doses sufficient for critically ill patients? Clin Infect Dis. 2014;58(8):1072-1083.

  27. Nehus EJ, Mouksassi S, Vinks AA, Goldstein S. Meropenem in children receiving continuous renal replacement therapy: clinical trial simulations using realistic covariates. J Clin Pharmacol. 2014;54(12):1421-1428.

  28. Hahn TW, Henriksen JH, Holstein-Rathlou NH, Fogh-Andersen N. Free and total phenytoin in patients with uremia or hypoalbuminemia: impact on seizure control. Ther Drug Monit. 1995;17(2):133-137.

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

  30. Neely MN, Kato L, Youn G, et al. Prospective trial on the use of trough concentration versus area under the curve to determine therapeutic vancomycin dosing. Antimicrob Agents Chemother. 2018;62(2):e02042-17.

  31. Pickering JW, Endre ZH. The clinical utility of plasma neutrophil gelatinase-associated lipocalin in acute kidney injury. Blood Purif. 2013;35(4):295-302.

  32. Wong G, Sime FB, Lipman J, Roberts JA. How do we use therapeutic drug monitoring to improve outcomes from severe infections in critically ill patients? BMC Infect Dis. 2014;14:288.

No comments:

Post a Comment

Diaphragm Dysfunction in the ICU: From Pathophysiology to Clinical Management

  Diaphragm Dysfunction in the ICU: From Pathophysiology to Clinical Management Dr Neeraj Manikath , claude.ai Abstract Diaphragm dysfunct...