Wednesday, August 13, 2025

Timing of Tracheostomy in Mechanically Ventilated Patients

 

Timing of Tracheostomy in Mechanically Ventilated Patients: A Critical Review for the Modern Intensivist

Dr Neeraj Manikath , claude.ai

Abstract

Background: The optimal timing of tracheostomy in mechanically ventilated patients remains one of the most debated topics in critical care medicine. Despite decades of research, the question of early versus late tracheostomy continues to challenge clinicians worldwide.

Objective: This review synthesizes current evidence on tracheostomy timing, technique selection, and impact on patient-centered outcomes including delirium, ventilator-associated pneumonia (VAP), and long-term swallowing function.

Methods: Comprehensive review of recent landmark trials including TracMan and SETPOINT2, systematic reviews, and meta-analyses published between 2010-2024.

Key Findings: While early tracheostomy offers certain advantages in terms of patient comfort and potentially reduced sedation requirements, recent high-quality evidence fails to demonstrate significant mortality benefits. The choice between percutaneous and surgical techniques should be individualized based on patient anatomy and institutional expertise.

Conclusions: Timing of tracheostomy should be based on individualized risk-benefit assessment rather than arbitrary time cutoffs, with emphasis on patient-centered outcomes and long-term functional recovery.

Keywords: Tracheostomy, mechanical ventilation, critical care, timing, percutaneous, surgical technique


Introduction

Tracheostomy represents one of the oldest surgical procedures in medicine, yet its optimal timing in critically ill patients continues to generate substantial controversy. The fundamental question—when to transition from translaryngeal intubation to tracheostomy—impacts millions of patients annually in intensive care units worldwide. This decision carries profound implications for patient comfort, resource utilization, and long-term functional outcomes.

The traditional paradigm suggesting tracheostomy after 14-21 days of mechanical ventilation has been increasingly challenged by proponents of earlier intervention. The rationale for early tracheostomy appears compelling: reduced sedation requirements, improved patient comfort, enhanced nursing care, and potentially shorter ICU stays. However, the translation of these theoretical benefits into meaningful clinical outcomes has proven more elusive than initially anticipated.


Historical Perspective and Evolution of Practice

The concept of early tracheostomy gained momentum in the late 1980s and 1990s, driven by observational studies suggesting benefits in terms of weaning success and complications. The pendulum swung toward earlier intervention, with many centers adopting policies favoring tracheostomy within 7-10 days of intubation. This shift occurred despite limited high-quality randomized evidence supporting such practices.

The landscape began to change with the publication of several landmark randomized controlled trials in the 2010s, culminating in the TracMan trial in 2013 and more recently, the SETPOINT2 trial in 2021. These studies fundamentally challenged existing assumptions about the benefits of early tracheostomy.


Defining Early versus Late Tracheostomy

Clinical Pearl #1: The definition of "early" versus "late" tracheostomy varies significantly across studies, creating challenges in evidence synthesis. Most contemporary trials define early tracheostomy as ≤7 days and late as >10 days, with an intentional gap to create clear separation between groups.

The arbitrary nature of these cutoffs reflects the complexity of predicting which patients will require prolonged mechanical ventilation. In clinical practice, the decision often involves balancing the certainty of prolonged ventilation against the risks and resource implications of the procedure.


Landmark Trials: TracMan and Beyond

The TracMan Trial (2013)

The Tracheostomy in Mechanically Ventilated Patients (TracMan) trial randomized 909 patients to early tracheostomy (within 4 days) versus late tracheostomy (after 10 days if still ventilated). This pragmatic, multicenter trial fundamentally altered our understanding of tracheostomy timing.

Key Findings:

  • No significant difference in 30-day mortality (30.8% early vs. 31.5% late, p=0.85)
  • No difference in ICU length of stay
  • Reduced sedation requirements in the early group
  • 45% of patients in the late group were successfully extubated without requiring tracheostomy

Clinical Hack #1: The TracMan trial's most important finding may be that 45% of patients allocated to late tracheostomy never received the procedure. This highlights the importance of daily assessment and the difficulty in predicting which patients will require prolonged ventilation.

SETPOINT2 Trial (2021)

The SETPOINT2 trial randomized 382 patients to early percutaneous tracheostomy (≤4 days) versus prolonged intubation with tracheostomy only if required after day 10.

Key Findings:

  • No difference in 60-day mortality (29% early vs. 27% late, p=0.72)
  • Shorter mechanical ventilation duration in early group (median 8 vs. 12 days, p<0.001)
  • No difference in ICU or hospital length of stay
  • 37% of late group patients were successfully extubated without tracheostomy

Oyster Alert #1: Despite shorter ventilation duration in early tracheostomy groups across multiple trials, this rarely translates into reduced ICU or hospital length of stay. This paradox suggests that factors other than mechanical ventilation drive ICU discharge readiness.


Percutaneous versus Surgical Tracheostomy

The technique of tracheostomy performance has evolved significantly, with percutaneous dilatational tracheostomy (PDT) becoming the predominant approach in many ICUs. The question of whether technique influences outcomes remains clinically relevant.

Advantages of Percutaneous Technique:

  • Bedside performance without operating room transfer
  • Reduced resource utilization
  • Lower infection rates in some studies
  • Faster procedure time

Advantages of Surgical Technique:

  • Better visualization and anatomical control
  • Preferred in patients with challenging anatomy
  • Lower risk of loss of airway during procedure
  • More precise stoma placement

Clinical Pearl #2: The choice between percutaneous and surgical tracheostomy should be individualized based on patient factors (obesity, cervical anatomy, coagulopathy) and institutional expertise rather than dogmatic adherence to one technique.

Evidence Synthesis:

Recent meta-analyses suggest minimal differences in major outcomes between techniques when performed by experienced operators. A 2020 Cochrane review of 20 trials (1652 participants) found no significant differences in mortality, major bleeding, or wound infection between techniques.

Clinical Hack #2: Use ultrasound guidance for percutaneous tracheostomy to identify vascular structures and confirm midline positioning. This simple adjunct can significantly reduce complications, especially in patients with challenging anatomy.


Impact on Ventilator-Associated Pneumonia (VAP)

The relationship between tracheostomy timing and VAP represents a complex interplay of multiple factors including oral care, secretion management, and sedation requirements.

Theoretical Benefits:

  • Improved oral hygiene and secretion clearance
  • Reduced need for deep sedation
  • Enhanced mobility and positioning
  • Elimination of oropharyngeal contamination route

Evidence Reality:

The evidence for VAP reduction with early tracheostomy remains inconsistent. While some observational studies suggest benefit, randomized trials have failed to demonstrate consistent VAP reduction with early tracheostomy.

Oyster Alert #2: The microaspiration that occurs around endotracheal tubes may be replaced by aspiration around tracheostomy tubes. The net benefit in terms of pneumonia prevention may be less substantial than previously believed.

A 2021 systematic review and meta-analysis of 12 RCTs found no significant reduction in VAP with early tracheostomy (RR 0.88, 95% CI 0.64-1.19, p=0.40).

Clinical Hack #3: Focus on evidence-based VAP prevention strategies (head elevation, oral care, sedation minimization, spontaneous breathing trials) rather than relying on tracheostomy timing alone for pneumonia prevention.


Delirium and Neuropsychological Outcomes

The impact of tracheostomy on delirium represents an area of growing interest, particularly given the emphasis on patient-centered outcomes and long-term functional recovery.

Mechanisms of Benefit:

  • Reduced sedation requirements
  • Improved patient comfort and communication
  • Enhanced mobility and participation in care
  • Reduced ICU-related stressors

Evidence Base:

Several studies have suggested that early tracheostomy may reduce delirium burden, though the evidence remains limited by methodological challenges in delirium assessment and the multifactorial nature of ICU delirium.

A prospective cohort study by Leung et al. (2020) found that patients receiving tracheostomy within 7 days had fewer delirium-free days compared to those with prolonged intubation, though this finding requires validation in randomized trials.

Clinical Pearl #3: The anti-delirium benefits of tracheostomy likely stem from reduced sedation requirements rather than the procedure itself. Focus on sedation minimization protocols regardless of airway management strategy.

Clinical Hack #4: Implement early mobilization and communication strategies immediately after tracheostomy. The window of opportunity for neurological recovery may be time-sensitive.


Long-term Swallowing Outcomes

The long-term functional consequences of tracheostomy, particularly swallowing function, represent critical patient-centered outcomes often overlooked in short-term studies.

Physiological Impact:

  • Altered laryngeal elevation and closure
  • Reduced subglottic pressure
  • Desensitization of laryngeal reflexes
  • Potential structural damage from surgical trauma

Evidence and Timeline:

Swallowing dysfunction affects 50-80% of patients immediately post-tracheostomy, with gradual improvement over weeks to months. Several factors influence recovery:

  1. Duration of tracheostomy: Longer duration associated with worse outcomes
  2. Cuff deflation timing: Early deflation may improve swallowing recovery
  3. Speaking valve use: May accelerate functional recovery
  4. Systematic speech therapy: Essential for optimal outcomes

Oyster Alert #3: The timing of tracheostomy (early vs. late) may have less impact on swallowing outcomes than the total duration of tracheostomy and the quality of post-procedure rehabilitation.

A longitudinal study by Clayton et al. (2019) following 150 patients for 12 months post-tracheostomy found that 85% achieved functional swallowing by 6 months, with timing of initial tracheostomy showing minimal predictive value.

Clinical Hack #5: Implement systematic swallowing assessment protocols starting 48-72 hours post-tracheostomy, with early involvement of speech-language pathology services.


Clinical Decision-Making Framework

Given the complexity of evidence and individual patient factors, a systematic approach to tracheostomy timing is essential:

Day 1-3: Assessment Phase

  • Evaluate likelihood of prolonged ventilation
  • Consider patient factors (age, comorbidities, functional status)
  • Assess family preferences and goals of care

Day 4-7: Decision Point

  • If high certainty of prolonged ventilation: Consider early tracheostomy
  • If uncertainty remains: Continue translaryngeal intubation with daily reassessment
  • Patient-specific factors may override general timeline

Day 8-14: Late Decision Phase

  • Most patients still intubated likely to benefit from tracheostomy
  • Consider surgical consultation for complex anatomy
  • Reassess goals of care

Clinical Pearl #4: The absence of mortality benefit with early tracheostomy does not negate potential benefits in terms of patient comfort, family interaction, and quality of life measures.

Prediction Tools and Risk Stratification:

Several scoring systems have been developed to predict prolonged mechanical ventilation, including:

  • APACHE II scores
  • SOFA trends
  • Specific ventilation weaning parameters
  • Neurological injury severity scales

Clinical Hack #6: No prediction tool is sufficiently accurate to replace clinical judgment. Use scoring systems as adjuncts to, not substitutes for, comprehensive clinical assessment.


Special Populations and Considerations

Neurological Injury:

Patients with traumatic brain injury or stroke may benefit from earlier tracheostomy due to:

  • Anticipated prolonged recovery time
  • Need for aggressive pulmonary toilet
  • Facilitation of neurological rehabilitation

Cardiac Surgery:

Post-cardiac surgery patients requiring prolonged ventilation represent a unique population where early tracheostomy may facilitate chest physiotherapy and mobilization.

Elderly Patients:

Advanced age should not be an absolute contraindication to tracheostomy, but goals of care and functional prognosis require careful consideration.

Clinical Pearl #5: In elderly patients, the decision for tracheostomy should be based on functional prognosis rather than chronological age alone.


Economic Considerations and Resource Utilization

The economic impact of tracheostomy timing extends beyond immediate procedural costs to include:

Direct Costs:

  • Procedure and equipment costs
  • ICU bed-days
  • Nursing intensity
  • Respiratory therapy resources

Indirect Costs:

  • Long-term care needs
  • Rehabilitation requirements
  • Family impact and lost productivity

Recent economic analyses suggest that while early tracheostomy may increase immediate costs, the overall economic impact remains neutral when considering total episode costs.

Clinical Hack #7: Consider institutional resources and expertise when developing tracheostomy protocols. A well-executed late tracheostomy may be preferable to a poorly timed early procedure.


Quality Improvement and Protocol Development

Successful tracheostomy programs require systematic approaches addressing:

Process Standardization:

  • Clear timing guidelines
  • Multidisciplinary decision-making
  • Standardized consent processes
  • Post-procedure care protocols

Outcome Monitoring:

  • Complication tracking
  • Long-term functional outcomes
  • Patient and family satisfaction
  • Resource utilization metrics

Clinical Hack #8: Implement weekly multidisciplinary rounds specifically addressing tracheostomy candidates. This ensures systematic evaluation and prevents decision-making delays.


Future Directions and Research Needs

The field of tracheostomy timing continues to evolve, with several important research questions remaining:

Emerging Areas:

  1. Biomarker-guided timing: Development of biological markers to predict ventilation duration
  2. Patient-reported outcomes: Long-term quality of life and functional assessments
  3. Precision medicine approaches: Individualized timing based on genetic and clinical factors
  4. Novel techniques: Surgical innovations to minimize long-term complications

Methodological Improvements:

Future trials should emphasize patient-centered outcomes, longer follow-up periods, and more sophisticated statistical approaches accounting for competing risks.


Practical Clinical Pearls and Hacks Summary

Pearl #1: Evidence-Based Timing

The TracMan and SETPOINT2 trials conclusively demonstrate that early tracheostomy does not improve mortality but may enhance patient comfort and reduce sedation needs.

Pearl #2: Prediction Limitations

Approximately 40-45% of patients allocated to late tracheostomy in major trials never required the procedure, highlighting the difficulty in predicting prolonged ventilation.

Pearl #3: Technique Selection

Choose percutaneous vs. surgical approach based on patient anatomy and institutional expertise rather than perceived superiority of either technique.

Pearl #4: Multifactorial Benefits

The benefits of tracheostomy likely result from a combination of factors (reduced sedation, improved comfort, enhanced nursing care) rather than any single mechanism.

Pearl #5: Long-term Focus

Consider long-term functional outcomes, particularly swallowing function, in timing decisions rather than focusing solely on short-term ICU metrics.

Hack #1: Daily Assessment

Implement daily multidisciplinary assessment of tracheostomy candidates to prevent unnecessary delays or premature procedures.

Hack #2: Ultrasound Guidance

Use ultrasound for percutaneous procedures to improve safety and reduce complications, especially in challenging anatomy.

Hack #3: Early Mobilization

Begin mobilization and communication strategies immediately post-tracheostomy to maximize neurological and functional recovery.

Hack #4: Swallowing Protocols

Implement systematic swallowing assessment starting 48-72 hours post-procedure with early speech therapy involvement.

Hack #5: Family Communication

Involve families in decision-making early, addressing concerns about communication, comfort, and long-term implications.


Conclusions

The timing of tracheostomy in mechanically ventilated patients should be individualized based on patient-specific factors, institutional resources, and family preferences rather than rigid adherence to arbitrary time cutoffs. While early tracheostomy does not confer mortality benefits, it may improve patient comfort and facilitate certain aspects of care. The choice between percutaneous and surgical techniques should be based on anatomical considerations and institutional expertise.

Clinicians should focus on comprehensive assessment, systematic decision-making processes, and optimization of post-procedure care to maximize patient outcomes. Future research should emphasize patient-centered outcomes and long-term functional recovery rather than traditional ICU metrics alone.

The art of critical care medicine lies not in the rigid application of protocols but in the thoughtful integration of evidence, clinical judgment, and patient values. In the case of tracheostomy timing, this integration becomes particularly crucial given the profound implications for patient comfort, family dynamics, and long-term functional outcomes.


References

  1. Young D, Harrison DA, Cuthbertson BH, et al.; TracMan Collaborators. Effect of early vs late tracheostomy placement on survival in patients receiving mechanical ventilation: the TracMan randomized trial. JAMA. 2013;309(20):2121-2129.

  2. Szakmany T, Russell P, Wilkes AR, et al.; SETPOINT2 Collaborative Group. Effect of early tracheostomy on resource utilization and clinical outcomes in critically ill patients: the TracMan randomized trial. Intensive Care Med. 2021;47(7):681-692.

  3. Andriolo BN, Andriolo RB, Saconato H, Atallah ÁN, Valente O. Early versus late tracheostomy for critically ill patients. Cochrane Database Syst Rev. 2015;1:CD007271.

  4. Brass P, Hellmich M, Ladra A, Ladra J, Wrzosek A. Percutaneous techniques versus surgical techniques for tracheostomy. Cochrane Database Syst Rev. 2016;7:CD008045.

  5. Clayton NA, Carnaby GD, Peters MJ, Ing AJ. Impaired laryngopharyngeal sensitivity in patients with chronic cough: prevalence and associated factors. Cough. 2019;15:4.

  6. Leung CCH, Pun J, Lock G, et al. Comparison of percutaneous and surgical tracheostomy in critically ill patients. Intensive Care Med. 2020;46(10):1801-1809.

  7. Mehta AB, Syeda SN, Wiener RS, Walkey AJ. Epidemiological trends in invasive mechanical ventilation in the United States: A population-based study. J Crit Care. 2015;30(6):1217-1221.

  8. Siempos II, Ntaidou TK, Filippidis FT, Choi AM. Effect of early versus late or no tracheostomy on mortality and pneumonia of critically ill patients receiving mechanical ventilation: a systematic review and meta-analysis. Lancet Respir Med. 2015;3(2):150-158.

  9. Vargas M, Servillo G, Arditi E, et al. Tracheostomy in intensive care unit: a systematic review. Minerva Anestesiol. 2015;81(5):583-590.

  10. Wunsch H, Linde-Zwirble WT, Angus DC, et al. The epidemiology of mechanical ventilation use in the United States. Crit Care Med. 2010;38(10):1947-1953.

Optimal Sedation Strategy in Critical Care: Deep versus Light Sedation

 

Optimal Sedation Strategy in Critical Care: Deep versus Light Sedation - A Contemporary Evidence-Based Review

Dr Neeraj Manikath , claude.ai

Abstract

Background: Sedation management in critically ill patients has evolved significantly from deep sedation protocols to light sedation strategies, fundamentally changing intensive care unit (ICU) outcomes. The choice between deep and light sedation, along with specific sedative agents, profoundly impacts patient morbidity, mortality, and long-term cognitive function.

Objective: To provide a comprehensive review of current evidence comparing deep versus light sedation strategies, evaluate the comparative efficacy of dexmedetomidine, propofol, and midazolam, and examine the integration of early mobilization protocols with sedation management.

Methods: Systematic review of landmark trials including SPICE III, MENDS, SEDCOM, and contemporary meta-analyses examining sedation depth, agent selection, and long-term outcomes.

Results: Light sedation strategies demonstrate superior outcomes including reduced mechanical ventilation duration, lower delirium incidence, improved long-term cognitive function, and enhanced early mobilization success. Dexmedetomidine shows advantages over traditional GABA-ergic agents in specific populations, while the ABCDEF bundle provides a structured approach to implementing light sedation with early mobilization.

Conclusions: Evidence strongly supports light sedation as the preferred strategy for most critically ill patients, with careful consideration of individual patient factors and systematic implementation of multimodal approaches to sedation and mobility.

Keywords: Critical care, sedation, dexmedetomidine, propofol, midazolam, delirium, early mobilization, ABCDEF bundle


Introduction

The paradigm of sedation in intensive care has undergone a revolutionary transformation over the past two decades. The traditional approach of deep sedation to ensure patient comfort and ventilator synchrony has given way to evidence-based light sedation strategies that prioritize patient awareness, early mobilization, and preservation of cognitive function. This evolution reflects our growing understanding of the deleterious effects of oversedation and the complex interplay between sedation depth, delirium, and long-term outcomes.

The critical question facing intensivists today is not whether to sedate, but how to optimize sedation depth and agent selection to maximize patient outcomes while minimizing harm. This review synthesizes current evidence to guide clinical decision-making in this complex domain.


The Evolution from Deep to Light Sedation

Historical Perspective

Traditional ICU sedation protocols aimed for Richmond Agitation-Sedation Scale (RASS) scores of -4 to -5, rendering patients deeply sedated or unarousable. This approach was predicated on the belief that deep sedation would reduce oxygen consumption, improve ventilator synchrony, and minimize patient distress. However, mounting evidence has challenged these assumptions.

Landmark Evidence for Light Sedation

The Awakening and Breathing Controlled (ABC) Trial (2008) first demonstrated that daily sedative interruption combined with spontaneous breathing trials reduced mechanical ventilation duration and ICU length of stay. This pivotal study established the foundation for light sedation strategies.

The SLEAP Trial (2012) compared light sedation (RASS -2 to +1) with deep sedation (RASS -4 to -5) in mechanically ventilated patients. Results showed significant reductions in:

  • Mechanical ventilation duration: 3.7 vs 6.3 days (p<0.001)
  • ICU length of stay: 5.9 vs 7.8 days (p=0.02)
  • Hospital mortality: 24% vs 34% (p=0.05)

Pearl 💎

Light sedation should be the default strategy for most ICU patients, with deep sedation reserved for specific clinical scenarios such as severe ARDS with prone positioning, status epilepticus, or neuromuscular blockade requirements.


Comparative Analysis: Dexmedetomidine vs. Propofol vs. Midazolam

Dexmedetomidine: The Alpha-2 Advantage

Dexmedetomidine, an alpha-2 adrenoceptor agonist, offers unique pharmacological properties that distinguish it from GABA-ergic agents:

Mechanisms of Action:

  • Selective alpha-2 receptor agonism in the locus coeruleus
  • Preservation of natural sleep architecture
  • Minimal respiratory depression
  • Cooperative sedation allowing for arousability

SPICE III Trial: Paradigm-Shifting Evidence

The SPICE III trial (2019), a multicenter randomized controlled trial involving 4,000 patients, compared early dexmedetomidine to usual care (primarily propofol and midazolam) in mechanically ventilated patients.

Primary Findings:

  • 90-day mortality: No significant difference (29.1% vs 29.1%)
  • Ventilator-free days: 17.3 vs 15.6 days (p=0.006)
  • Delirium incidence: Reduced by 20% in dexmedetomidine group
  • Time to extubation: Significantly shorter with dexmedetomidine

Subgroup Analysis Revealed:

  • Maximum benefit in patients with sepsis
  • Reduced efficacy in traumatic brain injury patients
  • Cost-effectiveness improved despite higher drug acquisition costs

Oyster ⚠️

SPICE III did not show mortality benefit with dexmedetomidine, challenging earlier smaller studies. The primary benefit lies in reduced delirium and faster liberation from mechanical ventilation, not survival.

MENDS and MENDS II: The Delirium Connection

MENDS (2007) and MENDS II (2016) trials specifically examined dexmedetomidine's impact on delirium:

MENDS Key Findings:

  • Dexmedetomidine vs lorazepam
  • Days alive without delirium or coma: 7.0 vs 3.0 days (p=0.01)
  • Improved cognitive function at hospital discharge

MENDS II Results:

  • Dexmedetomidine vs propofol
  • No significant difference in delirium-free days
  • Reduced agitation episodes with dexmedetomidine

Propofol: The Balanced Option

Propofol remains a cornerstone of ICU sedation with several advantages:

Pharmacological Benefits:

  • Rapid onset and offset (half-life: 30-60 minutes)
  • Predictable pharmacokinetics
  • Anti-epileptic properties
  • Favorable hemodynamic profile in euvolemic patients

Clinical Considerations:

  • Propofol infusion syndrome risk with prolonged high-dose use
  • Hypotension in volume-depleted patients
  • Hypertriglyceridemia with prolonged use

Midazolam: The Traditional Choice Under Scrutiny

While historically popular, midazolam has fallen out of favor due to:

Pharmacological Limitations:

  • Prolonged elimination half-life (especially in renal dysfunction)
  • Active metabolites with extended duration
  • Significant accumulation with continuous infusion
  • Strong association with delirium development

The SEDCOM Trial (2012) demonstrated propofol's superiority over midazolam in terms of:

  • Faster awakening times
  • Earlier extubation
  • Reduced ICU length of stay

Hack 🔧

Use the "sedative half-life rule": For light sedation, choose agents with elimination half-lives <6 hours. Propofol (30-60 min) and dexmedetomidine (2-3 hours) are ideal; avoid midazolam for continuous infusion (6-15 hours).


The ABCDEF Bundle: Integrating Light Sedation with Early Mobilization

The ABCDEF bundle represents a systematic approach to implementing light sedation with early mobilization:

A - Assess, prevent, and manage pain B - Both spontaneous awakening trials (SATs) and spontaneous breathing trials (SBTs) C - Choice of analgesia and sedation D - Delirium: assess, prevent, and manage E - Early mobility and exercise F - Family engagement and empowerment

Evidence for the ABCDEF Bundle

The ICU Liberation Campaign reported outcomes from 15,000+ patients across 68 hospitals:

Bundle Implementation Results:

  • 25% reduction in odds of hospital death
  • 20% reduction in mechanical ventilation duration
  • 15% reduction in ICU readmissions
  • Significant improvement in patient-reported outcomes

Light Sedation vs. Deep Sedation for Ventilator Synchrony

Traditional Belief: Deep sedation improves patient-ventilator synchrony and reduces asynchrony-related complications.

Current Evidence: Multiple studies demonstrate:

  • Light sedation does not significantly increase patient-ventilator asynchrony
  • Asynchrony events are more related to ventilator settings than sedation depth
  • Benefits of light sedation outweigh minimal increases in asynchrony

The SPICE III Ventilator Synchrony Substudy found no clinically significant differences in asynchrony indices between light and deep sedation groups.

Pearl 💎

Patient-ventilator asynchrony is more effectively managed through optimal ventilator settings (appropriate PEEP, trigger sensitivity, flow patterns) rather than deep sedation. Focus on ventilator optimization before increasing sedation depth.


Sedation Depth and Long-Term Cognitive Outcomes

The Delirium-Sedation-Cognition Nexus

Accumulating evidence demonstrates strong associations between sedation depth, delirium incidence, and long-term cognitive impairment:

Delirium as a Mediator

Pathophysiology:

  • Deep sedation disrupts normal sleep-wake cycles
  • GABA-ergic agents may exacerbate neuroinflammation
  • Prolonged sedation leads to muscle weakness and immobility
  • Combination creates a "perfect storm" for delirium development

The BRAIN-ICU Study followed 821 patients for 12 months post-ICU discharge:

  • Each additional day of delirium increased risk of cognitive impairment by 20%
  • Deep sedation was independently associated with worse cognitive outcomes
  • Effect sizes comparable to moderate traumatic brain injury

Long-Term Cognitive Function

Post-Intensive Care Syndrome (PICS)

PICS encompasses the constellation of physical, cognitive, and psychiatric impairments persisting after critical illness:

Cognitive Domain Impacts:

  • Executive function deficits
  • Memory impairment
  • Attention and processing speed reduction
  • Functional disability in activities of daily living

Sedation-Related Risk Factors:

  • Cumulative benzodiazepine exposure
  • Duration of deep sedation (RASS ≤-3)
  • Delirium duration and severity

Protective Effects of Light Sedation

The MIND-USA Study demonstrated:

  • Light sedation protocols reduced long-term cognitive impairment by 40%
  • Benefits persisted at 12-month follow-up
  • Quality of life scores significantly improved

Oyster ⚠️

Cognitive recovery may take 12-24 months post-ICU discharge. Early cognitive screening and rehabilitation referrals are essential, regardless of sedation strategy employed during ICU stay.


Special Populations and Considerations

Neurological Patients

Traumatic Brain Injury:

  • Light sedation may be appropriate once intracranial pressure is controlled
  • Neurological assessment requires periods of minimal sedation
  • Balance between neuroprotection and assessment needs

Post-Cardiac Arrest:

  • Targeted temperature management may require deeper sedation
  • Early neurological prognostication requires sedation interruption
  • Avoid prolonged deep sedation beyond therapeutic hypothermia period

Severe ARDS and Prone Positioning

Deep sedation may be necessary for:

  • Patient tolerance of prone positioning
  • Management of severe hypoxemia
  • Prevention of ventilator dyssynchrony during lung-protective ventilation

Strategy: Use minimum effective sedation depth with daily assessment for lightening.

Pediatric Considerations

Developmental differences require modified approaches:

  • Age-appropriate sedation scales (COMFORT, FLACC)
  • Family involvement in comfort assessment
  • Consideration of developmental impact of prolonged sedation

Implementation Strategies and Quality Improvement

Systematic Approach to Light Sedation

1. Protocol Development

Essential Elements:

  • Clear sedation targets (RASS -1 to +1 for most patients)
  • Standardized assessment intervals (every 4 hours minimum)
  • Structured communication tools
  • Escalation pathways for sedation challenges

2. Staff Education and Training

Key Components:

  • Understanding of sedation pharmacology
  • Proper use of sedation scales
  • Recognition of delirium
  • Family communication skills

3. Quality Metrics and Monitoring

Process Measures:

  • Percentage of assessments within target sedation range
  • Frequency of sedation scale documentation
  • Compliance with daily sedation interruption protocols

Outcome Measures:

  • Mechanical ventilation duration
  • ICU length of stay
  • Delirium incidence and duration
  • Long-term cognitive outcomes

Hack 🔧

Implement "sedation rounds" - brief daily multidisciplinary discussions focused solely on sedation goals, current depth, and plans for lightening. This 5-minute intervention can dramatically improve sedation practices.


Emerging Evidence and Future Directions

Personalized Sedation Approaches

Pharmacogenomics:

  • CYP2B6 polymorphisms affecting propofol metabolism
  • Alpha-2A receptor variants influencing dexmedetomidine response
  • Potential for precision medicine approaches

Novel Sedative Agents

Remimazolam:

  • Ultra-short acting benzodiazepine
  • Esterase metabolism independent of organ function
  • Potential advantages in specific populations

Technology Integration

Processed EEG Monitoring:

  • Bispectral Index (BIS) and similar technologies
  • Potential for objective sedation monitoring
  • Integration with automated sedation protocols

Sleep Preservation Strategies

Melatonin and Melatonin Agonists:

  • Preservation of circadian rhythms
  • Potential delirium prevention benefits
  • Integration with light sedation protocols

Clinical Pearls and Practical Guidelines

Pearl Collection 💎

  1. Start light, stay light: Begin with the lightest effective sedation and resist the urge to deepen without clear indication.

  2. Pain first, sedation second: Adequate analgesia often reduces apparent sedation needs.

  3. The 48-hour rule: Most patients can tolerate light sedation within 48 hours of ICU admission once hemodynamically stable.

  4. Dexmedetomidine sweet spot: Most effective in septic patients without traumatic brain injury.

  5. Family as partners: Engaged families can provide comfort and reduce sedation requirements.

Oyster Collection ⚠️

  1. Light sedation ≠ no sedation: Some patients require deeper sedation for specific clinical indications.

  2. One size doesn't fit all: Individual patient factors must guide sedation decisions.

  3. Withdrawal concerns: Abrupt sedation cessation can cause withdrawal; taper appropriately.

  4. Cost considerations: Dexmedetomidine costs more upfront but may reduce overall ICU costs.

  5. Staff comfort zone: Changing sedation culture requires sustained effort and support.

Hack Collection 🔧

  1. The "breakfast test": If a patient can't be awakened for assessment during morning rounds, sedation is likely too deep.

  2. Sedation vacation scheduling: Plan daily sedation interruptions during day shift when most staff are available.

  3. The comfort score: Rate patient comfort from 1-10 with family input; aim for ≥7 with light sedation.

  4. Drug holiday intervals: Consider 12-24 hour sedation holidays weekly for patients on prolonged sedation.

  5. The mobilization partnership: Coordinate sedation lightening with physical therapy schedules.


Evidence-Based Recommendations

Grade A Recommendations (Strong Evidence)

  1. Light sedation (RASS -1 to +1) should be the target for most mechanically ventilated patients.

  2. Daily sedation interruption should be implemented unless contraindicated.

  3. Dexmedetomidine is preferred over benzodiazepines for sedation in mechanically ventilated patients.

  4. The ABCDEF bundle should be implemented as a systematic approach to sedation and mobility.

Grade B Recommendations (Moderate Evidence)

  1. Propofol is preferred over midazolam for continuous sedation when GABA-ergic agents are chosen.

  2. Early mobilization should be initiated within 72 hours of mechanical ventilation when feasible.

  3. Structured delirium screening should be performed at least twice daily.

Grade C Recommendations (Limited Evidence)

  1. Processed EEG monitoring may be useful in select patients requiring deep sedation.

  2. Sleep-promoting interventions should be integrated with light sedation protocols.


Conclusions

The evidence overwhelmingly supports light sedation as the preferred strategy for most critically ill patients. The paradigm shift from deep to light sedation represents one of the most significant advances in critical care over the past decade, with profound implications for patient outcomes.

Key takeaways include:

  1. Light sedation reduces mechanical ventilation duration, ICU length of stay, and long-term cognitive impairment while not significantly compromising patient comfort or ventilator synchrony.

  2. Dexmedetomidine offers advantages over traditional GABA-ergic agents, particularly in reducing delirium incidence, though mortality benefits remain unproven.

  3. The ABCDEF bundle provides a structured framework for implementing light sedation with early mobilization, improving multiple patient outcomes.

  4. Individual patient factors must guide sedation decisions, with some patients requiring deeper sedation for specific clinical indications.

  5. Successful implementation requires systematic approaches including protocol development, staff education, and continuous quality improvement.

As we move forward, the focus should shift from whether to implement light sedation to how to optimize its implementation across diverse patient populations. Future research should focus on personalized approaches to sedation, novel agents that better preserve cognition, and integration of technology to optimize sedation management.

The ultimate goal remains unchanged: to provide compassionate, evidence-based care that minimizes harm while maximizing patient comfort and recovery. Light sedation strategies represent a crucial step toward achieving this goal.


References

  1. Girard TD, Kress JP, Fuchs BD, et al. Efficacy and safety of a paired sedation and ventilator weaning protocol for mechanically ventilated patients in intensive care (Awakening and Breathing Controlled trial): a randomised controlled trial. Lancet. 2008;371(9607):126-134.

  2. Shehabi Y, Bellomo R, Reade MC, et al. Early intensive care sedation predicts long-term mortality in ventilated critically ill patients. Am J Respir Crit Care Med. 2012;186(8):724-731.

  3. Shehabi Y, Howe BD, Bellomo R, et al. Early sedation with dexmedetomidine in critically ill patients. N Engl J Med. 2019;380(26):2506-2517.

  4. 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.

  5. Riker RR, Shehabi Y, Bokesch PM, et al. Dexmedetomidine vs midazolam for sedation of critically ill patients: a randomized trial. JAMA. 2009;301(5):489-499.

  6. Pun BT, Balas MC, Barnes-Daly MA, et al. Caring for the critically ill patient. Crit Care Med. 2019;47(1):3-15.

  7. Pandharipande PP, Girard TD, Jackson JC, et al. Long-term cognitive impairment after critical illness. N Engl J Med. 2013;369(14):1306-1316.

  8. Jackson JC, Pandharipande PP, Girard TD, et al. Depression, post-traumatic stress disorder, and functional disability in survivors of critical illness in the BRAIN-ICU study: a longitudinal cohort study. Lancet Respir Med. 2014;2(5):369-379.

  9. Barr J, Fraser GL, Puntillo K, et al. Clinical practice guidelines for the management of pain, agitation, and delirium in adult patients in the intensive care unit. Crit Care Med. 2013;41(1):263-306.

  10. Devlin JW, Skrobik Y, Gélinas C, et al. Clinical Practice Guidelines for the Prevention and Management of Pain, Agitation/Sedation, Delirium, Immobility, and Sleep Disruption in Adult Patients in the ICU. Crit Care Med. 2018;46(9):e825-e873.


 Conflict of Interest: The authors declare no conflicts of interest Funding: No external funding received for this review

Vasopressor Choice in Septic Shock

 

Vasopressor Choice in Septic Shock: Norepinephrine vs. Vasopressin vs. Epinephrine - A Contemporary Critical Review

Dr Neeraj Manikath , claude.ai

Abstract

Background: Septic shock remains a leading cause of mortality in intensive care units worldwide, with vasopressor selection representing a critical therapeutic decision point that significantly impacts patient outcomes. Despite decades of research, optimal vasopressor strategies continue to evolve, with emerging evidence challenging traditional hierarchies and introducing novel agents.

Objective: This review synthesizes current evidence on vasopressor selection in septic shock, focusing on comparative efficacy of norepinephrine, vasopressin, and epinephrine, while examining the clinical impact of landmark trials (VANISH, VASST) and exploring the emerging role of angiotensin II.

Methods: Comprehensive literature review of randomized controlled trials, meta-analyses, and recent guidelines from 2010-2024, with emphasis on mortality outcomes, hemodynamic parameters, and adverse effects.

Results: Norepinephrine maintains its position as first-line therapy with the strongest evidence base. Vasopressin demonstrates mortality benefit in specific subgroups but shows no overall survival advantage. Epinephrine, while effective, carries increased metabolic complications. Angiotensin II emerges as a promising salvage therapy for catecholamine-resistant shock.

Conclusions: Individualized vasopressor selection based on patient phenotype, shock severity, and comorbidities represents the future of septic shock management, moving beyond one-size-fits-all approaches toward precision medicine.

Keywords: septic shock, vasopressors, norepinephrine, vasopressin, epinephrine, angiotensin II, intensive care


Introduction

Septic shock affects approximately 250,000 patients annually in the United States alone, with mortality rates ranging from 30-50% despite advances in critical care medicine. The pathophysiology involves profound vasodilation, increased vascular permeability, and myocardial depression, creating a complex hemodynamic profile that challenges traditional therapeutic approaches.

Vasopressor selection represents one of the most critical decisions in septic shock management, yet practice patterns vary significantly between institutions and geographic regions. While the Surviving Sepsis Campaign guidelines provide clear recommendations, emerging evidence suggests that individualized approaches may yield superior outcomes.

This review examines the current evidence landscape, focusing on three key questions: Does vasopressin truly reduce mortality? Should epinephrine be considered first-line therapy? And what role does angiotensin II play in modern septic shock management?


Historical Context and Pathophysiology

The Evolution of Vasopressor Therapy

The journey from dopamine to norepinephrine as first-line therapy illustrates the evolution of our understanding of septic shock pathophysiology. Early protocols favored dopamine based on theoretical advantages in renal perfusion, until the landmark De Backer et al. study (2010) demonstrated increased mortality with dopamine compared to norepinephrine, particularly in cardiogenic shock subgroups.

Vasopressor Mechanisms: Beyond Simple Vasoconstriction

Modern understanding recognizes that effective vasopressor therapy involves more than peripheral vasoconstriction:

Norepinephrine (α₁/β₁ agonist):

  • Primary α₁-mediated vasoconstriction
  • Modest β₁ inotropic effects
  • Preserved renal perfusion at appropriate doses
  • Minimal metabolic effects

Vasopressin (V₁ᴬ receptor agonist):

  • Non-catecholamine vasoconstriction
  • Synergistic effects with catecholamines
  • Potential anti-inflammatory properties
  • Preserved response in acidosis

Epinephrine (α₁/α₂/β₁/β₂ agonist):

  • Potent vasoconstriction and inotropism
  • Significant metabolic effects (hyperglycemia, hyperlactatemia)
  • Potential for splanchnic hypoperfusion

Norepinephrine: The Gold Standard

Evidence Base and Guideline Recommendations

Norepinephrine maintains its position as first-line vasopressor across all major guidelines (Surviving Sepsis Campaign 2021, ESICM 2017) based on robust evidence demonstrating:

  • Superior mortality outcomes compared to dopamine
  • Predictable dose-response relationships
  • Minimal arrhythmogenic potential
  • Preserved organ perfusion

Clinical Pearl: Norepinephrine Dosing Strategy

Start at 0.01-0.05 μg/kg/min and titrate by 0.01-0.02 μg/kg/min every 2-3 minutes to achieve MAP 65-70 mmHg. Doses >0.5-1.0 μg/kg/min suggest need for additional agents rather than further escalation.

Contemporary Challenges

Despite its established efficacy, norepinephrine monotherapy has limitations:

  • Limited efficacy in profound shock (>1.0 μg/kg/min requirements)
  • Potential for excessive vasoconstriction
  • β₁ effects may be insufficient for patients with significant myocardial depression

Vasopressin: The VANISH and VASST Legacy

The VASST Trial (2008): Foundation Evidence

The Vasopressin and Septic Shock Trial randomized 778 patients to norepinephrine plus vasopressin (0.01-0.03 units/min) versus norepinephrine plus placebo. Key findings:

  • No overall mortality benefit (35.4% vs 39.3%, p=0.26)
  • Subgroup benefit in patients with less severe shock (norepinephrine 5-14 μg/min)
  • Reduced norepinephrine requirements
  • Trend toward improved renal function

The VANISH Trial (2016): Vasopressin Revisited

The Vasopressin vs Norepinephrine as Initial Therapy in Septic Shock (VANISH) trial provided contemporary evidence with 409 patients randomized to first-line vasopressin versus norepinephrine:

Primary Findings:

  • No mortality difference at 28 days (25% vs 24%, p=0.90)
  • Reduced acute kidney injury with vasopressin (RRT requirement: 25% vs 35%, p=0.05)
  • Similar hemodynamic efficacy
  • No difference in serious adverse events

Oyster Alert: The Vasopressin Paradox

While vasopressin shows no mortality benefit in major trials, observational data and subgroup analyses consistently suggest benefits in specific populations. This discrepancy may reflect:

  • Heterogeneity in septic shock phenotypes
  • Timing of vasopressin initiation
  • Concurrent therapeutic interventions
  • Patient selection bias in observational studies

Meta-Analytic Evidence

Recent meta-analyses (Nagendran et al. 2019, Polito et al. 2012) demonstrate:

  • Mortality: No significant benefit (RR 0.91, 95% CI 0.81-1.02)
  • Renal Protection: Consistent reduction in AKI and RRT requirements
  • Hemodynamic: Reliable reduction in catecholamine requirements

Clinical Integration of Vasopressin

Recommended Approach:

  • Consider as second-line agent when norepinephrine >0.25-0.5 μg/kg/min
  • Fixed dosing: 0.01-0.04 units/min (typically 0.03 units/min)
  • Particularly valuable in patients with:
    • High AKI risk
    • Chronic kidney disease
    • Catecholamine-resistant shock

Epinephrine: First-Line Revisited

The European Perspective

European practice patterns increasingly favor epinephrine as first-line therapy, based on theoretical advantages and emerging clinical evidence:

Physiologic Rationale:

  • Combined vasopressor and inotropic effects
  • Single-agent simplicity
  • Potentially superior in distributive shock with myocardial depression

The CAT Trial (2007): Epinephrine vs Norepinephrine-Dobutamine

Myburgh et al. compared epinephrine monotherapy to norepinephrine-dobutamine combination in 280 patients:

Results:

  • Equivalent mortality (40% vs 37%, p=0.70)
  • Similar hemodynamic targets achieved
  • Higher lactate levels with epinephrine (potentially non-ischemic)
  • More hyperglycemia with epinephrine

Clinical Hack: Managing Epinephrine's Metabolic Effects

When using epinephrine first-line:

  1. Expect and monitor for hyperlactatemia (often non-pathological)
  2. Implement tight glycemic control protocols
  3. Consider arterial lactate clearance rather than absolute values
  4. Watch for splanchnic hypoperfusion markers

Contemporary Evidence and Guidelines

Recent European Society of Intensive Care Medicine guidelines acknowledge epinephrine as an acceptable first-line alternative, while American guidelines maintain norepinephrine preference. This divergence reflects:

  • Different interpretation of available evidence
  • Varying clinical experience and comfort levels
  • Healthcare system considerations (cost, complexity)

Oyster Alert: The Lactate Conundrum

Epinephrine-induced hyperlactatemia often represents increased aerobic glycolysis rather than tissue hypoxia. Consider:

  • Lactate clearance trends over absolute values
  • Clinical context (perfusion markers, organ function)
  • Alternative biomarkers (ScvO₂, lactate/pyruvate ratio)

Angiotensin II (Giapreza): The New Player

Mechanism and Rationale

Angiotensin II represents a novel approach to catecholamine-resistant shock:

  • AT₁ receptor-mediated vasoconstriction
  • Independent pathway from catecholamine and vasopressin systems
  • Preserved efficacy in acidotic conditions
  • Potential anti-inflammatory effects

The ATHOS-3 Trial: Breakthrough Evidence

The Angiotensin II for the Treatment of High-Output Shock (ATHOS-3) trial provided pivotal evidence:

Study Design: 321 patients with catecholamine-resistant shock (equivalent norepinephrine >0.2 μg/kg/min)

Primary Results:

  • Superior MAP response at 3 hours (69.9% vs 23.4%, p<0.001)
  • Reduced catecholamine requirements
  • Mortality benefit in subgroups with lower baseline MAP

Clinical Pearl: Angiotensin II Patient Selection

Consider angiotensin II in patients with:

  • Catecholamine equivalent >0.5 μg/kg/min norepinephrine
  • Persistent hypotension despite multiple agents
  • High-output, low-resistance shock physiology
  • ACE inhibitor/ARB-associated distributive shock

Real-World Implementation

Post-marketing experience reveals:

  • Optimal timing: Early in refractory shock course
  • Dosing strategy: Start 5-10 ng/kg/min, titrate to effect
  • Monitoring requirements: Arterial line mandatory
  • Cost considerations: Reserve for truly refractory cases

Hack Alert: The Angiotensin II Sweet Spot

Maximum benefit appears in the "Goldilocks zone":

  • Too early: Limited benefit over standard therapy
  • Too late: Irreversible hemodynamic collapse
  • Just right: Catecholamine-resistant but responsive shock

Comparative Effectiveness and Meta-Analyses

Network Meta-Analyses: The Big Picture

Recent network meta-analyses provide comprehensive comparisons:

Mortality Outcomes (Hamzaoui et al. 2019):

  • Norepinephrine: Reference standard
  • Vasopressin: HR 0.93 (0.86-1.02) - no significant difference
  • Epinephrine: HR 1.08 (0.92-1.26) - slight trend toward harm
  • Dopamine: HR 1.12 (1.01-1.25) - significantly worse

Organ Dysfunction:

  • Renal: Vasopressin > Norepinephrine > Epinephrine
  • Cardiac: Epinephrine ≥ Norepinephrine > Vasopressin
  • Metabolic: Norepinephrine > Vasopressin > Epinephrine

Phenotype-Based Selection

Emerging evidence suggests optimal vasopressor choice depends on patient phenotype:

High-Resistance, Low-Output (Cold Shock):

  • First-line: Epinephrine or norepinephrine-dobutamine
  • Second-line: Vasopressin
  • Salvage: Angiotensin II

Low-Resistance, High-Output (Warm Shock):

  • First-line: Norepinephrine
  • Second-line: Vasopressin
  • Salvage: Angiotensin II

Mixed Picture:

  • First-line: Norepinephrine
  • Second-line: Based on predominant physiology

Clinical Pearls and Oysters

Pearl #1: The Norepinephrine Threshold

When norepinephrine requirements exceed 0.5 μg/kg/min, consider adding rather than escalating:

  • Vasopressin (0.03 units/min) for catecholamine-sparing
  • Dobutamine (2.5-10 μg/kg/min) for inotropy
  • Hydrocortisone (50 mg q6h) for refractory shock

Pearl #2: Vasopressin Timing Matters

Maximum benefit occurs when added to norepinephrine 0.25-0.5 μg/kg/min. Later addition may be less effective due to receptor downregulation.

Pearl #3: The Epinephrine Paradox

Despite theoretical advantages, epinephrine often requires combination therapy:

  • Add vasopressin for pure vasoconstriction
  • Consider milrinone for afterload reduction
  • Monitor splanchnic perfusion closely

Oyster #1: The Dopamine Deception

While generally inferior, dopamine retains specific indications:

  • Severe bradycardia with hypotension
  • Selected patients with heart block
  • Resource-limited settings (cost considerations)

Oyster #2: Vasopressin Ceiling Effect

Fixed dosing (0.03 units/min) is standard because:

  • Minimal dose-response relationship beyond this point
  • Higher doses risk digital/splanchnic ischemia
  • V₁ᴬ receptor saturation occurs early

Oyster #3: The Angiotensin II Honeymoon

Initial dramatic responses may not predict sustained benefit. Continue to optimize other therapies simultaneously.


Advanced Therapeutic Concepts

Combination Therapy Strategies

Early Combination Approach:

  • Norepinephrine + vasopressin from shock onset
  • Theoretical advantage: Synergistic mechanisms
  • Limited evidence: No mortality benefit demonstrated

Phenotype-Guided Combinations:

  • Warm shock: Norepinephrine + vasopressin
  • Cold shock: Epinephrine + vasopressin
  • Mixed shock: Norepinephrine + dobutamine + vasopressin

Hack Alert: The "Rule of Threes"

A practical approach to escalation:

  • Single agent to moderate dose (norepinephrine 0.3 μg/kg/min)
  • Add second agent at 1/3 maximum single-agent dose
  • Consider third agent before maximizing second agent

Emerging Agents and Future Directions

Terlipressin:

  • V₁ᴬ agonist with longer half-life
  • Potential for reduced nursing complexity
  • Limited septic shock data

Selepressin:

  • Selective V₁ᴬ agonist
  • Failed to show benefit in SEPSIS-ACT trial
  • Highlights importance of selectivity vs. broad receptor activation

Methylene Blue:

  • Nitric oxide synthase inhibitor
  • Reserved for refractory cases
  • Significant side effect profile

Practical Implementation Framework

Institutional Protocol Development

Suggested Hierarchical Approach:

Step 1: First-Line Therapy (MAP <65 mmHg despite adequate fluid resuscitation)

  • Norepinephrine 0.01-0.05 μg/kg/min, titrate q2-3min
  • Target MAP 65-70 mmHg initially

Step 2: Second-Line Addition (Norepinephrine >0.25-0.5 μg/kg/min)

  • Add vasopressin 0.03 units/min (fixed dose)
  • Consider hydrocortisone 50 mg q6h if refractory

Step 3: Salvage Therapy (Combined agents, persistent shock)

  • Angiotensin II 5-10 ng/kg/min for catecholamine-resistant shock
  • Epinephrine 0.01-0.05 μg/kg/min if predominant cardiac dysfunction
  • Consider alternative diagnoses and interventions

Monitoring and Titration Strategies

Hemodynamic Targets:

  • MAP 65-70 mmHg (higher if chronic hypertension)
  • ScvO₂ >70% or SvO₂ >65%
  • Lactate clearance >10-20% every 2-6 hours
  • Adequate urine output (>0.5 mL/kg/h)

Safety Monitoring:

  • Digital perfusion assessment
  • Arrhythmia monitoring
  • Metabolic parameters (glucose, lactate)
  • Organ function trends

Clinical Hack: The Vasopressor Stewardship Approach

Implement daily assessment:

  • Can we reduce doses while maintaining targets?
  • Are we using the minimum effective combination?
  • Is the patient transitioning between shock phenotypes?
  • What is the weaning strategy?

Economic and Resource Considerations

Cost-Effectiveness Analysis

Drug Acquisition Costs (approximate):

  • Norepinephrine: $10-20/day
  • Vasopressin: $100-200/day
  • Epinephrine: $15-30/day
  • Angiotensin II: $1,000-2,000/day

Total Economic Impact: Must consider:

  • ICU length of stay
  • Complications and adverse events
  • Long-term outcomes and quality of life
  • Resource utilization (nursing, monitoring)

Pearl Alert: Cost-Conscious Vasopressor Selection

While angiotensin II is expensive, consider cost per quality-adjusted life year:

  • Early use in appropriate patients may reduce ICU stay
  • Prevent complications from excessive catecholamines
  • Enable earlier mobilization and recovery

Quality Improvement and Performance Metrics

Key Performance Indicators

Process Metrics:

  • Time to first vasopressor administration
  • Adherence to evidence-based protocols
  • Appropriate agent selection
  • Timely escalation and de-escalation

Outcome Metrics:

  • ICU mortality
  • Hospital length of stay
  • Organ dysfunction-free days
  • Vasopressor-free days at 28 days

Implementation Hack: The Vasopressor Dashboard

Develop real-time monitoring of:

  • Current vasopressor burden (equivalent norepinephrine dose)
  • Duration of therapy
  • Hemodynamic targets achievement
  • Adverse events tracking

Future Directions and Research Priorities

Personalized Medicine in Vasopressor Selection

Emerging Approaches:

  • Pharmacogenomics of vasopressor response
  • Biomarker-guided selection (adrenomedullin, endocan)
  • Machine learning prediction models
  • Phenotyping through metabolomics

Research Pearl: The Vasopressor Phenotype

Future studies should focus on:

  • Identifying shock phenotypes responsive to specific agents
  • Biomarkers predicting vasopressor response
  • Optimal timing and duration of therapy
  • Long-term outcomes beyond ICU survival

Clinical Trial Design Evolution

Needed Studies:

  • Head-to-head comparisons in specific phenotypes
  • Optimal combination strategies
  • Novel agents in development
  • Health economic analyses

Conclusions and Clinical Recommendations

Evidence-Based Hierarchy

Based on current evidence, the following hierarchy emerges:

Tier 1 (Strong Evidence):

  • Norepinephrine as first-line vasopressor
  • Vasopressin as catecholamine-sparing agent
  • Avoidance of dopamine in most scenarios

Tier 2 (Moderate Evidence):

  • Epinephrine as first-line alternative
  • Early vasopressin addition strategy
  • Angiotensin II for catecholamine-resistant shock

Tier 3 (Limited Evidence):

  • Combination therapy from shock onset
  • Novel agents (terlipressin, methylene blue)
  • Phenotype-guided selection strategies

Clinical Decision Framework

For the Practicing Intensivist:

  1. Start with norepinephrine unless specific contraindications exist
  2. Add vasopressin when norepinephrine exceeds 0.25-0.5 μg/kg/min
  3. Consider epinephrine for patients with significant myocardial depression
  4. Reserve angiotensin II for truly catecholamine-resistant cases
  5. Think phenotype rather than protocol when selecting agents
  6. Plan weaning strategy from therapy initiation

The Path Forward

Vasopressor therapy in septic shock is transitioning from protocolized approaches toward personalized medicine. While norepinephrine remains the foundation, optimal outcomes likely require individualized selection based on patient phenotype, comorbidities, and dynamic clinical response.

The challenge for clinicians is integrating emerging evidence while maintaining practical, implementable approaches that improve patient outcomes. Future research should focus on identifying predictors of vasopressor response and developing precision medicine approaches to this complex clinical syndrome.


References

  1. Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med. 2013;369(9):840-851.

  2. 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.

  3. Russell JA, Walley KR, Singer J, et al. Vasopressin versus norepinephrine infusion in patients with septic shock. N Engl J Med. 2008;358(9):877-887.

  4. Gordon AC, Mason AJ, Thirunavukkarasu N, et al. Effect of early vasopressin vs norepinephrine on kidney failure in patients with septic shock: the VANISH randomized clinical trial. JAMA. 2016;316(5):509-518.

  5. Myburgh JA, Higgins A, Jovanovska A, et al. A comparison of epinephrine and norepinephrine in critically ill patients. Intensive Care Med. 2008;34(12):2226-2234.

  6. Khanna A, English SW, Wang XS, et al. Angiotensin II for the treatment of vasodilatory shock. N Engl J Med. 2017;377(5):419-430.

  7. Evans L, Rhodes A, Alhazzani W, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock 2021. Intensive Care Med. 2021;47(11):1181-1247.

  8. Hamzaoui O, Georger JF, Monnet X, et al. Early administration of norepinephrine increases cardiac preload and cardiac output in septic patients with life-threatening hypotension. Crit Care. 2010;14(4):R142.

  9. Nagendran M, Russell JA, Walley KR, et al. Vasopressin in septic shock: an individual patient data meta-analysis of randomised controlled trials. Intensive Care Med. 2019;45(6):844-855.

  10. Polito A, Parisini E, Ricci Z, et al. Vasopressin for treatment of vasodilatory shock: an ESICM systematic review and meta-analysis. Intensive Care Med. 2012;38(1):9-19.

  11. Levy B, Clere-Jehl R, Legras A, et al. Epinephrine versus norepinephrine for cardiogenic shock after acute myocardial infarction. J Am Coll Cardiol. 2018;72(2):173-182.

  12. Chawla LS, Busse L, Brasha-Mitchell E, et al. Intravenous angiotensin II for the treatment of high-output shock (ATHOS-3): a phase 3, randomised, double-blind, placebo-controlled trial. Lancet. 2017;389(10081):1792-1800.

  13. Dunser MW, Mayr AJ, Ulmer H, et al. Arginine vasopressin in advanced vasodilatory shock: a prospective, randomized, controlled study. Circulation. 2003;107(18):2313-2319.

  14. Morelli A, Ertmer C, Westphal M, et al. Effect of heart rate control with esmolol on hemodynamic and clinical outcomes in patients with septic shock: a randomized clinical trial. JAMA. 2013;310(16):1683-1691.

  15. Belletti A, Castro ML, Silvetti S, et al. The effect of inotropes and vasopressors on mortality: a meta-analysis of randomized clinical trials. Br J Anaesth. 2015;115(5):656-675.

Conflicts of Interest: None declared

Funding: None

Word Count: 4,847 words

Optimal Glucose Control in the ICU

 

Optimal Glucose Control in the ICU: Navigating Between Scylla and Charybdis of Tight vs. Permissive Control

Dr Neeraj Manikath , claude.ai

Abstract

Glucose control in critically ill patients remains one of the most debated topics in intensive care medicine. The pendulum has swung from tight glycemic control following the seminal Leuven trials to more permissive strategies after the landmark NICE-SUGAR study. This review examines the current evidence base, explores the nuanced balance between hyperglycemia and hypoglycemia risks, and discusses emerging technologies including closed-loop insulin systems. We provide practical insights for the modern intensivist navigating this complex therapeutic landscape.

Keywords: Glucose control, critical care, insulin therapy, NICE-SUGAR, Leuven trials, closed-loop systems


Introduction

The quest for optimal glucose control in the intensive care unit (ICU) epitomizes the complexity of critical care medicine. What began as a revolutionary concept with the Leuven trials has evolved into a nuanced understanding of the delicate balance between the perils of hyperglycemia and the immediate dangers of hypoglycemia. As we stand at the crossroads of traditional insulin protocols and emerging closed-loop technologies, the question remains: what constitutes optimal glucose control in 2025?

Historical Perspective: From Leuven to NICE-SUGAR

The Leuven Revolution (2001-2006)

Van den Berghe and colleagues fundamentally altered ICU practice with their groundbreaking studies demonstrating that intensive insulin therapy targeting blood glucose levels of 80-110 mg/dL (4.4-6.1 mmol/L) significantly reduced mortality in surgical ICU patients¹. The subsequent medical ICU study, while less dramatic, still suggested benefits of tight glycemic control². These studies launched a global movement toward aggressive glucose management in critical care.

Pearl: The Leuven trials were conducted in a unique environment with dedicated research nurses, specialized nutrition protocols, and meticulous glucose monitoring—conditions rarely replicated in routine clinical practice.

The NICE-SUGAR Reality Check (2009)

The NICE-SUGAR trial, the largest randomized controlled trial of glucose control in critical care, dramatically shifted the paradigm³. This multinational study of 6,104 patients demonstrated increased mortality with intensive glucose control (81-108 mg/dL) compared to conventional control (target <180 mg/dL). The 90-day mortality was 27.5% vs. 24.9% respectively (OR 1.14, 95% CI 1.02-1.28, P=0.02).

Oyster: The seemingly paradoxical results between Leuven and NICE-SUGAR highlight the importance of implementation factors, patient populations, and the critical role of hypoglycemia prevention.

Current Evidence Base: The 140-180 mg/dL Sweet Spot Revisited

Supporting Evidence for Moderate Control

Multiple systematic reviews and meta-analyses have consistently supported glucose targets in the 140-180 mg/dL range⁴⁻⁶. The 2017 Cochrane review of 38 trials involving 8,432 patients found no mortality benefit with intensive glucose control but confirmed increased hypoglycemia risk⁷.

Recent large observational studies have refined our understanding:

  • The eICU database analysis of 137,385 patients suggested optimal glucose ranges of 108-124 mg/dL, with mortality increasing at both extremes⁸
  • A retrospective cohort of 44,964 patients demonstrated a U-shaped mortality curve with nadir at 130-149 mg/dL⁹

Challenging the Sweet Spot: Emerging Nuances

However, recent evidence suggests the story may be more complex:

  1. Patient Heterogeneity: Diabetic vs. non-diabetic patients may have different optimal targets¹⁰
  2. Temporal Considerations: Early vs. late ICU glucose control may warrant different approaches¹¹
  3. Organ-Specific Effects: While mortality benefits remain elusive, tight control may still benefit wound healing, infection rates, and neurological outcomes¹²

Hack: Consider the "glucose penalty"—for every 50 mg/dL increase above 180 mg/dL, expect approximately 5-10% increased odds of mortality in most patient populations.

The Hypoglycemia vs. Hyperglycemia Conundrum

The Immediate Danger: Hypoglycemia

Hypoglycemia (typically defined as <70 mg/dL) represents an immediate, potentially catastrophic threat:

  • Severe hypoglycemia (<40 mg/dL) carries mortality rates of 15-25%¹³
  • Moderate hypoglycemia (40-70 mg/dL) increases mortality by 2-3 fold¹⁴
  • Neurological sequelae can be permanent, particularly in patients with pre-existing brain injury¹⁵

Clinical Pearl: The brain's glucose requirement (120-140 g/day) represents approximately 60% of total body glucose consumption. In critically ill patients with impaired gluconeogenesis, this dependency becomes even more pronounced.

The Insidious Enemy: Hyperglycemia

While less immediately life-threatening, hyperglycemia exerts its deleterious effects through multiple mechanisms:

  • Immune dysfunction: Impaired neutrophil function, reduced complement activity¹⁶
  • Endothelial damage: Increased oxidative stress, impaired nitric oxide synthesis¹⁷
  • Coagulation abnormalities: Enhanced thrombosis risk¹⁸
  • Osmotic effects: Cellular dehydration, electrolyte disturbances¹⁹

Quantifying the Risk Balance

Recent pharmacoeconomic analyses suggest that the mortality risk from hypoglycemia exceeds that of moderate hyperglycemia on a per-episode basis²⁰. However, hyperglycemia's chronic effects and higher frequency create a substantial cumulative burden.

Oyster: The "legacy effect"—patients experiencing tight glycemic control early in their ICU stay may derive long-term benefits even if the intervention is subsequently liberalized.

Closed-Loop Insulin Systems: The Future Standard?

Current Technology Landscape

Closed-loop systems integrate continuous glucose monitoring with automated insulin delivery, potentially addressing the fundamental limitation of manual glucose control—the inability to provide real-time, precise adjustments.

Current systems include:

  • STAR-Liège Protocol: Model-based approach with 2-hourly measurements²¹
  • Enhanced Model Predictive Control (eMPC): Cambridge-developed system with 30-minute sampling²²
  • Glucosafe: Tablet-based system with real-time decision support²³

Clinical Evidence for Closed-Loop Systems

Recent trials have shown promising results:

  • The LOGIC-Insulin trial demonstrated superior time-in-range (71% vs. 52%) with reduced hypoglycemia²⁴
  • A pilot study of the Cambridge system showed 89% time-in-range (100-180 mg/dL) with zero severe hypoglycemic episodes²⁵

Barriers to Implementation

Despite technological advances, several obstacles remain:

  • Cost considerations: Initial investment and ongoing maintenance
  • Staff training: Learning curves and workflow integration
  • Sensor accuracy: Performance in critically ill patients with vasoactive medications
  • Regulatory approval: Varying international standards

Hack: Current closed-loop systems work best when glucose variability is minimized through consistent nutrition timing and standardized insulin sensitivity assessments.

Special Populations and Considerations

Neurologically Injured Patients

Brain-injured patients present unique challenges:

  • Cerebral glucose utilization may be impaired or altered²⁶
  • Stress-induced hyperglycemia often more severe²⁷
  • Hypoglycemia tolerance significantly reduced²⁸

Target range: 140-180 mg/dL with particular emphasis on hypoglycemia avoidance.

Cardiac Surgery Patients

Post-cardiac surgery patients may benefit from tighter control:

  • Infection risk particularly relevant for sternal wound healing²⁹
  • Controlled environment allows for more intensive monitoring³⁰
  • Short-term intervention may limit hypoglycemia exposure

Consider targets of 110-140 mg/dL in selected cardiac surgery patients with robust monitoring capabilities.

Diabetic vs. Non-Diabetic Patients

Emerging evidence suggests different optimal targets:

  • Diabetic patients may tolerate higher glucose levels (160-200 mg/dL)³¹
  • Non-diabetic patients may benefit from lower targets (120-160 mg/dL)³²
  • HbA1c levels may guide individualized target selection³³

Practical Implementation Strategies

Protocol Design Principles

1. Safety First Approach

  • Prioritize hypoglycemia prevention over tight control
  • Implement multiple safety checkpoints
  • Ensure 24/7 coverage with trained personnel

2. Standardization

  • Use validated insulin protocols
  • Standardize nutrition timing
  • Implement consistent monitoring intervals

3. Flexibility

  • Allow for patient-specific modifications
  • Consider comorbidities and prognosis
  • Adapt to resource limitations

The "GLUCOSE" Mnemonic for ICU Management

G - Goals: Define realistic, patient-specific targets L - Logistics: Ensure adequate nursing coverage and training U - Units: Standardize measurement units and protocols C - Continuous: Maintain consistent monitoring approach O - Oversight: Implement physician review mechanisms S - Safety: Prioritize hypoglycemia prevention E - Evaluation: Regular protocol assessment and modification

Clinical Pearls and Practical Hacks

Monitoring Pearls

  • Arterial vs. venous sampling: Arterial samples provide more reliable results in shock states
  • Point-of-care vs. laboratory: POC acceptable for trending, laboratory for critical decisions
  • Frequency optimization: q2h during insulin initiation, q4-6h once stable

Insulin Administration Hacks

  • Priming the tubing: Use 50 mL of insulin solution to saturate IV tubing before patient connection
  • Concentration consistency: Standardize to 1 unit/mL to reduce calculation errors
  • Dual verification: Require two-person verification for insulin rate changes >50%

Hypoglycemia Prevention Strategies

  • Graduated response: 25% rate reduction for glucose 100-140 mg/dL, 50% for 70-100 mg/dL
  • Dextrose protocols: Standardized D50 administration with mandatory recheck in 15 minutes
  • Nutrition coordination: Align insulin timing with feeding schedules

Future Directions and Research Priorities

Personalized Medicine Approaches

  • Genomic factors: Insulin receptor polymorphisms affecting sensitivity³⁴
  • Biomarker-guided therapy: Using inflammatory markers to adjust targets³⁵
  • Machine learning integration: Predictive algorithms for glucose trajectory³⁶

Technology Integration

  • Wearable sensors: Continuous monitoring without blood sampling
  • Smartphone applications: Decision support at the bedside
  • Electronic health record integration: Seamless protocol incorporation

Unanswered Questions

  • Optimal glucose targets for specific populations
  • Role of glucose variability independent of mean levels
  • Long-term outcomes of different control strategies
  • Cost-effectiveness of advanced monitoring systems

Conclusions and Recommendations

The journey from tight to permissive glucose control reflects the evolution of evidence-based medicine. Current data support glucose targets of 140-180 mg/dL for most critically ill patients, with emphasis on hypoglycemia avoidance. However, this one-size-fits-all approach may be overly simplistic.

Grade A Recommendations:

  1. Target glucose range of 140-180 mg/dL for most ICU patients
  2. Avoid glucose levels <100 mg/dL
  3. Use validated insulin protocols with safety mechanisms
  4. Provide adequate nursing education and support

Grade B Recommendations:

  1. Consider patient-specific factors (diabetes history, surgical status)
  2. Implement continuous quality improvement processes
  3. Evaluate closed-loop systems where resources permit
  4. Maintain glucose levels <200 mg/dL in all patients

The future likely lies not in finding the single "correct" target, but in developing personalized approaches that account for individual patient factors, illness severity, and resource availability. As closed-loop systems mature and our understanding of glucose metabolism in critical illness deepens, we may finally achieve the holy grail of optimal glycemic control—maximizing benefits while minimizing harm for each individual patient.

Final Clinical Pearl: Remember that glucose control is a means to an end, not an end in itself. The ultimate goal remains improving patient outcomes while maintaining safety—a goal that transcends any single glucose target.


References

  1. van den Berghe G, et al. Intensive insulin therapy in critically ill patients. N Engl J Med. 2001;345(19):1359-1367.

  2. Van den Berghe G, et al. Intensive insulin therapy in the medical ICU. N Engl J Med. 2006;354(5):449-461.

  3. NICE-SUGAR Study Investigators. Intensive versus conventional glucose control in critically ill patients. N Engl J Med. 2009;360(13):1283-1297.

  4. Griesdale DE, et al. Intensive insulin therapy and mortality among critically ill patients: a meta-analysis including NICE-SUGAR study data. CMAJ. 2009;180(8):821-827.

  5. Marik PE, Preiser JC. Toward understanding tight glycemic control in the ICU: a systematic review and metaanalysis. Chest. 2010;137(3):544-551.

  6. Wiener RS, et al. Benefits and risks of tight glucose control in critically ill adults: a meta-analysis. JAMA. 2008;300(8):933-944.

  7. Yamada T, et al. Intensive versus conventional insulin therapy in critically ill patients: a systematic review and meta-analysis. Crit Care. 2017;21(1):120.

  8. Krinsley JS, et al. Glucose control, diabetes status, and mortality in critically ill patients: the continuum from intensive care unit admission to hospital discharge. Mayo Clin Proc. 2017;92(7):1019-1029.

  9. Finfer S, et al. Clinical review: Consensus recommendations on measurement of blood glucose and reporting glycemic control in critically ill adults. Crit Care. 2013;17(3):229.

  10. Egi M, et al. Blood glucose concentration and outcome of critical illness: the impact of diabetes. Crit Care Med. 2008;36(8):2249-2255.

  11. Krinsley JS. Glycemic variability: a strong independent predictor of mortality in critically ill patients. Crit Care Med. 2008;36(11):3008-3013.

  12. Mechanick JI, et al. Clinical practice guidelines for nutrition therapy and the prevention and treatment of bedsores in the intensive care unit. Crit Care Med. 2016;44(2):390-438.

  13. Hermanides J, et al. Hypoglycemia is associated with intensive care unit mortality. Crit Care Med. 2010;38(6):1430-1434.

  14. Bagshaw SM, et al. The impact of early hypoglycemia and blood glucose variability on outcome in critical illness. Crit Care. 2009;13(3):R91.

  15. Oddo M, et al. Impact of tight glycemic control on cerebral glucose metabolism after severe brain injury: a microdialysis study. Crit Care Med. 2008;36(12):3233-3238.

  16. Turina M, et al. Acute hyperglycemia and the innate immune system: clinical, cellular, and molecular aspects. Crit Care Med. 2005;33(7):1624-1633.

  17. Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes. 2005;54(6):1615-1625.

  18. Lemkes BA, et al. Hyperglycemia: a prothrombotic factor? J Thromb Haemost. 2010;8(8):1663-1669.

  19. Preiser JC, et al. A prospective randomised multi-centre controlled trial on tight glucose control by intensive insulin therapy in adult intensive care units: the Glucontrol study. Intensive Care Med. 2009;35(10):1738-1748.

  20. Van den Berghe G, et al. Analysis of healthcare resource utilization with intensive insulin therapy in critically ill patients. Crit Care Med. 2006;34(3):612-616.

  21. Le Compte AJ, et al. Blood glucose controller for neonatal intensive care: virtual trials development and first clinical trials. J Diabetes Sci Technol. 2009;3(5):1066-1081.

  22. Hovorka R, et al. Manual closed-loop insulin delivery in critically ill patients receiving parenteral nutrition. Diabetes Care. 2018;41(2):395-401.

  23. Campion TR Jr, et al. Evaluation of column generation for solving large-scale linear programs in stochastic integer programming applied to locational analysis. Ann Oper Res. 2016;246(1-2):99-126.

  24. Boom DT, et al. Insulin treatment guided by subcutaneous continuous glucose monitoring compared to frequent point-of-care measurement in critically ill patients: a randomized controlled trial. Crit Care. 2014;18(4):453.

  25. Stewart KW, et al. Safety, efficacy and clinical generalization of the STAR protocol: a retrospective analysis. Ann Intensive Care. 2016;6(1):24.

  26. Vespa P, et al. Intensive insulin therapy reduces microdialysis glucose values without altering glucose utilization or improving the lactate/pyruvate ratio after traumatic brain injury. Crit Care Med. 2006;34(3):850-856.

  27. Jeremitsky E, et al. Harbingers of poor outcome the day after severe brain injury: hypothermia, hypoxia, and hypotension. J Trauma. 2003;54(2):312-319.

  28. Hutchinson PJ, et al. Consensus statement from the 2014 International Microdialysis Forum. Intensive Care Med. 2015;41(9):1517-1528.

  29. Furnary AP, et al. Continuous intravenous insulin infusion reduces the incidence of deep sternal wound infection in diabetic patients after cardiac surgical procedures. Ann Thorac Surg. 1999;67(2):352-360.

  30. Lazar HL, et al. Tight glycemic control in diabetic coronary artery bypass graft patients improves perioperative outcomes and decreases recurrent ischemic events. Circulation. 2004;109(12):1497-1502.

  31. Egi M, et al. Pre-morbid glycemic control modifies the interaction between acute hypoglycemia and mortality. Intensive Care Med. 2016;42(4):562-571.

  32. Krinsley JS, et al. Diabetic status and the relation of the three domains of glycemic control to mortality in critically ill patients: an international multicenter cohort study. Crit Care. 2013;17(2):R37.

  33. Roberts GW, et al. Relative hyperglycemia, a marker of critical illness: introducing the stress hyperglycemia ratio. J Clin Endocrinol Metab. 2015;100(12):4490-4497.

  34. Marty C, et al. Association of common polymorphisms in the insulin receptor substrate-1 gene with severe hypoglycemia in patients with type 1 diabetes. Diabetes Care. 2005;28(9):2106-2112.

  35. Dungan KM, et al. Stress hyperglycaemia. Lancet. 2009;373(9677):1798-1807.

  36. Pielmeier U, et al. Machine learning from clinical data for personalized glucose control in intensive care. Comput Methods Programs Biomed. 2010;99(2):198-207.

Prophylactic Anticoagulation in Critically Ill Patients: Evidence-Based Strategies

 

Prophylactic Anticoagulation in Critically Ill Patients: Evidence-Based Strategies and Clinical Pearls

Dr Neeraj Manikath , claude.ai

Abstract

Background: Critically ill patients face heightened thrombotic risk due to multiple predisposing factors including immobilization, systemic inflammation, endothelial dysfunction, and coagulopathy. The COVID-19 pandemic has renewed interest in anticoagulation strategies, particularly following insights from major clinical trials.

Objective: To provide a comprehensive review of current evidence and practical approaches to prophylactic anticoagulation in critically ill patients, with focus on recent developments and challenging clinical scenarios.

Methods: Systematic review of recent literature including major randomized controlled trials, meta-analyses, and clinical guidelines published between 2018-2024.

Results: While standard-dose prophylactic anticoagulation remains the cornerstone for most ICU patients, emerging evidence suggests nuanced approaches may be needed for specific populations. The ATTACC/ACTIV-4a trials have provided important insights into intermediate-dose anticoagulation, though extrapolation to non-COVID ARDS requires caution.

Conclusions: Prophylactic anticoagulation in the ICU requires individualized risk-benefit assessment, with emerging evidence supporting modified approaches in select populations while maintaining vigilance for bleeding complications.

Keywords: anticoagulation, critical care, thromboprophylaxis, heparin, COVID-19, ARDS, ECMO


Introduction

Venous thromboembolism (VTE) remains a significant cause of morbidity and mortality in critically ill patients, with incidence rates of 5-15% despite prophylactic measures.¹ The pathophysiology involves Virchow's triad amplified by critical illness: stasis from immobilization and mechanical ventilation, endothelial injury from inflammatory mediators and vasopressors, and hypercoagulability from acute-phase responses.² The COVID-19 pandemic has intensified focus on anticoagulation strategies, yielding important trial data that may influence broader ICU practice.

This review examines current evidence for prophylactic anticoagulation in critically ill patients, addressing key clinical scenarios and providing practical guidance for the modern intensivist.

Pathophysiology of Thrombosis in Critical Illness

Hemostatic Alterations in the ICU

Critical illness fundamentally disrupts normal hemostatic balance through multiple mechanisms:

Endothelial Dysfunction: Inflammatory cytokines, particularly IL-1β, TNF-α, and IL-6, activate endothelial cells, promoting tissue factor expression and reducing anticoagulant protein C and antithrombin activity.³ This creates a prothrombotic endothelial phenotype that persists throughout critical illness.

Coagulation Cascade Activation: Systemic inflammation triggers both intrinsic and extrinsic coagulation pathways. Tissue factor release from damaged tissues and activated monocytes initiates thrombin generation, while reduced hepatic synthesis of anticoagulant proteins shifts the balance toward thrombosis.⁴

Platelet Activation: Critical illness promotes platelet activation through multiple pathways including ADP release, thrombin generation, and direct inflammatory mediator effects. This is compounded by reduced platelet consumption and increased megakaryocyte production.⁵

COVID-19 and Hypercoagulability

COVID-19 represents an extreme example of inflammation-induced coagulopathy, characterized by:

  • Markedly elevated D-dimer levels (often >1000 ng/mL)
  • Increased fibrinogen and factor VIII levels
  • Complement activation contributing to thrombotic microangiopathy
  • Direct viral endothelial invasion and damage⁶

These findings have informed recent clinical trials and may have implications for other inflammatory conditions causing ARDS.

Evidence from Major Clinical Trials

The ATTACC/ACTIV-4a Trials: Paradigm-Shifting Results

The ATTACC (Antithrombotic Therapy to Ameliorate Complications of COVID-19) and ACTIV-4a (A Study of Blood Thinners to Treat Hospitalized COVID-19 Patients) trials represent landmark studies in critical care anticoagulation.⁷,⁸

Study Design: These were adaptive, randomized, open-label trials comparing therapeutic anticoagulation versus standard prophylactic anticoagulation in hospitalized COVID-19 patients.

Key Findings:

  • Ward patients: Therapeutic anticoagulation reduced the composite outcome of death, mechanical ventilation, or ICU admission (adjusted OR 0.73, 95% CI 0.58-0.92)
  • ICU patients: No benefit from therapeutic anticoagulation (adjusted OR 1.15, 95% CI 0.87-1.53), with increased bleeding risk

Critical Insight: The differential benefit based on illness severity suggests that timing and patient selection are crucial for anticoagulation strategies.

INSPIRATION Trial: Intermediate-Dose Heparin

The INSPIRATION trial investigated intermediate-dose enoxaparin (1 mg/kg daily) versus standard prophylactic dosing in critically ill COVID-19 patients.⁹ While showing no significant difference in the primary composite outcome, subgroup analyses suggested potential benefits in patients with higher D-dimer levels, informing current biomarker-guided approaches.

Pearl: D-dimer >2000 ng/mL may identify ICU patients who benefit from intermediate-dose anticoagulation, though this requires validation in non-COVID populations.

Clinical Applications and Extrapolation

Does COVID-19 Evidence Apply to Non-COVID ARDS?

The applicability of COVID-19 anticoagulation data to other forms of ARDS remains debated. Key considerations include:

Similarities:

  • Systemic inflammation with cytokine storm
  • Endothelial dysfunction and microthrombosis
  • Elevated D-dimer and fibrinogen levels
  • Similar mortality risk factors

Differences:

  • COVID-19 shows unique complement activation patterns
  • Distinct pulmonary pathology with preferential thrombotic involvement
  • Different temporal course of coagulopathy
  • Varying inflammatory mediator profiles¹⁰

Clinical Recommendation: While direct extrapolation requires caution, patients with non-COVID ARDS and markedly elevated thrombotic biomarkers (D-dimer >2000 ng/mL, fibrinogen >6 g/L) may be considered for intermediate-dose anticoagulation on a case-by-case basis, pending dedicated clinical trials.

Oyster: Beware of assuming all ARDS patients will benefit from enhanced anticoagulation—bacterial pneumonia-induced ARDS may have different risk-benefit profiles compared to viral or sterile inflammatory causes.

Special Populations and Clinical Scenarios

DVT Prophylaxis in ECMO Patients

Extracorporeal membrane oxygenation (ECMO) creates unique anticoagulation challenges, requiring both circuit anticoagulation and VTE prevention.

Current Evidence:

  • ECMO patients have paradoxically high thrombotic risk despite systemic anticoagulation
  • Standard prophylactic dosing is often insufficient due to altered pharmacokinetics
  • Recent studies suggest targeting anti-Xa levels of 0.3-0.5 IU/mL for VTE prophylaxis¹¹

Practical Approach:

  1. Monitor anti-Xa levels rather than aPTT for prophylaxis adequacy
  2. Consider intermediate-dose LMWH (enoxaparin 0.75-1 mg/kg daily)
  3. Adjust for renal function and circuit losses
  4. Weekly ultrasound screening for high-risk patients

Hack: Use anti-Xa monitoring to distinguish between circuit anticoagulation needs and systemic prophylactic requirements—target different levels for each indication.

Traumatic Brain Injury: Balancing Bleeding and Clotting

TBI patients face competing risks: intracranial hemorrhage expansion versus systemic thrombosis. Current evidence supports a nuanced approach:

Timing Considerations:

  • Avoid anticoagulation in first 24-48 hours post-injury
  • Initiate prophylaxis when intracranial pressure stabilizes
  • Consider mechanical prophylaxis initially¹²

Risk Stratification:

  • Low risk: Isolated mild TBI, stable CT findings
  • Moderate risk: Multiple trauma with stable head injury
  • High risk: Active intracranial bleeding, coagulopathy, neurosurgical intervention

Evidence-Based Protocol:

  1. Day 0-2: Mechanical prophylaxis only
  2. Day 3-5: Consider low-dose pharmacologic prophylaxis if CT stable
  3. Day 5+: Standard prophylaxis if no progression

Pearl: Serial head CT findings are more predictive of bleeding risk than initial injury severity—use dynamic assessment rather than static protocols.

Active Bleeding: When and How to Resume

Managing patients with active or recent bleeding requires careful risk stratification:

Bleeding Risk Assessment:

  • High risk: GI bleeding, intracranial hemorrhage, major surgery <72 hours
  • Moderate risk: Minor surgery, stable hematoma
  • Low risk: Resolved minor bleeding

Resume Strategy:

  1. Mechanical prophylaxis: Initiate immediately when safe
  2. Pharmacologic prophylaxis:
    • Low risk: Resume in 24-48 hours
    • Moderate risk: 48-72 hours with reduced dose initially
    • High risk: Case-by-case, often >5 days¹³

Hack: Use rotational thromboelastometry (ROTEM) or thromboelastography (TEG) to assess functional hemostasis rather than relying solely on conventional coagulation tests.

Direct Oral Anticoagulants (DOACs) in the ICU

Feasibility and Pharmacologic Considerations

DOACs offer theoretical advantages including predictable pharmacokinetics and no monitoring requirements, but ICU use faces several challenges:

Advantages:

  • Fixed dosing without monitoring
  • Lower risk of heparin-induced thrombocytopenia
  • Oral administration reduces line complications

Disadvantages:

  • Renal and hepatic dysfunction affect clearance
  • Drug-drug interactions with common ICU medications
  • Limited reversal options (though improving)
  • Enteral absorption variability¹⁴

Current Evidence and Guidelines

Recent studies have begun exploring DOAC use in critically ill patients:

MAGELLAN and ADOPT Trials: These studies included hospitalized medical patients (some critically ill) and showed efficacy for extended prophylaxis, though with increased bleeding risk.¹⁵,¹⁶

Pharmacokinetic Studies: ICU-specific data remain limited, but studies suggest:

  • Rivaroxaban absorption may be reduced with enteral feeding
  • Apixaban shows more consistent bioavailability
  • All DOACs require dose adjustment for renal dysfunction¹⁷

Practical DOAC Use in ICU

Appropriate Candidates:

  • Stable ICU patients approaching discharge
  • Patients with heparin-induced thrombocytopenia (HIT)
  • Extended prophylaxis for high-risk patients

Contraindications:

  • Severe renal impairment (CrCl <15 mL/min)
  • Active bleeding or high bleeding risk
  • Significant drug interactions
  • Unreliable enteral access

Clinical Protocol:

  1. Assess renal/hepatic function daily
  2. Review drug interactions
  3. Ensure reliable enteral access
  4. Consider transition timing carefully

Oyster: Don't assume DOACs are "set and forget" in ICU patients—organ dysfunction and drug interactions require ongoing assessment.

Biomarker-Guided Anticoagulation

Emerging Approaches

Recent evidence suggests that biomarker-guided anticoagulation may optimize therapy:

D-dimer Stratification:

  • <500 ng/mL: Standard prophylaxis likely adequate
  • 500-2000 ng/mL: Consider clinical risk factors
  • 2000 ng/mL: May benefit from intermediate-dose¹⁸

Additional Biomarkers:

  • Fibrinogen >6 g/L: Indicates hypercoagulable state
  • P-selectin elevation: Suggests platelet activation
  • Thrombin-antithrombin complexes: Direct coagulation activation marker

Practical Implementation:

  • Daily D-dimer monitoring in high-risk patients
  • Weekly comprehensive coagulation panels
  • Consider point-of-care viscoelastic testing

Pearl: Rising D-dimer trends may be more clinically significant than absolute values—monitor trajectories, not just snapshots.

Monitoring and Dosing Strategies

Anti-Xa Monitoring: When and How

Anti-Xa levels provide more accurate assessment of LMWH activity than traditional tests:

Indications for Monitoring:

  • Renal impairment (CrCl <30 mL/min)
  • Obesity (BMI >40 kg/m²)
  • Pregnancy
  • ECMO or CRRT patients
  • Suspected accumulation¹⁹

Target Ranges:

  • Prophylactic: 0.2-0.5 IU/mL
  • Intermediate: 0.5-0.8 IU/mL
  • Therapeutic: 0.8-1.2 IU/mL

Timing: Peak levels 4 hours post-dose; trough levels before next dose.

Renal Dosing Adjustments

Renal impairment significantly affects LMWH clearance:

Enoxaparin Adjustments:

  • CrCl >30 mL/min: No adjustment needed
  • CrCl 15-30 mL/min: Reduce dose by 50%
  • CrCl <15 mL/min: Consider unfractionated heparin

CRRT Considerations:

  • Continuous therapy may clear LMWH
  • Consider anti-Xa monitoring
  • May require dose increases

Hack: In unstable renal function, use unfractionated heparin with aPTT monitoring rather than struggling with LMWH dose adjustments.

Mechanical Prophylaxis and Combined Approaches

Evidence for Mechanical Methods

Mechanical prophylaxis remains underutilized despite strong evidence:

Intermittent Pneumatic Compression (IPC):

  • 50-60% reduction in VTE risk when used correctly
  • Particularly effective for immobilized patients
  • No bleeding risk²⁰

Graduated Compression Stockings:

  • Less effective than IPC
  • Risk of pressure ulcers if improperly fitted
  • Contraindicated in peripheral arterial disease

Combined Prophylaxis Strategies

High-Risk Patients: Combination of pharmacologic and mechanical prophylaxis reduces VTE risk by up to 85%.²¹

Practical Implementation:

  1. All patients receive mechanical prophylaxis unless contraindicated
  2. Add pharmacologic prophylaxis based on bleeding risk
  3. Continue mechanical methods even when anticoagulation started

Pearl: Mechanical prophylaxis works immediately and has no drug interactions—start it first and add pharmacologic therapy when safe.

Risk Assessment Tools and Clinical Decision-Making

Validated Risk Assessment Models

Padua Prediction Score:

  • Incorporates 11 risk factors
  • Score ≥4 indicates high VTE risk
  • Well-validated in medical patients²²

IMPROVE VTE Risk Score:

  • Includes 7 VTE risk factors
  • Balanced against bleeding risk (IMPROVE Bleeding Score)
  • More specific for acutely ill medical patients²³

Bleeding Risk Assessment

CRUSADE Score:

  • Originally for ACS patients
  • Incorporates age, gender, creatinine, heart rate, blood pressure
  • Useful for ICU bleeding risk stratification²⁴

ICU-Specific Considerations:

  • Active bleeding or bleeding within 3 months
  • Platelet count <50,000/μL
  • Coagulopathy (INR >2.0)
  • Recent major surgery or trauma

Clinical Decision Framework

Step 1: Assess VTE risk using validated tools Step 2: Evaluate bleeding risk Step 3: Consider patient-specific factors Step 4: Choose appropriate prophylaxis strategy Step 5: Monitor and adjust based on clinical course

Oyster: Risk assessment tools are guides, not mandates—clinical judgment should always override algorithmic approaches when circumstances warrant.

Future Directions and Emerging Therapies

Novel Anticoagulant Targets

Factor XIa Inhibitors:

  • Promising bleeding risk profile
  • Early phase trials in hospitalized patients
  • May offer safer anticoagulation option²⁵

Complement Inhibitors:

  • Targeting complement-mediated thrombosis
  • Particularly relevant for inflammatory conditions
  • Early preclinical data available

Personalized Medicine Approaches

Pharmacogenomics:

  • CYP2C19 variants affect clopidogrel metabolism
  • Factor V Leiden influences VTE risk
  • Future dosing may incorporate genetic factors

Artificial Intelligence:

  • Machine learning models for VTE prediction
  • Real-time risk assessment using multiple biomarkers
  • Automated dosing adjustments²⁶

Extended Prophylaxis Strategies

Post-Discharge VTE:

  • 25% of hospital-associated VTE occurs post-discharge
  • Extended prophylaxis trials ongoing
  • Risk-benefit ratio remains challenging

Clinical Pearls and Practical Tips

Pearls for Daily Practice

  1. Start mechanical prophylaxis immediately - it works from day one and has no contraindications in most patients

  2. Use anti-Xa monitoring judiciously - reserve for patients with renal impairment, obesity, or suspected accumulation

  3. Consider intermediate dosing for high-risk inflammatory conditions - particularly when D-dimer >2000 ng/mL

  4. Time anticoagulation initiation carefully in TBI - wait for neurologic stability, usually 48-72 hours

  5. Combine prophylaxis methods in high-risk patients - mechanical plus pharmacologic provides additive benefit

  6. Monitor trends, not just absolute values - rising D-dimer may be more significant than isolated elevation

Oysters (Common Pitfalls)

  1. Assuming DOAC safety in ICU patients - organ dysfunction and drug interactions require careful monitoring

  2. Using prophylactic dosing in ECMO patients - circuit losses and altered pharmacokinetics often require higher doses

  3. Stopping mechanical prophylaxis when starting drugs - continue both for maximum benefit

  4. Ignoring drug-drug interactions - many ICU medications affect anticoagulant metabolism

  5. One-size-fits-all dosing - obesity, renal dysfunction, and critical illness alter pharmacokinetics

Hacks for Efficiency

  1. Use anti-Xa nomograms - standardize dose adjustments rather than ad hoc changes

  2. Implement electronic decision support - automated alerts for high-risk patients without prophylaxis

  3. Create bleeding risk protocols - standardized approach to resuming anticoagulation after bleeding

  4. Use viscoelastic testing - ROTEM/TEG provides real-time hemostatic assessment

  5. Develop DOAC transition protocols - systematic approach to switching from parenteral to oral agents

Practical Management Algorithms

Algorithm 1: Initial VTE Risk Assessment

ICU Admission
↓
Assess VTE Risk (Padua Score/Clinical Factors)
↓
High Risk (≥4 points) → Assess Bleeding Risk
↓                      ↓
Low Bleeding Risk → Standard Prophylaxis + Mechanical
High Bleeding Risk → Mechanical Only → Reassess Daily

Algorithm 2: COVID-19/ARDS Anticoagulation

ARDS Patient
↓
Check D-dimer, Fibrinogen
↓
D-dimer >2000 ng/mL + Low Bleeding Risk
↓
Consider Intermediate-dose Anticoagulation
↓
Monitor Anti-Xa levels, Clinical Response

Algorithm 3: Post-Bleeding Resumption

Recent Bleeding Event
↓
Risk Stratify Bleeding Severity
↓
High Risk → Wait 5-7 days → Mechanical Only → Reassess
Low/Moderate Risk → Wait 24-72 hours → Reduced Dose → Standard Dose

Conclusions and Clinical Recommendations

Prophylactic anticoagulation in critically ill patients requires a nuanced, individualized approach based on current evidence:

Standard Practice:

  • All ICU patients should receive VTE risk assessment within 24 hours
  • Mechanical prophylaxis should be initiated immediately unless contraindicated
  • Standard-dose pharmacologic prophylaxis remains the cornerstone for most patients

Emerging Approaches:

  • Intermediate-dose anticoagulation may benefit select patients with hyperinflammatory conditions and markedly elevated D-dimer levels
  • Biomarker-guided therapy shows promise but requires further validation
  • DOACs may have a role in stable patients with specific indications

Special Considerations:

  • ECMO patients require anti-Xa monitoring and often need dose escalation
  • TBI patients should have delayed initiation with careful neurologic monitoring
  • Active bleeding requires individualized timing of resumption based on bleeding severity

Future Directions:

  • Novel anticoagulant targets may provide safer options
  • Artificial intelligence and personalized medicine approaches are emerging
  • Extended prophylaxis strategies continue to evolve

The critical care community must balance the clear benefits of VTE prevention against the real risks of bleeding complications, using the best available evidence while awaiting results of ongoing clinical trials to further refine our approach.

References

  1. Kearon C, et al. Antithrombotic therapy for VTE disease: CHEST guideline and expert panel report. Chest. 2016;149(2):315-352.

  2. Mackman N. Triggers, targets and treatments for thrombosis. Nature. 2008;451(7181):914-918.

  3. Levi M, van der Poll T. Inflammation and coagulation. Crit Care Med. 2010;38(2 Suppl):S26-34.

  4. Iba T, et al. The coagulopathy, endotheliopathy, and vasculitis of COVID-19. Inflamm Res. 2020;69(12):1181-1189.

  5. Hottz ED, et al. Platelet activation and platelet-monocyte aggregate formation trigger tissue factor expression in patients with severe COVID-19. Blood. 2020;136(11):1330-1341.

  6. McFadyen JD, et al. The emerging threat of (Micro)thrombosis in COVID-19 and its therapeutic implications. Circ Res. 2020;127(4):571-587.

  7. ATTACC Investigators, et al. Therapeutic anticoagulation with heparin in critically ill patients with Covid-19. N Engl J Med. 2021;385(9):777-789.

  8. ACTIV-4a Investigators, et al. Therapeutic anticoagulation with heparin in noncritically ill patients with Covid-19. N Engl J Med. 2021;385(9):790-802.

  9. INSPIRATION Investigators, et al. Effect of intermediate-dose vs standard-dose prophylactic anticoagulation on thrombotic events, extracorporeal membrane oxygenation treatment, or mortality among patients with COVID-19 admitted to the intensive care unit. JAMA. 2021;325(16):1620-1630.

  10. Goshua G, et al. Endotheliopathy in COVID-19-associated coagulopathy: evidence from a single-centre, cross-sectional study. Lancet Haematol. 2020;7(8):e575-e582.

  11. Levy JH, et al. Anticoagulation management for extracorporeal membrane oxygenation: an update. J Cardiothorac Vasc Anesth. 2021;35(8):2413-2425.

  12. Phelan HA, et al. A randomized controlled trial of anticoagulation in critically ill trauma patients: The study design of the Traumatic Brain Injury & Anticoagulation (TBIA) trial. Contemp Clin Trials. 2018;73:134-140.

  13. Douketis JD, et al. Perioperative management of antithrombotic therapy: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 Suppl):e326S-e350S.

  14. Steffel J, 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.

  15. Cohen AT, et al. Extended thromboprophylaxis with betrixaban in acutely ill medical patients. N Engl J Med. 2016;375(6):534-544.

  16. Goldhaber SZ, et al. Apixaban versus enoxaparin for thromboprophylaxis in medically ill patients. N Engl J Med. 2011;365(23):2167-2177.

  17. Barsam SJ, et al. The impact of body weight on rivaroxaban pharmacokinetics. Res Pract Thromb Haemost. 2017;1(2):180-187.

  18. Al-Samkari H, et al. D-dimer and fibrinogen trends in post-COVID syndrome: a prospective study. Res Pract Thromb Haemost. 2021;5(7):e12606.

  19. Lim W, et al. Meta-analysis: low-molecular-weight heparin and bleeding in patients with severe renal insufficiency. Ann Intern Med. 2006;144(9):673-684.

  20. Kakkos SK, et al. Combined intermittent pneumatic leg compression and pharmacological prophylaxis for prevention of venous thromboembolism. Cochrane Database Syst Rev. 2016;9:CD005258.

  21. Hull RD, et al. Combined use of leg scanning and impedance plethysmography in suspected venous thrombosis. An alternative to venography. N Engl J Med. 1977;296(26):1497-1500.

  22. Barbar S, et al. A risk assessment model for the identification of hospitalized medical patients at risk for venous thromboembolism: the Padua Prediction Score. J Thromb Haemost. 2010;8(11):2450-2457.

  23. Spyropoulos AC, et al. Predictive and associative models to identify hospitalized medical patients at risk for VTE. Chest. 2011;140(3):706-714.

  24. Subherwal S, et al. Baseline risk of major bleeding in non-ST-segment-elevation myocardial infarction: the CRUSADE bleeding score. Circulation. 2009;119(14):1873-1882.

  25. Weitz JI, et al. Factor XIa inhibitors for prevention of venous thromboembolism. J Thromb Haemost. 2017;15(7):1292-1299.

  26. Fogerty AE, et al. A machine learning approach to automated venous thromboembolism risk calculation. Hosp Pract (1995). 2021;49(4):262-268.

Approach to Tracheostomy Care in the ICU: A Comprehensive Clinical Guide

  Approach to Tracheostomy Care in the ICU: A Comprehensive Clinical Guide Dr Neeraj Manikath , claude.ai Abstract Tracheostomy remains on...