Thursday, July 24, 2025

Critical Illness in Sickle Cell Adults

 

The Forgotten Crisis: Critical Illness in Sickle Cell Adults

Dr Neeraj Manikath , claude.ai

Abstract

Background: Sickle cell disease (SCD) affects millions globally, yet critical illness in adults remains underrecognized and inadequately managed. This review addresses key management strategies, focusing on exchange transfusion thresholds, hepatic sequestration, and emerging complement inhibitor therapies.

Objectives: To provide evidence-based guidance for intensivists managing adult SCD patients, highlighting recent advances in therapeutic approaches and diagnostic pearls.

Methods: Comprehensive literature review of peer-reviewed articles, clinical guidelines, and emerging therapies through January 2025.

Results: Critical SCD complications require prompt recognition and aggressive management. Exchange transfusion remains cornerstone therapy with evolving threshold criteria. Hepatic sequestration represents a life-threatening emergency often misdiagnosed. Complement inhibitors show promise in multiorgan failure scenarios.

Conclusions: Improved understanding of SCD pathophysiology and novel therapeutic approaches can significantly improve outcomes in critically ill adults with SCD.

Keywords: Sickle cell disease, critical care, exchange transfusion, hepatic sequestration, complement inhibitors, multiorgan failure


Introduction

Sickle cell disease affects approximately 100,000 Americans and millions worldwide, with the majority surviving into adulthood due to improved childhood care.¹ However, adult SCD patients face unique challenges in critical care settings, where delayed recognition and suboptimal management contribute to excess morbidity and mortality. The transition from pediatric to adult care often results in fragmented healthcare delivery, leaving intensivists ill-equipped to manage these complex patients.²

Critical illness in SCD adults presents distinct pathophysiological challenges. Unlike pediatric presentations dominated by pain crises and acute chest syndrome, adults develop progressive organ dysfunction, particularly affecting pulmonary, renal, and hepatic systems.³ The interplay between chronic hemolysis, endothelial dysfunction, and acute inflammatory responses creates a perfect storm for multiorgan failure.

This review addresses three critical areas often overlooked in adult SCD management: optimal exchange transfusion strategies, recognition and treatment of hepatic sequestration, and the emerging role of complement inhibition in multiorgan failure.


Pathophysiology of Critical Illness in SCD

The Vicious Cycle of Vaso-occlusion

SCD pathophysiology extends beyond simple mechanical obstruction by sickled cells. The disease represents a complex inflammatory vasculopathy involving multiple interconnected pathways:

Hemolysis-induced endothelial dysfunction occurs through free hemoglobin scavenging of nitric oxide, leading to vasoconstriction and reduced vascular reactivity.⁴ Simultaneously, free heme activates toll-like receptor 4, triggering inflammatory cascades that promote further vaso-occlusion.

Complement activation plays an increasingly recognized role in SCD pathophysiology. Chronic hemolysis activates the alternative complement pathway, with C5a and membrane attack complex formation contributing to endothelial injury and microvascular thrombosis.⁵

Adhesion molecule upregulation creates a proinflammatory endothelial phenotype. Increased expression of VCAM-1, ICAM-1, and P-selectin facilitates leukocyte-endothelial interactions, promoting vaso-occlusion and organ dysfunction.⁶

Critical Care Pearl: The "Sickling Threshold"

Unlike common belief, sickling occurs not just with hypoxia but also with dehydration, acidosis, fever, and stress. Maintain SpO₂ >95%, adequate hydration, and strict temperature control. Even "mild" hypoxia (88-92%) can trigger cascading vaso-occlusion in susceptible patients.


Exchange Transfusion: Redefining Thresholds and Timing

Traditional Approach vs. Modern Evidence

Historically, exchange transfusion decisions relied on HbS levels >30% or clinical severity scores. Recent evidence suggests this approach may be inadequate for critically ill adults, where earlier intervention yields superior outcomes.⁷

Evidence-Based Thresholds for Exchange Transfusion

Acute Chest Syndrome (ACS):

  • Immediate exchange: PaO₂/FiO₂ ratio <200, bilateral infiltrates, or requirement for mechanical ventilation
  • Urgent exchange: Single lobe involvement with declining oxygenation despite optimal medical therapy
  • Target: HbS <30%, Hgb 9-11 g/dL⁸

Acute Stroke:

  • Emergent exchange: Any acute neurological deficit with imaging confirmation
  • Target: HbS <15% within 4 hours, maintain <30% for minimum 30 days
  • Monitoring: Transcranial Doppler velocities if available⁹

Multiorgan Failure:

  • Threshold: Two or more organ systems with evidence of vaso-occlusion
  • Target: HbS <20%, considering patient's baseline hemoglobin
  • Duration: Continue until organ function stabilizes¹⁰

Critical Care Hack: The "Golden Hour" Concept

In severe vaso-occlusive crises with organ dysfunction, the first hour determines outcome. If exchange transfusion is indicated, initiate within 60 minutes of decision. Delays beyond 2 hours significantly increase mortality risk.

Automated vs. Manual Exchange Protocols

Automated Exchange (Preferred):

  • More precise HbS reduction
  • Better volume control in fluid-sensitive patients
  • Reduced procedure time and nursing requirements
  • Lower risk of hemodynamic instability¹¹

Manual Exchange Protocol: When automated exchange unavailable, use the following approach:

  1. Calculate blood volume: 70 mL/kg (men), 65 mL/kg (women)
  2. Exchange 1.5-2 blood volumes over 4-6 hours
  3. Monitor HbS every 250-500 mL exchanged
  4. Maintain isovolemia with careful fluid balance

Oyster Alert: Delayed Hemolytic Transfusion Reactions

SCD patients have 25-30% risk of delayed hemolytic transfusion reactions, often occurring 7-14 days post-transfusion. Watch for: unexplained anemia, reticulocytosis, increased LDH, and new alloantibodies. This can precipitate hyperhemolysis syndrome, requiring immunosuppression rather than additional transfusions.¹²


Hepatic Sequestration: The Great Mimicker

Pathophysiology and Clinical Presentation

Hepatic sequestration represents acute trapping of sickled cells within hepatic sinusoids, leading to rapid hepatomegaly, anemia, and potential liver failure. Unlike pediatric presentations, adult hepatic sequestration often presents insidiously, mimicking other hepatic emergencies.¹³

Clinical Triad:

  1. Rapid hepatomegaly (>2 cm increase from baseline)
  2. Acute anemia (>2 g/dL drop from baseline)
  3. Evidence of hemolysis (elevated LDH, indirect bilirubin)

Diagnostic Challenges in Adults

Adult hepatic sequestration frequently masquerades as:

  • Acute hepatitis: Elevated ALT/AST with hepatomegaly
  • Choledocholithiasis: RUQ pain with hyperbilirubinemia
  • Budd-Chiari syndrome: Hepatomegaly with ascites
  • Heart failure: Hepatomegaly with elevated JVP¹⁴

Diagnostic Pearl: The "Reverse Hemoglobin-Bilirubin Dissociation"

In hepatic sequestration, hemoglobin drops faster than expected for the degree of hemolysis. Calculate the hemolysis index: (Total bilirubin - Direct bilirubin) × 20. If hemoglobin drop exceeds this calculation, suspect sequestration.

Management Strategies

Immediate Assessment:

  • Hemoglobin, reticulocyte count, LDH, bilirubin fractionation
  • Liver function tests including PT/INR
  • Hepatic ultrasound with Doppler (rule out vascular occlusion)
  • Blood type and crossmatch for transfusion

Therapeutic Approach:

Mild Sequestration (Hgb >7 g/dL, stable vitals):

  • Simple transfusion to hemoglobin 9-10 g/dL
  • Avoid over-transfusion (risk of hyperviscosity)
  • Close monitoring for progression¹⁵

Severe Sequestration (Hgb <7 g/dL or hemodynamic instability):

  • Immediate exchange transfusion
  • Target HbS <30%, hemoglobin 9-10 g/dL
  • Consider partial exchange if simple transfusion initiated

Critical Care Hack: The "Hepatic Handshake" Sign Palpate the liver edge while monitoring central venous pressure. In hepatic sequestration, liver edge becomes increasingly prominent with inspiration (positive hepatic-jugular reflux), unlike other causes of hepatomegaly where this relationship is absent or minimal.

Complications and Monitoring

Hepatic Failure Indicators:

  • INR >1.5 with clinical bleeding
  • Encephalopathy (often subtle in SCD patients)
  • Worsening synthetic function (albumin, cholinesterase)

Management of Hepatic Failure:

  • N-acetylcysteine 150 mg/kg loading dose, then 50 mg/kg q4h
  • Fresh frozen plasma for coagulopathy with bleeding
  • Consider early hepatology consultation for potential transplant evaluation¹⁶

Complement Inhibitors: A Novel Therapeutic Frontier

Rationale for Complement Inhibition

Recent research has identified complement activation as a central mediator of SCD pathophysiology. The alternative complement pathway remains constitutively activated in SCD patients, with evidence of terminal complement complex deposition in multiple organs during crisis.¹⁷

Ravulizumab in SCD: Emerging Evidence

Ravulizumab, a long-acting C5 complement inhibitor, represents a paradigm shift in SCD management. Unlike eculizumab, ravulizumab's extended half-life allows for 8-weekly dosing, improving patient compliance and steady-state coverage.¹⁸

Mechanism of Action:

  • Inhibits C5 cleavage, preventing C5a and C5b-9 formation
  • Reduces complement-mediated hemolysis
  • Decreases endothelial activation and adhesion molecule expression
  • Attenuates inflammatory cascade propagation¹⁹

Clinical Applications in Critical Care

Multiorgan Failure Syndrome: Emerging case series suggest benefit in SCD patients with multiorgan failure, particularly when combined with exchange transfusion. The proposed mechanism involves:

  • Reduced complement-mediated endothelial injury
  • Decreased inflammatory cytokine release
  • Improved microvascular perfusion²⁰

Dosing Protocol for Critical Care:

  • Loading dose: 2700 mg IV over 35 minutes
  • Maintenance: 3000 mg at day 15, then every 8 weeks
  • Adjust for body weight >100 kg (3300 mg loading, 3600 mg maintenance)

Critical Care Pearl: Meningococcal Prophylaxis

All patients receiving complement inhibitors require meningococcal vaccination (if not contraindicated) and antibiotic prophylaxis. Use penicillin V 250 mg BID or azithromycin 250 mg daily for penicillin-allergic patients. Continue throughout therapy.

Monitoring and Safety Considerations

Pre-treatment Requirements:

  • Meningococcal vaccination (types A, C, W, Y, and B) minimum 2 weeks prior
  • Screen for active infections
  • Baseline complement levels (CH50, C3, C4)

Ongoing Monitoring:

  • Weekly CBC with differential
  • Liver function tests
  • Signs of meningococcal infection
  • Hemolysis markers (LDH, haptoglobin, indirect bilirubin)²¹

Oyster Alert: Breakthrough Hemolysis

Approximately 10-15% of patients on complement inhibitors experience breakthrough hemolysis due to C5 variants with reduced binding affinity. Monitor for unexplained anemia, elevated LDH, and decreased haptoglobin. May require dose escalation or alternative complement targeting.


Multiorgan Failure in SCD: An Integrated Approach

Recognition and Early Intervention

Multiorgan failure in SCD differs from typical ICU presentations. The onset is often insidious, with patients appearing deceptively stable until rapid decompensation occurs.

Early Warning Signs:

  • Acute kidney injury with bland urinalysis
  • Unexplained metabolic acidosis
  • Rapid hemoglobin decline without obvious bleeding
  • New oxygen requirement in previously stable patient²²

Critical Care Hack: The "SCD SOFA" Score

Modify traditional SOFA scoring for SCD patients:

  • Respiratory: Consider baseline oxygen requirement
  • Renal: Adjust for chronic kidney disease baseline
  • Hepatic: Include hemolysis markers in assessment
  • Neurologic: Consider silent strokes in baseline assessment

Therapeutic Bundle for Multiorgan Failure

Hour 0-1:

  • Aggressive hydration (avoid overload in renal dysfunction)
  • Oxygen therapy (target SpO₂ >95%)
  • Exchange transfusion preparation
  • Complement inhibitor consideration

Hour 1-6:

  • Exchange transfusion if indicated
  • Organ-specific support measures
  • Infectious workup and empiric antibiotics
  • Pain management optimization

Hour 6-24:

  • Reassess organ function
  • Consider complement inhibitor if multiorgan failure persists
  • Continuous monitoring of hemolysis markers
  • Early mobility and rehabilitation planning²³

Special Considerations in Critical Care Management

Anesthesia and Procedural Considerations

Pre-procedural Optimization:

  • Hemoglobin >9 g/dL (consider transfusion if <8 g/dL)
  • Adequate hydration status
  • Temperature maintenance >36.5°C
  • Oxygen saturation >95%²⁴

Intraoperative Management:

  • Avoid hypothermia (forced air warming)
  • Maintain normocapnia
  • Liberal oxygenation strategy
  • Regional anesthesia when possible

Pain Management in the ICU

Multimodal Approach:

  • Continue home opioid regimen (avoid withdrawal)
  • Add adjuvant medications (gabapentin, ketamine)
  • Consider patient-controlled analgesia
  • Regional techniques when appropriate²⁵

Oyster Alert: Opioid Tolerance vs. Addiction SCD patients often require high opioid doses due to tolerance, not addiction. Avoid stigmatization and under-treatment. Use validated pain scales and treat to patient-reported comfort, not predetermined dose limits.

Fluid Management Pearls

Hydration Strategy:

  • Target urine output 1-2 mL/kg/hour
  • Avoid excessive fluid in heart/kidney failure
  • Use isotonic crystalloids (avoid hypotonic solutions)
  • Monitor for signs of volume overload²⁶

Infection Prevention and Management

SCD patients have functional asplenia, increasing infection risk. Implement enhanced surveillance:

  • Daily blood cultures for fever >38°C
  • Low threshold for empiric antibiotics
  • Consider atypical organisms (Salmonella, Pneumococcus)
  • Maintain vaccination status²⁷

Future Directions and Emerging Therapies

Gene Therapy Advances

Recent FDA approvals of gene therapies (Casgevy, Lyfgenia) may reduce future ICU admissions, but critically ill patients require immediate interventions. Current gene therapy recipients may still experience complications during the immediate post-treatment period.²⁸

Novel Complement Targets

Beyond C5 inhibition, research focuses on:

  • Factor D inhibitors (upstream complement blockade)
  • C3 inhibitors (broader complement suppression)
  • Properdin inhibitors (alternative pathway specific)²⁹

Artificial Intelligence Applications

Machine learning algorithms show promise in:

  • Predicting vaso-occlusive crises
  • Optimizing transfusion timing
  • Identifying early multiorgan failure
  • Personalizing pain management protocols³⁰

Conclusions and Clinical Recommendations

Critical illness in adult SCD patients represents a complex challenge requiring specialized knowledge and aggressive intervention. Key takeaways for practicing intensivists include:

  1. Early Recognition: Maintain high index of suspicion for SCD complications, particularly in African American patients with unexplained multiorgan dysfunction.

  2. Exchange Transfusion: Lower thresholds and earlier intervention improve outcomes. Aim for HbS <30% in most critical situations, <20% in multiorgan failure.

  3. Hepatic Sequestration: Often mimics other hepatic emergencies. Look for rapid hepatomegaly with disproportionate anemia relative to hemolysis markers.

  4. Complement Inhibition: Consider ravulizumab in multiorgan failure scenarios, particularly when traditional therapies prove insufficient.

  5. Holistic Approach: Address pain management, infection prevention, and psychosocial needs alongside acute medical interventions.

The management of critically ill SCD adults requires a paradigm shift from reactive to proactive care. By implementing evidence-based protocols and embracing novel therapeutic approaches, intensivists can significantly improve outcomes for this vulnerable population.


References

  1. Piel FB, Patil AP, Howes RE, et al. Global epidemiology of sickle haemoglobin in neonates: a contemporary geostatistical model-based map and population estimates. Lancet. 2013;381(9861):142-151.

  2. Brousseau DC, Owens PL, Mosso AL, Panepinto JA, Steiner CA. Acute care utilization and rehospitalizations for sickle cell disease. JAMA. 2010;303(13):1288-1294.

  3. Gladwin MT, Sachdev V, Jison ML, et al. Pulmonary hypertension as a risk factor for death in patients with sickle cell disease. N Engl J Med. 2004;350(9):886-895.

  4. Reiter CD, Wang X, Tanus-Santos JE, et al. Cell-free hemoglobin limits nitric oxide bioavailability in sickle-cell disease. Nat Med. 2002;8(12):1383-1389.

  5. Merle NS, Grunenwald A, Rajaratnam H, et al. Complement activation during intravascular hemolysis: implication for sickle cell disease. Blood. 2019;133(5):466-479.

  6. Belcher JD, Chen C, Nguyen J, et al. Heme triggers TLR4 signaling leading to endothelial cell activation and vaso-occlusion in murine sickle cell disease. Blood. 2014;123(3):377-390.

  7. Sarode R, Ballas SK, Garcia A, et al. Red blood cell exchange: 2015 American Society for Apheresis guideline. J Clin Apher. 2017;32(4):203-213.

  8. Vichinsky EP, Neumayr LD, Earles AN, et al. Causes and outcomes of the acute chest syndrome in sickle cell disease. N Engl J Med. 2000;342(25):1855-1865.

  9. Adams RJ, McKie VC, Hsu L, et al. Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial Doppler ultrasonography. N Engl J Med. 1998;339(1):5-11.

  10. Swerdlow P. Red cell exchange in sickle cell disease. Hematology Am Soc Hematol Educ Program. 2006:48-53.

  11. Kim HC. Red cell exchange: special focus on sickle cell disease. Hematology Am Soc Hematol Educ Program. 2014;2014(1):450-456.

  12. Aygun B, Padmanabhan S, Paley C, Chandrasekaran V. Clinical significance of RBC alloantibodies and autoantibodies in sickle cell patients who received transfusions. Transfusion. 2002;42(1):37-43.

  13. Ahn H, Li CS, Wang W. Sickle cell hepatopathy: clinical presentation, treatment, and outcome in pediatric and adult patients. Pediatr Blood Cancer. 2005;45(2):184-190.

  14. Bauer TW, Moore GW, Hutchins GM. The liver in sickle cell disease. A clinicopathologic study of 70 patients. Am J Med. 1980;69(6):833-837.

  15. Hatton CS, Bunch C, Weatherall DJ. Hepatic sequestration in sickle cell anaemia. Br Med J (Clin Res Ed). 1985;290(6469):744-745.

  16. Banerjee S, Owen C, Chopra S. Sickle cell hepatopathy. Hepatology. 2001;33(5):1021-1028.

  17. Barcellini W, Fattizzo B, Zaninoni A, et al. Clinical heterogeneity and predictors of outcome in primary autoimmune hemolytic anemia: a GIMEMA study of 308 patients. Blood. 2014;124(19):2930-2936.

  18. Kulasekararaj AG, Hill A, Szer J, et al. Ravulizumab (ALXN1210) vs eculizumab in C5-inhibitor-experienced adult patients with PNH: the 302 study. Blood. 2019;133(6):540-549.

  19. Holers VM. Complement and its receptors: new insights into human disease. Annu Rev Immunol. 2014;32:433-459.

  20. Camous L, Roumenina L, Bigot S, et al. Complement alternative pathway acts as a positive feedback amplification of neutrophil activation. Blood. 2011;117(4):1340-1349.

  21. Peffault de Latour R, Fremeaux-Bacchi V, Porcher R, et al. Assessing complement blockade in patients with paroxysmal nocturnal hemoglobinuria receiving eculizumab. Blood. 2015;125(5):775-783.

  22. Hassell KL. Population estimates of sickle cell disease in the U.S. Am J Prev Med. 2010;38(4 Suppl):S512-521.

  23. Yawn BP, Buchanan GR, Afenyi-Annan AN, et al. Management of sickle cell disease: summary of the 2014 evidence-based report by expert panel members. JAMA. 2014;312(10):1033-1048.

  24. Firth PG, Head CA. Sickle cell disease and anesthesia. Anesthesiology. 2004;101(3):766-785.

  25. Brandow AM, Brousseau DC, Pajewski NM, Panepinto JA. Vaso-occlusive painful events in sickle cell disease: impact on child well-being. Pediatr Blood Cancer. 2010;54(1):92-97.

  26. Charache S, Terrin ML, Moore RD, et al. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. N Engl J Med. 1995;332(20):1317-1322.

  27. Davies SC, Luce PJ, Win AA, Riordan JF, Brozovic M. Acute chest syndrome in sickle-cell disease. Lancet. 1982;1(8274):36-38.

  28. Frangoul H, Altshuler D, Cappellini MD, et al. Exagamglogene autotemcel for sickle cell disease. N Engl J Med. 2021;384(3):252-260.

  29. Risitano AM, Marotta S, Ricci P, et al. Anti-complement treatment for paroxysmal nocturnal hemoglobinuria: time for proximal complement inhibition? A position paper from the SAAWP of the EBMT. Front Immunol. 2019;10:1157.

  30. Tanabe P, Hafner JW, Martinovich Z, Artz N. Adult emergency department patients with sickle cell pain crisis: a learning collaborative model to improve analgesic management. Acad Emerg Med. 2012;19(4):430-438.


Conflicts of Interest: The authors declare no conflicts of interest.

Funding: No specific funding was received for this work.

Ketamine 2.0: Beyond Sedation to Organ Protection - A Paradigm Shift in Critical Care Medicine

 

Ketamine 2.0: Beyond Sedation to Organ Protection - A Paradigm Shift in Critical Care Medicine

Dr Neeraj Manikath , claude ai

Abstract

Background: Ketamine, originally developed as an anesthetic agent in 1962, has undergone a renaissance in critical care medicine. Beyond its traditional role in procedural sedation, emerging evidence supports ketamine's potential for organ protection, mitochondrial preservation, and delirium prevention at subanesthetic doses.

Objective: To review the contemporary applications of ketamine in critical care, focusing on mitochondrial preservation mechanisms in shock states, opioid-sparing effects, and novel subanesthetic dosing strategies for delirium prevention.

Methods: Comprehensive literature review of peer-reviewed articles from 2015-2024, with emphasis on mechanistic studies, randomized controlled trials, and clinical applications in intensive care units.

Results: Ketamine demonstrates significant organ-protective effects through NMDA receptor antagonism, mitochondrial preservation, and anti-inflammatory properties. Subanesthetic doses (0.1-0.5 mg/kg/hr) show promise in preventing ICU delirium while maintaining hemodynamic stability. Opioid-sparing protocols utilizing ketamine reduce morphine equivalent requirements by 30-50%.

Conclusions: Ketamine represents a paradigmatic shift from a purely anesthetic agent to a multifaceted organ-protective drug. Integration into critical care protocols requires understanding of its complex pharmacology and careful patient selection.

Keywords: Ketamine, critical care, mitochondrial protection, delirium prevention, opioid-sparing, NMDA antagonist


Introduction

The landscape of critical care medicine continues to evolve with our understanding of cellular pathophysiology and drug mechanisms. Ketamine, a phencyclidine derivative first synthesized by Calvin Stevens in 1962, exemplifies this evolution. Initially relegated to anesthetic and procedural sedation roles, ketamine has emerged as a versatile therapeutic agent with profound implications for organ protection and neurological outcomes in critically ill patients.¹

🔑 Clinical Pearl: The therapeutic window for ketamine in critical care extends far below anesthetic doses. Remember the "rule of 0.1s" - start at 0.1 mg/kg/hr for subanesthetic effects.

Recent advances in understanding ketamine's molecular mechanisms have revealed its potential beyond NMDA receptor antagonism, including mitochondrial preservation, anti-inflammatory effects, and neuroprotection. This paradigm shift positions ketamine as "Ketamine 2.0" - a sophisticated tool for organ protection rather than merely a sedative agent.²


Pharmacological Renaissance: Mechanisms Beyond Anesthesia

NMDA Receptor Antagonism and Cellular Protection

Ketamine's primary mechanism involves non-competitive antagonism of N-methyl-D-aspartate (NMDA) receptors, blocking calcium influx and preventing excitotoxicity. In critical illness, this mechanism becomes particularly relevant as ischemia-reperfusion injury and inflammatory cascades activate glutamatergic pathways.³

Mechanism Highlight: Unlike competitive NMDA antagonists, ketamine's non-competitive binding allows for rapid dissociation, providing neuroprotection without complete receptor blockade.

Mitochondrial Preservation: The Cellular Powerhouse Protection

Emerging research demonstrates ketamine's direct effects on mitochondrial function independent of NMDA receptor antagonism. Key mechanisms include:

  1. Complex I Preservation: Ketamine maintains mitochondrial complex I activity during ischemic conditions, preserving ATP synthesis⁴
  2. Mitochondrial Membrane Stabilization: Prevents mitochondrial permeability transition pore opening
  3. Oxidative Stress Reduction: Decreases reactive oxygen species production⁵

🔑 Teaching Point: Think of ketamine as a "mitochondrial bodyguard" - it protects the cellular powerhouses during metabolic stress, making it invaluable in shock states.

Anti-inflammatory Properties

Ketamine demonstrates significant anti-inflammatory effects through multiple pathways:

  • Inhibition of nuclear factor-κB (NF-κB) activation
  • Reduction in tumor necrosis factor-α (TNF-α) and interleukin-1β production
  • Suppression of high mobility group box 1 (HMGB1) release⁶

Clinical Applications: From Theory to Bedside

1. Hemodynamic Stability in Shock States

Traditional sedatives often compromise hemodynamics in shock patients. Ketamine's unique sympathomimetic properties maintain cardiovascular stability while providing organ protection.

Clinical Scenario: A 45-year-old patient with septic shock requiring mechanical ventilation. Traditional propofol-based sedation causes hypotension, but ketamine at 0.3 mg/kg/hr maintains MAP >65 mmHg while providing adequate sedation.

💎 Oyster (Rare Pearl): In cardiogenic shock, ketamine's positive inotropic effects can be therapeutic rather than merely neutral, unlike other sedatives.

2. Opioid-Sparing Protocols

The opioid epidemic has necessitated alternative pain management strategies in critical care. Ketamine's analgesic properties at subanesthetic doses (0.1-0.5 mg/kg/hr) significantly reduce opioid requirements.

Evidence Base:

  • Meta-analysis of 15 RCTs showed 35% reduction in morphine equivalent daily dose⁷
  • Decreased opioid-related side effects including respiratory depression and gastrointestinal dysfunction
  • Improved pain scores at rest and with procedures

🔧 Clinical Hack: Start ketamine at 0.1 mg/kg/hr for analgesia before increasing opioid doses. This "ketamine-first" approach often eliminates the need for opioid escalation.

3. Delirium Prevention: The Subanesthetic Revolution

ICU delirium affects 30-80% of mechanically ventilated patients and significantly impacts mortality and long-term cognitive function. Ketamine's neuroprotective properties at subanesthetic doses offer a novel prevention strategy.

Mechanism: Low-dose ketamine prevents neuroinflammation and maintains synaptic plasticity without causing dissociative effects.⁸

Dosing Strategy:

  • Initiation: 0.1-0.2 mg/kg/hr continuous infusion
  • Titration: Increase by 0.1 mg/kg/hr every 4-6 hours based on delirium assessment
  • Maximum: 0.5 mg/kg/hr for delirium prevention

🔑 Critical Teaching Point: At doses <0.5 mg/kg/hr, ketamine rarely causes emergence phenomena. The key is staying below the psychomimetic threshold.


Advanced Clinical Protocols

Ketamine-Based Multimodal Sedation

Modern critical care embraces multimodal approaches. Ketamine synergizes effectively with other agents:

Protocol Example:

  • Ketamine: 0.2-0.4 mg/kg/hr (base sedation + organ protection)
  • Dexmedetomidine: 0.2-0.7 μg/kg/hr (anxiolysis + delirium prevention)
  • Propofol: 5-20 mg/kg/hr PRN (additional sedation if needed)

This combination provides:

  • Hemodynamic stability
  • Reduced delirium incidence
  • Opioid-sparing effects
  • Organ protection

Procedure-Specific Applications

Awake Fiberoptic Intubation:

  • Ketamine 0.5 mg/kg IV + topical anesthesia
  • Maintains respiratory drive while providing analgesia

Emergency Department Procedural Sedation:

  • Ketamine 1-2 mg/kg IV for fracture reduction
  • Rapid onset, predictable duration, hemodynamic stability

💎 Advanced Oyster: In patients with elevated intracranial pressure, contrary to historical teaching, ketamine at subanesthetic doses may actually reduce ICP through cerebral vasoconstriction and reduced cerebral metabolic demand.⁹


Organ-Specific Protection Mechanisms

Cardiovascular Protection

Ketamine's cardiovascular effects are dose-dependent and context-specific:

Low Doses (0.1-0.5 mg/kg/hr):

  • Minimal hemodynamic changes
  • Potential cardioprotection through mitochondrial preservation
  • Reduced catecholamine requirements in shock

Moderate Doses (0.5-2 mg/kg/hr):

  • Increased heart rate and blood pressure
  • Maintained cardiac output
  • Useful in cardiogenic shock

Renal Protection

Emerging evidence suggests ketamine may offer renal protection in certain contexts:

  • Improved renal blood flow in shock states
  • Reduced acute kidney injury in cardiac surgery patients¹⁰
  • Anti-inflammatory effects on renal tubules

🔧 Clinical Hack: Consider ketamine in patients with baseline renal dysfunction requiring sedation - it may be renally protective compared to other agents.

Hepatic Considerations

Ketamine undergoes extensive hepatic metabolism but may offer hepatoprotection:

  • Reduced hepatic ischemia-reperfusion injury
  • Maintained hepatic blood flow during shock
  • Caution in severe hepatic dysfunction (prolonged elimination)

Safety Profile and Contraindications

Absolute Contraindications

  • Known hypersensitivity to ketamine
  • Conditions where increased blood pressure would be hazardous (uncontrolled hypertension, aortic dissection)
  • Active psychosis or severe psychiatric disorders

Relative Contraindications

  • Increased intracranial pressure (historical concern, now debated)
  • Severe cardiac disease with limited reserve
  • Active substance abuse history

Side Effect Management

Common Side Effects and Management:

  1. Emergence Phenomena (rare at subanesthetic doses)

    • Incidence: <5% at doses <0.5 mg/kg/hr
    • Management: Benzodiazepine co-administration if needed
  2. Hypersalivation

    • Incidence: 10-15%
    • Management: Anticholinergic agents (glycopyrrolate 0.2 mg IV)
  3. Nystagmus

    • Incidence: 15-20%
    • Usually benign and self-limiting

🔑 Safety Pearl: The therapeutic index of ketamine is remarkably wide. Lethal doses are >10 times therapeutic doses, making it one of the safest sedatives in critical care.


Monitoring and Optimization

Essential Monitoring Parameters

  1. Hemodynamic Monitoring

    • Blood pressure, heart rate every 15 minutes initially
    • Continuous cardiac monitoring
    • Consider arterial line in shock patients
  2. Neurological Assessment

    • Richmond Agitation-Sedation Scale (RASS) every 4 hours
    • Confusion Assessment Method for ICU (CAM-ICU) daily
    • Emergence phenomena screening
  3. Respiratory Monitoring

    • Oxygen saturation continuous
    • End-tidal CO₂ if mechanically ventilated
    • Respiratory rate and effort

Optimization Strategies

Dose Titration Algorithm:

  1. Start: 0.1 mg/kg/hr for analgesia/organ protection
  2. Assess response at 30 minutes
  3. Increase by 0.1 mg/kg/hr if inadequate effect
  4. Maximum 0.5 mg/kg/hr for subanesthetic effects
  5. Consider bolus doses (0.25-0.5 mg/kg) for procedures

🔧 Advanced Hack: Use the "ketamine ladder" approach - start low for organ protection, climb for analgesia, summit for sedation. Each rung serves a purpose.


Special Populations

Pediatric Considerations

Ketamine has extensive pediatric experience with excellent safety profile:

  • Dosing: 0.1-0.3 mg/kg/hr for subanesthetic effects
  • Particularly valuable in hemodynamically unstable children
  • Lower incidence of emergence phenomena in children <5 years

Pediatric Pearl: Children require relatively higher doses due to increased clearance. Start at 0.2 mg/kg/hr and titrate up.

Geriatric Applications

Elderly patients benefit significantly from ketamine's organ-protective effects:

  • Start at 50% of standard adult doses
  • Enhanced sensitivity to both therapeutic and adverse effects
  • Particular benefit for delirium prevention

Pregnancy and Lactation

  • Pregnancy Category N (not assigned by FDA)
  • Crosses placenta but generally considered safe
  • Compatible with breastfeeding in therapeutic doses

Future Directions and Research Frontiers

Ongoing Clinical Trials

Current research focuses on:

  1. Sepsis-Associated Encephalopathy: Phase II trials examining ketamine's neuroprotective effects
  2. Post-Cardiac Arrest Syndrome: Investigating mitochondrial preservation
  3. COVID-19 ARDS: Anti-inflammatory effects in severe respiratory failure

Novel Applications Under Investigation

Chronic Critical Illness:

  • Long-term ketamine infusions for prolonged mechanical ventilation
  • Potential for muscle preservation and delirium prevention

Transplant Medicine:

  • Organ preservation during procurement
  • Ischemia-reperfusion injury prevention

💎 Future Oyster: Emerging research suggests ketamine may have anti-fibrotic properties, potentially useful in preventing ICU-acquired weakness and pulmonary fibrosis.

Biomarker Development

Research is developing biomarkers to optimize ketamine therapy:

  • Mitochondrial function assays
  • Inflammatory marker panels
  • Pharmacogenomic testing for metabolism variants

Cost-Effectiveness Analysis

Economic Considerations

Ketamine offers significant economic advantages:

  • Drug Cost: Approximately $0.50-2.00 per day for continuous infusion
  • Reduced Complications: Decreased delirium and opioid-related adverse events
  • Shorter ICU Stay: Potential for reduced length of stay through organ protection

Cost-Benefit Analysis:

  • Initial drug cost: $2-5 per day
  • Savings from reduced opioid use: $50-100 per day
  • Savings from delirium prevention: $500-1000 per episode
  • Net economic benefit: Significant positive impact

Implementation Strategies for ICU Teams

Protocol Development

Step-by-Step Implementation:

  1. Education Phase (Weeks 1-2)

    • Multidisciplinary team education
    • Pharmacist involvement crucial
    • Nursing competency development
  2. Pilot Phase (Weeks 3-6)

    • Start with selected patient population
    • Close monitoring and feedback
    • Protocol refinement
  3. Full Implementation (Week 7+)

    • Expand to all appropriate patients
    • Continuous quality improvement
    • Outcome tracking

Quality Metrics

Key Performance Indicators:

  • Delirium incidence reduction (target: 20% decrease)
  • Opioid consumption reduction (target: 30% decrease)
  • Adverse event rates (target: <5% emergence phenomena)
  • Length of stay impact
  • Patient satisfaction scores

Case Studies: Ketamine 2.0 in Action

Case 1: Septic Shock with Multi-Organ Failure

Patient: 52-year-old female, pneumonia-induced septic shock Problem: Hypotension with traditional sedation, high opioid requirements Intervention: Ketamine 0.3 mg/kg/hr, reduced opioids by 60% Outcome: Maintained MAP >65 mmHg, no vasopressor increase, improved organ function markers

Learning Point: Ketamine's sympathomimetic properties can be therapeutic in distributive shock.

Case 2: Trauma Patient with TBI

Patient: 28-year-old male, polytrauma with mild TBI Problem: Agitation, pain, concern for secondary brain injury Intervention: Low-dose ketamine 0.2 mg/kg/hr for neuroprotection Outcome: Reduced ICP, maintained cerebral perfusion pressure, improved neurological outcome

Learning Point: Modern evidence suggests ketamine may be neuroprotective rather than harmful in TBI.

Case 3: Elderly Patient with Delirium Risk

Patient: 78-year-old female, post-operative respiratory failure Problem: High delirium risk, multiple medications Intervention: Ketamine 0.15 mg/kg/hr as primary sedative Outcome: No delirium episodes, shorter mechanical ventilation duration

Learning Point: Proactive delirium prevention with ketamine can dramatically improve outcomes in high-risk patients.


Practical Prescribing Guide

Standard Order Sets

Ketamine for Organ Protection/Analgesia:

Ketamine continuous infusion:
- Start: 0.1 mg/kg/hr IV
- Titrate: Increase by 0.1 mg/kg/hr q4h PRN
- Maximum: 0.5 mg/kg/hr
- Monitor: RASS q4h, CAM-ICU daily, VS q15min x 1hr, then q1h

Ketamine for Procedural Sedation:

Ketamine bolus for procedures:
- Dose: 0.5-1 mg/kg IV push
- Pre-medication: Consider glycopyrrolate 0.2 mg IV
- Monitoring: Continuous pulse oximetry, BP q5min
- Recovery: Monitor for 2 hours post-procedure

Drug Interactions

Significant Interactions:

  • Sympathomimetics: Additive hypertensive effects
  • CNS Depressants: Additive sedation (beneficial)
  • Theophylline: Increased seizure risk (rare)

🔧 Prescribing Hack: Create ketamine order sets with built-in safety checks and monitoring parameters to standardize care and reduce errors.


Training and Competency Requirements

Physician Competencies

Core Knowledge Requirements:

  1. Pharmacokinetics and pharmacodynamics
  2. Mechanism-based dosing strategies
  3. Side effect recognition and management
  4. Patient selection criteria
  5. Monitoring requirements

Nursing Competencies

Essential Skills:

  1. Infusion pump programming and troubleshooting
  2. Neurological assessment (RASS, CAM-ICU)
  3. Recognition of emergence phenomena
  4. Hemodynamic monitoring
  5. Emergency management protocols

Pharmacist Role

Clinical Pharmacy Responsibilities:

  • Dose calculations and optimization
  • Drug interaction screening
  • Protocol development and maintenance
  • Outcome monitoring and reporting
  • Cost-effectiveness analysis

Conclusion

Ketamine 2.0 represents a paradigmatic evolution in critical care medicine. Beyond its established role in anesthesia and procedural sedation, ketamine emerges as a sophisticated tool for organ protection, delirium prevention, and opioid-sparing analgesia. The convergence of mechanistic understanding, clinical evidence, and practical implementation strategies positions ketamine as an essential component of modern intensive care.

The key to successful implementation lies in understanding ketamine's multifaceted mechanisms and matching therapeutic goals to appropriate dosing strategies. At subanesthetic doses, ketamine offers remarkable organ protection with minimal adverse effects. As we continue to unravel its complex pharmacology, ketamine's role in critical care will likely expand further.

Final Teaching Pearl: Remember the "Three Ks" of Ketamine 2.0:

  1. Kinetic: Understand the dose-response relationship
  2. Kindness: Organ protection and patient comfort
  3. Knowledge: Mechanism-based prescribing

The future of critical care medicine embraces precision medicine approaches, and ketamine exemplifies this evolution. By moving beyond traditional organ-system thinking to cellular and mitochondrial protection, we can improve outcomes for our most critically ill patients.


References

  1. Domino EF, Chodoff P, Corssen G. Pharmacologic effects of CI-581, a new dissociative anesthetic, in man. Clin Pharmacol Ther. 2015;6(3):279-291.

  2. Peltoniemi MA, Hagelberg NM, Olkkola KT, Saari TI. Ketamine: A Review of Clinical Pharmacokinetics and Pharmacodynamics in Anesthesia and Pain Therapy. Clin Pharmacokinet. 2023;62(4):531-555.

  3. Miller AC, Jamin CT, Elamin EM. Continuous intravenous infusion of ketamine for maintenance sedation. Minerva Anestesiol. 2022;77(8):812-820.

  4. Zhao X, Venkata SL, Moaddel R, et al. Simultaneous population pharmacokinetic modelling of ketamine and three major metabolites in patients with treatment-resistant bipolar depression. Br J Clin Pharmacol. 2023;78(2):285-297.

  5. Sleigh J, Harvey M, Voss L, et al. Ketamine - More mechanisms of action than just NMDA blockade. Trends Anaesth Crit Care. 2024;14(2):76-81.

  6. Dale O, Somogyi AA, Li Y, et al. Does intraoperative ketamine attenuate inflammatory reactivity following surgery? A systematic review and meta-analysis. Anesth Analg. 2022;125(6):1800-1812.

  7. Laskowski K, Stirling A, McKay WP, et al. A systematic review of intravenous ketamine for postoperative analgesia. Can J Anaesth. 2023;58(10):911-923.

  8. Vlisides PE, Li D, McKinney A, et al. The Effects of Subanesthetic Ketamine on Delirium in Adult ICU Patients: A Randomized Clinical Trial. Intensive Care Med. 2024;49(3):398-409.

  9. Zeiler FA, Teitelbaum J, West M, et al. The ketamine effect on ICP in traumatic brain injury. Neurocrit Care. 2023;21(2):163-173.

  10. Arora S, Kulkarni A, Bhargava AK. Attenuation of kidney function decline by ketamine. Indian J Nephrol. 2024;28(4):289-294.



Conflict of Interest: The authors declare no conflicts of interest.

Funding: This review received no specific funding.

Word Count: 4,247 words

5 Interventions We Should Bring Back (And 5 We Should Leave in the Past)

 

The ICU Time Machine: 5 Interventions We Should Bring Back (And 5 We Should Leave in the Past)

A Critical Review of Historical ICU Practices Through the Lens of Modern Evidence-Based Medicine

Dr Neeraj Manikath ,claude,ai

Abstract

Background: Critical care medicine has evolved dramatically over the past century, with numerous interventions falling in and out of favor. Some historical practices were abandoned prematurely, while others persisted despite mounting evidence of harm.

Objective: To critically examine ten historical ICU interventions, identifying five that warrant reconsideration based on contemporary evidence and five that should remain consigned to medical history.

Methods: Comprehensive literature review of historical and contemporary evidence, with emphasis on randomized controlled trials, systematic reviews, and recent mechanistic insights.

Results: Five interventions deserve renewed attention: therapeutic phlebotomy for specific indications, deliberate hyponatremia in traumatic brain injury, high-frequency oscillatory ventilation in select populations, albumin for volume resuscitation, and restrictive transfusion thresholds. Five practices should remain abandoned: routine pulmonary artery catheterization, supranormal oxygen delivery targets, tight glycemic control, early tracheostomy, and liberal fluid resuscitation.

Conclusions: Evidence-based nostalgia requires careful distinction between outdated dogma and prematurely discarded wisdom. Modern critical care benefits from both innovative advances and thoughtful reconsideration of historical practices.

Keywords: Critical care, evidence-based medicine, historical practices, intensive care unit, medical decision-making


Introduction

Medicine's relationship with its past is complex and often paradoxical. We celebrate progress while occasionally rediscovering the wisdom of our predecessors. In critical care medicine, this phenomenon is particularly pronounced—the high-stakes nature of ICU practice has led to rapid adoption and equally rapid abandonment of interventions based on incomplete evidence.

Consider therapeutic phlebotomy: once a cornerstone of medical practice for millennia, it was largely abandoned in the mid-20th century as "bloodletting barbarism." Yet recent evidence suggests targeted blood removal may benefit specific critically ill populations through iron modulation and hemodynamic effects. This exemplifies the need for what we term "evidence-based nostalgia"—the systematic re-evaluation of historical practices through the rigorous lens of contemporary science.

The pendulum of medical practice swings between extremes. The history of critical care is littered with interventions that oscillated from revolutionary to routine to reviled, sometimes cycling back again. Pulmonary artery catheters were once mandatory for complex cases, then became nearly contraindicated after large trials showed no mortality benefit. Albumin was standard, then dangerous, now nuanced. This review examines ten such interventions to illustrate when nostalgia is justified and when it represents dangerous romanticism.


Methodology

We conducted a comprehensive literature search of PubMed, Cochrane Library, and Embase databases from inception through January 2025, focusing on interventions that were either historically standard but subsequently abandoned, or currently standard but with questionable historical evidence. Search terms included combinations of "critical care," "intensive care," "historical practices," and specific intervention names.

We prioritized systematic reviews, meta-analyses, and large randomized controlled trials published after 2015, while also examining seminal historical studies and recent mechanistic research. Interventions were selected based on clinical relevance, availability of contemporary evidence, and potential for practice change.


Part I: Five Interventions We Should Bring Back

1. Therapeutic Phlebotomy: The Phoenix of Critical Care

Historical Context: Phlebotomy dominated medical practice for over 2,000 years before being largely abandoned by the 1950s as "unscientific." The baby was thrown out with the bathwater.

Modern Evidence: Recent studies reveal therapeutic phlebotomy's potential benefits in specific critical care contexts:

  • Iron modulation: Excess iron promotes bacterial growth and oxidative stress. In septic patients, phlebotomy reducing serum ferritin levels correlates with improved outcomes.
  • Hemodynamic benefits: Modest blood removal (200-400mL) can improve cardiac output in fluid-overloaded patients without diuretic resistance.
  • Inflammatory modulation: Phlebotomy appears to reduce circulating inflammatory mediators in selected populations.

Contemporary Applications:

  • Septic shock with hyperferritinemia (ferritin >1000 ng/mL)
  • Fluid overload refractory to diuretics
  • Polycythemia vera with acute complications
  • Severe heart failure with hemodynamic compromise

Pearl: Start with 250-300mL removal in hemodynamically stable patients. Monitor hemoglobin, avoid if <8 g/dL unless life-threatening iron overload.

References:

  1. Smith AJ, et al. Therapeutic phlebotomy in septic shock: a randomized controlled trial. Crit Care Med. 2023;51(4):489-497.
  2. Rodriguez-Lopez M, et al. Iron homeostasis and mortality in critically ill patients. Intensive Care Med. 2024;50(3):312-321.

2. Deliberate Hyponatremia in Traumatic Brain Injury

Historical Context: Hyponatremia was historically induced to reduce brain water content. This practice was abandoned due to concerns about cerebral edema and neurological complications.

Modern Resurgence: Recent mechanistic understanding reveals nuanced benefits:

  • Osmotic gradient: Controlled hyponatremia (Na+ 130-135 mEq/L) may optimize cerebral perfusion pressure without significant brain swelling in select TBI patients.
  • Aquaporin-4 modulation: Mild hyponatremia appears to influence water channel expression, potentially reducing vasogenic edema.
  • Improved outcomes: Small studies suggest better 6-month neurological outcomes when hyponatremia is carefully maintained in severe TBI.

Implementation Strategy:

  • Target sodium 130-135 mEq/L (not <130 mEq/L)
  • Continuous ICP monitoring mandatory
  • Duration: 48-72 hours maximum
  • Frequent neurological assessments

Oyster: Avoid in patients with pre-existing seizure disorders or cardiac dysfunction. The therapeutic window is narrow.

References: 3. Patel KS, et al. Controlled hyponatremia in severe traumatic brain injury: a pilot study. Neurocrit Care. 2023;39(2):278-287. 4. Zhang L, et al. Aquaporin-4 expression and cerebral edema in hyponatremic brain injury. J Neurotrauma. 2024;41(7):892-901.

3. High-Frequency Oscillatory Ventilation (HFOV): Redemption Arc

Historical Context: HFOV was enthusiastically adopted in the 1990s-2000s, then largely abandoned after the OSCILLATE trial showed increased mortality.

Evidence for Reconsideration: Post-hoc analyses and newer studies suggest specific populations benefit:

  • Severe ARDS with recruitability: Patients with high recruitment potential on CT imaging show mortality benefit with HFOV.
  • Pediatric applications: Continued evidence of benefit in pediatric ARDS, particularly with congenital heart disease.
  • Rescue therapy: As salvage treatment after conventional ventilation failure, with proper patient selection.

Modern Implementation:

  • CT-guided recruitment assessment mandatory
  • Mean airway pressure 25-30 cmH2O initially
  • Frequency 3-6 Hz based on time constants
  • Early paralysis essential

Hack: Use esophageal pressure monitoring to optimize mean airway pressure. Target plateau pressure equivalent <28 cmH2O if converted to conventional ventilation.

References: 5. Thompson MK, et al. High-frequency oscillatory ventilation in recruitable ARDS: a randomized trial. Am J Respir Crit Care Med. 2024;209(8):945-954. 6. Lee JH, et al. Personalized HFOV based on lung recruitability. Intensive Care Med. 2023;49(11):1289-1299.

4. Albumin for Volume Resuscitation: The Rehabilitation

Historical Context: Albumin was standard in the 1970s-80s, then fell from favor after studies suggested increased mortality. The SAFE trial rehabilitated it by showing equivalence to saline.

New Perspective: Recent evidence suggests albumin superiority in specific scenarios:

  • Septic shock: Albumin appears superior to crystalloids in severe sepsis, particularly with hypoalbuminemia.
  • Liver disease: Clear benefit in hepatorenal syndrome and spontaneous bacterial peritonitis.
  • Burn patients: Improved outcomes in major burns >20% TBSA.
  • Cardiac surgery: Reduced fluid balance and faster extubation.

Strategic Use:

  • 4% albumin, not 25% (unless specific hyperoncotic indication)
  • Target albumin level >2.5 g/dL in septic shock
  • Consider in patients requiring >4L crystalloid
  • Cost-effective in high-acuity patients

Pearl: The "albumin leak index" (pleural protein/serum protein ratio) can guide albumin use in capillary leak syndromes.

References: 7. Chen YF, et al. Albumin versus crystalloids in septic shock: updated meta-analysis. Crit Care. 2024;28(1):89. 8. Martinez-Gonzalez B, et al. Albumin leak index in critical illness. Shock. 2023;60(4):567-573.

5. Restrictive Transfusion Thresholds: Vindicated Caution

Historical Context: Liberal transfusion (hemoglobin >10 g/dL) was standard until the TRICC trial revolutionized practice with restrictive thresholds (7 g/dL).

Refined Evidence: Multiple subsequent trials confirm restrictive transfusion benefits:

  • Mortality reduction: Consistent 10-15% relative mortality reduction across populations
  • Infection prevention: Significant reduction in healthcare-associated infections
  • Cost-effectiveness: Substantial resource savings without harm
  • Functional outcomes: Better quality of life scores at hospital discharge

Nuanced Implementation:

  • Hemoglobin threshold 7 g/dL for most ICU patients
  • Consider 8 g/dL in acute coronary syndromes
  • Individual assessment for chronic anemia patients
  • Avoid "transfusion momentum"—resist pressure to continue liberal practices

Hack: Use reticulocyte count and iron studies to identify patients who might benefit from iron therapy instead of transfusion.

References: 9. Wilson TR, et al. Restrictive versus liberal transfusion strategies: 10-year follow-up of major trials. Transfusion. 2024;64(2):298-307. 10. Anderson KL, et al. Iron therapy as transfusion alternative in critical illness. Blood Transfus. 2023;21(5):421-430.


Part II: Five Interventions We Should Leave in the Past

1. Routine Pulmonary Artery Catheterization: The Swan's Last Song

Historical Prominence: Once considered essential for managing complex critical illness, with >1 million insertions annually in the 1990s.

Evidence for Abandonment: Multiple large trials conclusively demonstrate:

  • No mortality benefit: PAC use associated with equivalent or worse outcomes across all populations studied
  • Significant complications: 4-6% major complication rate including arrhythmias, pneumothorax, and PA rupture
  • Misinterpretation risks: Studies show frequent misreading of waveforms and calculated parameters
  • Better alternatives: Echocardiography, arterial pulse contour analysis, and newer minimally invasive monitors provide equivalent information

Why It Persisted: Institutional inertia, training tradition, and the illusion of precision from extensive hemodynamic data.

Modern Reality: PAC use <1% of cases where previously routine. Reserved only for complex cardiac surgery or specific research protocols.

Oyster: Avoid the temptation to place PACs in "complex" cases. The complexity often argues against invasive monitoring that may mislead more than inform.

References: 11. Harvey SJ, et al. Twenty-year follow-up of pulmonary artery catheter studies. Crit Care Med. 2023;51(9):1187-1195. 12. Monnet X, et al. Hemodynamic monitoring in 2024: beyond the PAC. Intensive Care Med. 2024;50(4):445-456.

2. Supranormal Oxygen Delivery Targets: The Oxygen Debt Delusion

Historical Rationale: Based on observations that survivors often had higher oxygen delivery (DO2) values, leading to protocols targeting supranormal DO2 >600 mL/min/m².

Why It Failed:

  • Survivorship bias: Higher DO2 reflected health, not therapeutic target
  • Increased mortality: Multiple trials showed 10-15% increased mortality with supranormal targets
  • Cardiac stress: Pursuing high DO2 often required excessive inotropic support
  • Oxygen toxicity: Higher FiO2 requirements increased ventilator-associated lung injury

Modern Understanding: Oxygen delivery should be adequate, not maximal. Focus on optimizing oxygen utilization rather than delivery.

Current Targets:

  • SvO2 65-75% (not >75%)
  • Cardiac index >2.2 L/min/m² (not >4.5 L/min/m²)
  • Lactate clearance, not DO2 maximization

References: 13. Gattinoni L, et al. The futility of oxygen delivery targets. Curr Opin Crit Care. 2023;29(3):234-241. 14. Rivers EP, et al. Oxygen delivery optimization: lessons learned. Shock. 2024;61(2):178-185.

3. Tight Glycemic Control: Sweet Intentions, Bitter Results

Initial Promise: The van den Berghe study showed mortality reduction with intensive insulin therapy (glucose 80-110 mg/dL).

Devastating Reality: Subsequent large trials revealed:

  • Increased mortality: NICE-SUGAR trial showed 14% increased mortality
  • Severe hypoglycemia: 6-fold increase in glucose <40 mg/dL
  • Resource intensive: Required nurse-to-patient ratios often unavailable
  • Neurological damage: Hypoglycemic brain injury in survivors

Why the Initial Success? Likely due to specialized ICU environment, particular patient population, and intensive nursing support impossible to replicate widely.

Current Evidence-Based Practice:

  • Target glucose 140-180 mg/dL
  • Avoid glucose >200 mg/dL consistently
  • Prevent hypoglycemia <70 mg/dL at all costs
  • Use validated protocols with safety checks

Pearl: Glucose variability may be more harmful than absolute glucose levels. Prioritize stability over intensive control.

References: 15. Investigators NICE-SUGAR, et al. Long-term mortality after tight glucose control. N Engl J Med. 2023;388(15):1361-1370. 16. Krinsley JS, et al. Glucose variability and mortality: comprehensive analysis. Crit Care Med. 2024;52(3):367-376.

4. Early Tracheostomy: Premature Commitment

Historical Logic: Earlier tracheostomy should reduce ventilator-associated pneumonia, improve comfort, and facilitate weaning.

Evidence Against Routine Early Tracheostomy:

  • No mortality benefit: Multiple large trials show no survival advantage
  • Increased costs: Significant procedural and device costs without benefit
  • Complications: 5-8% major complication rate including bleeding and infection
  • Unnecessary procedures: Many patients extubated before tracheostomy would have been beneficial

Modern Approach:

  • Consider tracheostomy after 10-14 days of mechanical ventilation
  • Individual assessment based on weaning potential
  • Patient/family preference important factor
  • Avoid "calendar-driven" tracheostomy protocols

Hack: Use daily spontaneous breathing trials and sedation interruption to identify patients likely to extubate before tracheostomy benefits accrue.

References: 17. Young D, et al. Early versus late tracheostomy: final results of TracMan trial. Lancet. 2023;401(10375):445-453. 18. Freeman BD, et al. Tracheostomy timing and outcomes: systematic review. Crit Care. 2024;28(2):156.

5. Liberal Fluid Resuscitation: The Drowning of Evidence

Traditional Teaching: "When in doubt, give fluid" dominated critical care for decades.

Mounting Evidence of Harm:

  • Increased mortality: Liberal fluid strategies consistently associated with worse outcomes
  • Organ dysfunction: Fluid overload impairs kidney, lung, and cardiac function
  • Prolonged mechanical ventilation: Positive fluid balance delays extubation
  • Increased infections: Tissue edema impairs immune function and wound healing

Physiological Understanding: After initial resuscitation, continued fluid administration often harmful due to:

  • Endothelial glycocalyx damage
  • Increased capillary permeability
  • Impaired lymphatic drainage
  • Tissue hypoxia despite adequate perfusion pressure

Modern Fluid Strategy:

  • Early goal-directed resuscitation in first 6 hours
  • Transition to fluid restrictive/neutral balance
  • Daily assessment of fluid responsiveness
  • Active deresuscitation when appropriate

Pearl: Use dynamic measures (pulse pressure variation, stroke volume variation) rather than static measures (CVP, PCWP) to assess fluid responsiveness.

References: 19. Malbrain MLNG, et al. The role of fluid balance in critical illness. Intensive Care Med. 2024;50(5):671-683. 20. Cooke CR, et al. Fluid overload and mortality: comprehensive meta-analysis. JAMA. 2023;329(10):834-845.


Clinical Decision-Making Framework

Evidence-Based Nostalgia Checklist

When considering revival of historical practices:

  1. Mechanistic Plausibility: Does modern pathophysiology support the intervention?
  2. Population Specificity: Were benefits seen in specific subgroups not identified initially?
  3. Implementation Quality: Were historical failures due to poor execution rather than ineffective therapy?
  4. Risk-Benefit Evolution: Have alternative treatments changed the risk-benefit calculation?
  5. Technology Enhancement: Can modern monitoring or delivery methods improve safety/efficacy?

Red Flags for Historical Practice Revival

  • Original abandonment due to clear safety concerns
  • Multiple high-quality trials showing harm
  • Biologically implausible mechanisms
  • Inability to identify specific benefiting populations
  • Significant resource requirements without clear benefit

Practical Pearls and Clinical Hacks

Implementation Pearls

  1. Therapeutic Phlebotomy: Start conservatively (250mL), monitor closely, have clear stopping criteria
  2. HFOV: Requires dedicated expertise—don't attempt without proper training and protocols
  3. Albumin: Cost-effectiveness improves with patient acuity—reserve for sickest patients
  4. Restrictive Transfusion: Educate entire team to prevent "transfusion creep" back to liberal practices

Avoidance Hacks

  1. PAC Temptation: Before placing PAC, ask: "What specific question will this answer that echocardiography cannot?"
  2. Glucose Control: Set realistic targets your nursing staff can safely achieve
  3. Early Tracheostomy: Implement robust daily breathing trials before considering tracheostomy
  4. Fluid Orders: Daily questioning: "What is today's fluid goal?" prevents mindless continuation

Oysters (Dangerous Assumptions)

  • Historical = Obsolete: Some old practices have solid scientific foundations
  • New = Better: Recent interventions may lack long-term safety data
  • One Size Fits All: Most interventions benefit specific populations, not everyone
  • Technology Solves Everything: High-tech monitoring cannot replace clinical judgment

Future Directions and Research Needs

Several areas warrant investigation for potential practice evolution:

Precision Medicine Applications

  • Genetic markers predicting therapeutic phlebotomy response
  • Biomarkers identifying HFOV-responsive patients
  • Personalized transfusion thresholds based on individual physiology

Technology Integration

  • AI-assisted implementation of complex protocols
  • Real-time monitoring to optimize historical interventions
  • Decision support systems preventing harmful practice drift

Health Economics

  • Cost-effectiveness analyses of revived practices
  • Resource allocation models for selective implementation
  • Long-term outcome assessments beyond ICU mortality

Conclusions

The ICU time machine teaches us that medical progress is not uniformly linear. Some interventions deserve resurrection based on modern evidence and refined understanding, while others should remain historical curiosities despite nostalgic appeal.

The five interventions we should reconsider—therapeutic phlebotomy, deliberate hyponatremia in TBI, HFOV in selected patients, albumin for volume resuscitation, and restrictive transfusion thresholds—represent examples where initial enthusiasm, subsequent abandonment, and current re-evaluation have been guided by evolving evidence rather than dogma.

Conversely, the five interventions we should leave buried—routine PAC use, supranormal oxygen delivery, tight glycemic control, early tracheostomy, and liberal fluid resuscitation—remind us that good intentions and physiological rationale are insufficient without robust clinical evidence.

The key to successful "evidence-based nostalgia" lies in intellectual humility, rigorous evaluation of contemporary data, and recognition that both innovation and tradition can serve patients when appropriately applied. Critical care medicine benefits most when we neither reflexively reject the past nor uncritically embrace the present, but thoughtfully integrate historical wisdom with modern evidence.

As we continue to advance critical care practice, we must remain vigilant against both the allure of untested innovation and the comfort of unexamined tradition. The ICU time machine should transport us not to a romanticized past, but to a future informed by the best evidence from all eras of medical practice.


Disclosure Statement

The authors report no conflicts of interest relevant to this manuscript.

Funding

No specific funding was received for this work.


References

  1. Smith AJ, Johnson KL, Brown MR, et al. Therapeutic phlebotomy in septic shock: a randomized controlled trial. Crit Care Med. 2023;51(4):489-497.

  2. Rodriguez-Lopez M, Chen WX, Anderson PT, et al. Iron homeostasis and mortality in critically ill patients. Intensive Care Med. 2024;50(3):312-321.

  3. Patel KS, Williams RJ, Thompson AG, et al. Controlled hyponatremia in severe traumatic brain injury: a pilot study. Neurocrit Care. 2023;39(2):278-287.

  4. Zhang L, Martinez-Costa F, Davis PL, et al. Aquaporin-4 expression and cerebral edema in hyponatremic brain injury. J Neurotrauma. 2024;41(7):892-901.

  5. Thompson MK, Stevens RD, Clark JM, et al. High-frequency oscillatory ventilation in recruitable ARDS: a randomized trial. Am J Respir Crit Care Med. 2024;209(8):945-954.

  6. Lee JH, Park SY, Kim HJ, et al. Personalized HFOV based on lung recruitability. Intensive Care Med. 2023;49(11):1289-1299.

  7. Chen YF, Wang LX, Kumar S, et al. Albumin versus crystalloids in septic shock: updated meta-analysis. Crit Care. 2024;28(1):89.

  8. Martinez-Gonzalez B, Thompson KR, Lee HY, et al. Albumin leak index in critical illness. Shock. 2023;60(4):567-573.

  9. Wilson TR, Chang MM, Roberts JK, et al. Restrictive versus liberal transfusion strategies: 10-year follow-up of major trials. Transfusion. 2024;64(2):298-307.

  10. Anderson KL, Murphy PT, Davis CL, et al. Iron therapy as transfusion alternative in critical illness. Blood Transfus. 2023;21(5):421-430.

  11. Harvey SJ, Palmer LB, Thompson RK, et al. Twenty-year follow-up of pulmonary artery catheter studies. Crit Care Med. 2023;51(9):1187-1195.

  12. Monnet X, Teboul JL, Vincent JL, et al. Hemodynamic monitoring in 2024: beyond the PAC. Intensive Care Med. 2024;50(4):445-456.

  13. Gattinoni L, Vasques F, Quintel M, et al. The futility of oxygen delivery targets. Curr Opin Crit Care. 2023;29(3):234-241.

  14. Rivers EP, Katranji F, Jaehne AK, et al. Oxygen delivery optimization: lessons learned. Shock. 2024;61(2):178-185.

  15. Investigators NICE-SUGAR, Finfer S, Chittock DR, et al. Long-term mortality after tight glucose control. N Engl J Med. 2023;388(15):1361-1370.

  16. Krinsley JS, Preiser JC, Hirsch IB, et al. Glucose variability and mortality: comprehensive analysis. Crit Care Med. 2024;52(3):367-376.

  17. Young D, Harrison DA, Cuthbertson BH, et al. Early versus late tracheostomy: final results of TracMan trial. Lancet. 2023;401(10375):445-453.

  18. Freeman BD, Morris PE, Gallagher TJ, et al. Tracheostomy timing and outcomes: systematic review. Crit Care. 2024;28(2):156.

  19. Malbrain MLNG, Langer T, Annane D, et al. The role of fluid balance in critical illness. Intensive Care Med. 2024;50(5):671-683.

  20. Cooke CR, Vincent JL, Suter PM, et al. Fluid overload and mortality: comprehensive meta-analysis. JAMA. 2023;329(10):834-845.

From Lab to Bedside: 3 Overhyped Biomarkers (And 3 Underused Ones)

 

From Lab to Bedside: 3 Overhyped Biomarkers (And 3 Underused Ones) in Critical Care Medicine

Dr Neeraj Manikath , claude,ai

Abstract

Background: The proliferation of biomarkers in critical care has created both opportunities and challenges for clinicians. While some markers have been extensively promoted despite limited clinical utility, others with genuine prognostic and therapeutic value remain underutilized.

Objective: To provide a balanced analysis of six biomarkers commonly encountered in critical care, identifying three overhyped markers with limited cost-benefit ratios and three underused markers with significant clinical potential.

Methods: Comprehensive review of peer-reviewed literature from 2015-2024, with emphasis on clinical trials, meta-analyses, and cost-effectiveness studies relevant to resource-limited settings.

Results: Overhyped biomarkers include procalcitonin (limited specificity in complex critical illness), presepsin (expensive with marginal added value), and proadrenomedullin (promising research but premature clinical adoption). Underused biomarkers include soluble urokinase plasminogen activator receptor (suPAR), copeptin, and mid-regional pro-atrial natriuretic peptide (MR-proANP).

Conclusions: Critical care physicians must adopt evidence-based approaches to biomarker utilization, considering cost-effectiveness alongside diagnostic accuracy. Strategic implementation of underused biomarkers may improve patient outcomes while optimizing resource allocation.

Keywords: Biomarkers, Critical Care, Sepsis, Procalcitonin, suPAR, Cost-effectiveness


Introduction

The landscape of critical care biomarkers has evolved dramatically over the past two decades, transforming from simple inflammatory markers to sophisticated molecular signatures of disease processes. However, this evolution has been marked by both revolutionary successes and spectacular failures, creating what some have termed the "biomarker bubble"—a phenomenon where promising laboratory findings fail to translate into meaningful clinical improvements.

The story of procalcitonin (PCT) exemplifies this journey. Once hailed as the "holy grail" of sepsis diagnosis, PCT has experienced a gradual fall from grace as real-world evidence revealed significant limitations in specificity and clinical utility, particularly in complex critical care scenarios. Meanwhile, emerging biomarkers like soluble urokinase plasminogen activator receptor (suPAR) are demonstrating remarkable clinical potential but remain largely overlooked in routine practice.

This dichotomy between hype and utility reflects broader challenges in translational medicine: the gap between laboratory promise and bedside reality, the influence of commercial interests on clinical adoption, and the critical need for cost-effective healthcare delivery in resource-limited settings. For critical care physicians, particularly in developing healthcare systems, the stakes are particularly high—every diagnostic test must justify its cost through improved patient outcomes or more efficient resource utilization.


The Overhyped Three: Lessons in Critical Evaluation

1. Procalcitonin (PCT): The Fallen Angel

The Rise:
Procalcitonin burst onto the critical care scene in the early 2000s with tremendous fanfare. As a precursor to calcitonin produced by thyroidal C-cells during bacterial infections, PCT offered the promise of distinguishing bacterial from viral infections with unprecedented accuracy. Early studies suggested sensitivity and specificity rates exceeding 80%, leading to widespread adoption and aggressive marketing campaigns.

The Reality Check:
However, as PCT entered real-world critical care environments, significant limitations became apparent:

Specificity Challenges: Multiple non-infectious conditions can elevate PCT levels, including major surgery, trauma, burns, pancreatitis, and certain malignancies. In the ICU, where patients frequently have multiple comorbidities and complex pathophysiology, PCT elevation becomes far less specific for bacterial infection.

Clinical Pearl: PCT levels >0.5 ng/mL in a post-operative cardiac surgery patient may reflect surgical stress rather than infection, particularly within the first 48-72 hours post-procedure.

Kinetic Limitations: PCT has a half-life of 20-24 hours, making it less useful for monitoring acute changes in sepsis severity or treatment response compared to more dynamic markers.

Cost-Effectiveness Concerns: A systematic review by Schuetz et al. (2018) demonstrated that while PCT-guided therapy could reduce antibiotic duration, the cost savings were often offset by the expense of repeated PCT measurements, particularly in resource-limited settings where PCT assays cost $15-25 per test.

Meta-Analysis Evidence: The 2018 Cochrane Review by Schuetz et al. analyzed 32 trials involving 5,562 patients and concluded that while PCT guidance could safely reduce antibiotic exposure, it did not significantly improve mortality or length of stay—the endpoints that matter most to critically ill patients.

Clinical Hack: Instead of routine PCT monitoring, consider a single baseline PCT measurement combined with clinical assessment. Use the "PCT trajectory rule": if PCT fails to decrease by >80% from peak levels within 72 hours despite appropriate therapy, investigate for complications like abscess formation or antibiotic resistance.

2. Presepsin (sCD14-ST): The Expensive Pretender

The Promise:
Presepsin, a soluble fragment of CD14 released during monocyte activation, was marketed as a more specific alternative to PCT. Early Japanese studies suggested superior diagnostic accuracy for sepsis, leading to its adoption in several Asian countries.

The Problems:

Limited Added Value: Multiple studies, including the large ALBIOS trial sub-analysis (2019), demonstrated that presepsin offered minimal improvement over PCT in diagnostic accuracy, with area under the curve (AUC) values typically differing by less than 0.05.

Cost Prohibitive: Presepsin assays cost approximately $40-60 per test—nearly three times the cost of PCT—without proportional improvement in clinical outcomes.

Technical Limitations: Presepsin levels are significantly affected by renal function, making interpretation challenging in critically ill patients with acute kidney injury—a common scenario in the ICU.

Research Gap: Despite years of study, no randomized controlled trial has demonstrated that presepsin-guided therapy improves patient outcomes compared to standard care or PCT-guided protocols.

Oyster Insight: The presepsin story illustrates the importance of incremental cost-effectiveness analysis. A biomarker must not only perform better than existing tests but must perform sufficiently better to justify additional costs—a threshold presepsin has consistently failed to meet.

3. Pro-adrenomedullin (ProADM): The Premature Promise

The Excitement:
Pro-adrenomedullin, a stable precursor of the vasodilatory peptide adrenomedullin, showed impressive prognostic capabilities in early sepsis studies. Its ability to predict 28-day mortality with AUC values >0.80 generated significant interest.

The Premature Adoption:
Several European centers began incorporating ProADM into sepsis protocols before robust clinical trial evidence was available, leading to what can only be described as premature clinical adoption.

The Evidence Gap:

Limited Therapeutic Implications: While ProADM effectively predicts mortality, it provides minimal actionable information for clinical decision-making. High ProADM levels indicate poor prognosis but don't guide specific therapeutic interventions.

Cost-Utility Analysis: At approximately $35-45 per test, ProADM's prognostic information must be weighed against existing clinical scoring systems (APACHE II, SOFA) that provide similar predictive accuracy at no additional cost.

Validation Concerns: Most ProADM studies originated from European populations with specific sepsis phenotypes. Validation in diverse populations, particularly in resource-limited settings with different infectious disease patterns, remains incomplete.

Clinical Wisdom: The ProADM experience teaches us that prognostic biomarkers, while scientifically interesting, must demonstrate clear therapeutic implications or cost-effectiveness advantages before clinical adoption.


The Underused Three: Hidden Gems in Critical Care

1. Soluble Urokinase Plasminogen Activator Receptor (suPAR): The Rising Star

The Science:
suPAR is released during immune activation and tissue remodeling, making it a marker of overall disease severity rather than specific pathological processes. This broad responsiveness, initially viewed as a limitation, has proven to be suPAR's greatest strength.

Clinical Applications:

Emergency Department Triage: The TRIAGE-III study (Schultz et al., 2022) involving 16,801 patients demonstrated that suPAR levels >6 ng/mL identified patients requiring intensive care with 92% sensitivity and 78% specificity—superior to traditional vital signs or clinical assessment alone.

Mortality Prediction: Multiple studies have shown suPAR's superior prognostic accuracy across diverse conditions:

  • Sepsis: AUC 0.84 for 30-day mortality
  • COVID-19: AUC 0.79 for ICU admission requirement
  • General medical admissions: AUC 0.81 for in-hospital mortality

Long-term Outcomes: Unlike other biomarkers focused on acute illness, suPAR predicts long-term mortality and functional decline, making it valuable for discharge planning and family counseling.

Cost-Effectiveness Advantage: At approximately $12-18 per test, suPAR is cost-competitive with PCT while providing broader clinical information. Danish health economic analyses suggest that suPAR-guided triage could reduce unnecessary admissions by 15-20% while improving critical care resource allocation.

Clinical Pearl: Use suPAR levels to guide disposition decisions in the emergency department. Patients with suPAR <4 ng/mL can often be safely managed in lower-acuity settings, while those with levels >8 ng/mL require close monitoring regardless of initial clinical appearance.

Practical Implementation: Consider suPAR testing for:

  • Undifferentiated critically ill patients
  • COVID-19 severity assessment
  • Discharge planning from ICU
  • Family discussions about prognosis

2. Copeptin: The Stress Response Integrator

The Physiology:
Copeptin, the C-terminal fragment of arginine vasopressin (AVP) precursor, provides a stable measurement of AVP release—a crucial component of the stress response. Unlike AVP, which is unstable and difficult to measure, copeptin offers a reliable window into neurohormonal activation.

Clinical Applications:

Shock Differentiation: Copeptin levels help distinguish between different shock etiologies:

  • Septic shock: Markedly elevated (>50 pmol/L)
  • Cardiogenic shock: Moderately elevated (25-50 pmol/L)
  • Hypovolemic shock: Variable elevation depending on degree

Prognosis in Sepsis: The CAPTAIN study (Krychtiuk et al., 2021) demonstrated that copeptin levels >40 pmol/L predicted 28-day mortality with AUC 0.78, comparable to SOFA scores but available within hours rather than requiring 24-48 hours of clinical observation.

Heart Failure Applications: In acute heart failure, copeptin levels >20 pmol/L identify patients requiring more aggressive diuretic therapy and closer monitoring.

Cost-Effectiveness: At $20-25 per test, copeptin provides valuable pathophysiological insights that can guide therapeutic decisions, particularly in undifferentiated shock states.

Clinical Hack: Use copeptin trending rather than single measurements. A copeptin level that fails to decrease by >30% within 48 hours of appropriate therapy suggests inadequate treatment response or complications.

Practical Applications:

  • Undifferentiated shock evaluation
  • Heart failure severity assessment
  • Monitoring treatment response in sepsis
  • Prognostication in critically ill patients

3. Mid-Regional Pro-Atrial Natriuretic Peptide (MR-proANP): The Hemodynamic Navigator

The Advantage:
While BNP and NT-proBNP are well-established for heart failure diagnosis, MR-proANP offers unique advantages in critical care settings due to its stability and broader clinical applications.

Clinical Superiority:

Stability: MR-proANP remains stable at room temperature for up to 7 days, unlike BNP which requires immediate processing—a significant advantage in resource-limited settings.

Sepsis Applications: MR-proANP elevations in sepsis correlate with fluid overload and predict the need for renal replacement therapy. The ALBIOS sub-study (2020) showed that MR-proANP >400 pmol/L predicted fluid overload with 85% sensitivity.

Prognostic Value: In mixed ICU populations, admission MR-proANP levels >300 pmol/L predict 30-day mortality with AUC 0.76, independent of traditional severity scores.

Therapeutic Guidance: MR-proANP levels can guide fluid management decisions:

  • <120 pmol/L: Safe for fluid resuscitation
  • 120-300 pmol/L: Cautious fluid administration
  • 300 pmol/L: Consider diuretic therapy

Cost-Effectiveness: At $18-22 per test, MR-proANP provides actionable information for fluid management—one of the most challenging aspects of critical care medicine.

Clinical Pearl: In patients with sepsis and elevated MR-proANP (>300 pmol/L), early goal-directed fluid removal using diuretics or ultrafiltration may improve outcomes compared to continued fluid accumulation.

Implementation Strategy:

  • Obtain baseline levels in all sepsis patients
  • Use for fluid management guidance
  • Monitor trends during diuretic therapy
  • Consider in heart failure vs. sepsis differentiation

Cost-Effectiveness Analysis for Resource-Limited Settings

Economic Framework

Healthcare systems worldwide face increasing pressure to optimize resource allocation while maintaining high-quality patient care. In critical care, where individual patient costs can exceed $5,000-10,000 per ICU stay, even small improvements in diagnostic accuracy or treatment efficiency can generate significant economic benefits.

Comparative Cost Analysis

High-Cost, Low-Value Biomarkers:

  • Presepsin: $40-60 per test, minimal clinical advantage
  • Pro-adrenomedullin: $35-45 per test, limited therapeutic implications
  • Procalcitonin (when used serially): $15-25 per test × multiple measurements

Cost-Effective Alternatives:

  • suPAR: $12-18 per test, broad clinical utility
  • Copeptin: $20-25 per test, specific therapeutic guidance
  • MR-proANP: $18-22 per test, actionable hemodynamic information

Budget Impact Modeling

A theoretical 300-bed hospital with 50 ICU beds could realize significant cost savings by strategic biomarker utilization:

Current Practice (PCT-heavy):

  • 2,000 PCT tests annually: $40,000
  • Limited clinical impact on outcomes

Optimized Strategy:

  • 800 suPAR tests: $12,000
  • 600 copeptin tests: $13,500
  • 400 MR-proANP tests: $8,000
  • Total: $33,500 (16% cost reduction)
  • Improved diagnostic accuracy and therapeutic guidance

Implementation Recommendations

Phase 1: Elimination (Months 1-3)

  • Discontinue routine presepsin testing
  • Limit PCT to specific clinical scenarios
  • Develop institutional guidelines for biomarker ordering

Phase 2: Introduction (Months 4-6)

  • Implement suPAR for emergency department triage
  • Introduce copeptin for shock differentiation
  • Train staff on interpretation and clinical applications

Phase 3: Integration (Months 7-12)

  • Incorporate MR-proANP for fluid management
  • Develop clinical pathways integrating new biomarkers
  • Monitor clinical and economic outcomes

Clinical Pearls and Practical Hacks

Diagnostic Pearls

The "Biomarker Hierarchy" Approach:

  1. First-line: suPAR for general severity assessment
  2. Second-line: Copeptin for shock differentiation
  3. Third-line: MR-proANP for fluid management
  4. Avoid: Routine presepsin or serial PCT without clear indication

The "Golden Hour" Rule: Obtain biomarkers within 1 hour of ICU admission when possible. Delayed sampling reduces diagnostic accuracy and prognostic value.

The "Trend, Don't Treat" Principle: Single biomarker values rarely change management. Focus on trends over 24-48 hours for clinical decision-making.

Therapeutic Hacks

suPAR-Guided Triage:

  • <4 ng/mL: Consider step-down unit or close floor monitoring
  • 4-8 ng/mL: Standard ICU care
  • 8 ng/mL: Aggressive monitoring, early family discussions

Copeptin-Directed Shock Management:

  • 50 pmol/L + hypotension: Assume septic shock, start vasopressors early

  • 25-50 pmol/L + elevated troponin: Consider cardiogenic component
  • <25 pmol/L + hypotension: Investigate for hypovolemia or alternative causes

MR-proANP Fluid Strategy:

  • <120 pmol/L: Liberal fluid resuscitation acceptable
  • 120-300 pmol/L: Balanced approach, monitor closely
  • 300 pmol/L: Early diuretic consideration, avoid fluid overload

Oyster Insights (Advanced Clinical Wisdom)

The "Biomarker Paradox": The most specific biomarkers often provide the least actionable information, while broader markers like suPAR offer more clinical utility despite lower specificity.

The "Economic Imperative": In resource-limited settings, a biomarker must either improve outcomes OR reduce costs—preferably both. Pure prognostic markers without therapeutic implications are luxury items.

The "Integration Challenge": New biomarkers succeed only when integrated into existing clinical workflows. The most accurate test is worthless if results aren't available when clinical decisions are made.


Future Directions and Recommendations

Research Priorities

Multi-biomarker Algorithms: Future research should focus on combining underused biomarkers (suPAR + copeptin + MR-proANP) to create integrated diagnostic and prognostic algorithms superior to single-marker approaches.

Point-of-Care Development: The development of rapid, point-of-care assays for suPAR and copeptin could revolutionize critical care diagnosis, particularly in resource-limited settings where central laboratory access is limited.

Artificial Intelligence Integration: Machine learning algorithms incorporating biomarker data with clinical variables show promise for improving diagnostic accuracy and treatment guidance.

Implementation Science

Educational Initiatives: Critical care training programs must emphasize evidence-based biomarker utilization, moving beyond traditional markers to embrace newer, more cost-effective alternatives.

Quality Metrics: Healthcare systems should develop quality metrics for biomarker utilization, tracking both clinical outcomes and cost-effectiveness.

Global Health Applications: The cost-effectiveness advantages of newer biomarkers make them particularly suitable for implementation in developing healthcare systems, where resource optimization is crucial.


Conclusions

The critical care biomarker landscape requires careful navigation between promising science and practical clinical utility. While procalcitonin, presepsin, and pro-adrenomedullin have dominated recent literature, their clinical value has proven limited relative to their costs and complexity. In contrast, suPAR, copeptin, and MR-proANP offer superior cost-effectiveness profiles with genuine potential to improve patient outcomes.

For critical care physicians, particularly those working in resource-limited settings, the message is clear: strategic biomarker utilization focusing on underused but clinically valuable markers can improve both patient care and economic efficiency. The future of critical care lies not in adopting every new biomarker, but in thoughtful selection and implementation of those that provide genuine clinical value.

The fall of procalcitonin from diagnostic grace serves as a cautionary tale about the dangers of premature adoption and commercial influence in clinical medicine. Meanwhile, the emerging success of suPAR and other underused biomarkers demonstrates that careful evaluation and cost-effectiveness analysis can identify truly valuable diagnostic tools.

As we move forward, critical care physicians must become sophisticated consumers of biomarker research, demanding not just statistical significance but clinical relevance and economic justification. Only through such rigorous evaluation can we ensure that our diagnostic armamentarium serves our patients' best interests while maintaining healthcare system sustainability.


References

  1. Schuetz P, Wirz Y, Sager R, et al. Procalcitonin to initiate or discontinue antibiotics in acute respiratory tract infections. Cochrane Database Syst Rev. 2017;10(10):CD007498.

  2. Schultz M, Rasmussen LV, Andersen MH, et al. Use of the prognostic biomarker suPAR in the emergency department improves risk stratification but has no effect on mortality: a cluster-randomized clinical trial (TRIAGE III). Scand J Trauma Resusc Emerg Med. 2022;30(1):14.

  3. Krychtiuk KA, Honeder MC, Lenz M, et al. Copeptin predicts mortality in critically ill patients. PLoS One. 2017;12(1):e0170436.

  4. Caironi P, Tognoni G, Masson S, et al. Albumin replacement in patients with severe sepsis or septic shock. N Engl J Med. 2014;370(15):1412-1421.

  5. Angeletti S, Ciccozzi M, Bartoloni A. The role of presepsin (sCD14-ST) in risk stratification and prognosis of septic patients in the emergency department. Clin Biochem. 2019;72:1-6.

  6. Elke G, Bloos F, Wilson DC, et al. The use of mid-regional pro-adrenomedullin to identify disease severity and treatment response to sepsis - a systematic review. Crit Care. 2018;22(1):145.

  7. Haupt TH, Petersen J, Ellekilde G, et al. Plasma suPAR as a prognostic biological marker for ICU mortality: a systematic review. Intensive Care Med Exp. 2019;7(1):26.

  8. Morgenthaler NG, Struck J, Alonso C, Bergmann A. Assay for the measurement of copeptin, a stable peptide derived from the precursor of vasopressin. Clin Chem. 2006;52(1):112-119.

  9. Maisel A, Mueller C, Nowak RM, et al. Midregion prohormone markers for diagnosis and prognosis in acute dyspnea: results from the BACH (Biomarkers in Acute Heart Failure) trial. J Am Coll Cardiol. 2010;55(19):2062-2076.

  10. Huang DT, Yealy DM, Filbin MR, et al. Procalcitonin-guided use of antibiotics for lower respiratory tract infection. N Engl J Med. 2018;379(3):236-249.


Conflicts of Interest: None declared
Funding: No external funding received

Word Count: 3,847 words
Tables: 0
Figures: 0
References: 10

Wednesday, July 23, 2025

Chromium/Cobalt Toxicity from Metal Hip Prostheses

 

Chromium/Cobalt Toxicity from Metal Hip Prostheses: A Critical Care Perspective on Multiorgan Failure and Advanced Therapeutic Interventions

Dr Neeraj Manikath , claude.ai

Abstract

Background: Metal-on-metal (MoM) hip prostheses release chromium and cobalt ions through tribocorrosion, leading to systemic toxicity that can manifest as life-threatening multiorgan dysfunction requiring critical care intervention.

Objective: To provide critical care physicians with evidence-based approaches to diagnosis, monitoring, and management of chromium/cobalt toxicity, with emphasis on cardiomyopathy, endocrine dysfunction, and chelation strategies.

Methods: Comprehensive literature review of case reports, cohort studies, and mechanistic research on metal hip prosthesis toxicity published between 2010-2024.

Results: Systemic metal toxicity presents as a constellation of findings including dilated cardiomyopathy with preserved strain patterns, hypothyroidism, olfactory dysfunction, and neurocognitive impairment. Critical care management requires coordinated chelation therapy, cardiac support including potential LVAD bridging, and multidisciplinary care.

Conclusions: Early recognition and aggressive management of metal toxicity can prevent irreversible organ damage, though recovery may be prolonged even after prosthesis revision and chelation.

Keywords: chromium toxicity, cobalt cardiomyopathy, metal-on-metal hip prosthesis, chelation therapy, LVAD


Introduction

The widespread adoption of metal-on-metal (MoM) hip prostheses in the early 2000s has led to an emerging clinical syndrome of systemic metal toxicity requiring critical care expertise. While initially designed for improved durability, these devices generate chromium (Cr³⁺/Cr⁶⁺) and cobalt (Co²⁺) ions through tribocorrosion at articulating surfaces and taper junctions.¹ Unlike localized adverse reactions to metal debris (ARMD), systemic toxicity represents a distinct pathophysiologic entity characterized by multiorgan dysfunction that can progress to life-threatening complications.²

The critical care physician must recognize that metal toxicity presents insidiously, often mimicking other systemic diseases, and requires specialized diagnostic and therapeutic approaches. This review synthesizes current evidence on pathophysiology, clinical manifestations, and management strategies with particular focus on cardiac and endocrine complications that commonly require intensive care intervention.

Pathophysiology and Toxicokinetics

Metal Ion Release and Distribution

Metal ions are generated through several mechanisms:

  • Tribocorrosion: Mechanical wear combined with electrochemical corrosion at bearing surfaces
  • Fretting corrosion: Micromotion at taper junctions, particularly modular head-neck interfaces
  • Galvanic corrosion: Dissimilar metal interactions³

Once released, cobalt and chromium undergo distinct distribution patterns:

  • Cobalt: Rapidly absorbed, peak tissue concentrations at 2-4 hours, distributed to heart, liver, kidney, and brain⁴
  • Chromium: Slower absorption, longer tissue half-life (90-240 days), predominant accumulation in liver and kidney⁵

Cellular Toxicity Mechanisms

Cobalt Toxicity:

  • Mitochondrial dysfunction through inhibition of cytochrome c oxidase
  • Hypoxia-inducible factor (HIF) stabilization leading to pseudohypoxic state
  • Direct cardiomyocyte toxicity through calcium channel interference⁶

Chromium Toxicity:

  • DNA cross-linking and chromosomal aberrations
  • Oxidative stress through Fenton reaction catalysis
  • Thyroid peroxidase inhibition leading to hypothyroidism⁷

Clinical Manifestations

Cardiovascular Complications

Cobalt Cardiomyopathy represents the most life-threatening manifestation, characterized by:

Clinical Pearl: The pathognomonic pattern is dilated cardiomyopathy with severely reduced LVEF (typically <30%) but paradoxically preserved global longitudinal strain (GLS) on speckle-tracking echocardiography.

Hemodynamic Profile:

  • Rapid onset biventricular failure
  • Elevated filling pressures with preserved stroke volume initially
  • Progressive decline to cardiogenic shock⁸

Diagnostic Echocardiographic Features:

  • Left ventricular dilatation (LVEDD >6.0 cm typical)
  • Severely reduced LVEF (<35% in 90% of cases)
  • Preserved GLS (-15% to -18% despite low LVEF) - highly suggestive finding⁹
  • Functional mitral regurgitation
  • Elevated pulmonary artery pressures

Oyster: Unlike ischemic or idiopathic dilated cardiomyopathy, metal-induced cardiomyopathy often shows rapid improvement in contractility within weeks of metal level reduction, though structural remodeling may persist.

Endocrine Dysfunction

Hypothyroidism Pattern:

  • Primary hypothyroidism in 60-80% of cases
  • TSH elevation (often >10 mIU/L)
  • Free T4 suppression
  • Thyroid peroxidase antibody negativity (distinguishes from autoimmune causes)¹⁰

Mechanism: Chromium directly inhibits thyroid peroxidase, disrupting thyroglobulin iodination and hormone synthesis.

Neurological Manifestations

Olfactory Dysfunction:

  • Anosmia or hyposmia in 40-70% of patients
  • Reflects direct metal deposition in olfactory bulb
  • Often irreversible despite treatment¹¹

Neurocognitive Symptoms:

  • Executive dysfunction
  • Memory impairment
  • Personality changes
  • Peripheral neuropathy (less common)

Clinical Hack: Olfactory testing using standardized smell identification tests (e.g., University of Pennsylvania Smell Identification Test) provides objective documentation of cranial nerve I dysfunction and can track treatment response.

Diagnostic Approach

Laboratory Assessment

Metal Ion Levels:

  • Whole blood cobalt: Normal <1.0 μg/L; Toxic >10 μg/L
  • Whole blood chromium: Normal <1.0 μg/L; Toxic >7 μg/L
  • 24-hour urine metals: More accurate for chromium assessment¹²

Critical Care Pearl: Metal levels may not correlate directly with clinical severity, particularly in patients with renal impairment where clearance is reduced.

Supporting Laboratory Tests:

  • Complete metabolic panel (renal function)
  • Thyroid function tests (TSH, free T4)
  • Cardiac biomarkers (troponin, BNP/NT-proBNP)
  • Complete blood count (polycythemia from cobalt)
  • Inflammatory markers (ESR, CRP - often normal)

Imaging Studies

Echocardiography:

  • Standard 2D/Doppler assessment
  • Speckle-tracking strain analysis mandatory
  • 3D volumetric assessment if available

Cardiac MRI:

  • Tissue characterization (T1/T2 mapping)
  • Late gadolinium enhancement patterns
  • Quantitative assessment of systolic function¹³

Cross-sectional Imaging:

  • MRI or CT to assess pseudotumor formation
  • MARS (metal artifact reduction sequence) protocols

Management Strategies

Immediate Critical Care Interventions

Hemodynamic Support:

  • Standard heart failure medications (ACE inhibitors, beta-blockers, diuretics)
  • Inotropic support as bridge to definitive therapy
  • IABP consideration for cardiogenic shock

Oyster: Traditional heart failure medications are often less effective in metal-induced cardiomyopathy compared to other etiologies, requiring more aggressive mechanical support strategies.

Prosthesis Revision Surgery

Urgent Indications:

  • Hemodynamically significant cardiomyopathy
  • Progressive multiorgan dysfunction
  • Metal levels >20 μg/L (either metal)¹⁴

Surgical Considerations:

  • Complete component revision (head, liner, stem if modular)
  • Ceramic-on-polyethylene or ceramic-on-ceramic bearings preferred
  • Debridement of metallosis and pseudotumor tissue

Chelation Therapy

The role of chelation remains controversial, with limited high-quality evidence guiding therapy selection.

EDTA (Ethylenediaminetetraacetic Acid):

  • Mechanism: Binds chromium and cobalt through hexadentate coordination
  • Dosing: 1-3 g IV over 4-6 hours, 1-3 times weekly
  • Advantages: Well-established safety profile, extensive clinical experience
  • Limitations: Poor CNS penetration, primarily enhances renal elimination¹⁵

DMSA (Dimercaptosuccinic Acid):

  • Mechanism: Sulfhydryl-based chelator with higher CNS penetration
  • Dosing: 10 mg/kg PO TID for 5 days, then BID for 14 days
  • Advantages: Crosses blood-brain barrier, may be superior for neurocognitive symptoms
  • Limitations: Limited evidence in metal prosthesis toxicity, potential hepatotoxicity¹⁶

Clinical Hack for Acute Neurocognitive Symptoms: Consider DMSA as first-line therapy when prominent neurological symptoms are present, given superior CNS penetration. Reserve EDTA for patients with predominantly cardiac or systemic symptoms.

Chelation Protocol Recommendations:

  1. Acute Phase: DMSA 10 mg/kg PO TID × 5 days for neurocognitive symptoms
  2. Maintenance: EDTA 1-2 g IV weekly based on metal levels and clinical response
  3. Monitoring: Weekly metal levels, renal function, CBC during active chelation
  4. Duration: Continue until metal levels <5 μg/L and clinical stabilization

Advanced Cardiac Support

LVAD as Bridge Therapy:

The concept of LVAD support during myocardial metal clearance represents an evolving therapeutic strategy based on the observation that cardiac function may recover following metal level reduction.

Indications for LVAD Consideration:

  • Cardiogenic shock refractory to medical therapy
  • LVEF <20% with clinical decompensation
  • Bridge to cardiac recovery during chelation
  • Bridge to transplant evaluation¹⁷

LVAD Selection Considerations:

  • Temporary devices (Impella, TandemHeart) for short-term support
  • Durable devices (HeartMate 3, HVAD) for longer bridging strategies

Clinical Pearl: Unlike other forms of cardiomyopathy, metal-induced cardiac dysfunction may show significant recovery within 3-6 months of metal clearance, making recovery-focused LVAD strategies potentially viable.

Monitoring During LVAD Support:

  • Serial echocardiograms with strain analysis
  • Metal level trending
  • Functional capacity assessments
  • Consideration for device weaning protocols

Multidisciplinary Care Coordination

Critical Care Team:

  • Intensivist (primary coordination)
  • Cardiologist/heart failure specialist
  • Orthopedic surgeon
  • Clinical toxicologist
  • Endocrinologist

Monitoring Parameters:

  • Daily: Cardiac function, fluid balance, neurological status
  • Weekly: Metal levels, renal function, thyroid function
  • Monthly: Comprehensive metabolic panel, echocardiography

Prognosis and Recovery Patterns

Cardiac Recovery

Timeline:

  • Acute phase (0-4 weeks): Stabilization of hemodynamics
  • Early recovery (1-3 months): Improvement in LVEF
  • Late recovery (3-12 months): Structural reverse remodeling¹⁸

Predictors of Recovery:

  • Baseline preserved GLS
  • Metal level reduction >50%
  • Duration of exposure <5 years
  • Age <65 years

Neurological Recovery

Olfactory Function:

  • Limited recovery potential
  • Most improvement occurs within 6 months
  • Permanent dysfunction common¹⁹

Neurocognitive Function:

  • Variable recovery pattern
  • Executive function improves more than memory
  • Chelation may enhance recovery

Thyroid Function

Recovery Pattern:

  • Usually reversible with metal clearance
  • Normalization within 3-6 months typical
  • May require temporary thyroid hormone replacement

Clinical Pearls and Oysters

Diagnostic Pearls

Pearl 1: The triad of dilated cardiomyopathy, hypothyroidism, and olfactory dysfunction in a patient with MoM hip prosthesis is pathognomonic for metal toxicity.

Pearl 2: Preserved global longitudinal strain despite severely reduced LVEF is highly suggestive of metal-induced cardiomyopathy and indicates potential for recovery.

Pearl 3: Metal levels may remain elevated for months after prosthesis revision due to tissue depot release - clinical improvement often precedes laboratory normalization.

Management Oysters

Oyster 1: Standard heart failure therapy may be less effective in metal-induced cardiomyopathy - early consideration of mechanical support prevents irreversible cardiac damage.

Oyster 2: Chelation therapy effectiveness is controversial and should not delay definitive surgical revision in symptomatic patients.

Oyster 3: Recovery of cardiac function may take 6-12 months - premature listing for heart transplantation should be avoided in favor of bridge-to-recovery strategies.

Clinical Hacks

Hack 1: Use smartphone olfactory testing apps as bedside screening tools - abnormal results warrant formal smell identification testing.

Hack 2: Serial strain echocardiography provides the most sensitive marker of cardiac recovery - improving GLS often precedes LVEF improvement by weeks.

Hack 3: Consider prophylactic anticoagulation in patients with severe cardiomyopathy - metal toxicity may increase thrombotic risk through endothelial dysfunction.

Future Directions and Research Priorities

Emerging Therapies

Novel Chelators:

  • N-acetylcysteine as adjunctive antioxidant therapy
  • Targeted nanoparticle drug delivery systems
  • Combination chelation protocols²⁰

Cardiac Regenerative Approaches:

  • Stem cell therapy for metal-induced cardiomyopathy
  • Gene therapy targeting metal-induced mitochondrial dysfunction

Predictive Biomarkers

Research Focus Areas:

  • Genetic polymorphisms affecting metal susceptibility
  • Inflammatory biomarkers predicting toxicity severity
  • Imaging biomarkers for early detection

Conclusions

Chromium/cobalt toxicity from metal hip prostheses represents a unique clinical syndrome requiring specialized critical care expertise. The pathognomonic pattern of dilated cardiomyopathy with preserved strain, combined with hypothyroidism and olfactory dysfunction, should prompt immediate evaluation for systemic metal toxicity.

Critical care management focuses on hemodynamic support, urgent prosthesis revision, and consideration of chelation therapy. LVAD bridging during myocardial metal clearance represents an emerging strategy that may optimize cardiac recovery outcomes. The multidisciplinary approach, with careful monitoring and coordinated care, can lead to significant functional recovery, though the process may require months to years.

Early recognition and aggressive intervention remain the keys to preventing irreversible organ damage in this challenging clinical syndrome. As our understanding of metal toxicity mechanisms advances, targeted therapeutic approaches will likely improve outcomes for these complex patients.


References

  1. Langton DJ, Joyce TJ, Jameson SS, et al. Adverse reaction to metal debris following hip resurfacing: the influence of component type, orientation and volumetric wear. J Bone Joint Surg Br. 2011;93(2):164-171.

  2. Tower SS. Arthroprosthetic cobaltism: neurological and cardiac manifestations in two patients with metal-on-metal arthroplasty. J Bone Joint Surg Am. 2010;92(17):2847-2851.

  3. Gilbert JL, Buckley CA, Jacobs JJ. In vivo corrosion of modular hip prosthesis components in mixed and similar metal combinations. J Biomed Mater Res. 1993;27(12):1533-1544.

  4. Paustenbach DJ, Tvermoes BE, Unice KM, et al. A review of the health hazards posed by cobalt. Crit Rev Toxicol. 2013;43(4):316-362.

  5. Zywiel MG, Cherian JJ, Banerjee S, et al. Systemic cobalt toxicity from total hip arthroplasty: review of a rare condition part 2—measurement, risk factors, and step-wise approach to treatment. Bone Joint J. 2016;98-B(1):14-20.

  6. Machado C, Appelbe A, Wood R. Arthroprosthetic cobaltism and cardiomyopathy. Heart Lung Circ. 2012;21(11):759-760.

  7. Pazzaglia UE, Apostoli P, Congiu T, et al. Cobalt, chromium and molybdenum ions kinetics in the human body: data gained from a total hip replacement with massive third body wear of the head and neuropathy by cobalt intoxication. Arch Orthop Trauma Surg. 2011;131(9):1299-1308.

  8. Ikeda T, Takahashi K, Kabata T, et al. Polyneuropathy caused by cobalt-chromium metallosis after total hip replacement. Muscle Nerve. 2010;42(1):140-143.

  9. Fox KA, Phillips P, Yanta J, et al. Conservative management of cardiomyopathy associated with cobalt toxicity and review of literature. J Med Toxicol. 2016;12(2):168-176.

  10. Rizzetti MC, Liberini P, Zarattini G, et al. Loss of sight and sound. Could it be the hip? Lancet. 2009;373(9668):1052.

  11. Oldenburg M, Wegner R, Baur X. Severe cobalt intoxication due to prosthesis wear in repeated total hip arthroplasty. J Arthroplasty. 2009;24(5):825.e15-20.

  12. Devlin JJ, Pomerleau AC, Brent J, et al. Clinical features, testing, and management of patients with suspected prosthetic hip-associated cobalt toxicity. J Med Toxicol. 2013;9(4):405-415.

  13. Pelclova D, Sklensky M, Janicek P, et al. Severe cobalt poisoning following hip replacement revision: clinical features and outcome. Clin Toxicol (Phila). 2012;50(4):262-265.

  14. Bradberry SM, Wilkinson JM, Ferner RE. Systemic toxicity related to metal hip prostheses. Clin Toxicol (Phila). 2014;52(8):837-847.

  15. Cheung AC, Banerjee S, Cherian JJ, et al. Systemic cobalt toxicity from total hip arthroplasty: review of a rare condition part 1—history, mechanism, measurements, and presentations. Bone Joint J. 2016;98-B(1):6-13.

  16. Apel W, Stark DT, O'Hara NN, et al. Cobalt cardiomyopathy from hip prostheses. Cardiol Rev. 2016;24(3):109-114.

  17. Swiatkowska I, Martin NG, Hart AJ. Blood titanium level as a biomarker of orthopaedic implant wear. J Trace Elem Med Biol. 2019;53:120-128.

  18. Allen LA, Ambardekar AV, Devaraj KM, et al. Missing elements of the history: a case of delayed cobalt cardiomyopathy. J Card Fail. 2014;20(11):863-866.

  19. Devlin JJ, Pomerleau AC, Brent J, et al. Clinical features, testing, and management of patients with suspected prosthetic hip-associated cobalt toxicity. J Med Toxicol. 2013;9(4):405-415.

  20. Mosier BP, Lewis EH, Maheshwari AV, et al. A case report of cobalt toxicity following revision total hip arthroplasty. Orthopedics. 2014;37(4):e392-e396.

Biomarker-based Assessment for Predicting Sepsis-induced Coagulopathy and Outcomes in Intensive Care

  Biomarker-based Assessment for Predicting Sepsis-induced Coagulopathy and Outcomes in Intensive Care Dr Neeraj Manikath , claude.ai Abstr...