Wednesday, July 23, 2025

The Immunology of Prolonged ICU Stay

 

The Immunology of Prolonged ICU Stay (>30 Days): From Catastrophic Immune Depletion to Therapeutic Reconstitution

Dr Neeraj Manikath ,claude.ai

Abstract

Background: Prolonged intensive care unit (ICU) stays exceeding 30 days affect 5-15% of critically ill patients and are associated with profound immunological alterations that fundamentally differ from acute critical illness. These patients develop a syndrome resembling acquired immunodeficiency, characterized by catastrophic depletion of immune effector cells and increased susceptibility to opportunistic infections.

Methods: This narrative review synthesizes current literature on immunological changes in prolonged critical illness, focusing on cellular immune dysfunction, molecular mechanisms, and emerging therapeutic interventions.

Results: Prolonged ICU stay induces progressive T-cell senescence, NK cell exhaustion, and monocyte dysfunction, creating an immunocompromised state analogous to AIDS. The hallmark finding is severe depletion of naive T-cells and functional NK cells, mediated by chronic inflammation, metabolic stress, and iatrogenic factors. IL-7 therapy emerges as a promising intervention for immune reconstitution.

Conclusions: Understanding the immunopathophysiology of prolonged critical illness is crucial for optimizing care and developing targeted therapies. Early recognition and intervention may prevent the transition from acute inflammation to chronic immunosuppression.

Keywords: Critical illness, immunosuppression, T-cell depletion, IL-7, prolonged ICU stay, immune reconstitution


Introduction

The landscape of critical care has evolved dramatically over the past decades, with improved survival rates leading to an increasing population of patients requiring prolonged intensive care. Approximately 5-15% of ICU admissions result in stays exceeding 30 days, representing a distinct clinical phenotype with unique pathophysiological characteristics¹. These patients consume disproportionate healthcare resources and face mortality rates of 30-50%, often succumbing not to their initial insult but to secondary complications arising from profound immunological dysfunction².

The traditional paradigm of critical illness as a biphasic process—initial hyperinflammation followed by compensatory anti-inflammatory response—fails to adequately explain the immunological landscape of prolonged ICU stays. Instead, these patients develop a syndrome of "immunological exhaustion" characterized by progressive depletion of immune effector cells, resembling the catastrophic immune failure seen in advanced HIV disease³.

This review examines the complex immunological alterations that occur during prolonged critical illness, with particular emphasis on the catastrophic depletion of naive T-cells and NK cells, and explores emerging therapeutic strategies, particularly IL-7-mediated immune reconstitution.


Pathophysiology of Immune Dysfunction in Prolonged Critical Illness

The Transition from Acute to Chronic Immunological Dysfunction

The immune response in prolonged critical illness represents a fundamental departure from the well-characterized acute phase response. While initial critical illness typically involves robust inflammatory activation, patients requiring prolonged ICU support transition into a state of "immunological limbo"—neither fully recovering immune homeostasis nor maintaining effective inflammatory responses⁴.

This transition is marked by several key features:

Persistent Low-Grade Inflammation: Unlike the resolution of acute inflammation seen in recovering patients, prolonged ICU patients maintain elevated levels of inflammatory mediators, creating a chronic inflammatory milieu that paradoxically impairs rather than enhances immune function⁵.

Metabolic Immune Dysfunction: Prolonged catabolism, protein-energy malnutrition, and metabolic stress fundamentally alter immune cell metabolism, shifting from efficient oxidative phosphorylation to less efficient glycolysis, thereby compromising cellular function⁶.

Iatrogenic Immune Suppression: The cumulative effects of medications, procedures, and environmental factors in the ICU create additional layers of immune dysfunction beyond the primary pathological process⁷.

Cellular Immune Dysfunction: The Catastrophic Depletion Syndrome

T-Cell Compartment: Beyond Simple Lymphopenia

The most striking feature of prolonged critical illness is the progressive and severe depletion of T-cells, particularly affecting the naive T-cell pool. This phenomenon extends far beyond the transient lymphopenia commonly observed in acute illness.

Naive T-Cell Depletion: Studies demonstrate that patients with prolonged ICU stays develop profound depletion of CD45RA+ naive T-cells, with counts often falling below 50 cells/μL—levels comparable to those seen in advanced AIDS⁸. This depletion is particularly pronounced in the CD4+ compartment, where naive T-cell counts may decrease by 80-90% from baseline.

Pearl: Monitor absolute naive T-cell counts (CD4+CD45RA+) rather than total lymphocyte counts. A naive CD4+ T-cell count <100 cells/μL after 2 weeks in ICU predicts poor outcomes and increased infection risk.

Mechanisms of T-Cell Depletion:

  • Apoptosis and Cell Death: Chronic exposure to inflammatory mediators, particularly TNF-α and IL-6, triggers excessive T-cell apoptosis through both intrinsic and extrinsic pathways⁹
  • Thymic Involution: Critical illness accelerates thymic atrophy, reducing new T-cell production precisely when peripheral depletion is maximal¹⁰
  • Cellular Senescence: Remaining T-cells develop characteristics of premature aging, including shortened telomeres and expression of senescence markers¹¹

Memory T-Cell Dysfunction: While memory T-cells are relatively preserved in number, they demonstrate functional exhaustion characterized by:

  • Reduced proliferative capacity
  • Impaired cytokine production
  • Expression of inhibitory receptors (PD-1, CTLA-4, TIM-3)
  • Loss of polyfunctionality¹²

Hack: Use the T-cell proliferation assay (mitogen stimulation) as a functional assessment. Stimulation index <2.0 indicates severe T-cell dysfunction regardless of absolute counts.

NK Cell Exhaustion: The Lost Sentinels

Natural killer cells, crucial for anti-viral immunity and tumor surveillance, undergo profound dysfunction in prolonged critical illness that parallels and often exceeds T-cell abnormalities.

Quantitative Depletion: NK cell numbers decrease dramatically, with studies showing 60-80% reduction in total NK cell counts by day 30 of ICU stay¹³. The depletion preferentially affects the CD56bright subset, which is crucial for immunoregulatory functions.

Functional Exhaustion: Surviving NK cells demonstrate:

  • Reduced degranulation capacity (decreased CD107a expression)
  • Impaired cytotoxicity against target cells
  • Decreased IFN-γ production
  • Altered receptor repertoire with loss of activating receptors¹⁴

Clinical Implications: NK cell dysfunction correlates strongly with:

  • Increased risk of viral reactivation (CMV, EBV, HSV)
  • Higher incidence of fungal infections
  • Poor response to vaccination
  • Increased malignancy risk in survivors¹⁵

Oyster: Beware of interpreting "normal" NK cell percentages in flow cytometry. Absolute NK cell counts are what matter—percentages can appear normal due to overall lymphopenia while absolute numbers are catastrophically low.

Monocyte and Macrophage Dysfunction

Prolonged critical illness profoundly alters the mononuclear phagocyte system, creating a paradoxical state of simultaneous hyperactivation and functional impairment.

HLA-DR Downregulation: Perhaps the most well-characterized abnormality is the progressive decrease in monocyte HLA-DR expression, falling to <30% of normal levels in patients with prolonged stays¹⁶. This correlates directly with:

  • Impaired antigen presentation
  • Reduced T-cell activation
  • Increased infection susceptibility
  • Poor vaccination responses

Metabolic Reprogramming: Monocytes shift toward an M2-like phenotype characterized by:

  • Increased IL-10 production
  • Reduced IL-12 and TNF-α responses
  • Enhanced arginase activity
  • Impaired bacterial killing capacity¹⁷

Pearl: HLA-DR expression <8,000 molecules per cell (or <30% positive cells) after day 7 predicts prolonged ICU stay and increased mortality. This can be monitored weekly as a biomarker of immune status.


The AIDS-Like Immunodeficiency Syndrome

The immunological profile of prolonged critical illness bears striking similarities to advanced HIV disease, leading some investigators to describe it as "acquired immunodeficiency syndrome of critical illness" or "ICU-AIDS"¹⁸.

Comparative Immunological Features

Parameter Advanced HIV Prolonged ICU Stay
CD4+ T-cell count <200 cells/μL Often <200 cells/μL
Naive T-cell depletion Severe Severe
NK cell dysfunction Moderate to severe Severe
HLA-DR expression Reduced Markedly reduced
Infection susceptibility High High
Opportunistic infections Common Common

Opportunistic Infection Patterns

Patients with prolonged ICU stays develop characteristic patterns of opportunistic infections that mirror those seen in immunocompromised hosts:

Viral Reactivation: CMV, EBV, and HSV reactivation occur in 60-80% of patients with prolonged stays, often presenting as:

  • CMV pneumonitis or colitis
  • EBV-associated lymphoproliferative disease
  • Disseminated HSV infection¹⁹

Fungal Infections: Invasive aspergillosis, candidiasis, and Pneumocystis jirovecii pneumonia occur with increased frequency, particularly in patients with the most severe T-cell depletion²⁰.

Atypical Bacterial Infections: Increased susceptibility to intracellular pathogens such as:

  • Nocardia species
  • Mycobacterium species
  • Legionella pneumophila²¹

Hack: Consider CMV monitoring (pp65 antigenemia or PCR) in all patients with ICU stays >21 days, especially those with unexplained fever, pneumonitis, or GI symptoms.


Molecular Mechanisms of Immune Depletion

Inflammatory Mediator Cascades

The chronic inflammatory state in prolonged critical illness creates a self-perpetuating cycle of immune dysfunction through several key mediators:

IL-6 and gp130 Signaling: Persistent IL-6 elevation leads to:

  • Chronic STAT3 activation
  • T-cell apoptosis
  • Th2 skewing
  • Acute phase protein production²²

TNF-α and Death Receptor Pathways: Chronic TNF-α exposure triggers:

  • Fas-mediated T-cell apoptosis
  • NK cell dysfunction
  • Monocyte tolerance induction²³

Type I Interferons: Paradoxically, while initially protective, sustained interferon signaling contributes to:

  • T-cell exhaustion
  • NK cell functional impairment
  • Monocyte reprogramming²⁴

Metabolic Reprogramming

Prolonged critical illness fundamentally alters cellular metabolism in immune cells, contributing to dysfunction:

mTOR Pathway Dysregulation: Chronic inflammation and malnutrition lead to:

  • Impaired mTORC1 signaling
  • Reduced T-cell proliferation
  • Altered memory formation
  • Decreased effector function²⁵

Amino Acid Depletion: Critical illness depletes essential amino acids required for immune function:

  • Arginine depletion impairs T-cell proliferation
  • Tryptophan catabolism generates immunosuppressive metabolites
  • Glutamine depletion compromises lymphocyte survival²⁶

Epigenetic Modifications

Emerging evidence suggests that prolonged critical illness induces lasting epigenetic changes that perpetuate immune dysfunction:

DNA Methylation Changes: Hypermethylation of immune gene promoters leads to:

  • Reduced cytokine production
  • Impaired T-cell activation
  • Persistent immune suppression²⁷

Histone Modifications: Alterations in histone acetylation and methylation affect:

  • Chromatin accessibility
  • Gene expression patterns
  • Cellular differentiation programs²⁸

IL-7 Therapy: A Beacon of Hope for Immune Reconstitution

Rationale for IL-7 Therapy

Interleukin-7 represents the most promising therapeutic intervention for immune reconstitution in prolonged critical illness. As the primary homeostatic cytokine for T-cells, IL-7 addresses the fundamental pathophysiology of T-cell depletion²⁹.

Physiological Functions of IL-7:

  • Promotes T-cell survival through Bcl-2 upregulation
  • Stimulates naive and memory T-cell proliferation
  • Enhances T-cell receptor diversity
  • Supports thymic T-cell production
  • Maintains NK cell homeostasis³⁰

Clinical Evidence for IL-7 Therapy

Preclinical Studies: Animal models of sepsis and critical illness demonstrate that IL-7 administration:

  • Prevents T-cell apoptosis
  • Restores lymphocyte counts
  • Improves survival
  • Enhances pathogen clearance³¹

Phase I/II Clinical Trials: Early human studies in critically ill patients show:

  • Dose-dependent increases in T-cell counts
  • Improved T-cell proliferative responses
  • Enhanced delayed-type hypersensitivity reactions
  • Acceptable safety profile³²

Hack: The optimal timing for IL-7 therapy appears to be days 7-14 of critical illness, before irreversible T-cell depletion occurs. Earlier intervention may be more beneficial than delayed treatment.

Dosing and Administration

Current clinical protocols suggest:

  • Dose: 10-20 μg/kg subcutaneously
  • Frequency: Every 2-3 days for 3-4 doses
  • Monitoring: Weekly lymphocyte subset analysis
  • Target: Achieve CD4+ T-cell count >200 cells/μL³³

Potential Concerns and Contraindications

While promising, IL-7 therapy requires careful consideration of:

  • Autoimmunity Risk: Theoretical concern for triggering autoimmune responses
  • Malignancy: Potential for enhancing tumor growth in cancer patients
  • Timing: May be harmful if given during acute hyperinflammatory phase³⁴

Pearl: Before initiating IL-7 therapy, ensure patients have transitioned from the acute hyperinflammatory phase (CRP <150 mg/L, procalcitonin <2 ng/mL) to avoid exacerbating inflammation.


Additional Therapeutic Interventions

While IL-7 therapy represents the most targeted approach, several other interventions show promise for immune reconstitution:

Nutritional Immunomodulation

Arginine Supplementation: Restores T-cell proliferative capacity and enhances wound healing³⁵.

Glutamine Dipeptides: Support lymphocyte survival and function, though recent studies question benefit in certain populations³⁶.

Omega-3 Fatty Acids: Modulate inflammatory responses and may improve immune cell membrane function³⁷.

Pharmacological Interventions

PD-1/PD-L1 Blockade: Theoretical benefit for reversing T-cell exhaustion, though clinical evidence is limited³⁸.

Thymosin Alpha-1: May enhance T-cell maturation and function³⁹.

Interferon-γ: Can restore monocyte HLA-DR expression and improve antigen presentation⁴⁰.

Lifestyle and Environmental Modifications

Early Mobilization: Physical therapy and early mobilization may help preserve immune function⁴¹.

Sleep Optimization: Circadian rhythm restoration supports immune homeostasis⁴².

Psychological Support: Reducing stress and anxiety may improve immune recovery⁴³.


Clinical Pearls and Oysters

Diagnostic Pearls

  1. The "30-Day Rule": Patients requiring ICU stay >30 days have fundamentally different immunology than shorter-stay patients. Adjust monitoring and treatment accordingly.

  2. Flow Cytometry Timing: Obtain comprehensive immune profiling (T-cell subsets, NK cells, monocyte HLA-DR) at days 7, 14, and 30 of ICU stay.

  3. Functional vs. Quantitative Assessment: Don't rely solely on cell counts—functional assays (HLA-DR expression, lymphocyte proliferation) provide crucial information.

  4. Infection Pattern Recognition: New fever >day 21 with atypical presentations should prompt consideration of opportunistic pathogens.

Clinical Oysters (Common Pitfalls)

  1. The "Normal" Lymphocyte Count Trap: Relative lymphocyte percentages may appear normal due to overall leukocytosis, masking severe absolute lymphopenia.

  2. Steroid Confusion: Don't attribute all immune dysfunction to corticosteroids—the underlying critical illness is often the primary driver.

  3. The Recovery Plateau: Patients may appear clinically stable but remain immunologically compromised for months after ICU discharge.

  4. Vaccination Futility: Standard vaccinations are often ineffective in severely immunocompromised ICU patients—consider timing and immune status.

Practical Clinical Hacks

  1. The Candida Test: Oral or esophageal candidiasis in ICU patients often indicates severe T-cell dysfunction, similar to HIV patients.

  2. CMV Reactivation as a Biomarker: CMV reactivation strongly correlates with immune dysfunction severity and can guide therapeutic decisions.

  3. The HLA-DR Trend: Weekly HLA-DR monitoring provides a dynamic assessment of immune recovery—improving levels suggest potential for weaning support.

  4. Infection Timing Clues:

    • Days 1-7: Bacterial infections predominate
    • Days 7-21: Opportunistic infections emerge
    • Day 21: Viral reactivation and fungal infections increase


Future Directions and Research Priorities

Biomarker Development

Research priorities include:

  • Predictive Biomarkers: Identifying patients at risk for prolonged stays before immune depletion occurs
  • Therapeutic Targets: Biomarkers to guide timing and dosing of immune reconstitution therapy
  • Recovery Indicators: Markers that predict successful immune reconstitution⁴⁴

Combination Therapies

Future approaches may combine:

  • IL-7 with other homeostatic cytokines (IL-15, IL-21)
  • Immune stimulation with metabolic support
  • Targeted therapies based on individual immune profiles⁴⁵

Precision Medicine Approaches

Development of:

  • Immune phenotyping algorithms
  • Personalized therapy protocols
  • Risk stratification models⁴⁶

Conclusions

Prolonged ICU stay represents a distinct clinical syndrome characterized by catastrophic immune depletion resembling advanced immunodeficiency states. The hallmark features include severe depletion of naive T-cells and NK cells, creating vulnerability to opportunistic infections and poor outcomes.

Understanding this immunopathophysiology is crucial for modern critical care practice. The traditional focus on organ support must be expanded to include immune system restoration. IL-7 therapy emerges as the most promising intervention for immune reconstitution, though optimal timing, dosing, and patient selection remain areas of active investigation.

Clinicians must recognize that immune dysfunction in prolonged critical illness extends far beyond the acute phase and may persist for months after ICU discharge. This recognition should inform approaches to infection prevention, vaccination strategies, and long-term follow-up care.

The field stands at a crucial juncture where improved understanding of immune dysfunction mechanisms is translating into targeted therapeutic interventions. Future success will depend on early recognition of at-risk patients, timely intervention before irreversible immune depletion occurs, and individualized approaches based on immune phenotyping.

As we continue to improve survival from critical illness, optimizing immune recovery becomes paramount for ensuring not just survival, but meaningful recovery for our most vulnerable patients.


References

  1. Damuth E, et al. Long-term survival of critically ill patients treated with prolonged mechanical ventilation: a systematic review and meta-analysis. Lancet Respir Med. 2015;3(7):544-553.

  2. Kahn JM, et al. Long-term acute care hospital utilization after critical illness. JAMA. 2010;303(22):2253-2259.

  3. Hotchkiss RS, et al. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis. 2013;13(3):260-268.

  4. Venet F, Monneret G. Advances in the understanding and treatment of sepsis-induced immunosuppression. Nat Rev Nephrol. 2018;14(2):121-137.

  5. Kellum JA, et al. Understanding the inflammatory cytokine response in pneumonia and sepsis: results of the Genetic and Inflammatory Markers of Sepsis (GenIMS) Study. Arch Intern Med. 2007;167(15):1655-1663.

  6. Buck MD, et al. T cell metabolism drives immunity. J Exp Med. 2015;212(9):1345-1360.

  7. Vincent JL, et al. The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. Intensive Care Med. 1996;22(7):707-710.

  8. Drewry AM, et al. Persistent lymphopenia after diagnosis of sepsis predicts mortality. Shock. 2014;42(5):383-391.

  9. Hotchkiss RS, et al. Apoptotic cell death in patients with sepsis, shock, and multiple organ dysfunction. Crit Care Med. 1999;27(7):1230-1251.

  10. Gruver AL, et al. Immunosenescence of ageing. J Pathol. 2007;211(2):144-156.

  11. Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014;69 Suppl 1:S4-9.

  12. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15(8):486-499.

  13. Giamarellos-Bourboulis EJ, et al. Early changes of CD4-positive lymphocytes and NK cells in patients with severe Gram-negative sepsis. Crit Care. 2006;10(6):R166.

  14. Forel JM, et al. Immune alterations in sepsis and critical illness. Ann Intensive Care. 2020;10(1):24.

  15. Roquilly A, et al. Hydrocortisone therapy for patients with multiple trauma: the randomized controlled HYPOLYTE study. JAMA. 2011;305(12):1201-1209.

  16. Monneret G, et al. Monitoring immune dysfunctions in the septic patient: a new skin for the old ceremony. Mol Med. 2008;14(1-2):64-78.

  17. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11(11):723-737.

  18. Hotchkiss RS, et al. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013;13(12):862-874.

  19. Papazian L, et al. Cytomegalovirus reactivation in ICU patients. Intensive Care Med. 2016;42(1):28-37.

  20. Vandewoude KH, et al. Clinical relevance of Aspergillus isolation from respiratory tract samples in critically ill patients. Crit Care. 2006;10(1):R31.

  21. Torres A, et al. International ERS/ESICM/ESCMID/ALAT guidelines for the management of hospital-acquired pneumonia and ventilator-associated pneumonia. Eur Respir J. 2017;50(3):1700582.

  22. Hunter CA, Jones SA. IL-6 as a keystone cytokine in health and disease. Nat Immunol. 2015;16(5):448-457.

  23. Wajant H, et al. Tumor necrosis factor signaling. Cell Death Differ. 2003;10(1):45-65.

  24. McNab F, et al. Type I interferons in infectious disease. Nat Rev Immunol. 2015;15(2):87-103.

  25. Buck MD, et al. T cell metabolism drives immunity. J Exp Med. 2015;212(9):1345-1360.

  26. Cruzat V, et al. Glutamine: metabolism and immune function, supplementation and clinical translation. Nutrients. 2018;10(11):1564.

  27. Saeed S, et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science. 2014;345(6204):1251086.

  28. Netea MG, et al. Trained immunity: A program of innate immune memory in health and disease. Science. 2016;352(6284):aaf1098.

  29. Mackall CL, et al. Distinctions between CD8+ and CD4+ T-cell regenerative pathways result in prolonged T-cell subset imbalance after intensive chemotherapy. Blood. 1997;89(10):3700-3707.

  30. Fry TJ, Mackall CL. The many faces of IL-7: from lymphopoiesis to peripheral T cell maintenance. J Immunol. 2005;174(11):6571-6576.

  31. Unsinger J, et al. IL-7 promotes T cell viability, trafficking, and functionality and improves survival in sepsis. J Immunol. 2010;184(7):3768-3779.

  32. Francois B, et al. Interleukin-7 restores lymphocytes in septic shock: the IRIS-7 randomized clinical trial. JCI Insight. 2018;3(5):e98960.

  33. Venet F, et al. IL-7 restores lymphocyte functions in septic patients. J Immunol. 2012;189(10):5073-5081.

  34. Kasten KR, et al. T cells are potent early mediators of the host response to sepsis. Shock. 2010;34(4):327-336.

  35. Popovic PJ, et al. Arginine and immunity. J Nutr. 2007;137(6 Suppl 2):1681S-1686S.

  36. Heyland D, et al. A randomized trial of glutamine and antioxidants in critically ill patients. N Engl J Med. 2013;368(16):1489-1497.

  37. Calder PC. Marine omega-3 fatty acids and inflammatory processes: Effects, mechanisms and clinical relevance. Biochim Biophys Acta. 2015;1851(4):469-484.

  38. Brahmamdam P, et al. Delayed administration of anti-PD-1 antibody reverses immune dysfunction and improves survival during sepsis. J Leukoc Biol. 2010;88(2):233-240.

  39. Romani L, et al. Thymosin alpha 1 activates dendritic cells for antifungal Th1 resistance through toll-like receptor signaling. Blood. 2004;103(11):4232-4239.

  40. Docke WD, et al. Monocyte deactivation in septic patients: restoration by IFN-gamma treatment. Nat Med. 1997;3(6):678-681.

  41. Schweickert WD, et al. Early physical and occupational therapy in mechanically ventilated, critically ill patients: a randomised controlled trial. Lancet. 2009;373(9678):1874-1882.

  42. Trammell RA, Toth LA. Markers of sleep and wake in inbred mice. J Sleep Res. 2011;20(3):441-451.

  43. Corrigan JJ, et al. Relationships between post-traumatic stress disorder, depression, and functional health in survivors of critical illness. Crit Care Med. 2017;45(7):1083-1089.

  44. Boomer JS, et al. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA. 2011;306(23):2594-2605.

  45. Cheron A, et al. Lack of recovery in monocyte human leukocyte antigen-DR expression is independently associated with the development of sepsis after major trauma. Crit Care. 2010;14(6):R208.

  46. Davenport EE, et al. Genomic landscape of the individual host response and outcomes in sepsis: a prospective cohort study. Lancet Respir Med. 2016;4(4):259-271.

No comments:

Post a Comment

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...