Subscribe to RSS
DOI: 10.1055/a-2762-8278
Limitations in the Design of Critical Care Studies and Suggestions for Future Research Directions
Authors
Abstract
Glucocorticoid (GC) therapy has been a cornerstone of critical care; however, its full potential has been constrained by fixed-dose regimens and trial designs that predate current insights into the dynamic, phase-specific functions of glucocorticoid receptor α (GRα). This study shifts focus from mechanistic pathways to the clinical implications of phase-adaptive care, emphasizing how GC therapy can be optimized through individualized, response-guided strategies tailored to illness trajectory and biological variability. Rather than reiterating GRα's mechanistic role, which is discussed in Chapter 3, this work highlights its practical relevance in therapeutic decision-making across the three sequential phases of critical illness: priming, modulatory, and restorative. In this clinically oriented framework, phase-specific treatment adjustments are informed by real-time changes in systemic stress markers, immune dynamics, and metabolic indicators. Earlier randomized controlled trials were instrumental in establishing safety but often failed to account for evolving physiological demands or receptor variability, contributing to inconsistent outcomes. To bridge this translational gap, this study proposes the integration of response-guided protocols utilizing accessible clinical biomarkers—such as C-reactive protein, interleukin-6, D-dimer, and lactate—allowing for adaptive dosing and tapering strategies aligned with patient-specific recovery patterns. Moving beyond pharmacologic dosing, the study outlines adjunctive clinical strategies—including targeted micronutrient supplementation and microbiome-supportive therapies—not as theoretical possibilities but as practical co-interventions that can be incorporated into intensive care unit protocols. Furthermore, it explores how artificial intelligence-enabled clinical decision systems and adaptive trial designs can operationalize precision care by dynamically stratifying patients and tailoring interventions to shifting biological profiles. Together, these applied strategies support a transition from static treatment paradigms to a precision medicine model in critical care—one that aligns GC therapy with individualized recovery trajectories, maximizes therapeutic responsiveness, and reduces treatment-related risks through multimodal, phase-responsive interventions.
Keywords
Critical Illness - glucocorticoid treatment - phase-specific therapy - homeostatic correction - biomarker-guided treatment - micronutrient and microbiome integrationContributors' Statement
G.U.M. conceived and wrote the manuscript; J.M.S. authored the section on artificial intelligence; and S.L. authored the section on redesigning clinical trials for complex, multisystem conditions.
Note
A comprehensive literature review was conducted using Google Scholar and the Consensus database to identify relevant articles published between 1995 and 2025. Manual screening of references from key review articles and clinical guidelines was also performed to ensure completeness and relevance to this translational synthesis.
Publication History
Received: 28 May 2025
Accepted: 02 December 2025
Article published online:
15 January 2026
© 2026. Thieme. All rights reserved.
Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA
-
References
- 1 Meduri GU. Factors influencing glucocorticoid treatment response: mechanism-based strategies to overcome glucocorticoid resistance and restore GRα function. Semin Respir Crit Care Med 2026
- 2 Meduri GU. Glucocorticoids and GRα signaling in critical illness: phase-specific homeostatic corrections across systems. Semin Respir Crit Care Med 2025
- 3 Meduri GU. Synergistic glucocorticoids, vitamins, and microbiome strategies for gut protection in critical illness. Explor Endocr Metab Dis 2025; 2: 1014
- 4 Meduri GU. An historical review of glucocorticoid treatment in Sepsis. Disease pathophysiology and the design of treatment investigation. Sepsis 1999; 3: 21-38
- 5 Meduri GU, Bridges L, Siemieniuk RAC, Kocak M. An exploratory reanalysis of the randomized trial on efficacy of corticosteroids as rescue therapy for the late phase of acute respiratory distress syndrome. Crit Care Med 2018; 46 (06) 884-891
- 6 Meduri G.U. Glucocorticoids and GRα Signaling in Critical Illness: Phase-Specific Homeostatic Corrections Across Systems. Review. Seminars in Respiratory and Critical Care Medicine 2026
- 7 Meduri GU, Psarra A-MG. The glucocorticoid system: a multifaceted regulator of mitochondrial function, endothelial homeostasis, and intestinal barrier integrity. Semin Respir Crit Care Med 2026
- 8 Meduri GU, Tolley EA, Chinn A, Stentz F, Postlethwaite A. Procollagen types I and III aminoterminal propeptide levels during acute respiratory distress syndrome and in response to methylprednisolone treatment. Am J Respir Crit Care Med 1998; 158 (5 Pt 1): 1432-1441
- 9 Nathens AB, Neff MJ, Jurkovich GJ. et al. Randomized, prospective trial of antioxidant supplementation in critically ill surgical patients. Ann Surg 2002; 236 (06) 814-822
- 10 Shrestha DB, Budhathoki P, Sedhai YR. et al. Vitamin C in critically ill patients: an updated systematic review and meta-analysis. Nutrients 2021; 13 (10) 3564
- 11 Notz Q, Herrmann J, Schlesinger T. et al. Clinical significance of micronutrient supplementation in critically ill COVID-19 patients with severe ARDS. Nutrients 2021; 13 (06) 2113
- 12 Berger MM. Do micronutrient deficiencies contribute to mitochondrial failure in critical illness?. Curr Opin Clin Nutr Metab Care 2020; 23 (02) 102-110
- 13 Hakiminia B, Alikiaie B, Khorvash F, Mousavi S. Targeting mitochondrial and brain injury markers in acquired brain injuries: a randomized, double-blind, placebo-controlled study with melatonin. Adv Pharm Bull 2022; 12 (01) 118-127
- 14 Kellermann L, Jensen KB, Bergenheim F. et al. Mucosal vitamin D signaling in inflammatory bowel disease. Autoimmun Rev 2020; 19 (11) 102672
- 15 Żmijewski MA. Nongenomic activities of vitamin D. Nutrients 2022; 14 (23) 5104
- 16 Dimitrov V, Bouttier M, Boukhaled G. et al. Hormonal vitamin D up-regulates tissue-specific PD-L1 and PD-L2 surface glycoprotein expression in humans but not mice. J Biol Chem 2017; 292 (50) 20657-20668
- 17 Xu H, Wang Y, Jurutka PW. et al. 16α-Hydroxytrametenolic acid from Poria cocos improves intestinal barrier function through glucocorticoid receptor mediated PI3K/Akt/NF-κB pathway. J Agric Food Chem 2019; 67 (39) 10871-10879
- 18 Sirisinha S. The pleiotropic role of vitamin A in regulating mucosal immunity. Asian Pac J Allergy Immunol 2015; 33 (02) 71-89
- 19 Carr AC, Maggini S. Vitamin C and immune function. Nutrients 2017; 9 (11) 1211
- 20 Mocchegiani E, Costarelli L, Giacconi R. et al. Vitamin E-gene interactions in aging and inflammatory age-related diseases: implications for treatment. A systematic review. Ageing Res Rev 2014; 14 (14) 81-101
- 21 Hanna M, Jaqua E, Nguyen V, Clay J. B Vitamins: Functions and Uses in Medicine. Perm J 2022; 26 (02) 89-97
- 22 Wesselink E, Koekkoek WAC, Grefte S, Witkamp RF, van Zanten ARH. Feeding mitochondria: potential role of nutritional components to improve critical illness convalescence. Clin Nutr 2019; 38 (03) 982-995
- 23 Lee JG, Jang JY, Baik SM. Selenium as an antioxidant: roles and clinical applications in critically ill and trauma patients: a narrative review. Antioxidants 2025; 14 (03) 294
- 24 Touyz RM, de Baaij JHF, Hoenderop JGJ. Magnesium disorders. N Engl J Med 2024; 390 (21) 1998-2009
- 25 McMillan DC, Maguire D, Talwar D. Relationship between nutritional status and the systemic inflammatory response: micronutrients. Proc Nutr Soc 2019; 78 (01) 56-67
- 26 Koekkoek KWA, Berger MM. An update on essential micronutrients in critical illness. Curr Opin Crit Care 2023; 29 (04) 315-329
- 27 Meduri GU, Confalonieri M, Chaudhuri D, Rochwerg B, Meibohm B. Prolonged glucocorticoid treatment in ARDS: pathobiological rationale and pharmacological principles. In: Fink G. , ed. Handbook of Stress: Stress, Immunology and Inflammation Academic Press; 2024. :289–323:chap 24. vol. Encyclopedia of Stress
- 28 Scheschowitsch K, Leite JA, Assreuy J. New insights in glucocorticoid receptor signaling—more than just a ligand-binding receptor. Front Endocrinol (Lausanne) 2017; 8: 16
- 29 Eisenstein AB, Hartroft PM. Alterations in the rat adrenal cortex induced by sodium deficiency: steroid hormone secretion. Endocrinology 1957; 60 (05) 634-640
- 30 Hayashi R, Wada H, Ito K, Adcock IM. Effects of glucocorticoids on gene transcription. Eur J Pharmacol 2004; 500 (1–3) 51-62
- 31 Zhang Y, Leung DYM, Goleva E. Vitamin D enhances glucocorticoid action in human monocytes: involvement of granulocyte-macrophage colony-stimulating factor and mediator complex subunit 14. J Biol Chem 2013; 288 (20) 14544-14553
- 32 Lucock M. Folic acid: nutritional biochemistry, molecular biology, and role in disease processes. Mol Genet Metab 2000; 71 (1–2): 121-138
- 33 Mohammadi H, Talebi S, Ghavami A. et al. Effects of zinc supplementation on inflammatory biomarkers and oxidative stress in adults: a systematic review and meta-analysis of randomized controlled trials. J Trace Elem Med Biol 2021; 68: 126857
- 34 Padayatty SJ, Katz A, Wang Y. et al. Vitamin C as an antioxidant: evaluation of its role in disease prevention. J Am Coll Nutr 2003; 22 (01) 18-35
- 35 Burton GW, Traber MG. Vitamin E: antioxidant activity, biokinetics, and bioavailability. Annu Rev Nutr 1990; 10 (01) 357-382
- 36 Choi EJ, Jeon CH, Park DH, Kwon TH. Allithiamine exerts therapeutic effects on sepsis by modulating metabolic flux during dendritic cell activation. Mol Cells 2020; 43 (11) 964-973
- 37 Theron A, Anderson R, Grabow G, Meiring JL. In vitro and in vivo stimulation of neutrophil migration and lymphocyte transformation by thiamine related to inhibition of the peroxidase/H2O2/halide system. Clin Exp Immunol 1981; 44 (02) 295-303
- 38 Wang T-T, Nestel FP, Bourdeau V. et al. Cutting edge: 1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. J Immunol 2004; 173 (05) 2909-2912
- 39 Dimitrov V, Barbier C, Ismailova A. et al. Vitamin D-regulated gene expression profiles: species-specificity and Cell-specific effects on metabolism and immunity. Endocrinology 2021; 162 (02) bqaa218
- 40 Greulich T, Regner W, Branscheidt M. et al. Altered blood levels of vitamin D, cathelicidin and parathyroid hormone in patients with sepsis-a pilot study. Anaesth Intensive Care 2017; 45 (01) 36-45
- 41 Ang A, Pullar JM, Currie MJ, Vissers MCM. Vitamin C and immune cell function in inflammation and cancer. Biochem Soc Trans 2018; 46 (05) 1147-1159
- 42 Sorice A, Guerriero E, Capone F, Colonna G, Castello G, Costantini S. Ascorbic acid: its role in immune system and chronic inflammation diseases. Mini Rev Med Chem 2014; 14 (05) 444-452
- 43 Sae-Khow K, Tachaboon S, Wright HL. et al. Defective neutrophil function in patients with sepsis is mostly restored by ex vivo ascorbate incubation. J Inflamm Res 2020; 13: 263-274
- 44 Kim Y, Kim H, Bae S. et al. Vitamin C is an essential factor on the anti-viral immune responses through the production of interferon-α/β at the initial stage of influenza A virus (H3N2) infection. Immune Netw 2013; 13 (02) 70-74
- 45 Combs Jr GF, McClung JP. The vitamins: fundamental aspects in nutrition and health. Edition 5. Chapter 11 Thiamin. Academic press; 2016: 298-314
- 46 Collie JTB, Greaves RF, Jones OAH, Lam Q, Eastwood GM, Bellomo R. Vitamin B1 in critically ill patients: needs and challenges. Clin Chem Lab Med 2017; 55 (11) 1652-1668
- 47 Ryan ZC, Craig TA, Folmes CD. et al. 1α, 25-dihydroxyvitamin D3 regulates mitochondrial oxygen consumption and dynamics in human skeletal muscle cells. J Biol Chem 2016; 291 (03) 1514-1528
- 48 Luo G, Xie ZZ, Liu FY, Zhang GB. Effects of vitamin C on myocardial mitochondrial function and ATP content in hypoxic rats. Chung Kuo Yao Li Hsueh Pao 1998; 19 (04) 351-355
- 49 Charoenngam N, Holick MF. Immunologic effects of vitamin D on human health and disease. Nutrients 2020; 12 (07) 2097
- 50 Dancer RC, Parekh D, Lax S. et al. Vitamin D deficiency contributes directly to the acute respiratory distress syndrome (ARDS). Thorax 2015; 70 (07) 617-624
- 51 Tyml K. Vitamin C and microvascular dysfunction in systemic inflammation. Antioxidants 2017; 6 (03) 49
- 52 Oudemans-van Straaten HM, Elbers PWG, Spoelstra-de Man AME. How to give vitamin C a cautious but fair chance in severe sepsis. Chest 2017; 151 (06) 1199-1200
- 53 Marik PE. Glucocorticosteroids as adjunctive therapy for acute respiratory distress syndrome and sepsis? Yes, but not as monotherapy. Crit Care Med 2017; 45 (05) 910-911
- 54 Carr AC, Shaw GM, Fowler AA, Natarajan R. Ascorbate-dependent vasopressor synthesis: a rationale for vitamin C administration in severe sepsis and septic shock?. Crit Care 2015; 19 (01) 418
- 55 Mohammed BM, Fisher BJ, Kraskauskas D. et al. Vitamin C: a novel regulator of neutrophil extracellular trap formation. Nutrients 2013; 5 (08) 3131-3151
- 56 Yadav UC, Kalariya NM, Srivastava SK, Ramana KV. Protective role of benfotiamine, a fat-soluble vitamin B1 analogue, in lipopolysaccharide-induced cytotoxic signals in murine macrophages. Free Radic Biol Med 2010; 48 (10) 1423-1434
- 57 Bozic I, Savic D, Laketa D. et al. Benfotiamine attenuates inflammatory response in LPS stimulated BV-2 microglia. PLoS One 2015; 10 (02) e0118372
- 58 Menezes RR, Godin AM, Rodrigues FF. et al. Thiamine and riboflavin inhibit production of cytokines and increase the anti-inflammatory activity of a corticosteroid in a chronic model of inflammation induced by complete Freund's adjuvant. Pharmacol Rep 2017; 69 (05) 1036-1043
- 59 Bagnoud M, Hoepner R, Pistor M. et al. Vitamin D augments glucocorticosteroid efficacy via inhibition of mTORc1. Acta Neuropathol 2019; 138 (03) 443-456
- 60 Kassel O, Sancono A, Krätzschmar J, Kreft B, Stassen M, Cato AC. Glucocorticoids inhibit MAP kinase via increased expression and decreased degradation of MKP-1. EMBO J 2001; 20 (24) 7108-7116
- 61 Zhang Y, Leung DY, Richers BN. et al. Vitamin D inhibits monocyte/macrophage proinflammatory cytokine production by targeting MAPK phosphatase-1. J Immunol 2012; 188 (05) 2127-2135
- 62 Stio M, Martinesi M, Bruni S. et al. The Vitamin D analogue TX 527 blocks NF-kappaB activation in peripheral blood mononuclear cells of patients with Crohn's disease. J Steroid Biochem Mol Biol 2007; 103 (01) 51-60
- 63 Ojaimi S, Skinner NA, Strauss BJ, Sundararajan V, Woolley I, Visvanathan K. Vitamin D deficiency impacts on expression of toll-like receptor-2 and cytokine profile: a pilot study. J Transl Med 2013; 11 (01) 176
- 64 Zold E, Szodoray P, Nakken B. et al. Alfacalcidol treatment restores derailed immune-regulation in patients with undifferentiated connective tissue disease. Autoimmun Rev 2011; 10 (03) 155-162
- 65 Hwang J, You H, Kwon DH, Son Y, Lee GY, Han SN. Transcriptome analysis of T cells from Ldlr−/− mice and effects of in vitro vitamin D treatment. J Nutr Biochem 2023; 2023: 109510
- 66 Patak P, Willenberg HS, Bornstein SR. Vitamin C is an important cofactor for both adrenal cortex and adrenal medulla. Endocr Res 2004; 30 (04) 871-875
- 67 Moskowitz A, Andersen LW, Huang DT. et al. Ascorbic acid, corticosteroids, and thiamine in sepsis: a review of the biologic rationale and the present state of clinical evaluation. Crit Care 2018; 22 (01) 283
- 68 Okamoto K, Tanaka H, Makino Y, Makino I. Restoration of the glucocorticoid receptor function by the phosphodiester compound of vitamins C and E, EPC-K1 (L-ascorbic acid 2-[3,4-dihydro-2,5,7,8-tetramethyl-2-(4,8,12-trimethyltridecyl)-2H-1-benzopyran-6-yl hydrogen phosphate] potassium salt), via a redox-dependent mechanism. Biochem Pharmacol 1998; 56 (01) 79-86
- 69 Chen Y, Luo G, Yuan J. et al. Vitamin C mitigates oxidative stress and tumor necrosis factor-alpha in severe community-acquired pneumonia and LPS-induced macrophages. Mediators Inflamm 2014; 2014: 426740
- 70 Bowie AG, O'Neill LA. Vitamin C inhibits NF-κ B activation by TNF via the activation of p38 mitogen-activated protein kinase. J Immunol 2000; 165 (12) 7180-7188
- 71 Donnino MW, Andersen LW, Chase M. et al; Center for Resuscitation Science Research Group. Randomized, double-blind, placebo-controlled trial of thiamine as a metabolic resuscitator in septic shock: a pilot study. Crit Care Med 2016; 44 (02) 360-367
- 72 Schreiber SN. The transcriptional coactivator PGC-1 [alpha] as a modulator of ERR [alpha] and GR signaling: function in mitochondrial biogenesis. University_of_Basel; 2004
- 73 Latham CM, Brightwell CR, Keeble AR. et al. Vitamin D promotes skeletal muscle regeneration and mitochondrial health. Front Physiol 2021; 12: 660498
- 74 Jain SK, Micinski D. Vitamin D upregulates glutamate cysteine ligase and glutathione reductase, and GSH formation, and decreases ROS and MCP-1 and IL-8 secretion in high-glucose exposed U937 monocytes. Biochem Biophys Res Commun 2013; 437 (01) 7-11
- 75 Fisher BJ, Seropian IM, Kraskauskas D. et al. Ascorbic acid attenuates lipopolysaccharide-induced acute lung injury. Crit Care Med 2011; 39 (06) 1454-1460
- 76 Zheng S, Yang J, Hu X. et al. Vitamin D attenuates lung injury via stimulating epithelial repair, reducing epithelial cell apoptosis and inhibits TGF-β induced epithelial to mesenchymal transition. Biochem Pharmacol 2020; 177: 113955
- 77 Mohammed BM, Fisher BJ, Kraskauskas D. et al. Vitamin C promotes wound healing through novel pleiotropic mechanisms. Int Wound J 2016; 13 (04) 572-584
- 78 Vissers MC, Hampton MB. The role of oxidants and vitamin C on neutrophil apoptosis and clearance. Biochem Soc Trans 2004; 32 (Pt3): 499-501
- 79 Sambon M, Wins P, Bettendorff L. Neuroprotective effects of thiamine and precursors with higher bioavailability: focus on benfotiamine and dibenzoylthiamine. Int J Mol Sci 2021; 22 (11) 5418
- 80 Fabri M, Stenger S, Shin D-M. et al. Vitamin D is required for IFN-γ-mediated antimicrobial activity of human macrophages. Sci Transl Med 2011; 3 (104) 104ra102
- 81 Gioda CR, Capettini LS, Cruz JS, Lemos VS. Thiamine deficiency leads to reduced nitric oxide production and vascular dysfunction in rats. Nutr Metab Cardiovasc Dis 2014; 24 (02) 183-188
- 82 Ascher E, Gade PV, Hingorani A. et al. Thiamine reverses hyperglycemia-induced dysfunction in cultured endothelial cells. Surgery 2001; 130 (05) 851-858
- 83 Moss C. Thiamin status and supplementation in the management of diabetes mellitus and its vascular comorbidities. Vitamins & Minerals 2013 2013.
- 84 Sassi F, Tamone C, D'Amelio P. Vitamin D: nutrient, hormone, and immunomodulator. Nutrients 2018; 10 (11) 1656
- 85 Rao Z, Chen X, Wu J. et al. Vitamin D receptor inhibits NLRP3 activation by impeding its BRCC3-mediated deubiquitination. Front Immunol 2019; 10: 2783
- 86 Berger MM, Shenkin A, Schweinlin A. et al. ESPEN micronutrient guideline. Clin Nutr 2022; 41 (06) 1357-1424
- 87 Berger MM, Amrein K, Barazzoni R. et al. The science of micronutrients in clinical practice - report on the ESPEN symposium. Clin Nutr 2024; 43 (01) 268-283
- 88 Menger J, Lee Z-Y, Notz Q. et al. Administration of vitamin D and its metabolites in critically ill adult patients: an updated systematic review with meta-analysis of randomized controlled trials. Crit Care 2022; 26 (01) 268
- 89 Manzanares W, Dhaliwal R, Jiang X, Murch L, Heyland DK. Antioxidant micronutrients in the critically ill: a systematic review and meta-analysis. Crit Care 2012; 16 (02) R66
- 90 Jaff S, Zeraattalab-Motlagh S, Amiri Khosroshahi R, Gubari M, Mohammadi H, Djafarian K. The effect of selenium therapy in critically ill patients: an umbrella review of systematic reviews and meta-analysis of randomized controlled trials. Eur J Med Res 2023; 28 (01) 104
- 91 Gu W-J, Duan X-J, Liu X-Z. et al. Association of magnesium sulfate use with mortality in critically ill patients with sepsis: a retrospective propensity score-matched cohort study. Br J Anaesth 2023; 131 (05) 861-870
- 92 Marik PE, Khangoora V, Rivera R, Hooper MH, Catravas J. Hydrocortisone, vitamin c, and thiamine for the treatment of severe sepsis and septic shock: a retrospective before-after study. Chest 2017; 151 (06) 1229-1238
- 93 Gunst J, Egi M, Van den Berghe G. Nutrition and metabolic control for ICU patients. Intensive Care Med 2025; 51 (06) 1150-1152
- 94 Angus DC, Derde L, Al-Beidh F. et al; Writing Committee for the REMAP-CAP Investigators. Effect of hydrocortisone on mortality and organ support in patients with severe COVID-19: the REMAP-CAP COVID-19 corticosteroid domain randomized clinical trial. JAMA 2020; 324 (13) 1317-1329
- 95 Wang H, Yee D. I-SPY 2: a neoadjuvant adaptive clinical trial designed to improve outcomes in high-risk breast cancer. Curr Breast Cancer Rep 2019; 11 (04) 303-310
- 96 Fowler III AA, Truwit JD, Hite RD. et al. Effect of vitamin c infusion on organ failure and biomarkers of inflammation and vascular injury in patients with sepsis and severe acute respiratory failure: the CITRIS-ALI randomized clinical trial. JAMA 2019; 322 (13) 1261-1270
- 97 Branco RG, Garcia PC. Ferritin and C-reactive protein as markers of systemic inflammation in sepsis. Pediatr Crit Care Med 2017; 18 (02) 194-196
- 98 Cui Z, Merritt Z, Assa A. et al. Early and significant reduction in C-reactive protein levels after corticosteroid therapy is associated with reduced mortality in patients with COVID-19. J Hosp Med 2021; 16 (03) 142-148
- 99 Ayuningtyas LV, Hermosaningtyas AA, Airlangga P. et al. Comparison of changes in inflammation markers NLR, CRP, and LCR after corticosteroid therapy in severe and critical COVID-19 patients. Trends Sci 2023;
- 100 Singh N, Kumar R, Kumar S, Prasad N, Muni S, Kumari N. The trend of C-reactive protein after corticosteroid therapy in COVID-19 patients admitted to IGIMS, Patna. Cureus 2024; 16 (01) e51499
- 101 Wu WF, Fang Q, He GJ. Efficacy of corticosteroid treatment for severe community-acquired pneumonia: a meta-analysis. Am J Emerg Med 2018; 36 (02) 179-184
- 102 Ito K, Hanazawa T, Tomita K, Barnes PJ, Adcock IM. Oxidative stress reduces histone deacetylase 2 activity and enhances IL-8 gene expression: role of tyrosine nitration. Biochem Biophys Res Commun 2004; 315 (01) 240-245
- 103 Cloonan SM, Choi AM. Mitochondria in lung disease. J Clin Invest 2016; 126 (03) 809-820
- 104 Liu J, Liu Y, Xiang P. et al. Neutrophil-to-lymphocyte ratio predicts critical illness patients with 2019 coronavirus disease in the early stage. J Transl Med 2020; 18 (01) 206
- 105 Yan X, Li F, Wang X. et al. Neutrophil to lymphocyte ratio as prognostic and predictive factor in patients with coronavirus disease 2019: a retrospective cross-sectional study. J Med Virol 2020; 92 (11) 2573-2581
- 106 Zhang B, Han Y, Chen X. et al. Association of monocyte-to-lymphocyte and neutrophil-to-lymphocyte ratios with persistent critical illness in patients with severe trauma. J Trauma Nurs 2022; 29 (05) 240-251
- 107 Takahashi W, Nakada TA, Yazaki M, Oda S. Interleukin-6 levels act as a diagnostic marker for infection and a prognostic marker in patients with organ dysfunction in intensive care units. Shock 2016; 46 (03) 254-260
- 108 Iwase S, Nakada TA, Hattori N. et al. Interleukin-6 as a diagnostic marker for infection in critically ill patients: A systematic review and meta-analysis. Am J Emerg Med 2019; 37 (02) 260-265
- 109 Meduri GU, Headley S, Kohler G. et al. Persistent elevation of inflammatory cytokines predicts a poor outcome in ARDS. Plasma IL-1 beta and IL-6 levels are consistent and efficient predictors of outcome over time. Chest 1995; 107 (04) 1062-1073
- 110 Villar J, Ferrando C, Martínez D. et al; dexamethasone in ARDS network. Dexamethasone treatment for the acute respiratory distress syndrome: a multicentre, randomised controlled trial. Lancet Respir Med 2020; 8 (03) 267-276
- 111 Meduri GU, Tolley EA, Chrousos GP, Stentz F. Prolonged methylprednisolone treatment suppresses systemic inflammation in patients with unresolving acute respiratory distress syndrome: evidence for inadequate endogenous glucocorticoid secretion and inflammation-induced immune cell resistance to glucocorticoids. Am J Respir Crit Care Med 2002; 165 (07) 983-991
- 112 Raess N, Schuetz P, Cesana-Nigro N. et al. Influence of prednisone on inflammatory biomarkers in community-acquired pneumonia: secondary analysis of a randomized trial. J Clin Pharmacol 2021; 61 (11) 1406-1414
- 113 Shorr AF, Thomas SJ, Alkins SA, Fitzpatrick TM, Ling GS. D-dimer correlates with proinflammatory cytokine levels and outcomes in critically ill patients. Chest 2002; 121 (04) 1262-1268
- 114 Kernan KF, Carcillo JA. Hyperferritinemia and inflammation. Int Immunol 2017; 29 (09) 401-409
- 115 Chen H, Xie J, Su N. et al. Corticosteroid therapy is associated with improved outcome in critically ill patients with COVID-19 with hyperinflammatory phenotype. Chest 2021; 159 (05) 1793-1802
- 116 Huang I, Pranata R, Lim MA, Oehadian A, Alisjahbana B. C-reactive protein, procalcitonin, D-dimer, and ferritin in severe coronavirus disease-2019: a meta-analysis. Ther Adv Respir Dis 2020; 14: 1753466620937175
- 117 Horvat CM, Fabio A, Nagin DS. et al; on behalf of the Eunice Kennedy Shriver National Institute of Child Health and Human Development Collaborative Pediatric Critical Care Research Network. Mortality risk in pediatric sepsis based on C-reactive protein and ferritin levels. Pediatr Crit Care Med 2022; 23 (12) 968-979
- 118 Schnaubelt S, Tihanyi D, Strassl R. et al. Hemophagocytic lymphohistiocytosis in COVID-19: Case reports of a stepwise approach. Medicine (Baltimore) 2021; 100 (12) e25170
- 119 Marty P, Roquilly A, Vallée F. et al. Lactate clearance for death prediction in severe sepsis or septic shock patients during the first 24 hours in intensive care unit: an observational study. Ann Intensive Care 2013; 3 (01) 3-3
- 120 Bruno RR, Wernly B, Binneboessel S. et al. Failure of lactate clearance predicts the outcome of critically ill septic patients. Diagnostics (Basel) 2020; 10 (12) 1105
- 121 Harrington JS, Choi AMK, Nakahira K. Mitochondrial DNA in sepsis. Curr Opin Crit Care 2017; 23 (04) 284-290
- 122 Krychtiuk KA, Ruhittel S, Hohensinner PJ. et al. Mitochondrial DNA and toll-like receptor-9 are associated with mortality in critically ill patients. Crit Care Med 2015; 43 (12) 2633-2641
- 123 Supinski GS, Schroder EA, Callahan LA. Mitochondria and critical illness. Chest 2020; 157 (02) 310-322
- 124 Lubkin DT, Bishawi M, Barbas AS, Brennan TV, Kirk AD. Extracellular mitochondrial DNA and N-formyl peptides in trauma and critical illness: a systematic review. Crit Care Med 2018; 46 (12) 2018-2028
- 125 Faust H, Reilly J, Anderson B. et al. Plasma mitochondrial DNA levels are associated with acute respiratory distress syndrome in trauma and sepsis patients. Chest 2019;
- 126 Reincke M, Allolio B, Würth G, Winkelmann W, Würth G. The hypothalamic-pituitary-adrenal axis in critical illness: response to dexamethasone and corticotropin-releasing hormone. J Clin Endocrinol Metab 1993; 77 (01) 151-156
- 127 Van den Berghe G, Téblick A, Langouche L, Gunst J. The hypothalamus-pituitary-adrenal axis in sepsis- and hyperinflammation-induced critical illness: gaps in current knowledge and future translational research directions. EBioMedicine 2022; 84: 104284
- 128 de Jong MF, Molenaar N, Beishuizen A, Groeneveld AB. Diminished adrenal sensitivity to endogenous and exogenous adrenocorticotropic hormone in critical illness: a prospective cohort study. Crit Care 2015; 19 (01) 1
- 129 Venkatesh B, Mortimer RH, Couchman B, Hall J. Evaluation of random plasma cortisol and the low dose corticotropin test as indicators of adrenal secretory capacity in critically ill patients: a prospective study. Anaesth Intensive Care 2005; 33 (02) 201-209
- 130 Annane D, Pastores SM, Rochwerg B. et al. Guidelines for the diagnosis and management of critical illness-related corticosteroid insufficiency (CIRCI) in critically ill patients (part I): Society of Critical Care Medicine (SCCM) and European Society of Intensive Care Medicine (ESICM) 2017. Intensive Care Med 2017; 43 (12) 1751-1763
- 131 Peeters B, Meersseman P, Vander Perre S. et al. Adrenocortical function during prolonged critical illness and beyond: a prospective observational study. Intensive Care Med 2018; 44 (10) 1720-1729
- 132 Schuetz P, Christ-Crain M, Schild U. et al. Effect of a 14-day course of systemic corticosteroids on the hypothalamic-pituitary-adrenal-axis in patients with acute exacerbation of chronic obstructive pulmonary disease. BMC Pulm Med 2008; 8 (08) 1-1
- 133 Nawab QU, Golden E, Confalonieri M, Umberger R, Meduri GU. Corticosteroid treatment in severe community-acquired pneumonia: duration of treatment affects control of systemic inflammation and clinical improvement. Intensive Care Med 2011; 37 (09) 1553-1554
- 134 Ho KS, Narasimhan B, Difabrizio L. et al. Impact of corticosteroids in hospitalised COVID-19 patients. BMJ Open Respir Res 2021; 8 (01) e000766
- 135 Takeshita Y, Terada J, Hirasawa Y. et al. Development of a novel score model to predict hyperinflammation in COVID-19 as a forecast of optimal steroid administration timing. Front Med (Lausanne) 2022; 9: 935255
- 136 Smit JM, Van Der Zee PA, Stoof SCM. et al. Predicting benefit from adjuvant therapy with corticosteroids in community-acquired pneumonia: a data-driven analysis of randomised trials. Lancet Respir Med 2025; 13 (03) 221-233
- 137 Klowak JA, Bijelić V, Barrowman N, Menon K. Genomics of Pediatric Septic Shock Investigators. The association of corticosteroids and pediatric sepsis biomarker risk model (PERSEVERE)-II biomarker risk stratification with mortality in pediatric septic shock. Pediatr Crit Care Med 2023; 24 (03) 186-193
- 138 Wong HR, Atkinson SJ, Cvijanovich NZ. et al. Combining prognostic and predictive enrichment strategies to identify children with septic shock responsive to corticosteroids. Crit Care Med 2016; 44 (10) e1000-e1003
- 139 Forel JM, Guervilly C, Hraiech S. et al. Type III procollagen is a reliable marker of ARDS-associated lung fibroproliferation. Intensive Care Med 2015; 41 (01) 1-11
- 140 Daenen K, Boyd A, Huijben JA. et al. Inflammatory biomarkers demonstrate predictive capacity for mortality in COVID-19-related ARDS patients receiving high-dose corticosteroids: a longitudinal analysis. J Inflamm Res 2025; 18: 2395-2408
- 141 See KC. Personalizing care for critically ill adults using omics: a concise review of potential clinical applications. Cells 2023; 12 (04) 541
- 142 Babu M, Snyder M. Multi-omics profiling for health. Mol Cell Proteomics 2023; 22 (06) 100561
- 143 Balakrishna A, Sonny A. Personalized. Crit Care Med 2020; 207-230
- 144 Vodovotz Y. Towards systems immunology of critical illness at scale: from single cell 'omics to digital twins. Trends Immunol 2023; 44 (05) 345-355
- 145 Meduri GU, Leeper KV, Tolley E. et al. Inflammatory cytokine response to high-dose corticosteroids in late ARDS. Chest 1993; 104: 12S
- 146 Ayroldi E, Riccardi C. Glucocorticoid-induced leucine zipper (GILZ): a new important mediator of glucocorticoid action. FASEB J 2009; 23 (11) 3649-3658
- 147 Shah S, King EM, Chandrasekhar A, Newton R. Roles for the mitogen-activated protein kinase (MAPK) phosphatase, DUSP1, in feedback control of inflammatory gene expression and repression by dexamethasone. J Biol Chem 2014; 289 (19) 13667-13679
- 148 Hoppstädter J, Ammit AJ. Role of dual-specificity phosphatase 1 in glucocorticoid-driven anti-inflammatory responses. Front Immunol 2019; 10: 1446
- 149 Vago JP, Tavares LP, Garcia CC. et al. The role and effects of glucocorticoid-induced leucine zipper in the context of inflammation resolution. J Immunol 2015; 194 (10) 4940-4950
- 150 Mittelstadt PR, Ashwell JD. Inhibition of AP-1 by the glucocorticoid-inducible protein GILZ. J Biol Chem 2001; 276 (31) 29603-29610
- 151 Barnes PJ. How corticosteroids control inflammation: Quintiles Prize Lecture 2005. Br J Pharmacol 2006; 148 (03) 245-254
- 152 Newton R, Seybold J, Kuitert LM, Bergmann M, Barnes PJ. Repression of cyclooxygenase-2 and prostaglandin E2 release by dexamethasone occurs by transcriptional and post-transcriptional mechanisms involving loss of polyadenylated mRNA. J Biol Chem 1998; 273 (48) 32312-32321
- 153 Abraham SM, Lawrence T, Kleiman A. et al. Antiinflammatory effects of dexamethasone are partly dependent on induction of dual specificity phosphatase 1. J Exp Med 2006; 203 (08) 1883-1889
- 154 Vago JP, Nogueira CR, Tavares LP. et al. Annexin A1 modulates natural and glucocorticoid-induced resolution of inflammation by enhancing neutrophil apoptosis. J Leukoc Biol 2012; 92 (02) 249-258
- 155 Perretti M, D'Acquisto F. Annexin A1 and glucocorticoids as effectors of the resolution of inflammation. Nat Rev Immunol 2009; 9 (01) 62-70
- 156 Strausbaugh HJ, Rosen SD. A potential role for annexin 1 as a physiologic mediator of glucocorticoid-induced L-selectin shedding from myeloid cells. J Immunol 2001; 166 (10) 6294-6300
- 157 Purvis GSD, Solito E, Thiemermann C. Annexin-A1: therapeutic potential in microvascular disease. Front Immunol 2019; 10: 938
- 158 Chen L, Lv F, Pei L. Annexin 1: a glucocorticoid-inducible protein that modulates inflammatory pain. Eur J Pain 2014; 18 (03) 338-347
- 159 Solito E, Mulla A, Morris JF, Christian HC, Flower RJ, Buckingham JC. Dexamethasone induces rapid serine-phosphorylation and membrane translocation of annexin 1 in a human folliculostellate cell line via a novel nongenomic mechanism involving the glucocorticoid receptor, protein kinase C, phosphatidylinositol 3-kinase, and mitogen-activated protein kinase. Endocrinology 2003; 144 (04) 1164-1174
- 160 Sawmynaden P, Perretti M. Glucocorticoid upregulation of the annexin-A1 receptor in leukocytes. Biochem Biophys Res Commun 2006; 349 (04) 1351-1355
- 161 Kelly L, McGrath S, Rodgers L. et al. Annexin-A1: the culprit or the solution?. Immunology 2022; 166 (01) 2-16
- 162 Yang YH, Morand E, Leech M. Annexin A1: potential for glucocorticoid sparing in RA. Nat Rev Rheumatol 2013; 9 (10) 595-603
- 163 Harhay MO, Wagner J, Ratcliffe SJ. et al. Outcomes and statistical power in adult critical care randomized trials. Am J Respir Crit Care Med 2014; 189 (12) 1469-1478
- 164 Vincent JL, Marini JJ, Pesenti A. Do trials that report a neutral or negative treatment effect improve the care of critically ill patients? No. Intensive Care Med 2018; 44 (11) 1989-1991
- 165 Friedrich JO, Harhay MO, Angus DC. et al; International Forum for Acute Care Trialists (InFACT). Mortality as a measure of treatment effect in clinical trials recruiting critically ill patients. Crit Care Med 2023; 51 (02) 222-230
- 166 Granholm A, Anthon CT, Kjær MN. et al. Patient-important outcomes other than mortality in contemporary ICU trials: a scoping review. Crit Care Med 2022; 50 (10) e759-e771
- 167 Bernard GR, Luce JM, Sprung CL. et al. High-dose corticosteroids in patients with the adult respiratory distress syndrome. N Engl J Med 1987; 317 (25) 1565-1570
- 168 Hernán MA, Hsu J, Healy B. A second chance to get causal inference right: a classification of data science tasks. Chance 2019; 32 (01) 42-49
- 169 Van Veen D, Van Uden C, Blankemeier L. et al. Adapted large language models can outperform medical experts in clinical text summarization. Nat Med 2024; 30 (04) 1134-1142
- 170 Smit JM, Krijthe JH, van Bommel J. Causal Inference for ICU Collaborators. The future of artificial intelligence in intensive care: moving from predictive to actionable AI. Intensive Care Med 2023; 49 (09) 1114-1116
- 171 Chen JH, Asch SM. Machine learning and prediction in medicine—beyond the peak of inflated expectations. N Engl J Med 2017; 376 (26) 2507-2509
- 172 Maley JH, Wanis KN, Young JG, Celi LA. Mortality prediction models, causal effects, and end-of-life decision making in the intensive care unit. BMJ Health Care Inform 2020; 27 (03) e100220
- 173 Miguel A, Hernan R, James M. Causal inference:. what if. CRC PRESS; 2023
- 174 Feuerriegel S, Frauen D, Melnychuk V. et al. Causal machine learning for predicting treatment outcomes. Nat Med 2024; 30 (04) 958-968
- 175 The National Heart Lung. Network BIACT. Higher versus lower positive end-expiratory pressures in patients with ARDS. N Engl J Med 2004; 351: 327-336
- 176 Neto AS, Tomlinson G, Sahetya SK. et al. Higher PEEP for acute respiratory distress syndrome: a Bayesian meta-analysis of randomised clinical trials. Crit Care Resusc 2023; 23 (02) 171-182
- 177 Smit J, Krijthe J, Van Bommel J. et al. The heterogeneous effect of high PEEP strategies on survival in acute respiratory distress syndrome: preliminary results of a data-driven analysis of randomized trials. MedRxiv 2025 https://doi.org/10.1101/2025.01.23.25320649
- 178 Bica I, Alaa AM, Lambert C, van der Schaar M. From real-world patient data to individualized treatment effects using machine learning: current and future methods to address underlying challenges. Clin Pharmacol Ther 2021; 109 (01) 87-100
- 179 Kent DM, Paulus JK, van Klaveren D. et al. The predictive approaches to treatment effect heterogeneity (PATH) statement. Ann Intern Med 2020; 172 (01) 35-45
- 180 Santacruz CA, Pereira AJ, Celis E, Vincent J-L. Which multicenter randomized controlled trials in critical care medicine have shown reduced mortality? A systematic review. Crit Care Med 2019; 47 (12) 1680-1691
- 181 Briel M, Meade M, Mercat A. et al. Higher vs lower positive end-expiratory pressure in patients with acute lung injury and acute respiratory distress syndrome: systematic review and meta-analysis. JAMA 2010; 303 (09) 865-873
- 182 Schandelmaier S, Briel M, Varadhan R. et al. Development of the instrument to assess the credibility of effect modification analyses (ICEMAN) in randomized controlled trials and meta-analyses. CMAJ 2020; 192 (32) E901-E906
- 183 Dahabreh IJ, Kazi DS. Toward personalizing care: assessing heterogeneity of treatment effects in randomized trials. JAMA 2023; 329 (13) 1063-1065
- 184 Smit JM, Jonkman AH, Krijthe JH. Sub-phenotyping in critical care: a valuable strategy or methodologically fragile path?. Intensive Care Med Exp 2025; 13 (01) 59
- 185 Munroe ES, Spicer A, Castellvi-Font A. et al; Platform of Randomized Adaptive Clinical Trials in Critical Illness (PRACTICAL) investigators, Evidence-based Individualized Treatment Effects (EvITE) Group. Evidence-based personalised medicine in critical care: a framework for quantifying and applying individualised treatment effects in patients who are critically ill. Lancet Respir Med 2025; 13 (06) 556-568
- 186 Harrell F. Viewpoints on heterogeneity of treatment effect and precision medicine. Library Catalog; 2018 . Accessed December 30, 2025 at: www.fharrell.com
- 187 Selby JV, Maas CC, Fireman BH, Kent DM. Potential clinical impact of predictive modeling of heterogeneous treatment effects: scoping review of the impact of the PATH Statement. medRxiv 2025; 2024.05 . 06.24306774.
- 188 Martin-Loeches I, Torres A, Nagavci B. et al. ERS/ESICM/ESCMID/ALAT guidelines for the management of severe community-acquired pneumonia. Intensive Care Med 2023; 49 (06) 615-632
- 189 Chaudhuri D, Nei AM, Rochwerg B. et al. 2024 focused update: guidelines on use of corticosteroids in sepsis, acute respiratory distress syndrome, and community-acquired pneumonia. Crit Care Med 2024; 52 (05) e219-e233
- 190 Reade MC, Delaney A, Bailey MJ. et al. Prospective meta-analysis using individual patient data in intensive care medicine. Intensive Care Med 2010; 36 (01) 11-21
- 191 Briel M, Spoorenberg SMC, Bos WJW, Christ-Crain M. Reply to Stern et al. Clin Infect Dis 2018; 67 (09) 1467-1468
- 192 Buell KG, Spicer AB, Casey JD. et al. Individualized treatment effects of oxygen targets in mechanically ventilated critically ill adults. JAMA 2024; 331 (14) 1195-1204
- 193 Goligher EC, Lawler PR, Jensen TP. et al; REMAP-CAP, ATTACC, and ACTIV-4a Investigators. Heterogeneous treatment effects of therapeutic-dose heparin in patients hospitalized for COVID-19. JAMA 2023; 329 (13) 1066-1077
- 194 Bouras M, Roquilly A, Mahe P. et al. Cortisol total/CRP ratio for the prediction of hospital-acquired pneumonia and initiation of corticosteroid therapy in traumatic brain-injured patients. Critical Care 2019 2019 23.
- 195 Torres A, Ceccato A, Ferrer M. et al. Effect of corticosteroids on C-reactive protein in patients with severe community-acquired pneumonia and high inflammatory response: the effect of lymphopenia. J Clin Med 2019; 8 (09) 1461
- 196 Zhuo H, Liu K, Anderson B. et al. Plasma sTNFR1 and IL8 for prognostic enrichment in sepsis trials: a prospective cohort study. Crit Care 2019-12-01 2019; 23: 400
- 197 Kox M, Monneret G, Rimmele T. et al. Monocytic HLA-DR expression kinetics in septic shock patients with different pathogens, sites of infection and adverse outcomes. Crit Care 2020; 24 (01) 110
- 198 Joshi I, Carney W, Rock E. Utility of monocyte HLA-DR and rationale for therapeutic GM-CSF in sepsis immunoparalysis. Front Immunol 2023; 14: 1130214