Pharmacological principles guiding prolonged glucocorticoid treatment in ARDS

Gianfranco Umberto Meduri, Djillali Annane, Marco Confalonieri, George P Chrousos, Bram Rochwerg, Amanda Busby, Barbara Ruaro, Bernd Meibohm, Gianfranco Umberto Meduri, Djillali Annane, Marco Confalonieri, George P Chrousos, Bram Rochwerg, Amanda Busby, Barbara Ruaro, Bernd Meibohm

Abstract

Current literature addressing the pharmacological principles guiding glucocorticoid (GC) administration in ARDS is scant. This paucity of information may have led to the heterogeneity of treatment protocols and misinterpretation of available findings. GCs are agonist compounds that bind to the GC receptor (GR) producing a pharmacological response. Clinical efficacy depends on the magnitude and duration of exposure to GR. We updated the meta-analysis of randomized trials investigating GC treatment in ARDS, focusing on treatment protocols and response. We synthesized the current literature on the role of the GR in GC therapy including genomic and non-genomic effects, and integrated current clinical pharmacology knowledge of various GCs, including hydrocortisone, methylprednisolone and dexamethasone. This review addresses the role dosage, timing of initiation, mode of administration, duration, and tapering play in achieving optimal response to GC therapy in ARDS. Based on RCTs' findings, GC plasma concentration-time profiles, and pharmacodynamic studies, optimal results are most likely achievable with early intervention, an initial bolus dose to achieve close to maximal GRα saturation, followed by a continuous infusion to maintain high levels of response throughout the treatment period. In addition, patients receiving similar GC doses may experience substantial between-patient variability in plasma concentrations affecting clinical response. GC should be dose-adjusted and administered for a duration targeting clinical and laboratory improvement, followed by dose-tapering to achieve gradual recovery of the suppressed hypothalamic-pituitary-adrenal (HPA) axis. These findings have practical clinical relevance. Future RCTs should consider these pharmacological principles in the study design and interpretation of findings.

Keywords: Acute respiratory distress syndrome; Duration; Glucocorticoid; Glucocorticoid receptor; Pharmacodynamic; Randomized trial; Receptor affinity.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Fig. 1
Fig. 1
Relations on natural logarithmic scales between mean levels of nuclear NF-κB and nuclear GC-GRα during the natural progression of ARDS, and in response to prolonged methylprednisolone treatment. Mean intracellular changes of nuclear GC-activated GRα and NF-κB were observed by exposing peripheral blood mononuclear cells of a healthy volunteer to ARDS patient plasma samples collected longitudinally (days 1, 3, 5, and 8) and after randomization to methylprednisolone treatment (randomization day (R) and post-randomization days 3, 5, 7, and 10). The mean values of nuclear NF-κB are plotted against the mean nuclear GC-GRα levels. Improvers had a pre-defined improvement in lung injury score [45] and/or gas exchange component by day 7. The left panel shows ARDS patients with adaptive and maladaptive responses. In improvers, an inverse relationship was observed between these two transcription factors, with the longitudinal direction of the interaction shifting leftwards (decreased NF-κB) and upward (increased GC-GRα). Conversely, in non-improvers, NF-κB increased over time, while GC-GRα showed no significant changes. We defined the first interaction as GC-GRα-driven, and the second interaction as NF-κB-driven [80]. The right panel shows non-improvers-survivors randomized after day 8 of ARDS to methylprednisolone (n = 11) vs. placebo (n = 6). After natural logarithmic transformation and adjustment for repeated measurements, partial correlations among responses to plasma from the methylprednisolone group were − 0.92 (p < 0.0001) both for nuclear NF-κB and nuclear GRα. For responses to plasma from the placebo group, no significant relationship was found between nuclear NF-κB and nuclear GRα (r = 0.11; p = 0.70) [7]. Prolonged methylprednisolone treatment was associated with upregulation in all measurements of GC-GRα-activity leading to reduction in NF-κB DNA-binding and transcription of inflammatory cytokines. Glucocorticoid treatment changed the longitudinal direction of systemic inflammation from dysregulated (NF-κB-driven, maladaptive response) to regulated (GRα-driven, adaptive response) with significant improvement in indices of alveolar-capillary membrane permeability and markers of inflammation, hemostasis, and tissue repair. Reproduced with permission from reference [5]
Fig. 2
Fig. 2
Glucocorticoid receptor α as a cellular rheostat of homeostatic corrections. The glucocorticoid receptor α (GRα) acts as a cellular rheostat to ensure that a proper response is elicited by the neuroendocrine and immune systems throughout the three phases of homeostatic corrections. The serial sequence of regulatory functions includes the: (1) activation and reinforcement of innate immunity (ready-reinforce), (2) downregulation of pro-inflammatory transcription factors (repress), and (3) promotion of disease resolution (resolve- restore).by switching production from pro-inflammatory to pro-resolving mediators, while at the same stimulating antifibrotic and antioxidant molecules that limit tissue damage and fibrosis, to help achieve optimal restoration of anatomy and function of the affected tissues, and (4) parallel support of adaptive immunity. Modified with permission from reference.[4]. TLR2 toll-like receptor 2, purinergic receptor P2Y2R; NLRP3 NOD-like receptor pyrin containing 3, APR acute phase response, TF transcription factor, NF-κB nuclear factor-κB, AP-1 activator protein-1, AnxA1 annexinA1, AnxA1 receptor, ALXR A4 lipoxin receptor, GILZ glucocorticoid-induced leucine zipper, TGFβ transforming growth factor β
Fig. 3
Fig. 3
Concentration–time profiles for different dosing regimens that have been used in the treatment of early ARDS. Glucocorticoid average plasma concentrations were simulated on the basis of published pharmacokinetic parameters for hydrocortisone (HC), methylprednisolone (MP) and dexamethasone (DX) and converted to MP equivalent concentrations using their reported relative receptor affinity (RRA) and fraction unbound to plasma proteins (fu) [8, 9]. Pharmacokinetic parameters used for the simulations were as previously described: Clearance 18, 21, and 17 L/h; volume of distribution 33, 64, and 103 L for HC, MP and DX, respectively. The conversion to MP equivalent concentrations for HC and DX was performed according to the relationship: MP equivalent plasma concentration = HC or DX concentration × (fu,HC or DX/fu,MP) × (RRAHC or DX/RRAMP). The applied value for RRA and fu were 9, 42, 100 and 0.20, 0.23, 0.32 for HC, MP and DX, respectively, as previously reported [8, 9]. fu determines the fraction of the drug concentration that is not bound to plasma proteins and is thus available to enter cells and interact with GC receptors, i.e., the fraction of the concentration that is pharmacologically active
Fig. 4
Fig. 4
Methylprednisolone pharmacokinetics in ARDS patients. a Time-dependent increase in methylprednisolone clearance [CLt (L/Hr)] in patients treated with a 1 mg/kg loading dose, followed by a 1 mg/kg/day continuous infusion. The high systemic inflammatory state may be responsible for the impaired methylprednisolone metabolism observed in early ARDS at the beginning of therapy. High levels of pro-inflammatory cytokines are known to inhibit the expression and activity of hepatic drug metabolizing enzymes, including multiple cytochrome P450 isozymes relevant for the metabolism of methylprednisolone. Inflammation resolves progressively during continuous therapy, leading to re-establishment of homeostatic conditions of drug metabolizing enzyme systems. This is reflected by a time-dependent increase in methylprednisolone clearance. It took about 2 days (41.1 h) of methylprednisolone therapy to achieve 50% of the improvement in clearance towards the re-establishment of homeostasis.[34]. b Methylprednisolone plasma concentration–time profile in ARDS patients receiving the aforementioned dosing regimen. Methylprednisolone clearance is impaired during time period 1 and its concentrations are high enough to trigger genomic and non-genomic GC effects. This most likely establishes initial control of the generalized inflammatory state [33]. Inflammatory control is at least partially established during period 2 within 2 days of therapy, leading to an increased hepatic methylprednisolone clearance, secondary to re-established drug metabolizing activity. The concentrations are maintained around 203 ± 147 ng/mL during period 3, exerting prolonged sustained anti-inflammatory activity. Reproduced with permission from Yates et al. [34]. Importantly, ARDS patients receiving similar GC doses experience a substantial variability in the resulting plasma concentrations due to between-patient variability; this may affect nati-inflammatory response to treatment
Fig. 5
Fig. 5
Protocol for prolonged methylprednisolone treatment in patients with early ARDS. This protocol was recommended in the 2017 guidelines of the Multispecialty Task Force of the Society of Critical Care Medicine and the European Society of Intensive Care Medicine (Supplemental Digital Content 5, https://links.lww.com/CCM/C918) [35] Vitamin supplementation was not part of the recommended protocol. The dosage is adjusted to ideal body weight and rounded up to the nearest 10 mg (e.g., 77 mg rounded up to 80 mg). Thereafter I would state that 80 mg is an example for a patient with an ideal body weight of 77. Day 0, intravenous bolus (80 mg in 50 cc normal saline) over 30 min. Day 0-to ICU discharge: infusion is done by adding the daily dosage to 240 cc of normal saline and running it at 10 cc/h. If necessary, infusion can be changed to bolus every 6 h (1/4 daily dose) or in the last 6 days to every 12 h (1/2 daily dose). If on day 3–5 there is no improvement or even worsening oxygenation indices the condition is considered “unresolving ARDS”. In this case, a protocol of similar duration of treatment, but with double the daily dose of methylprednisolone (starting with 160 mg/day) is initiated. If the patient is extubated before day 14, the methylprednisolone infusion is advanced to day 15 of drug therapy and tapered according to schedule. Oral administration should be delayed to 5 days after extubation, because enteral absorption of methylprednisolone, and likely other GCs, is compromised for days after extubation. Rapid tapering can be associated with rebound systemic inflammation in the presence of suppressed adrenal function with worsening of lung physiology and an increased mortality risk [60]. If patients worsen significantly, then GC treatment should be restarted again, and after improvement followed by slow tapering, to comply with the Food and Drug Administration’s package insert warnings (Reference ID: 3,032,293) [61]. Vitamin supplementation: ascorbic acid 1.5 g every 6 h mixed in 100 ml saline solution × 4 doses per day; thiamine 100 mg every 12 h mixed in 100 ml dextrose 5% in water × 2 doses per day; vitamin D 480,000 IU dose (30 ml) × 1 dose. Recheck vitamin D level on day 5; if low, supplement 96,000 IU/day for 5 days

References

    1. Villar J, Ferrando C, Martinez D, Ambros A, Munoz T, Soler JA, Aguilar G, Alba F, Gonzalez-Higueras E, Conesa LA, Martin-Rodriguez C, Diaz-Dominguez FJ, Serna-Grande P, Rivas R, Ferreres J, Belda J, Capilla L, Tallet A, Anon JM, Fernandez RL, Gonzalez-Martin JM, dexamethasone in An Dexamethasone treatment for the acute respiratory distress syndrome: a multicentre, randomised controlled trial. Lancet Respir Med. 2020;8:267–276. doi: 10.1016/S2213-2600(19)30417-5.
    1. Group WHOREAfC-TW, Sterne JAC, Murthy S, Diaz JV, Slutsky AS, Villar J, Angus DC, Annane D, Azevedo LCP, Berwanger O, Cavalcanti AB, Dequin PF, Du B, Emberson J, Fisher D, Giraudeau B, Gordon AC, Granholm A, Green C, Haynes R, Heming N, Higgins JPT, Horby P, Juni P, Landray MJ, Le Gouge A, Leclerc M, Lim WS, Machado FR, McArthur C, Meziani F, Moller MH, Perner A, Petersen MW, Savovic J, Tomazini B, Veiga VC, Webb S,Marshall JC, (2020) Association between administration of systemic corticosteroids and mortality among critically Ill patients with COVID-19: a meta-analysis. JAMA 324(13):1330–1341. 10.1001/jama.2020.17023
    1. Park JH, Lee HK. Re-analysis of single cell transcriptome reveals that the NR3C1-CXCL8-neutrophil axis determines the severity of COVID-19. Front Immunol. 2020;11:2145. doi: 10.3389/fimmu.2020.02145.
    1. Meduri GU,Chrousos GP, (2020) General adaptation in critical illness: glucocorticoid receptor-alpha master regulator of homeostatic corrections. Front Endocrinol (Lausanne) 11:161. 10.3389/fendo.2020.00161
    1. Meduri GU, Annane D, Chrousos GP, Marik PE, Sinclair SE. Activation and regulation of systemic inflammation in ARDS: Rationale for prolonged glucocorticoid therapy. Chest. 2009;136:1631–1643. doi: 10.1378/chest.08-2408.
    1. Stringer J (2017) Basic concepts in pharmacology: what you need to know for each drug class. In: Editor (eds) Book Basic concepts in pharmacology: what you need to know for each drug class. McGraw Hill Professional
    1. 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;165:983–991. doi: 10.1164/ajrccm.165.7.2106014.
    1. Derendorf H, Möllmann H, Hochhaus G, Meibohm B, Barth J. Clinical PK/PD modelling as a tool in drug development of corticosteroids. Int J Clin Pharmacol Ther. 1997;35:481–488.
    1. Derendorf H, Hochhaus G, Mollmann H, Barth J, Krieg M, Tunn S, Mollmann C. Receptor-based pharmacokinetic-pharmacodynamic analysis of corticosteroids. J Clin Pharmacol. 1993;33:115–123. doi: 10.1002/j.1552-4604.1993.tb03930.x.
    1. Lapp HE, Bartlett AA, Hunter RG. Stress and glucocorticoid receptor regulation of mitochondrial gene expression. J Mol Endocrinol. 2019;62:R121–R128. doi: 10.1530/JME-18-0152.
    1. Buttgereit F, da Silva JA, Boers M, Burmester GR, Cutolo M, Jacobs J, Kirwan J, Kohler L, Van Riel P, Vischer T, Bijlsma JW. Standardised nomenclature for glucocorticoid dosages and glucocorticoid treatment regimens: current questions and tentative answers in rheumatology. Ann Rheum Dis. 2002;61:718–722. doi: 10.1136/ard.61.8.718.
    1. Jiang CL, Liu L, Tasker JG. Why do we need nongenomic glucocorticoid mechanisms? Front Neuroendocrinol. 2014;35:72–75. doi: 10.1016/j.yfrne.2013.09.005.
    1. Charmandari E, Nicolaides NC, Chrousos GP. Adrenal insufficiency. Lancet. 2014;383:2152–2167. doi: 10.1016/S0140-6736(13)61684-0.
    1. Dendoncker K, Libert C. Glucocorticoid resistance as a major drive in sepsis pathology. Cytokine Growth Factor Rev. 2017;35:85–96. doi: 10.1016/j.cytogfr.2017.04.002.
    1. Dejager L, Vandevyver S, Petta I, Libert C. Dominance of the strongest: inflammatory cytokines versus glucocorticoids. Cytokine Growth Factor Rev. 2014;25:21–33. doi: 10.1016/j.cytogfr.2013.12.006.
    1. Meduri GU, Yates CR. Systemic inflammation-associated glucocorticoid resistance and outcome of ARDS. Ann N Y Acad Sci. 2004;1024:24–53. doi: 10.1196/annals.1321.004.
    1. Annane D, Pastores S, Arlt W, Balk R, Beishuizen A, Briegel J, Carcillo J, Christ-Crain M, Cooper M, Marik P. Critical illness-related corticosteroid insufficiency (CIRCI): a narrative review from a multispecialty task force of the society of critical care medicine (SCCM) and the European Society of Intensive Care Medicine (ESICM) Crit Care Med. 2017;45:2089–2099. doi: 10.1097/CCM.0000000000002724.
    1. Wang XQ, Zhou X, Zhou Y, Rong L, Gao L, Xu W. Low-dose dexamethasone alleviates lipopolysaccharide-induced acute lung injury in rats and upregulates pulmonary glucocorticoid receptors. Respirology. 2008;13:772–780. doi: 10.1111/j.1440-1843.2008.01344.x.
    1. Zhang Y, Zuo W, Rong Q, Teng G, Y Z, Glucocorticoid receptor expression on acute lung injury induced by endotoxin in rats. World J Emerg Med. 2010;1:65–69.
    1. Czock D, Keller F, Rasche FM, Haussler U. Pharmacokinetics and pharmacodynamics of systemically administered glucocorticoids. Clin Pharmacokinet. 2005;44:61–98. doi: 10.2165/00003088-200544010-00003.
    1. Mollmann H, Balbach S, Hochhaus G, Barth J,Derendorf H, (1995) Pharmacokinetic-pharmacodynamic correlations of corticosteroids. Handbook of pharmacokinetic/pharmacodynamic correlation: 323–362
    1. Croxtall JD, Van Hal PTW, Choudhury Q, Gilroy DW, Flower RJ. Different glucocorticoids vary in their genomic and non-genomic mechanism of action in A549 cells. Br J Pharmacol. 2002;135:511–519. doi: 10.1038/sj.bjp.0704474.
    1. Ayyar VS, Jusko WJ. Transitioning from basic toward systems pharmacodynamic models: lessons from corticosteroids. Pharmacol Rev. 2020;72:414–438. doi: 10.1124/pr.119.018101.
    1. Meduri GU, Headley AS, Golden E, Carson SJ, Umberger RA, Kelso T, Tolley EA. Effect of prolonged methylprednisolone therapy in unresolving acute respiratory distress syndrome: a randomized controlled trial. JAMA. 1998;280:159–165. doi: 10.1001/jama.280.2.159.
    1. Steinberg KP, Hudson LD, Goodman RB, Hough CL, Lanken PN, Hyzy R, Thompson BT, Ancukiewicz M, National Heart L, Blood Institute Acute Respiratory Distress Syndrome Clinical Trials N Efficacy and safety of corticosteroids for persistent acute respiratory distress syndrome. N Engl J Med. 2006;354:1671–1684. doi: 10.1056/NEJMoa051693.
    1. Meduri GU, Golden E, Freire AX, Taylor E, Zaman M, Carson SJ, Gibson M, Umberger R. Methylprednisolone infusion in early severe ARDS: results of a randomized controlled trial. Chest. 2007;131:954–963. doi: 10.1378/chest.06-2100.
    1. Rezk N, Ibrahim A. Effects of methylprednisolone in early ARDS. Egypt J Chest Dis Tuberc. 2013;62:167–172. doi: 10.1016/j.ejcdt.2013.02.013.
    1. Meduri GU, Bridges L, Shih MC, Marik PE, Siemieniuk RAC, Kocak M. Prolonged glucocorticoid treatment is associated with improved ARDS outcomes: analysis of individual patients’ data from four randomized trials and trial-level meta-analysis of the updated literature. Intensive Care Med. 2016;42:829–840. doi: 10.1007/s00134-015-4095-4.
    1. Confalonieri M, Urbino R, Potena A, Piattella M, Parigi P, Puccio G, Della Porta R, Giorgio C, Blasi F, Umberger R, Meduri GU. Hydrocortisone infusion for severe community-acquired pneumonia: a preliminary randomized study. Am J Respir Crit Care Med. 2005;171:242–248. doi: 10.1164/rccm.200406-808OC.
    1. Annane D, Sebille V, Bellissant E, Ger-Inf-05 Study G Effect of low doses of corticosteroids in septic shock patients with or without early acute respiratory distress syndrome. Crit Care Med. 2006;34:22–30. doi: 10.1097/01.CCM.0000194723.78632.62.
    1. Sabry NA, Omar EE-D. Corticosteroids and ICU course of community acquired pneumonia in Egyptian settings. Pharmacol Pharm. 2011;2:73–81. doi: 10.4236/pp.2011.22009.
    1. Liu L, Li J, Huang YZ, Liu SQ, Yang CS, Guo FM, Qiu HB, Yang Y. The effect of stress dose glucocorticoid on patients with acute respiratory distress syndrome combined with critical illness-related corticosteroid insufficiency. Zhonghua Nei Ke Za Zhi. 2012;51:599–603.
    1. Morgan ET. Regulation of cytochromes P450 during inflammation and infection. Drug Metab Rev. 1997;29:1129–1188. doi: 10.3109/03602539709002246.
    1. Yates CR, Vysokanov A, Mukherjee A, Ludden TM, Tolley EA, Meduri GU, Dalton JT. Time-variant increase in methylprednisolone clearance in patients with acute respiratory distress syndrome: a population pharmocokinetic study. J Clin Pharmacol. 2001;41:1–10. doi: 10.1177/00912700122010276.
    1. Annane D, Pastores S, Rochwerg B, Arlt W, Balk R, Beishuizen A, Briegel J, Carcillo J, Christ-Crain M, Cooper M. 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) Crit Care Med. 2017;45:2078–2088. doi: 10.1097/CCM.0000000000002737.
    1. Volbeda M, Wetterslev J, Gluud C, Zijlstra JG, van der Horst IC, Keus F. Glucocorticosteroids for sepsis: systematic review with meta-analysis and trial sequential analysis. Intensive Care Med. 2015;41:1220–1234. doi: 10.1007/s00134-015-3899-6.
    1. Jantz MA, Sahn SA. Corticosteroids in acute respiratory failure. Am J Respir Crit Care Med. 1999;160:1079–1100. doi: 10.1164/ajrccm.160.4.9901075.
    1. Teng D, Pang QF, Yan WJ, Zhao Xin W, Xu CY. The harmful effect of prolonged high-dose methylprednisolone in acute lung injury. Int Immunopharmacol. 2013;15:223–226. doi: 10.1016/j.intimp.2012.12.004.
    1. Takaki M, Ichikado K, Kawamura K, Gushima Y, Suga M. The negative effect of initial high-dose methylprednisolone and tapering regimen for acute respiratory distress syndrome: a retrospective propensity matched cohort study. Crit Care. 2017;21:135. doi: 10.1186/s13054-017-1723-0.
    1. Kido T, Muramatsu K, Asakawa T, Otsubo H, Ogoshi T, Oda K, Kubo T, Fujino Y, Matsuda S, Mayumi T. The relationship between high-dose corticosteroid treatment and mortality in acute respiratory distress syndrome: a retrospective and observational study using a nationwide administrative database in Japan. BMC Pulmonary Med. 2018;18:28. doi: 10.1186/s12890-018-0597-5.
    1. Meduri GU, Kanangat S, Bronze M, Patterson DR, Meduri CU, Pak C, Schaberg DR. Effects of methylprednisolone on intracellular bacterial growth. Clin Diagn Lab Immunol. 2001;8:1156–1163. doi: 10.1128/CDLI.8.6.1156-1163.2001.
    1. Tongyoo S, Permpikul C, Mongkolpun W, Vattanavanit V, Udompanturak S, Kocak M, Meduri GU. Hydrocortisone treatment in early sepsis-associated acute respiratory distress syndrome: results of a randomized controlled trial. Crit Care. 2016;20:329. doi: 10.1186/s13054-016-1511-2.
    1. Annane D, Bellissant E, Bollaert PE, Briegel J, Keh D, Kupfer Y, Pirracchio R,Rochwerg B, (2019) Corticosteroids for treating sepsis in children and adults. Cochrane Database Syst Rev 12(12):CD002243. 10.1002/14651858.CD002243.pub4
    1. Busillo JM, Cidlowski JA. The five Rs of glucocorticoid action during inflammation: ready, reinforce, repress, resolve, and restore. Trends Endocrinol Metab: TEM. 2013;24:109–119. doi: 10.1016/j.tem.2012.11.005.
    1. Murray JF, Matthay MA, Luce JM, Flick MR. An expanded definition of the adult respiratory distress syndrome. Am Rev Respir Dis. 1988;138:720–723. doi: 10.1164/ajrccm/138.3.720.
    1. Vincent JL, Moreno R, Takala J, Willatts S, De Mendonca A, Bruining H, Reinhart CK, Suter PM, Thijs LG. The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. On behalf of the working group on sepsis-related problems of the european society of intensive care medicine. Intensive Care Med. 1996;22:707–710. doi: 10.1007/BF01709751.
    1. Horby P, Lim WS, Emberson J, Mafham M, Bell J, Linsell L, Staplin N, Brightling C, Ustianowski A,Elmahi E, (2020) Dexamethasone in hospitalized patients with Covid-19—Preliminary Report. N Engl J Med. 10.1056/NEJMoa2021436
    1. Müller B, Peri G, Doni A, Perruchoud AP, Landmann R, Pasqualini F, Mantovani A. High circulating levels of the IL-1 type II decoy receptor in critically ill patients with sepsis: association of high decoy receptor levels with glucocorticoid administration. J Leukoc Biol. 2002;72:643–649.
    1. de Kruif MD, Lemaire LC, Giebelen IA, Struck J, Morgenthaler NG, Papassotiriou J, Elliott PJ, van der Poll T. The influence of corticosteroids on the release of novel biomarkers in human endotoxemia. Intensive Care Med. 2008;34:518–522. doi: 10.1007/s00134-007-0955-x.
    1. Perren A, Cerutti B, Lepori M, Senn V, Capelli B, Duchini F, Domenighetti G. Influence of steroids on procalcitonin and C-reactive protein in patients with COPD and community-acquired pneumonia. Infection. 2008;36:163–166. doi: 10.1007/s15010-007-7206-5.
    1. Rinaldi S, Adembri C, Grechi S, De Gaudio AR. Low-dose hydrocortisone during severe sepsis: effects on microalbuminuria. Crit Care Med. 2006;34:2334–2339. doi: 10.1097/.
    1. Furst D, Saag K Glucocorticoid withdrawal. Uptodate. 2014 Mar [citado 25 Sep 2014]. In: Editor (eds) Book Glucocorticoid withdrawal. Uptodate. 2014 Mar [citado 25 Sep 2014]
    1. Dinsen S, Baslund B, Klose M, Rasmussen AK, Friis-Hansen L, Hilsted L, Feldt-Rasmussen U. Why glucocorticoid withdrawal may sometimes be as dangerous as the treatment itself. Eur J Internal Med. 2013;24:714–720. doi: 10.1016/j.ejim.2013.05.014.
    1. Schuetz P, Leuppi JD, Bingisser R, Bodmer M, Briel M, Drescher T, Duerring U, Henzen C, Leibbrandt Y, Maier S, Miedinger D, Mueller B, Scherr A, Schindler C, Stoeckli R, Viatte S, von Garnier C, Tamm M, Rutishauser J. Prospective analysis of adrenal function in patients with acute exacerbations of COPD: the Reduction in the Use of Corticosteroids in Exacerbated COPD (REDUCE) trial. Eur J Endocrinol. 2015;173:19–27. doi: 10.1530/EJE-15-0182.
    1. Hakkinen PJ, Schmoyer RL, Witschi HP. Potentiation of butylated-hydroxytoluene-induced acute lung damage by oxygen. Effects of prednisolone and indomethacin. Am Rev Respir Dis. 1983;128:648–651.
    1. Kehrer JP, Klein-Szanto AJ, Sorensen EM, Pearlman R, Rosner MH. Enhanced acute lung damage following corticosteroid treatment. Am Rev Respir Dis. 1984;130:256–261. doi: 10.1164/arrd.1984.130.2.256.
    1. Keh DBT, Weber-Cartens S, Schulz C, Ahlers O, Bercker S, Volk HD, Doecke WD, Falke KJ, Gerlach H. Immunologic and hemodynamic effects of “low-dose” hydrocortisone in septic shock: a double-blind, randomized, placebo-controlled, crossover study. Am J Respir Crit Care Med. 2003;167:512–520. doi: 10.1164/rccm.200205-446OC.
    1. Gagnon S, Boota AM, Fischl MA, Baier H, Kirksey OW, La Voie L. Corticosteroids as adjunctive therapy for severe Pneumocystis carinii pneumonia in the acquired immunodeficiency syndrome. A double-blind, placebo-controlled trial. N Engl J Med. 1990;323:1444–1450. doi: 10.1056/NEJM199011223232103.
    1. Nawab Q, Golden E, Confalonieri M, Umberger R, Meduri G. Corticosteroid treatment in severe community-acquired pneumonia: duration of treatment affects control of systemic inflammation and clinical improvement. Intensive Care Med. 2011;37:1153–1554. doi: 10.1007/s00134-011-2274-5.
    1. 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;46:884–891. doi: 10.1097/CCM.0000000000003021.
    1. Administration TFaD (Revised October 2011) SOLU-MEDROL ® (methylprednisolone sodium succinate for injection, USP). In: Editor (ed)^(eds) Book SOLU-MEDROL ® (methylprednisolone sodium succinate for injection, USP). pp. 1–18
    1. Amrein K, Oudemans-van Straaten HM, Berger MM. Vitamin therapy in critically ill patients: focus on thiamine, vitamin C, and vitamin D. Intensive Care Med. 2018;44:1940–1944. doi: 10.1007/s00134-018-5107-y.
    1. Moskowitz A, Andersen LW, Huang DT, Berg KM, Grossestreuer AV, Marik PE, Sherwin RL, Hou PC, Becker LB, Cocchi MN, Doshi P, Gong J, Sen A, Donnino MW. Ascorbic acid, corticosteroids, and thiamine in sepsis: a review of the biologic rationale and the present state of clinical evaluation. Crit Care. 2018;22:283. doi: 10.1186/s13054-018-2217-4.
    1. Fowler AA, 3rd, Truwit JD, Hite RD, Morris PE, DeWilde C, Priday A, Fisher B, Thacker LR, 2nd, Natarajan R, Brophy DF, Sculthorpe R, Nanchal R, Syed A, Sturgill J, Martin GS, Sevransky J, Kashiouris M, Hamman S, Egan KF, Hastings A, Spencer W, Tench S, Mehkri O, Bindas J, Duggal A, Graf J, Zellner S, Yanny L, McPolin C, Hollrith T, Kramer D, Ojielo C, Damm T, Cassity E, Wieliczko A, Halquist M. 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;322:1261–1270. doi: 10.1001/jama.2019.11825.
    1. Takano K, Yamamoto S, Tomita K, Takashina M, Yokoo H, Matsuda N, Takano Y, Hattori Y. Successful treatment of acute lung injury with pitavastatin in septic mice: potential role of glucocorticoid receptor expression in alveolar macrophages. J Pharmacol Exp Ther. 2011;336:381–390. doi: 10.1124/jpet.110.171462.
    1. Komatsubara M, Hara T, Hosoya T, Toma K, Tsukamoto-Yamauchi N, Iwata N, Inagaki K, Wada J, Otsuka F, (2017) Melatonin regulates catecholamine biosynthesis by modulating bone morphogenetic protein and glucocorticoid actions. J Steroid Biochem Mol Biol 165:182–189. 10.1016/j.jsbmb.2016.06.002
    1. Hong J, Guo W-z, Yan Z-h, Di L, Lu C-l. Influence of drug treatment on glucocorticoid receptor levels in patients with coronary heart disease. Chin Med J. 2010;123:1685–1689.
    1. Mantzarlis K, Tsolaki V,Zakynthinos E, (2017) Role of oxidative stress and mitochondrial dysfunction in sepsis and potential therapies. Oxid Med Cell Longev 2017:5985209. 10.1155/2017/5985209
    1. Souffriau J, Libert C, (2018) Mechanistic insights into the protective impact of zinc on sepsis. Cytokine Growth Factor Rev 39:92–101. 10.1016/j.cytogfr.2017.12.002
    1. Erol N, Saglam L, Saglam YS, Erol HS, Altun S, Aktas MS,Halici MB, (2019) The protection potential of antioxidant vitamins against acute respiratory distress syndrome: a rat trial. Inflammation 42(5):1585–1594. 10.1007/s10753-019-01020-2
    1. Siemieniuk RA, Meade MO, Alonso-Coello P, Briel M, Evaniew N, Prasad M, Alexander PE, Fei Y, Vandvik PO, Loeb M, Guyatt GH. Corticosteroid therapy for patients hospitalized with community-acquired pneumonia: a systematic review and meta-analysis. Ann Intern Med. 2015;163:519–528. doi: 10.7326/M15-0715.
    1. Arabi MY, Meduri GU, Chrousos GP, (2020) The ten reasons why corticosteroid therapy also reduces mortality in severe COVID‑19. Intensive Care Med. 10.1007/s00134-020-06223-y
    1. Lamontagne F, Agoritsas T, Macdonald H, Leo Y-S, Diaz J, Agarwal A, Appiah JA, Arabi Y, Blumberg L, Calfee CS, (2020) A living WHO guideline on drugs for covid-19. BMJ 370:m3379. 10.1136/bmj.m3379
    1. Meduri GU, Schwingshackl A, Hermans G. Prolonged glucocorticoid treatment in ARDS: impact on intensive care unit-acquired weakness. Front Pediatr. 2016;4:69. doi: 10.3389/fped.2016.00069.
    1. Meduri GU. Clinical review: a paradigm shift: the bidirectional effect of inflammation on bacterial growth. Clinical implications for patients with acute respiratory distress syndrome. Crit Care. 2002;6:24–29. doi: 10.1186/cc1450.
    1. Kaufmann I, Briegel J, Schliephake F, Hoelzl A, Chouker A, Hummel T, Schelling G, Thiel M. Stress doses of hydrocortisone in septic shock: beneficial effects on opsonization-dependent neutrophil functions. Intensive Care Med. 2008;34:344–349. doi: 10.1007/s00134-007-0868-8.
    1. Spencer-Segal JL, Hyzy RC, Iwashyna TJ, Standiford TJ. Psychiatric symptoms in survivors of acute respiratory distress syndrome. Effects of age, sex, and immune modulation. Ann Am Thorac Soc. 2017;14:960–967. doi: 10.1513/AnnalsATS.201606-468OC.
    1. Schelling G, Briegel J, Roozendaal B, Stoll C, Rothenhausler HB, Kapfhammer HP. The effect of stress doses of hydrocortisone during septic shock on posttraumatic stress disorder in survivors. Biol Psychiatry. 2001;50:978–985. doi: 10.1016/S0006-3223(01)01270-7.
    1. Reddy K, Sinha P, O'Kane CM, Gordon AC, Calfee CS, McAuley DF. Subphenotypes in critical care: translation into clinical practice. Lancet Respir Med. 2020;8:631–643. doi: 10.1016/S2213-2600(20)30124-7.
    1. Meduri GU, Muthiah MP, Carratu P, Eltorky M, Chrousos GP. Nuclear factor-kappaB- and glucocorticoid receptor alpha- mediated mechanisms in the regulation of systemic and pulmonary inflammation during sepsis and acute respiratory distress syndrome. Evidence for inflammation-induced target tissue resistance to glucocorticoids. NeuroImmunoModulation. 2005;12:321–338. doi: 10.1159/000091126.

Source: PubMed

3
Suscribir