Physiologic effects of stress dose corticosteroids in in-hospital cardiac arrest (CORTICA): A randomized clinical trial

Spyros D Mentzelopoulos, Evanthia Pappa, Sotirios Malachias, Charikleia S Vrettou, Achilleas Giannopoulos, George Karlis, George Adamos, Ioannis Pantazopoulos, Aikaterini Megalou, Zafeiris Louvaris, Vassiliki Karavana, Epameinondas Aggelopoulos, Gerasimos Agaliotis, Marielen Papadaki, Aggeliki Baladima, Ismini Lasithiotaki, Fotini Lagiou, Prodromos Temperikidis, Aggeliki Louka, Andreas Asimakos, Marios Kougias, Demosthenes Makris, Epameinondas Zakynthinos, Maria Xintara, Maria-Eirini Papadonta, Aikaterini Koutsothymiou, Spyros G Zakynthinos, Eleni Ischaki, Spyros D Mentzelopoulos, Evanthia Pappa, Sotirios Malachias, Charikleia S Vrettou, Achilleas Giannopoulos, George Karlis, George Adamos, Ioannis Pantazopoulos, Aikaterini Megalou, Zafeiris Louvaris, Vassiliki Karavana, Epameinondas Aggelopoulos, Gerasimos Agaliotis, Marielen Papadaki, Aggeliki Baladima, Ismini Lasithiotaki, Fotini Lagiou, Prodromos Temperikidis, Aggeliki Louka, Andreas Asimakos, Marios Kougias, Demosthenes Makris, Epameinondas Zakynthinos, Maria Xintara, Maria-Eirini Papadonta, Aikaterini Koutsothymiou, Spyros G Zakynthinos, Eleni Ischaki

Abstract

Aim: Postresuscitation hemodynamics are associated with hospital mortality/functional outcome. We sought to determine whether low-dose steroids started during and continued after cardiopulmonary resuscitation (CPR) affect postresuscitation hemodynamics and other physiological variables in vasopressor-requiring, in-hospital cardiac arrest.

Methods: We conducted a two-center, randomized, double-blind trial of patients with adrenaline (epinephrine)-requiring cardiac arrest. Patients were randomized to receive either methylprednisolone 40 mg (steroids group) or normal saline-placebo (control group) during the first CPR cycle post-enrollment. Postresuscitation shock was treated with hydrocortisone 240 mg daily for 7 days maximum and gradual taper (steroids group), or saline-placebo (control group). Primary outcomes were arterial pressure and central-venous oxygen saturation (ScvO2) within 72 hours post-ROSC.

Results: Eighty nine of 98 controls and 80 of 86 steroids group patients with ROSC were treated as randomized. Primary outcome data were collected from 100 patients with ROSC (control, n = 54; steroids, n = 46). In intention-to-treat mixed-model analyses, there was no significant effect of group on arterial pressure, marginal mean (95% confidence interval) for mean arterial pressure, steroids vs. control: 74 (68-80) vs. 72 (66-79) mmHg] and ScvO2 [71 (68-75)% vs. 69 (65-73)%], cardiac index [2.8 (2.5-3.1) vs. 2.9 (2.5-3.2) L/min/m2], and serum cytokine concentrations [e.g. interleukin-6, 89.1 (42.8-133.9) vs. 75.7 (52.1-152.3) pg/mL] determined within 72 hours post-ROSC (P = 0.12-0.86). There was no between-group difference in body temperature, echocardiographic variables, prefrontal blood flow index/cerebral autoregulation, organ failure-free days, and hazard for poor in-hospital/functional outcome, and adverse events (P = 0.08->0.99).

Conclusions: Our results do not support the use of low-dose corticosteroids in in-hospital cardiac arrest.Trial Registration:ClinicalTrials.gov number: NCT02790788 ( https://www.clinicaltrials.gov ).

Keywords: Heart Arrest; Hemodynamics; Hydrocortisone; Methylprednisolone; Post-cardiac arrest syndrome.

Conflict of interest statement

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

© 2022 The Authors.

Figures

Fig. 1
Fig. 1
The study flow chart. ROSC, return of spontaneous circulation; ALS, advance life support; ICU, intensive care unit; CPR, cardiopulmonary resuscitation; PRS, postresuscitation shock. *, Pertains to any confirmed deviation from the ALS algorithms presented in references 25 and 26; two patients (control, n = 1) were excluded after randomization. , Consent was refused within 48 hours of ROSC in all cases; four of these patients were initially randomized to the control group. ‡, Four patients were started on open label, stress-dose hydrocortisone within 24 hours post-ROSC; in 3 of these patients, the prescribed hydrocortisone dose differed from the dose specified by the study protocol; study treatment was withheld in another 3 patients within 24 hours post-ROSC; consequently, a total of 6 patients were not treated as randomized. §, Nine patients were started on open label stress-dose hydrocortisone within 24 hours post-ROSC; consequently, these patients were not treated as randomized.
Fig. 2
Fig. 2
Time course of mean arterial pressure (A), log-transformed vasopressor infusion rate in patients who received vasopressors and no./total no. (%) of patient-days of vasopressor use (B), and log-transformed interleukin (IL)-6 (C) in the Steroids and Control group. Data are presented as mean (95% confidence interval). ITT analysis: intention-to-treat mixed-model analysis corresponding to the first 72 hours postresuscitation; EXPL: exploratory mixed-model analyses corresponding to days 1–10 (A, B) or to days 1–7 (C) postresuscitation. In B, the nos./total nos. (%) of patient days of vasopressor use were compared by Fisher’s exact test. ROSC, return of spontaneous circulation. AIC: Akaike information criterion; %VE: percent variance explained, which reflects the squared Pearson correlation coefficient determined by linear regression with observed variable values as dependent variable and mixed model-estimated variable values as independent variable. A, B, and C: The sequences of numbers just above the horizontal axes represent nos. of patients participating in the analysis at the respective follow-up time points. B: The numbers on the top of the bars reflect numbers of patients not receiving any vasopressor support at the corresponding time-points of follow-up. †, Additional model information: A, ITT and EXPL analysis: effect of center and insulin infusion rate, P = 0.17 to 0.52. ‡, Additional model information: B, ITT and EXPL analysis: effect of center, insulin infusion rate, and blood glucose, P = 0.22 to 0.71. §, Additional model information: C, ITT and EXPL analysis: effect of center P = 0.10 to 0.11. †,‡,§, Log-transformed values were actually used in all analyses, because data exhibited skewed distributions; in A, we present the actually observed/recorded values of mean arterial pressure in mmHg, solely for the purpose of a simplified and clear presentation.
Fig. 3
Fig. 3
Probability of survival with a Cerebral Performance Category (CPC) score of 1 or 2 until day 60 after the return of spontaneous circulation (ROSC), which was identical to survival to hospital discharge with a CPC score of 1 or 2; intention-to-treat (ITT) analysis. “No. Alive:” reflects total number of participants minus (1) those who died before the corresponding time point; and (2) those in whom neurologic failure (i.e. Glasgow Coma Scale score of ≤9) was confirmed (before the corresponding time point) and was followed either by death before hospital discharge (without any intervening neurologic failure free day) or by determination of a CPC score of 3 or 4 at hospital discharge (again, without any intervening neurologic failure free day). Therefore, “No. Alive” reflects patients who could still achieve hospital discharge with a CPC score of 1 or 2.

References

    1. Mentzelopoulos S.D., Zakynthinos S.G. Post-cardiac arrest syndrome: pathological processes, biomarkers and vasopressor support, and potential therapeutic targets. Resuscitation. 2017;121:A12–A14.
    1. Jozwiak M., Bougouin W., Geri G., Grimaldi D., Cariou A. Post-resuscitation shock: recent advances in pathophysiology and treatment. Ann Intensive Care. 2020;10:170.
    1. Fink K., Schwarz M., Feldbrügge L., et al. Severe endothelial injury and subsequent repair in patients after successful cardiopulmonary resuscitation. Crit Care. 2010;14:R104.
    1. Reis C., Akyol O., Araujo C., et al. Pathophysiology and the Monitoring Methods for Cardiac Arrest Associated Brain Injury. Int J Mol Sci. 2017;18:129.
    1. González-Ibarra F.P., Varon J., López-Meza E.G. Therapeutic hypothermia: critical review of the molecular mechanisms of action. Front Neurol. 2011;2:4.
    1. Cooper M.S., Stewart P.M. Corticosteroid insufficiency in acutely ill patients. N Engl J Med. 2003;348:727–734.
    1. Hékimian G., Baugnon T., Thuong M., et al. Cortisol levels and adrenal reserve after successful cardiac arrest resuscitation. Shock. 2004;22:116–119.
    1. Pantazopoulos I.N., Zakynthinos S.G., Mentzelopoulos S.D. Corticosteroids and inflammation after cardiac arrest. Resuscitation. 2016;99:e7–e8.
    1. Vaahersalo J., Skrifvars M.B., Pulkki K., et al. FINNRESUSCI Laboratory Study Group. Admission interleukin-6 is associated with post resuscitation organ dysfunction and predicts long-term neurological outcome after out-of-hospital ventricular fibrillation. Resuscitation. 2014;85:1573–1579.
    1. Kim J.J., Hyun S.Y., Hwang S.Y., et al. Hormonal responses upon return of spontaneous circulation after cardiac arrest: a retrospective cohort study. Crit Care. 2011;15:R53.
    1. Adrie C., Adib-Conquy M., Laurent I., et al. Successful cardiopulmonary resuscitation after cardiac arrest as a “sepsis-like” syndrome. Circulation. 2002;106:562–568.
    1. Adrie C., Laurent I., Monvhi M., Cariou A., Dhainou J.F., Spaulding C. Postresuscitation disease after cardiac arrest: a sepsis-like syndrome? Curr Opin Crit Care. 2004;10:208–212.
    1. Keh D., Boehnke T., Weber-Cartens S., et al. 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.
    1. Barnes P.J., Adcock I. Anti-inflammatory actions of steroids: molecular mechanisms. Trends Pharmacol Sci. 1993;14:436–441.
    1. Annane D., Bellissant E., Sebille V., et al. Impaired pressor sensitivity to noradrenaline in septic shock patients with and without impaired adrenal function reserve. Br J Clin Pharmacol. 1998;46:589–597.
    1. Annane D., Renault A., Brun-Buisson C., et al. Hydrocortisone plus Fludrocortisone for Adults with Septic Shock. N Engl J Med. 2018;378:809–818.
    1. Venkatesh B., Finfer S., Cohen J., et al. ADRENAL Trial Investigators and the Australian-New Zealand Intensive Care Society Clinical Trials Group. Adjunctive Glucocorticoid Therapy in Patients with Septic Shock. N Engl J Med. 2018;378:797–808.
    1. Koliantzaki I., Zakynthinos S.G., Mentzelopoulos S.D. In: Resuscitation. Gullo A., Ristagno G., editors. Milano; Springer: 2014. The Potential Contribution of Corticosteroids to Positive Cardiac Arrest Outcomes; pp. 143–155.
    1. Mentzelopoulos S.D., Zakynthinos S.G., Tzoufi M., et al. Vasopressin, epinephrine, and corticosteroids for in-hospital cardiac arrest. Arch Intern Med. 2009;169:15–24.
    1. Mentzelopoulos S.D., Malachias S., Chamos C., et al. Vasopressin, steroids, and epinephrine and neurologically favorable survival after in-hospital cardiac arrest: a randomized clinical trial. JAMA. 2013;310:270–279.
    1. Finkel M.S., Oddis C.V., Jacob T.D., Watkins S.C., Hattler B.G., Simmons R.L. Negative inotropic effects of cytokines on the heart mediated by nitric oxide. Science. 1992;257:387–389.
    1. Kröll S., El-Gindi J., Thanabalasundaram G., Panpumthong P., Schrot S., Hartmann C., Galla H.J. Control of the blood-brain barrier by glucocorticoids and the cells of the neurovascular unit. Ann N Y Acad Sci. 2009;1165:228–239.
    1. Kozler P., Riljak V., Pokorný J. Methylprednisolone reduces axonal impairment in the experimental model of brain oedema. Neuro Endocrinol Lett. 2011;32:831–835.
    1. Trzeciak S., Jones A.E., Kilgannon J.H., et al. Significance of arterial hypotension after resuscitation from cardiac arrest. Crit Care Med. 2009;37:2895–2903.
    1. Link M.S., Berkow L.C., Kudenchuk P.J., et al. Part 7: Adult Advanced Cardiovascular Life Support: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation. 2015;132(18 Suppl 2):S444–S464. Erratum in: Circulation. 2015;132:e385.
    1. Soar J., Nolan J.P., Böttiger B.W., et al. European Resuscitation Council Guidelines for Resuscitation 2015: Section 3. Adult advanced life support. Resuscitation. 2015;95:100–147.
    1. Vincent J.L., Moreno R., Takala J., et al. 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.
    1. Cummins R.O., Chamberlain D., Hazinski M.F., et al. Recommended guidelines for reviewing, reporting, and conducting research on in-hospital resuscitation: the in-hospital “Utstein style.”. Circulation. 1997;95:2213–2239.
    1. Boushel R., Langberg H., Olesen J., et al. Regional blood flow during exercise in humans measured by near – infrared spectroscopy and indocyanine green. J Appl Physiol. 2000;89:1868–1878.
    1. Tatro J.B. Endogenous antipyretics. Clin Infect Dis. 2000;31(Suppl 5):S190–S201.
    1. Tuure L., Hämäläinen M., Nummenmaa E., Moilanen T., Moilanen E. Downregulation of microsomal prostaglandin E synthase-1 (mPGES-1) expression in chondrocytes is regulated by MAP kinase phosphatase-1 (MKP-1) Int Immunopharmacol. 2019;71:139–143.
    1. Bro-Jeppesen J., Hassager C., Wanscher M., et al. Targeted temperature management at 33°C versus 36°C and impact on systemic vascular resistance and myocardial function after out-of-hospital cardiac arrest: a sub-study of the Target Temperature Management Trial. Circ Cardiovasc Interv. 2014;7:663–672.
    1. Annane D. Glucocorticoids in the treatment of severe sepsis and septic shock. Curr Opin Crit Care. 2005;11:449–453.
    1. Baumann G., Dingman J.F. Distribution, blood transport, and degradation of antidiuretic hormone in man. J Clin Invest. 1976;57:1109–1116.
    1. Brown H., Prescott R. In: Applied Mixed Models in Medicine. 2nd ed. Brown H., Prescott R., editors. John Wiley and Sons Ltd; Chichester, West Sussex, England: 2006. Normal Mixed Models; pp. 33–106.
    1. Fergusson D., Aaron S.D., Guyatt G., Hébert P. Post-randomisation exclusions: the intention to treat principle and excluding patients from analysis. BMJ. 2002;325:652–654.
    1. Ono M., Brady K., Easley R.B., et al. Duration and magnitude of blood pressure below cerebral autoregulation threshold during cardiopulmonary bypass is associated with major morbidity and operative mortality. J Thorac Cardiovasc Surg. 2014;147:483–489.
    1. Ameloot K., Genbrugge C., Meex I., et al. An observational near-infrared spectroscopy study on cerebral autoregulation in post-cardiac arrest patients: time to drop 'one-size-fits-all' hemodynamic targets? Resuscitation. 2015;90:121–126.
    1. Sondergaard S., Parkin G., Aneman A. Central venous pressure: soon an outcome-associated matter. Curr Opin Anaesthesiol. 2016;29:179–185.
    1. Åneman A., Wilander P., Zoerner F., Lipcsey M., Chew M.S. Vasopressor Responsiveness Beyond Arterial Pressure: A Conceptual Systematic Review Using Venous Return Physiology. Shock. 2021;56:352–359.
    1. Bro-Jeppesen J., Kjaergaard J., Stammet P., et al. TTM-Trial Investigators. Predictive value of interleukin-6 in post-cardiac arrest patients treated with targeted temperature management at 33 °C or 36 °C. Resuscitation. 2016;98:1–8.
    1. Kilgannon J.H., Roberts B.W., Jones A.E., et al. Arterial blood pressure and neurologic outcome after resuscitation from cardiac arrest. Crit Care Med. 2014;42:2083–2091.
    1. Laurikkala J., Wilkman E., Pettilä V., et al. Mean arterial pressure and vasopressor load after out-of-hospital cardiac arrest: Associations with one-year neurologic outcome. Resuscitation. 2016;105:116–122.
    1. Roberts B.W., Kilgannon J.H., Hunter B.R., et al. Association Between Elevated Mean Arterial Blood Pressure and Neurologic Outcome After Resuscitation From Cardiac Arrest: Results From a Multicenter Prospective Cohort Study. Crit Care Med. 2019;47:93–100.
    1. Donnino M.W., Andersen L.W., Berg K.M., et al. Collaborating Authors from the Beth Israel Deaconess Medical Center’s Center for Resuscitation Science Research Group. Corticosteroid therapy in refractory shock following cardiac arrest: a randomized, double-blind, placebo-controlled, trial. Crit Care. 2016;20:82.
    1. Mentzelopoulos S.D., Mongardon N., Xanthos T., Zakynthinos S.G. Possible significance of hemodynamic and immunomodulatory effects of early stress-dose steroids in cardiac arrest. Crit Care. 2016;20:211.
    1. Skyschally A., Haude M., Dörge H., et al. Glucocorticoid treatment prevents progressive myocardial dysfunction resulting from experimental coronary microembolization. Circulation. 2004;109:2337–2342.
    1. Ullian M.E. The role of corticosteriods in the regulation of vascular tone. Cardiovasc Res. 1999;41:55–64.
    1. Divald A., Powell S.R. Proteasome mediates removal of proteins oxidized during myocardial ischemia. Free Radic Biol Med. 2006;40:156–164.
    1. Calise J., Powell S.R. The ubiquitin proteasome system and myocardial ischemia. Am J Physiol Heart Circ Physiol. 2013;304:H337–H349.
    1. Vassiliou A.G., Floros G., Jahaj E., et al. Decreased glucocorticoid receptor expression during critical illness. Eur J Clin Invest. 2019;49:e13073.
    1. Grafton S.T., Longstreth W.T., Jr. Steroids after cardiac arrest: a retrospective study with concurrent, nonrandomized controls. Neurology. 1988;38:1315–1316.
    1. Jastremski M., Sutton-Tyrrell K., Vaagenes P., Abramson N., Heiselman D., Safar P. Glucocorticoid treatment does not improve neurological recovery following cardiac arrest. Brain Resuscitation Clinical Trial I Study Group. JAMA. 1989;262:3427–3430.
    1. Niimura T., Zamami Y., Koyama T., et al. Hydrocortisone administration was associated with improved survival in Japanese patients with cardiac arrest. Sci Rep. 2017;7:17919.
    1. Tsai M.S., Chuang P.Y., Huang C.H., et al. Postarrest Steroid Use May Improve Outcomes of Cardiac Arrest Survivors. Crit Care Med. 2019;47:167–175.
    1. Li Y., Zhang J., Cai N., He F. Efficacy and safety of corticosteroid therapy in patients with cardiac arrest: a systematic review of randomised controlled trials. Eur J Clin Pharmacol. 2020;76:1631–1638.
    1. Sahebnasagh A., Najmeddin F., Najafi A., et al. Efficacy of Glucocorticoid Administration in Patients with Cardiac Arrest: A Systematic Review of Clinical Studies. Curr Med Chem. 2021 doi: 10.2174/0929867328666210531145617. Epub ahead of print.
    1. Andersen L.W., Isbye D., Kjærgaard J., et al. Effect of Vasopressin and Methylprednisolone vs Placebo on Return of Spontaneous Circulation in Patients With In-Hospital Cardiac Arrest: A Randomized Clinical Trial. JAMA. 2021;326:1586–1594.
    1. Limbourg F.P., Liao J.K. Nontranscriptional actions of the glucocorticoid receptor. J Mol Med (Berl) 2003;81:168–174.
    1. Czock D., Keller F., Rasche F.M., Häussler U. Pharmacokinetics and pharmacodynamics of systemically administered glucocorticoids. Clin Pharmacokinet. 2005;44:61–98.
    1. Christ M., Meyer C., Sippel K., Wehling M. Rapid aldosterone signaling in vascular smooth muscle cells: involvement of phospholipase C, diacylglycerol and protein kinase C alpha. Biochem Biophys Res Commun. 1995;213:123–129.
    1. Yang S., Zhang L. Glucocorticoids and vascular reactivity. Curr Vasc Pharmacol. 2004;2:1–12.
    1. Williams S., Ghosh C. Neurovascular glucocorticoid receptors and glucocorticoids: implications in health, neurological disorders and drug therapy. Drug Discov Today. 2020;25:89–106.
    1. Fruchter O., Kino T., Zoumakis E., Alesci S., De Martino M., Chrousos G., Hochberg Z. The human glucocorticoid receptor (GR) isoform beta differentially suppresses GR{alpha}-induced transactivation stimulated by synthetic glucocorticoids. J Clin Endocrinol Metab. 2005;90:3505–3509.
    1. Rajapandi T., Greene L.E., Eisenberg E. The molecular chaperones Hsp90 and Hsc70 are both necessary and sufficient to activate hormone binding by glucocorticoid receptor. J Biol Chem. 2000;275:22597–22604.

Source: PubMed

3
購読する