Skip to main content

Association between dexamethasone treatment and the host response in COVID-19 patients admitted to the general ward

Abstract

Dexamethasone improves clinical outcomes in COVID-19 patients requiring supplementary oxygen. We investigated possible mechanisms of action by comparing sixteen plasma host response biomarkers in general ward patients before and after implementation of dexamethasone as standard of care. 48 patients without and 126 patients with dexamethasone treatment were sampled within 48 h of admission. Endothelial cell and coagulation activation biomarkers were comparable. Dexamethasone treatment was associated with lower plasma interleukin (IL)-6 and IL-1 receptor antagonist levels, whilst other inflammation parameters were not affected. These data argue against modification of vascular-procoagulant responses as an early mechanism of action of dexamethasone in COVID-19.

Introduction

Dexamethasone decreases the risk of progression to invasive mechanical ventilation and mortality in hospitalized patients with coronavirus disease 2019 (COVID-19) requiring supplementary oxygen [1, 2]. For this reason, dexamethasone became standard of care in the Netherlands for hospitalized COVID-19 patients requiring oxygen in August 2020. Data on the mechanism of action underlying the beneficial effect of dexamethasone in COVID-19 is limited.

COVID-19 is associated with endothelial dysfunction and coagulation activation, accompanied by hyperinflammation [3]. In sepsis, corticosteroids inhibit inflammation and endothelial cell activation [4]. However, systemic use of glucocorticoids has been linked to venous thromboembolism, particularly pulmonary embolism [5]. Here, we sought to evaluate the early effect of dexamethasone on endothelial, coagulation and inflammatory responses after hospitalization for COVID-19 by comparing biomarker levels in patients admitted in the era before and after implementation of dexamethasone as standard therapy.

Methods

Patient enrolment was done in one secondary and two tertiary hospitals from March 2020 to May 2020 (first wave of the Dutch COVID-19 outbreak, no dexamethasone) and from October 2020 to March 2021 (second wave, dexamethasone 6 mg daily for up to 10 days). Samples were included from the ELDER-BIOME study (NCT02928367) and the Amsterdam UMC COVID-19 biobank. Patients were eligible if they were admitted to a general ward with COVID-19 (confirmed by SARS-CoV-2 PCR), required oxygen support and had provided written informed consent. Exclusion criteria were readmission, transfer from another hospital, participation in an intervention trial and chronic steroid use. EDTA blood was obtained within 48 h of admission, and if possible 3–4 days after admission. Sixteen biomarkers were measured by Luminex multiplex assay (R&D Systems Inc., Minneapolis, United States) and stratified according to three pathophysiological domains: “endothelial cell activation and function”, “coagulation activation” and “systemic inflammation”. Analyses were performed using R statistical software (version 4.0.2). Biomarker data were log-transformed. Normally distributed data were analysed by Student’s t-test, nonparametric continuous data by Mann–Whitney U test, and categorical data by Fisher exact test. To correct for possible confounding factors, a multiple linear regression analysis was conducted using age, gender, length of symptoms, the time between admission and sampling, comorbidities (cardiovascular, pulmonary, diabetes, malignancy, chronic kidney disease, immune suppression), and disease severity scores (4C Mortality, CURB and MEWS score) as covariates. All significance testing of biomarkers was multiple testing corrected using the Benjamini-Hochberg (BH) method.

Results

Of the 174 patients, 48 patients were enrolled during the first wave (not treated with dexamethasone) and 126 during the second wave (treated with dexamethasone before sampling; Table 1). The median time between the first dose of dexamethasone and sampling was 19 h [interquartile range (IQR): 15–35]. The time between hospital admission and study sampling did not differ between groups (median [IQR] of 1 [1, 2] day). Age, body mass index and chronic comorbidities were similar between groups, whilst the proportion of females was higher in the first wave. Baseline vital signs, clinical and radiology severity scores did not differ between groups. None of the patients was vaccinated, and none received anti-IL-6 antibody treatment. Routine laboratory measurements at baseline showed no differences, except for higher platelet counts in wave 2. Outcome parameters in terms of pulmonary embolism, intensive care unit admission, hospital length of stay and mortality were comparable between groups.

Table 1 Patient characteristics

Our primary objective was to compare host response biomarker levels in plasma obtained within 48 h of admission, i.e., after initiation of dexamethasone treatment in wave 2. Biomarker levels reflecting endothelial cell activation and function or activation of the coagulation system did not differ between patients with or without dexamethasone treatment (Fig. 1). However, patients treated with dexamethasone had lower plasma interleukin (IL)-6 concentrations (median [IQR] 7.92 [4.63–13.89] vs 13.81 [8.06–25.01] pg/mL in untreated patients, p = 0.01) and lower plasma IL-1 receptor antagonist concentrations (1.08 [0.80–1.97] vs 2.04 [1.11–3.26] ng/mL, p < 0.01). After correction for possible confounding, dexamethasone treatment was a significant predictor of not only plasma IL-6 and IL-1 receptor antagonist concentrations, but also of IL-8. Other markers of systemic inflammation or cytokines were not significantly different between waves. In a secondary analysis, we analyzed biomarkers in a subset of patients [13 (27%) from wave 1 and 40 (32%) from wave 2] from whom follow-up samples were obtained 3–4 days after admission. At day 3–4, plasma IL-6 and IL-1 receptor antagonist concentrations were not different between groups, whilst thrombomodulin (endothelial injury marker) and soluble TREM-1 (inflammation marker) were higher in dexamethasone-treated patients (Additional file 1: Table).

Fig. 1
figure 1

Biomarkers in hospitalized COVID-19 ward patients stratified according to treatment with dexamethasone prior to sampling. Data are expressed as box-whisker diagrams representing the median and 1.5 IQR as whiskers, and individual data points. Dotted lines indicate median values obtained in 21 age-matched outpatient clinic non-infectious controls. * Indicates BH-corrected P < 0.05. IL: interleukin, IL-1RA: interleukin-1 receptor antagonist, MMP-8: matrix metalloproteinase-8, RAGE: receptor for advanced glycation end-products, TREM-1: triggering receptor expressed on myeloid cells-1, VCAM-1: vascular cell adhesion molecule-1

Discussion

We here compared the plasma concentrations of 16 host response biomarkers providing insight into dysregulation of key pathways implicated in the pathogenesis of COVID-19 in dexamethasone-treated and untreated patients. Unlike hydrocortisone in sepsis [4], dexamethasone treatment was not associated with attenuated endothelial responses in COVID-19, and dexamethasone did not impact coagulation activation. Dexamethasone treatment was associated with lower plasma IL-6 and IL-1 receptor antagonist levels early after initiation.

A recent observational study in 20 dexamethasone-treated and 12 dexamethasone-untreated COVID-19 patients reported a decrease in angiopoietin-2 and receptor for advanced glycation end-products after dexamethasone treatment [6], biomarkers that were not different between treatment groups in our study. However, unlike our study, this previous investigation involved critically ill patients, had a small sample size and had major baseline differences.

Remarkably, the plasma concentrations of thrombomodulin and TREM-1 were higher at day 3–4 in patients treated with dexamethasone. A clear explanation for this unexpected finding is not available. Dexamethasone did not affect the plasma levels of thrombomodulin in healthy subjects [7], and decreased TREM-1 expression and release from pro-monocytic cells [8].

Our study has strengths and limitations. Data on the effects of dexamethasone on the immune response to SARS-CoV-2 infection are scarce. Whilst patient groups were largely comparable, results are not from a controlled clinical trial. Nevertheless, selection or indication bias is estimated to be minimal due to dexamethasone treatment becoming standard of care from a specific moment in time. The alpha variant (B.1.1.7) became dominant once we terminated enrolling patients in March 2021, minimizing the influence of different SARS-CoV-2 variants. Additionally, correction for demographics, length of symptoms, comorbidity, and diseases severity did not change the results. We compared systemic host responses; analyses of pulmonary responses are not feasible in patients who are not intubated. Our primary analysis focused on host response parameters shortly after hospital admission; nonetheless, measurements in a subgroup 3–4 days after admission did not disclose a mechanism of protective action of dexamethasone either. Notably, in community-acquired pneumonia, dexamethasone influenced plasma cytokine levels early after treatment initiation [9] while in a controlled human inflammation model a single prednisolone dose acutely inhibited cytokine release and endothelial cell activation [10].

Our data argue against modification of vascular-procoagulant responses as an early mechanism of action of dexamethasone in COVID-19. The results of this study could be a rationale for further exploration of the mechanism of action of dexamethasone in COVID-19 in future research.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

BH:

Benjamini-Hochberg

COVID-19:

Coronavirus disease 2019

ICU:

Intensive care unit

IL:

Interleukin

IL-1RA:

Interleukin-1 receptor antagonist

IQR:

Interquartile range

MEWS:

Modified early warning score

MMP-8:

Matrix metalloproteinase-8

qSOFA:

Quick sequential organ failure assessment

RAGE:

Receptor for advanced glycation end-products

TREM-1:

Triggering receptor expressed on myeloid cells-1

VCAM-1:

Vascular cell adhesion molecule-1

References

  1. Group RC, Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, et al. Dexamethasone in hospitalized patients with COVID-19. N Engl J Med. 2021;384(8):693–704.

    Article  Google Scholar 

  2. Cano EJ, Fonseca Fuentes X, Corsini Campioli C, O’Horo JC, Abu Saleh O, Odeyemi Y, et al. Impact of corticosteroids in coronavirus disease 2019 outcomes: systematic review and meta-analysis. Chest. 2021;159(3):1019–40.

    Article  CAS  Google Scholar 

  3. Lazzaroni MG, Piantoni S, Masneri S, Garrafa E, Martini G, Tincani A, et al. Coagulation dysfunction in COVID-19: the interplay between inflammation, viral infection and the coagulation system. Blood Rev. 2021;46: 100745.

    Article  CAS  Google Scholar 

  4. Heming N, Sivanandamoorthy S, Meng P, Bounab R, Annane D. Immune effects of corticosteroids in sepsis. Front Immunol. 2018;9:1736.

    Article  Google Scholar 

  5. Johannesdottir SA, Horvath-Puho E, Dekkers OM, Cannegieter SC, Jorgensen JO, Ehrenstein V, et al. Use of glucocorticoids and risk of venous thromboembolism: a nationwide population-based case-control study. JAMA Intern Med. 2013;173(9):743–52.

    Article  CAS  Google Scholar 

  6. Kim WY, Kweon OJ, Cha MJ, Baek MS, Choi SH. Dexamethasone may improve severe COVID-19 via ameliorating endothelial injury and inflammation: a preliminary pilot study. PLoS ONE. 2021;16(7): e0254167.

    Article  CAS  Google Scholar 

  7. Jilma B, Blann AD, Stohlawetz P, Eichler HG, Kautzky-Willer A, Wagner OF. Dexamethasone lowers circulating E-selectin and ICAM-1 in healthy men. J Lab Clin Med. 2000;135(3):270–4.

    Article  CAS  Google Scholar 

  8. Mihailidou I, Pelekanou A, Pistiki A, Spyridaki A, Tzepi IM, Damoraki G, et al. Dexamethasone down-regulates expression of triggering receptor expressed on myeloid cells-1: evidence for a TNFalpha-related effect. Front Public Health. 2013;1:50.

    Article  Google Scholar 

  9. Remmelts HH, Meijvis SC, Biesma DH, van Velzen-Blad H, Voorn GP, Grutters JC, et al. Dexamethasone downregulates the systemic cytokine response in patients with community-acquired pneumonia. Clin Vaccine Immunol. 2012;19(9):1532–8.

    Article  CAS  Google Scholar 

  10. de Kruif MD, Lemaire LC, Giebelen IA, van Zoelen MA, Pater JM, van den Pangaart PS, et al. Prednisolone dose-dependently influences inflammation and coagulation during human endotoxemia. J Immunol. 2007;178(3):1845–51.

    Article  Google Scholar 

  11. Knight SR, Ho A, Pius R, Buchan I, Carson G, Drake TM, et al. Risk stratification of patients admitted to hospital with covid-19 using the ISARIC WHO Clinical Characterisation Protocol: development and validation of the 4C Mortality Score. BMJ. 2020;370: m3339.

    Article  Google Scholar 

Download references

Acknowledgements

Amsterdam UMC COVID-19 biobank study group

Michiel A. van Agtmael (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Anne Geke Algera (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Brent Appelman (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Frank E.H.P. van Baarle (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Diane J.C. Bax (Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands), Martijn Beudel (Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands), Harm Jan Bogaard (Department of Pulmonology, Amsterdam UMC, Amsterdam, The Netherlands), Marije Bomers (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Peter I. Bonta (Department of Pulmonology, Amsterdam UMC, Amsterdam, The Netherlands), Lieuwe D.J. Bos (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Michela Botta (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Justin de Brabander (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Godelieve J. de Bree (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Sanne de Bruin (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Marianne Bugiani (Department of Medical Microbiology, Amsterdam UMC, Amsterdam, The Netherlands), Esther B. Bulle (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Osoul Chouchane (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Alex P.M. Cloherty (Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands), David T.P. Buis (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Maurits C.F.J. de Rotte (Department of Clinical Chemistry, Amsterdam UMC, Amsterdam, The Netherlands), Mirjam Dijkstra (Department of Clinical Chemistry, Amsterdam UMC, Amsterdam, The Netherlands), Dave A. Dongelmans (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Romein W.G. Dujardin (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Paul E. Elbers (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Lucas M. Fleuren (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Suzanne E. Geerlings (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Theo B.H. Geijtenbeek (Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands), Armand R.J. Girbes (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Bram Goorhuis (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Martin P. Grobusch (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Florianne M.J. Hafkamp (Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands), Laura A. Hagens (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Jörg Hamann (Amsterdam UMC Biobank Core Facility, Amsterdam UMC, Amsterdam, The Netherlands), Vanessa C. Harris (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Robert Hemke (Department of Radiology, Amsterdam UMC, Amsterdam, The Netherlands), Sabine M. Hermans (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Leo M.A. Heunks (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Markus W. Hollmann (Department of Anesthesiology, Amsterdam UMC, Amsterdam, The Netherlands), Janneke Horn (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Joppe W. Hovius (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Menno D. de Jong (Department of Medical Microbiology, Amsterdam UMC, Amsterdam, The Netherlands), Rutger Koning (Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands), Endry H.T. Lim (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Niels van Mourik (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Jeannine Nellen (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Esther J. Nossent (Department of Pulmonology, Amsterdam UMC, Amsterdam, The Netherlands), Frederique Paulus (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Edgar Peters (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Dan A.I. Piña-Fuentes (Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands), Tom van der Poll (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Bennedikt Preckel (Department of Anesthesiology, Amsterdam UMC, Amsterdam, The Netherlands), Jan M. Prins (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Jorinde Raasveld (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Tom D.Y. Reijnders (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Michiel Schinkel (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Femke A.P. Schrauwen (Department of Clinical Chemistry, Amsterdam UMC, Amsterdam, The Netherlands), Marcus J. Schultz (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Alex R. Schuurman (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Jaap Schuurmans (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Kim Sigalof (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Marleen A. Slim (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Patrick Smeele (Department of Pulmonology, Amsterdam UMC, Amsterdam, The Netherlands), Marry R. Smit (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Cornelis S. Stijnis (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Charlotte E. Teunissen (Neurochemical Laboratory, Amsterdam UMC, Amsterdam, The Netherlands), Patrick Thoral (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Anissa M. Tsonas (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Pieter R. Tuinman (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Marc van der Valk (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Denise P. Veelo (Department of Anesthesiology, Amsterdam UMC, Amsterdam, The Netherlands), Carolien Volleman (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Heder de Vries (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Lonneke A. Vught (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Michèle van Vugt (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Dorien Wouters (Department of Clinical Chemistry, Amsterdam UMC, Amsterdam, The Netherlands), Koos Zwinderman (Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, Amsterdam, The Netherlands), Matthijs C. Brouwer (Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands), W. Joost Wiersinga (Department of Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands), Alexander P.J. Vlaar (Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands), Diederik van de Beek (Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands).

Funding

JDB, EM and van CVL are in part funded by European Union H2020 (FAIR project, Grant 847786). The funder had no role in the design and conduct of the study: collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

The authors designed the study together and were involved in collecting the data. JDB, EM and TVDP did the analyses and drafted the manuscript. The other authors revised the initial draft. All authors approved the final version of the manuscript. JDB, EM, CL, OC, TR, AS and TVDP had access to the data. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Justin de Brabander.

Ethics declarations

Ethics approval and consent to participate

The ethical board of Amsterdam UMC location AMC approved the collection of data for the study purposes (NL57847.018.16, 06/09/2016). The study has been performed according to the Declaration of Helsinki. The paper reflects the authors’ own research and analysis in a truthful and complete manner.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Table S1

. Host response biomarkers at day 3-4 of hospital admission. Results are presented as median [interquartile range]. IL interleukin, IL-1RA interleukin-1 receptor antagonist, MMP-8 matrix metalloproteinase-8, RAGE receptor for advanced glycation end-products, TREM-1 triggering receptor expressed on myeloid cells-1, VCAM-1 vascular cell adhesion molecule-1. * Reasons for missingness were discharged (41.3%), transfer to another hospital (5.8%), initiation of trial medication (10.8%) and no sampling (39.7%).

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

de Brabander, J., Michels, E.H.A., van Linge, C.C.A. et al. Association between dexamethasone treatment and the host response in COVID-19 patients admitted to the general ward. Respir Res 23, 145 (2022). https://doi.org/10.1186/s12931-022-02060-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12931-022-02060-3