Skip to main content
Top
Published in: Virology Journal 1/2022

Open Access 01-12-2022 | Anakinra | Review

COVID-19 infection: an overview on cytokine storm and related interventions

Authors: Soheila Montazersaheb, Seyed Mahdi Hosseiniyan Khatibi, Mohammad Saeid Hejazi, Vahideh Tarhriz, Afsaneh Farjami, Faramarz Ghasemian Sorbeni, Raheleh Farahzadi, Tohid Ghasemnejad

Published in: Virology Journal | Issue 1/2022

Login to get access

Abstract

Coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has posed a significant threat to global health. This virus affects the respiratory tract and usually leads to pneumonia in most patients and acute respiratory distress syndrome (ARDS) in 15% of cases. ARDS is one of the leading causes of death in patients with COVID-19 and is mainly triggered by elevated levels of pro-inflammatory cytokines, referred to as cytokine storm. Interleukins, such as interleukin-6 (1L-6), interleukin-1 (IL-1), interleukin-17 (IL-17), and tumor necrosis factor-alpha (TNF-α) play a very significant role in lung damage in ARDS patients through the impairments of the respiratory epithelium. Cytokine storm is defined as acute overproduction and uncontrolled release of pro-inflammatory markers, both locally and systemically. The eradication of COVID-19 is currently practically impossible, and there is no specific treatment for critically ill patients with COVID-19; however, suppressing the inflammatory response may be a possible strategy. In light of this, we review the efficacy of specific inhibitors of IL6, IL1, IL-17, and TNF-α for treating COVID-19-related infections to manage COVID-19 and improve the survival rate for patients suffering from severe conditions.

Graphical Abstract

Literature
4.
go back to reference Grasselli G, et al. Pathophysiology of COVID-19-associated acute respiratory distress syndrome—authors’ reply. Lancet Respir Med. 2021;9(1):e5–6.PubMedCrossRef Grasselli G, et al. Pathophysiology of COVID-19-associated acute respiratory distress syndrome—authors’ reply. Lancet Respir Med. 2021;9(1):e5–6.PubMedCrossRef
6.
go back to reference Pearce L, Davidson SM, Yellon DM. The cytokine storm of COVID-19: a spotlight on prevention and protection. Expert Opin Ther Targets. 2020;24(8):723–30.PubMedCrossRef Pearce L, Davidson SM, Yellon DM. The cytokine storm of COVID-19: a spotlight on prevention and protection. Expert Opin Ther Targets. 2020;24(8):723–30.PubMedCrossRef
7.
go back to reference Yavuz S, Ünal S. Anti-viral treatment of COVID-19. Turk J Med Sci. 2020;50(SI-1):611–9.CrossRef Yavuz S, Ünal S. Anti-viral treatment of COVID-19. Turk J Med Sci. 2020;50(SI-1):611–9.CrossRef
9.
go back to reference Prompetchara E, Ketloy C, Palaga T. Immune responses in COVID-19 and potential vaccines: lessons learned from SARS and MERS epidemic. Asian Pac J Allergy Immunol. 2020;38(1):1–9.PubMed Prompetchara E, Ketloy C, Palaga T. Immune responses in COVID-19 and potential vaccines: lessons learned from SARS and MERS epidemic. Asian Pac J Allergy Immunol. 2020;38(1):1–9.PubMed
10.
go back to reference Channappanavar R, Perlman, S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. In: Seminars in immunopathology. Springer; 2017. Channappanavar R, Perlman, S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. In: Seminars in immunopathology. Springer; 2017.
11.
go back to reference Wang Y, et al. Unique epidemiological and clinical features of the emerging 2019 novel coronavirus pneumonia (COVID-19) implicate special control measures. J Med Virol. 2020;92(6):568–76.PubMedCrossRef Wang Y, et al. Unique epidemiological and clinical features of the emerging 2019 novel coronavirus pneumonia (COVID-19) implicate special control measures. J Med Virol. 2020;92(6):568–76.PubMedCrossRef
12.
go back to reference Caricchio R, et al. Preliminary predictive criteria for COVID-19 cytokine storm. Ann Rheum Dis. 2021;80(1):88–95.PubMedCrossRef Caricchio R, et al. Preliminary predictive criteria for COVID-19 cytokine storm. Ann Rheum Dis. 2021;80(1):88–95.PubMedCrossRef
13.
14.
go back to reference McGonagle D, et al. The role of cytokines including interleukin-6 in COVID-19 induced pneumonia and macrophage activation syndrome-like disease. Autoimmun Rev. 2020;19(6):102537.PubMedPubMedCentralCrossRef McGonagle D, et al. The role of cytokines including interleukin-6 in COVID-19 induced pneumonia and macrophage activation syndrome-like disease. Autoimmun Rev. 2020;19(6):102537.PubMedPubMedCentralCrossRef
18.
19.
21.
go back to reference Adhikari SP, et al. Epidemiology, causes, clinical manifestation and diagnosis, prevention and control of coronavirus disease (COVID-19) during the early outbreak period: a scoping review. Infect Dis Poverty. 2020;9(1):1–12.CrossRef Adhikari SP, et al. Epidemiology, causes, clinical manifestation and diagnosis, prevention and control of coronavirus disease (COVID-19) during the early outbreak period: a scoping review. Infect Dis Poverty. 2020;9(1):1–12.CrossRef
23.
go back to reference Wang D, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus–infected pneumonia in Wuhan, China. JAMA. 2020;323(11):1061–9.PubMedPubMedCentralCrossRef Wang D, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus–infected pneumonia in Wuhan, China. JAMA. 2020;323(11):1061–9.PubMedPubMedCentralCrossRef
24.
go back to reference Aggarwal G, et al. Association of cardiovascular disease with coronavirus disease 2019 (COVID-19) severity: a meta-analysis. Curr Probl Cardiol. 2020;45:100617.PubMedPubMedCentralCrossRef Aggarwal G, et al. Association of cardiovascular disease with coronavirus disease 2019 (COVID-19) severity: a meta-analysis. Curr Probl Cardiol. 2020;45:100617.PubMedPubMedCentralCrossRef
26.
go back to reference Mao L, et al. Neurological manifestations of hospitalized patients with COVID-19 in Wuhan, China: a retrospective case series study. MedRxiv; 2020. Mao L, et al. Neurological manifestations of hospitalized patients with COVID-19 in Wuhan, China: a retrospective case series study. MedRxiv; 2020.
27.
go back to reference Aghagoli G, et al. Neurological involvement in COVID-19 and potential mechanisms: a review. Neurocrit Care. 2020;34:1062–71.CrossRef Aghagoli G, et al. Neurological involvement in COVID-19 and potential mechanisms: a review. Neurocrit Care. 2020;34:1062–71.CrossRef
29.
go back to reference Joseph A, et al. Acute kidney injury in patients with SARS-CoV-2 infection. Ann Intensive Care. 2020;10(1):1–8.CrossRef Joseph A, et al. Acute kidney injury in patients with SARS-CoV-2 infection. Ann Intensive Care. 2020;10(1):1–8.CrossRef
31.
go back to reference Zhou Y, et al. Aberrant pathogenic GM-CSF+ T cells and inflammatory CD14+ CD16+ monocytes in severe pulmonary syndrome patients of a new coronavirus. BioRxiv; 2020. Zhou Y, et al. Aberrant pathogenic GM-CSF+ T cells and inflammatory CD14+ CD16+ monocytes in severe pulmonary syndrome patients of a new coronavirus. BioRxiv; 2020.
32.
go back to reference Zhu J, et al. Clinical characteristics of 3062 COVID-19 patients: a meta-analysis. J Med Virol. 2020;92(10):1902–14.PubMedCrossRef Zhu J, et al. Clinical characteristics of 3062 COVID-19 patients: a meta-analysis. J Med Virol. 2020;92(10):1902–14.PubMedCrossRef
33.
go back to reference Kaya G, Kaya A, Saurat J-H. Clinical and histopathological features and potential pathological mechanisms of skin lesions in COVID-19: review of the literature. Dermatopathology. 2020;7(1):3–16.PubMedPubMedCentralCrossRef Kaya G, Kaya A, Saurat J-H. Clinical and histopathological features and potential pathological mechanisms of skin lesions in COVID-19: review of the literature. Dermatopathology. 2020;7(1):3–16.PubMedPubMedCentralCrossRef
34.
go back to reference Beyerstedt S, Casaro EB, Rangel ÉB. COVID-19: angiotensin-converting enzyme 2 (ACE2) expression and tissue susceptibility to SARS-CoV-2 infection. Eur J Clin Microbiol Infect Dis. 2021;40:905–19.PubMedPubMedCentralCrossRef Beyerstedt S, Casaro EB, Rangel ÉB. COVID-19: angiotensin-converting enzyme 2 (ACE2) expression and tissue susceptibility to SARS-CoV-2 infection. Eur J Clin Microbiol Infect Dis. 2021;40:905–19.PubMedPubMedCentralCrossRef
35.
go back to reference Zou X, et al. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front Med. 2020;14:185–92.PubMedCrossRef Zou X, et al. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front Med. 2020;14:185–92.PubMedCrossRef
37.
go back to reference Li CK, et al. T cell responses to whole SARS coronavirus in humans. J Immunol. 2008;181(8):5490–500.PubMedCrossRef Li CK, et al. T cell responses to whole SARS coronavirus in humans. J Immunol. 2008;181(8):5490–500.PubMedCrossRef
38.
go back to reference Yang X, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;8(5):475–81.PubMedPubMedCentralCrossRef Yang X, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;8(5):475–81.PubMedPubMedCentralCrossRef
39.
41.
go back to reference Liu Y, et al. Elevated plasma levels of selective cytokines in COVID-19 patients reflect viral load and lung injury. Natl Sci Rev. 2020;7(6):1003–11.PubMedPubMedCentralCrossRef Liu Y, et al. Elevated plasma levels of selective cytokines in COVID-19 patients reflect viral load and lung injury. Natl Sci Rev. 2020;7(6):1003–11.PubMedPubMedCentralCrossRef
42.
44.
go back to reference Kishimoto T. Interleukin-6: discovery of a pleiotropic cytokine. Arthritis Res Ther. 2006;8(2):1–6. Kishimoto T. Interleukin-6: discovery of a pleiotropic cytokine. Arthritis Res Ther. 2006;8(2):1–6.
45.
go back to reference Rostamian A, et al. Interleukin-6 as a Potential Predictor of COVID-19 disease severity in hospitalized patients and its association with clinical laboratory routine tests. Immunoregulation. 2020;3(1):29–36.CrossRef Rostamian A, et al. Interleukin-6 as a Potential Predictor of COVID-19 disease severity in hospitalized patients and its association with clinical laboratory routine tests. Immunoregulation. 2020;3(1):29–36.CrossRef
46.
go back to reference Coomes EA, Haghbayan H. Interleukin-6 in COVID-19: a systematic review and meta-analysis. Rev Med Virol. 2020;30(6):1–9.PubMedCrossRef Coomes EA, Haghbayan H. Interleukin-6 in COVID-19: a systematic review and meta-analysis. Rev Med Virol. 2020;30(6):1–9.PubMedCrossRef
48.
go back to reference Chen J-J, et al. Interleukin-6 signaling blockade treatment for cytokine release syndrome in COVID-19. Exp Ther Med. 2021;21(1):1–1. Chen J-J, et al. Interleukin-6 signaling blockade treatment for cytokine release syndrome in COVID-19. Exp Ther Med. 2021;21(1):1–1.
49.
go back to reference Rose-John S. Coordination of interleukin-6 biology by membrane bound and soluble receptors. In: Mackiewicz A, Kurpisz M, Żeromski J, editors. Progress in basic and clinical immunology. Berlin: Springer; 2001. p. 145–51.CrossRef Rose-John S. Coordination of interleukin-6 biology by membrane bound and soluble receptors. In: Mackiewicz A, Kurpisz M, Żeromski J, editors. Progress in basic and clinical immunology. Berlin: Springer; 2001. p. 145–51.CrossRef
50.
go back to reference Wolf J, Rose-John S, Garbers CJC. Interleukin-6 and its receptors: a highly regulated and dynamic system. Cytokine. 2014;70(1):11–20.PubMedCrossRef Wolf J, Rose-John S, Garbers CJC. Interleukin-6 and its receptors: a highly regulated and dynamic system. Cytokine. 2014;70(1):11–20.PubMedCrossRef
51.
go back to reference Lacroix M, et al. Novel insights into interleukin 6 (IL-6) cis-and trans-signaling pathways by differentially manipulating the assembly of the IL-6 signaling complex. J Biol Chem. 2015;290(45):26943–53.PubMedPubMedCentralCrossRef Lacroix M, et al. Novel insights into interleukin 6 (IL-6) cis-and trans-signaling pathways by differentially manipulating the assembly of the IL-6 signaling complex. J Biol Chem. 2015;290(45):26943–53.PubMedPubMedCentralCrossRef
54.
go back to reference Neurath MF, Finotto S. IL-6 signaling in autoimmunity, chronic inflammation and inflammation-associated cancer. Cytokine Growth Factor Rev. 2011;22(2):83–9.PubMedCrossRef Neurath MF, Finotto S. IL-6 signaling in autoimmunity, chronic inflammation and inflammation-associated cancer. Cytokine Growth Factor Rev. 2011;22(2):83–9.PubMedCrossRef
55.
go back to reference Heink S, et al. Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH 17 cells. Nat Immunol. 2017;18(1):74–85.PubMedCrossRef Heink S, et al. Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH 17 cells. Nat Immunol. 2017;18(1):74–85.PubMedCrossRef
57.
go back to reference Tanaka T, Narazaki M, Kishimoto T. Anti-interleukin-6 receptor antibody, tocilizumab, for the treatment of autoimmune diseases. FEBS Lett. 2011;585(23):3699–709.PubMedCrossRef Tanaka T, Narazaki M, Kishimoto T. Anti-interleukin-6 receptor antibody, tocilizumab, for the treatment of autoimmune diseases. FEBS Lett. 2011;585(23):3699–709.PubMedCrossRef
58.
59.
go back to reference Jones SA, et al. IL-6 transsignaling: the in vivo consequences. J Interferon Cytokine Res. 2005;25(5):241–53.PubMedCrossRef Jones SA, et al. IL-6 transsignaling: the in vivo consequences. J Interferon Cytokine Res. 2005;25(5):241–53.PubMedCrossRef
60.
go back to reference Kaly L, Rosner I. Tocilizumab: a novel therapy for non-organ-specific autoimmune diseases. Best Pract Res Clin Rheumatol. 2012;26(1):157–65.PubMedCrossRef Kaly L, Rosner I. Tocilizumab: a novel therapy for non-organ-specific autoimmune diseases. Best Pract Res Clin Rheumatol. 2012;26(1):157–65.PubMedCrossRef
61.
go back to reference Commission CNH. Chinese clinical guidance for COVID-19 pneumonia diagnosis and treatment. J Tianjin Univ Tradit Chin Med. 2020;39(02):121–7. Commission CNH. Chinese clinical guidance for COVID-19 pneumonia diagnosis and treatment. J Tianjin Univ Tradit Chin Med. 2020;39(02):121–7.
62.
go back to reference Campochiaro C, et al. efficacy and safety of tocilizumab in severe COVID-19 patients: a single-centre retrospective cohort study. Eur J Intern Med. 2020;76:43–9.PubMedPubMedCentralCrossRef Campochiaro C, et al. efficacy and safety of tocilizumab in severe COVID-19 patients: a single-centre retrospective cohort study. Eur J Intern Med. 2020;76:43–9.PubMedPubMedCentralCrossRef
63.
go back to reference Price CC, et al. Tocilizumab treatment for cytokine release syndrome in hospitalized patients with coronavirus disease 2019: survival and clinical outcomes. Chest. 2020;158(4):1397–408.PubMedCrossRef Price CC, et al. Tocilizumab treatment for cytokine release syndrome in hospitalized patients with coronavirus disease 2019: survival and clinical outcomes. Chest. 2020;158(4):1397–408.PubMedCrossRef
65.
go back to reference Toniati P, et al. tocilizumab for the treatment of severe COVID-19 pneumonia with hyperinflammatory syndrome and acute respiratory failure: a single center study of 100 patients in Brescia, Italy. Autoimmun Rev. 2020;19:102568.PubMedPubMedCentralCrossRef Toniati P, et al. tocilizumab for the treatment of severe COVID-19 pneumonia with hyperinflammatory syndrome and acute respiratory failure: a single center study of 100 patients in Brescia, Italy. Autoimmun Rev. 2020;19:102568.PubMedPubMedCentralCrossRef
66.
go back to reference Canziani LM, et al. Interleukin-6 receptor blocking with intravenous tocilizumab in COVID-19 severe acute respiratory distress syndrome: a retrospective case-control survival analysis of 128 patients. J Autoimmun. 2020;114:102511.PubMedPubMedCentralCrossRef Canziani LM, et al. Interleukin-6 receptor blocking with intravenous tocilizumab in COVID-19 severe acute respiratory distress syndrome: a retrospective case-control survival analysis of 128 patients. J Autoimmun. 2020;114:102511.PubMedPubMedCentralCrossRef
67.
go back to reference Raimondo MG, et al. Profile of sarilumab and its potential in the treatment of rheumatoid arthritis. Drug Des Dev Ther. 2017;11:1593.CrossRef Raimondo MG, et al. Profile of sarilumab and its potential in the treatment of rheumatoid arthritis. Drug Des Dev Ther. 2017;11:1593.CrossRef
68.
go back to reference Bae S-C, Lee Y. Comparative efficacy and tolerability of sarilumab 150 and 200 mg in patients with active rheumatoid arthritis. Z Rheumatol. 2018;77(5):421–8.PubMedCrossRef Bae S-C, Lee Y. Comparative efficacy and tolerability of sarilumab 150 and 200 mg in patients with active rheumatoid arthritis. Z Rheumatol. 2018;77(5):421–8.PubMedCrossRef
69.
go back to reference Benucci M, et al. COVID-19 pneumonia treated with Sarilumab: a clinical series of eight patients. J Med Virol. 2020;92(11):2368–70.PubMedCrossRef Benucci M, et al. COVID-19 pneumonia treated with Sarilumab: a clinical series of eight patients. J Med Virol. 2020;92(11):2368–70.PubMedCrossRef
70.
go back to reference Wu K-C, Lin C-J. The regulation of drug-metabolizing enzymes and membrane transporters by inflammation: evidences in inflammatory diseases and age-related disorders. J Food Drug Anal. 2019;27(1):48–59.PubMedCrossRef Wu K-C, Lin C-J. The regulation of drug-metabolizing enzymes and membrane transporters by inflammation: evidences in inflammatory diseases and age-related disorders. J Food Drug Anal. 2019;27(1):48–59.PubMedCrossRef
71.
go back to reference Lee EBJI. A review of sarilumab for the treatment of rheumatoid arthritis. Immunotherapy. 2018;10(1):57–65.PubMedCrossRef Lee EBJI. A review of sarilumab for the treatment of rheumatoid arthritis. Immunotherapy. 2018;10(1):57–65.PubMedCrossRef
73.
go back to reference Lescure F-X, et al. Sarilumab in patients admitted to hospital with severe or critical COVID-19: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med. 2021;9:522–32.PubMedPubMedCentralCrossRef Lescure F-X, et al. Sarilumab in patients admitted to hospital with severe or critical COVID-19: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med. 2021;9:522–32.PubMedPubMedCentralCrossRef
74.
go back to reference Khiali S, Rezagholizadeh A, Entezari-Maleki T. A comprehensive review on sarilumab in COVID-19. Expert Opin Biol Ther. 2020;21:615–26.PubMedCrossRef Khiali S, Rezagholizadeh A, Entezari-Maleki T. A comprehensive review on sarilumab in COVID-19. Expert Opin Biol Ther. 2020;21:615–26.PubMedCrossRef
75.
go back to reference Deisseroth A, et al. FDA approval: siltuximab for the treatment of patients with multicentric Castleman disease. Clin Cancer Res. 2015;21(5):950–4.PubMedCrossRef Deisseroth A, et al. FDA approval: siltuximab for the treatment of patients with multicentric Castleman disease. Clin Cancer Res. 2015;21(5):950–4.PubMedCrossRef
76.
go back to reference Davis CC, Shah KS, Lechowicz MJ. Clinical development of siltuximab. Curr Oncol Rep. 2015;17(7):29.PubMedCrossRef Davis CC, Shah KS, Lechowicz MJ. Clinical development of siltuximab. Curr Oncol Rep. 2015;17(7):29.PubMedCrossRef
77.
go back to reference Villaescusa L, Zaragozá F, Gayo-Abeleira I, Zaragozá C. A New Approach to the Management of COVID-19. Antagonists of IL-6: Siltuximab. Adv Ther. 2022;24:1–23. Villaescusa L, Zaragozá F, Gayo-Abeleira I, Zaragozá C. A New Approach to the Management of COVID-19. Antagonists of IL-6: Siltuximab. Adv Ther. 2022;24:1–23.
78.
go back to reference Rose-John S. Interleukin-6 signalling in health and disease. F1000Research. 2020;9:1013.CrossRef Rose-John S. Interleukin-6 signalling in health and disease. F1000Research. 2020;9:1013.CrossRef
79.
go back to reference Honore PM, et al. Inhibiting IL-6 in COVID-19: we are not sure. Crit Care. 2020;24(1):1–3.CrossRef Honore PM, et al. Inhibiting IL-6 in COVID-19: we are not sure. Crit Care. 2020;24(1):1–3.CrossRef
80.
go back to reference Barkhausen T, et al. Selective blockade of interleukin-6 trans-signaling improves survival in a murine polymicrobial sepsis model. Read Online Crit Care Med Soc Crit Care Med. 2011;39(6):1407–13. Barkhausen T, et al. Selective blockade of interleukin-6 trans-signaling improves survival in a murine polymicrobial sepsis model. Read Online Crit Care Med Soc Crit Care Med. 2011;39(6):1407–13.
81.
go back to reference Palau V, Riera M, Soler MJ. ADAM17 inhibition may exert a protective effect on COVID-19. Nephrol Dial Transplant. 2020;35(6):1071–2.PubMedCrossRef Palau V, Riera M, Soler MJ. ADAM17 inhibition may exert a protective effect on COVID-19. Nephrol Dial Transplant. 2020;35(6):1071–2.PubMedCrossRef
82.
83.
go back to reference Salvarani C, et al. Effect of tocilizumab vs standard care on clinical worsening in patients hospitalized with COVID-19 pneumonia: a randomized clinical trial. JAMA Intern Med. 2021;181(1):24–31.PubMedCrossRef Salvarani C, et al. Effect of tocilizumab vs standard care on clinical worsening in patients hospitalized with COVID-19 pneumonia: a randomized clinical trial. JAMA Intern Med. 2021;181(1):24–31.PubMedCrossRef
84.
go back to reference Stone JH, et al. efficacy of tocilizumab in patients hospitalized with Covid-19. N Engl J Med. 2020;383(24):2333–44.PubMedCrossRef Stone JH, et al. efficacy of tocilizumab in patients hospitalized with Covid-19. N Engl J Med. 2020;383(24):2333–44.PubMedCrossRef
85.
go back to reference Hermine O, et al. effect of tocilizumab vs usual care in adults hospitalized with COVID-19 and moderate or severe pneumonia: a randomized clinical trial. JAMA Intern Med. 2021;181(1):32–40.PubMedCrossRef Hermine O, et al. effect of tocilizumab vs usual care in adults hospitalized with COVID-19 and moderate or severe pneumonia: a randomized clinical trial. JAMA Intern Med. 2021;181(1):32–40.PubMedCrossRef
87.
go back to reference Cavalli G, et al. Interleukin 1α: a comprehensive review on the role of IL-1α in the pathogenesis and targeted treatment of autoimmune and inflammatory diseases. Autoimmun Rev. 2021;20:102763.PubMedCrossRef Cavalli G, et al. Interleukin 1α: a comprehensive review on the role of IL-1α in the pathogenesis and targeted treatment of autoimmune and inflammatory diseases. Autoimmun Rev. 2021;20:102763.PubMedCrossRef
88.
go back to reference Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009;27:519–50.PubMedCrossRef Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009;27:519–50.PubMedCrossRef
89.
go back to reference Burzynski LC, et al. The coagulation and immune systems are directly linked through the activation of interleukin-1α by thrombin. Immunity. 2019;50(4):1033–42.PubMedPubMedCentralCrossRef Burzynski LC, et al. The coagulation and immune systems are directly linked through the activation of interleukin-1α by thrombin. Immunity. 2019;50(4):1033–42.PubMedPubMedCentralCrossRef
90.
go back to reference Kim B, et al. The interleukin-1α precursor is biologically active and is likely a key alarmin in the IL-1 family of cytokines. Front Immunol. 2013;4:391.PubMedPubMedCentralCrossRef Kim B, et al. The interleukin-1α precursor is biologically active and is likely a key alarmin in the IL-1 family of cytokines. Front Immunol. 2013;4:391.PubMedPubMedCentralCrossRef
91.
go back to reference Zheng Y, et al. Intracellular interleukin-1 receptor 2 binding prevents cleavage and activity of interleukin-1α, controlling necrosis-induced sterile inflammation. Immunity. 2013;38(2):285–95.PubMedPubMedCentralCrossRef Zheng Y, et al. Intracellular interleukin-1 receptor 2 binding prevents cleavage and activity of interleukin-1α, controlling necrosis-induced sterile inflammation. Immunity. 2013;38(2):285–95.PubMedPubMedCentralCrossRef
92.
go back to reference Wang X, et al. RNA viruses promote activation of the NLRP3 inflammasome through a RIP1-RIP3-DRP1 signaling pathway. Nat Immunol. 2014;15(12):1126–33.PubMedCrossRef Wang X, et al. RNA viruses promote activation of the NLRP3 inflammasome through a RIP1-RIP3-DRP1 signaling pathway. Nat Immunol. 2014;15(12):1126–33.PubMedCrossRef
93.
94.
go back to reference Bester J, Matshailwe C, Pretorius EJC. Simultaneous presence of hypercoagulation and increased clot lysis time due to IL-1β, IL-6 and IL-8. Cytokine. 2018;110:237–42.PubMedCrossRef Bester J, Matshailwe C, Pretorius EJC. Simultaneous presence of hypercoagulation and increased clot lysis time due to IL-1β, IL-6 and IL-8. Cytokine. 2018;110:237–42.PubMedCrossRef
95.
go back to reference Biondi-Zoccai G, et al. Atherothrombosis prevention and treatment with anti-interleukin-1 agents. Curr Atheroscler Rep. 2020;22(1):1–7. Biondi-Zoccai G, et al. Atherothrombosis prevention and treatment with anti-interleukin-1 agents. Curr Atheroscler Rep. 2020;22(1):1–7.
96.
go back to reference Park WY, et al. Cytokine balance in the lungs of patients with acute respiratory distress syndrome. Am J Respir Crit Care Med. 2001;164(10):1896–903.PubMedCrossRef Park WY, et al. Cytokine balance in the lungs of patients with acute respiratory distress syndrome. Am J Respir Crit Care Med. 2001;164(10):1896–903.PubMedCrossRef
97.
go back to reference Ramírez J, Cañete JD. Anakinra for the treatment of rheumatoid arthritis: a safety evaluation. Expert Opin Drug Saf. 2018;17(7):727–32.PubMedCrossRef Ramírez J, Cañete JD. Anakinra for the treatment of rheumatoid arthritis: a safety evaluation. Expert Opin Drug Saf. 2018;17(7):727–32.PubMedCrossRef
98.
go back to reference Cavalli G, et al. Interleukin-1 blockade with high-dose anakinra in patients with COVID-19, acute respiratory distress syndrome, and hyperinflammation: a retrospective cohort study. Lancet Rheumatol. 2020;2(6):e325–31.PubMedPubMedCentralCrossRef Cavalli G, et al. Interleukin-1 blockade with high-dose anakinra in patients with COVID-19, acute respiratory distress syndrome, and hyperinflammation: a retrospective cohort study. Lancet Rheumatol. 2020;2(6):e325–31.PubMedPubMedCentralCrossRef
99.
go back to reference Balkhair A, et al. Anakinra in hospitalized patients with severe COVID-19 pneumonia requiring oxygen therapy: results of a prospective, open-label, interventional study. Int J Infect Dis. 2021;103:288–96.PubMedCrossRef Balkhair A, et al. Anakinra in hospitalized patients with severe COVID-19 pneumonia requiring oxygen therapy: results of a prospective, open-label, interventional study. Int J Infect Dis. 2021;103:288–96.PubMedCrossRef
101.
go back to reference Rondeau J-M, et al. The molecular mode of action and species specificity of canakinumab, a human monoclonal antibody neutralizing IL-1β. London: Taylor & Francis; 2015.CrossRef Rondeau J-M, et al. The molecular mode of action and species specificity of canakinumab, a human monoclonal antibody neutralizing IL-1β. London: Taylor & Francis; 2015.CrossRef
105.
go back to reference Generali D, et al. Canakinumab as treatment for COVID-19-related pneumonia: a prospective case-control study. Int J Infect Dis. 2021;104:433–40.PubMedCrossRef Generali D, et al. Canakinumab as treatment for COVID-19-related pneumonia: a prospective case-control study. Int J Infect Dis. 2021;104:433–40.PubMedCrossRef
107.
go back to reference Cavalli G, et al. Interleukin-1 and interleukin-6 inhibition compared with standard management in patients with COVID-19 and hyperinflammation: a cohort study. Lancet Rheumatol. 2021;3(4):e253–61.PubMedPubMedCentralCrossRef Cavalli G, et al. Interleukin-1 and interleukin-6 inhibition compared with standard management in patients with COVID-19 and hyperinflammation: a cohort study. Lancet Rheumatol. 2021;3(4):e253–61.PubMedPubMedCentralCrossRef
108.
go back to reference Dinarello CA. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood J Am Soc Hematol. 2011;117(14):3720–32. Dinarello CA. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood J Am Soc Hematol. 2011;117(14):3720–32.
109.
110.
go back to reference Jin W, Dong C. IL-17 cytokines in immunity and inflammation. Emerg Microbes Infect. 2013;2(1):1–5.CrossRef Jin W, Dong C. IL-17 cytokines in immunity and inflammation. Emerg Microbes Infect. 2013;2(1):1–5.CrossRef
111.
go back to reference Raucci F, et al. Interleukin-17A (IL-17A), a key molecule of innate and adaptive immunity, and its potential involvement in COVID-19-related thrombotic and vascular mechanisms. Autoimmun Rev. 2020;19(7):102572.PubMedPubMedCentralCrossRef Raucci F, et al. Interleukin-17A (IL-17A), a key molecule of innate and adaptive immunity, and its potential involvement in COVID-19-related thrombotic and vascular mechanisms. Autoimmun Rev. 2020;19(7):102572.PubMedPubMedCentralCrossRef
112.
go back to reference Liu WJ, et al. T-cell immunity of SARS-CoV: implications for vaccine development against MERS-CoV. Antivir Res. 2017;137:82–92.PubMedCrossRef Liu WJ, et al. T-cell immunity of SARS-CoV: implications for vaccine development against MERS-CoV. Antivir Res. 2017;137:82–92.PubMedCrossRef
114.
go back to reference Rokni M, et al. Cytokines and COVID-19: friends or foes? Hum Vaccines Immunotherap. 2020;16(10):2363–5.CrossRef Rokni M, et al. Cytokines and COVID-19: friends or foes? Hum Vaccines Immunotherap. 2020;16(10):2363–5.CrossRef
115.
116.
go back to reference Hou W, et al. Interleukin-6 (IL-6) and IL-17 synergistically promote viral persistence by inhibiting cellular apoptosis and cytotoxic T cell function. J Virol. 2014;88(15):8479–89.PubMedPubMedCentralCrossRef Hou W, et al. Interleukin-6 (IL-6) and IL-17 synergistically promote viral persistence by inhibiting cellular apoptosis and cytotoxic T cell function. J Virol. 2014;88(15):8479–89.PubMedPubMedCentralCrossRef
117.
go back to reference Shibabaw T. Inflammatory cytokine: IL-17A signaling pathway in patients present with COVID-19 and current treatment strategy. J Inflamm Res. 2020;13:673.PubMedPubMedCentralCrossRef Shibabaw T. Inflammatory cytokine: IL-17A signaling pathway in patients present with COVID-19 and current treatment strategy. J Inflamm Res. 2020;13:673.PubMedPubMedCentralCrossRef
119.
go back to reference Bulat V, et al. Potential role of IL-17 blocking agents in the treatment of severe COVID-19? Br J Clin Pharmacol. 2020;87:1578–81.PubMedCrossRef Bulat V, et al. Potential role of IL-17 blocking agents in the treatment of severe COVID-19? Br J Clin Pharmacol. 2020;87:1578–81.PubMedCrossRef
120.
go back to reference Sahu U, et al. Interleukin-17: a multifaceted cytokine in viral infections. J Cell Physiol. 2021;236:8000–19.PubMedCrossRef Sahu U, et al. Interleukin-17: a multifaceted cytokine in viral infections. J Cell Physiol. 2021;236:8000–19.PubMedCrossRef
121.
go back to reference Wu D, Yang XO. TH17 responses in cytokine storm of COVID-19: an emerging target of JAK2 inhibitor Fedratinib. J Microbiol Immunol Infect. 2020;53(3):368–70.PubMedPubMedCentralCrossRef Wu D, Yang XO. TH17 responses in cytokine storm of COVID-19: an emerging target of JAK2 inhibitor Fedratinib. J Microbiol Immunol Infect. 2020;53(3):368–70.PubMedPubMedCentralCrossRef
123.
go back to reference Kalliolias GD, Ivashkiv LB. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatol. 2016;12(1):49–62.PubMedCrossRef Kalliolias GD, Ivashkiv LB. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatol. 2016;12(1):49–62.PubMedCrossRef
124.
go back to reference Bradley J. TNF-mediated inflammatory disease. J Pathol J Pathol Soc G B Irel. 2008;214(2):149–60. Bradley J. TNF-mediated inflammatory disease. J Pathol J Pathol Soc G B Irel. 2008;214(2):149–60.
126.
go back to reference Patel S, Wadhwa M. Therapeutic use of specific tumour necrosis factor inhibitors in inflammatory diseases including COVID-19. Biomed Pharmacother. 2021;140:111785.PubMedPubMedCentralCrossRef Patel S, Wadhwa M. Therapeutic use of specific tumour necrosis factor inhibitors in inflammatory diseases including COVID-19. Biomed Pharmacother. 2021;140:111785.PubMedPubMedCentralCrossRef
127.
go back to reference Leija-Martínez JJ, et al. IL-17A and TNF-α as potential biomarkers for acute respiratory distress syndrome and mortality in patients with obesity and COVID-19. Med Hypotheses. 2020;144:109935.PubMedPubMedCentralCrossRef Leija-Martínez JJ, et al. IL-17A and TNF-α as potential biomarkers for acute respiratory distress syndrome and mortality in patients with obesity and COVID-19. Med Hypotheses. 2020;144:109935.PubMedPubMedCentralCrossRef
Metadata
Title
COVID-19 infection: an overview on cytokine storm and related interventions
Authors
Soheila Montazersaheb
Seyed Mahdi Hosseiniyan Khatibi
Mohammad Saeid Hejazi
Vahideh Tarhriz
Afsaneh Farjami
Faramarz Ghasemian Sorbeni
Raheleh Farahzadi
Tohid Ghasemnejad
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Virology Journal / Issue 1/2022
Electronic ISSN: 1743-422X
DOI
https://doi.org/10.1186/s12985-022-01814-1

Other articles of this Issue 1/2022

Virology Journal 1/2022 Go to the issue