The Role of Cytokines including Interleukin-6 in COVID-19 induced Pneumonia and Macrophage Activation Syndrome-Like Disease

Dennis McGonagle, Kassem Sharif, Anthony O'Regan, Charlie Bridgewood, Dennis McGonagle, Kassem Sharif, Anthony O'Regan, Charlie Bridgewood

Abstract

Severe COVID-19 associated pneumonia patients may exhibit features of systemic hyper-inflammation designated under the umbrella term of macrophage activation syndrome (MAS) or cytokine storm, also known as secondary haemophagocytic lymphohistocytosis (sHLH). This is distinct from HLH associated with immunodeficiency states termed primary HLH -with radically different therapy strategies in both situations. COVID-19 infection with MAS typically occurs in subjects with adult respiratory distress syndrome (ARDS) and historically, non-survival in ARDS was linked to sustained IL-6 and IL-1 elevation. We provide a model for the classification of MAS to stratify the MAS-like presentation in COVID-19 pneumonia and explore the complexities of discerning ARDS from MAS. We discuss the potential impact of timing of anti-cytokine therapy on viral clearance and the impact of such therapy on intra-pulmonary macrophage activation and emergent pulmonary vascular disease.

Crown Copyright © 2020. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
Classical MAS Organ Pattern versus COVID-19 Related Picture. Panel A). The MAS/sHLH picture is associated with viral and other infections that does not have a special tropism for pulmonary tissues. The hyper-activation of T-cells is associated with lymphoid organ hyperplasia with hepatosplenomegaly and adenopathy. The resulting excessive reticuloendothelial system activity manifests as bone marrow haemophagocytosis and deranged liver function tests, deranged lipid profiles as a result of liver Kupffer cell dysfunction. Systemically circulating activated macrophages contribute to disseminated intravascular coagulation (DIC). A similar picture can develop in primary HLH that represents a rare group of immunodeficiency, linked to impaired cytolytic activity due to perforin pathway loss of function, with expansion of immune cells and the lymphoid organs that house them. In primary HLH, cytokine antagonism used with anti-microbial therapy may temporarily help prior to definitive bone marrow transplantation. Pulmonary involvement may be a feature of both pHLH and sHLH. Panel B). There is evidence for MAS/sHLH emerging in the COVID-19 setting that is supported by the abnormal laboratory parameters without reporting of the classical organomegaly pattern described in Fig. 1A. Hyperactivation and over-zealous immune activity appears to be more confined to the lung parenchyma and immediately adjacent bronchial alveolar lymphoid tissue and is associated with ARDS development. Pulmonary haemophagocytosis has has been occasionally reported in human coronavirus infection but not yet been reported in COVID-19 associated pneumonia [56]. .It is proposed that severe widespread alveolar and interstitial inflammation extends to the closely juxtaposed pulmonary vasculature. The normal circulatory fibrinogen levels and regional fibrinolysis with elevated D-dimer formation seen in early COVID-19 pneumonia is not a feature of typical acute onset MAS/HLH. This MAS-like intra-pulmonary inflammation might influence a propensity towards severe local vascular dysfunction including micro-thrombosis and haemorrhage resulting in a lung centric pulmonary intravascular coagulopathy (PIC) presentation rather than a DIC presentation
Fig. 2
Fig. 2
Hyper-cytokinaemic Overlaps Between ARDS and MAS. Elevated cytokine levels or hypercytokinaemia are associated with the current COVID-19 infection [20] and independently found in the MAS spectrum and many other settings including sepsis [17]. Cytokines including IL-6 may be substantially elevated in isolated ARDS. The magnitude of cytokine level changes may not be reliable in the differentiation from other cause of elevated cytokine levels including severe infection or tissue destruction and dependable cut-off values do not exist. Superimposed second viral or bacterial infections in either setting may further drive systemic cytokine responses. Therefore, phase 3 studies based on elevated IL-6 levels may encompass a group of ARDS cases without the bone fide MAS phenotype [57].
Fig. 3
Fig. 3
Relative Innate and Adaptive Immune Mechanisms in HLH Spectrum Pathology. Immunodeficiency State (Primary HLH) Vs Immunocompetency State (MAS/sHLH. COVID-19 Immunodeficiency State Vs Immunocompetency State. The HLH responses can occur in the setting of both immunodeficiency states and in immunocompetence settings and following adaptive immune system engineering (Panel A and B in blue). In humans with defects in NK and CD8+ T-cells, the inability to kill virally infected cells results in primary HLH with hyper-cytokinaemia that can't be cured with anti-cytokine strategies as genetic defect typically in perforin pathway machinery makes pathogen elimination impossible [58] (Panel A). In CAR-T cell therapy hyper proliferating engineered T-cells may drive MAS/sHLH that only lasts for the duration of the presence of detectable tumour antigen (Panel B). Similar mechanisms may occur in sJIA or autoimmune diseases, but this remains to be fully defined. This model is based on the immunological disease continuum modified for gain or loss of function in innate or adaptive immunity (Reference 43). When COVID-19 hyper-inflammatory or cytokine storm reactions are viewed through the lens of primary and secondary HLH, it is noteworthy that virally induced immunosuppression may play a key role (Panel C). Coronavirus family members including COVID-19 pneumonia is associated with robust interferon suppression, blood lymphopenia including NK cell abnormalities. Such scenarios likely drive macrophage infiltration and the “second wave” of non-type-1 interferon pathway cytokines including IL-6, IL-1, IL-18, INFγ, GM-CSF and others that lead to a blood hyper-cytokinaemic picture (Panel B). It remains to be seen how many cases of COVID-19 related fatalities have mutations in genes associated with the primary HLH disorders and thus closely resemble primary HLH (Panel A). Based on persistent viral shedding from the respiratory tract during CORVID-19 related disease, then an exaggerated immune response including IFN driven and T-cell driven responses that clears the virus but induces pulmonary immune system collateral damage and ARDS offers an explanation for the more typical MAS/sHLH phenotype. Viral clearance in the face of marked hyper-inflammation would be expected in the face of brisk anti-viral antigen responses.
Fig. 4
Fig. 4
IL-6 in Viral Pneumoia and Potential Role in Type II Pneumocyte COVID-19 infection. Panel A). Some experimental models of viral pulmonary infection triggering ARDS show that IL-6 may have a protective role with its blockade increasing mortality including reduction and phagocytosis and increased fibrosis depending on model. Panel B). Bacterial, rather than viral infections, originating outside the lung that also trigger ARDS but in this setting IL-6 blockade may be beneficial. The latter is more akin to classical MAS/sHLH where pulmonary pathology, when present, may be due to exogenous factors. However, the fact that IL-6 is detrimental in murine virally induced ARDS cannot be extrapolated to the novel COVID-19 virus infection that shows a tropism for type 2 pneumocytes (Panel C). The COVID-19 suppression of first line interferon responses and abrogation of T-cell responses suggests a role for type 2 pneunocyte gp130 receptor expression and IL-6 in pulmonary pathology [59]. Also in experimental models, IL-6 may either suppress or facilitate viral replication depending on the virus [49], so data on COVID-19 related disease is eagerly awaited.

References

    1. Cao B., Wang Y., Wen D., Liu W., Wang J., Fan G. A trial of Lopinavir–Ritonavir in adults hospitalized with severe Covid-19. New Engl J Med. 2020
    1. Mehta P., McAuley D.F., Brown M., Sanchez E., Tattersall R.S., Manson J.J. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet. 2020
    1. Schulert G.S., Yasin S., Carey B., Chalk C., Do T., Schapiro A.H. Systemic Juvenile Idiopathic arthritis-associated lung disease: characterization and risk factors. Arthritis Rheumatol (Hoboken, NJ) 2019;71:1943–1954.
    1. Martini A.J.A.R. Vol. 12. 2012. Systemic juvenile idiopathic arthritis; pp. 56–59.
    1. Behrens E.M.J.A.R. Vol. 7. 2008. Macrophage activation syndrome in rheumatic disease: what is the role of the antigen presenting cell? pp. 305–308.
    1. De Benedetti F., Brunner H.I., Ruperto N., Kenwright A., Wright S., Calvo I. Randomized trial of tocilizumab in systemic juvenile idiopathic arthritis. N Engl J Med. 2012;367:2385–2395.
    1. Feist E., Quartier P., Fautrel B., Schneider R., Sfriso P., Efthimiou P. Efficacy and safety of canakinumab in patients with Still's disease: exposure-response analysis of pooled systemic juvenile idiopathic arthritis data by age groups. Clin Exp Rheumatol. 2018;36:668–675.
    1. Favalli E.G., Ingegnoli F., De Lucia O., Cincinelli G., Cimaz R., Caporali R.J.A.R. COVID-19 infection and rheumatoid arthritis: Faraway, so close! Autoimmun Rev. 2020;102523
    1. Crayne C.B., Albeituni S., Nichols K.E., Cron R.Q. The immunology of macrophage activation syndrome. Front Immunol. 2019;10 119–119.
    1. Martinez F.O., Sica A., Mantovani A., Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–461.
    1. Carter S.J., Tattersall R.S., Ramanan A.V. Macrophage activation syndrome in adults: recent advances in pathophysiology, diagnosis and treatment. Rheumatology (Oxford) 2019;58:5–17.
    1. Ravelli A., De Benedetti F., Viola S., Martini A. Macrophage activation syndrome in systemic juvenile rheumatoid arthritis successfully treated with cyclosporine. J Pediatr. 1996;128:275–278.
    1. Davi S., Consolaro A., Guseinova D., Pistorio A., Ruperto N., Martini A. An international consensus survey of diagnostic criteria for macrophage activation syndrome in systemic juvenile idiopathic arthritis. J Rheumatol. 2011;38:764–768.
    1. de Benedetti F., Massa M., Robbioni P., Ravelli A., Burgio G.R., Martini A. Correlation of serum interleukin-6 levels with joint involvement and thrombocytosis in systemic juvenile rheumatoid arthritis. Arthritis Rheum. 1991;34:1158–1163.
    1. Poddighe D., Cavagna L., Brazzelli V., Bruni P., Marseglia G.L.J.A.R. Vol. 13. 2014. A hyper-ferritinemia syndrome evolving in recurrent macrophage activation syndrome, as an onset of amyopathic juvenile dermatomyositis: a challenging clinical case in light of the current diagnostic criteria; pp. 1142–1148.
    1. Colafrancesco S., Priori R., Alessandri C., Astorri E., Perricone C., Blank M. Vol. 60. 2014. sCD163 in AOSD: a biomarker for macrophage activation related to hyperferritinemia; pp. 177–183.
    1. Rosário C., Zandman-Goddard G., Meyron-Holtz E.G., D'Cruz D.P., Shoenfeld Y.J.B.M. Vol. 11. 2013. The hyperferritinemic syndrome: macrophage activation syndrome, Still's disease, septic shock and catastrophic antiphospholipid syndrome; p. 185.
    1. Sharif K., Vieira Borba V., Zandman-Goddard G., Shoenfeld Y.J.C., E. Immunology . Vol. 191. 2018. Eppur Si Muove: ferritin is essential in modulating inflammation; pp. 149–150.
    1. Bracaglia C., Prencipe G., De Benedetti F. Macrophage activation syndrome: different mechanisms leading to a one clinical syndrome. Pediatr Rheumatol Online J. 2017;15 5–5.
    1. Wu C., Chen X., Cai Y., Xia J.A., Zhou X., Xu S. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 Pneumonia in Wuhan, China. JAMA Intern Med. 2020
    1. Wan S., Yi Q., Fan S., Lv J., Zhang X., Guo L. Characteristics of lymphocyte subsets and cytokines in peripheral blood of 123 hospitalized patients with 2019 novel coronavirus pneumonia (NCP) medRxiv. 2020 2020.02.10.20021832.
    1. Vastert S.J., Jamilloux Y., Quartier P., Ohlman S., Osterling Koskinen L., Kullenberg T. Anakinra in children and adults with Still's disease. Rheumatology (Oxford) 2019;58 vi9-vi22.
    1. Quartier P., Allantaz F., Cimaz R., Pillet P., Messiaen C., Bardin C. A multicentre, randomised, double-blind, placebo-controlled trial with the interleukin-1 receptor antagonist anakinra in patients with systemic-onset juvenile idiopathic arthritis. ANAJIS trialAnn Rheum Dis. 2011;70:747–754.
    1. Yokota S., Imagawa T., Mori M., Miyamae T., Aihara Y., Takei S. Efficacy and safety of tocilizumab in patients with systemic-onset juvenile idiopathic arthritis: a randomised, double-blind, placebo-controlled, withdrawal phase III trial. Lancet (London, England) 2008;371:998–1006.
    1. Yokota S., Itoh Y., Morio T., Sumitomo N., Daimaru K., Minota S. Macrophage activation syndrome in patients with systemic juvenile idiopathic arthritis under treatment with Tocilizumab. J Rheumatol. 2015;42:712–722.
    1. Yao X.H., Li T.Y., He Z.C., Ping Y.F., Liu H.W., Yu S.C. A pathological report of three COVID-19 cases by minimally invasive autopsies. Zhonghua Bing Li Xue Za Zhi. 2020;49 E009.
    1. Tisoncik J.R., Korth M.J., Simmons C.P., Farrar J., Martin T.R., Katze M.G. Into the eye of the cytokine storm. Microbiol Mol Biol Rev. 2012;76:16–32.
    1. Zhou F., Yu T., Du R., Fan G., Liu Y., Liu Z. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020
    1. Meduri G.U., Headley S., Kohler G., Stentz F., Tolley E., Umberger R. Vol. 107. 1995. Persistent elevation of inflammatory cytokines predicts a poor outcome in ARDS: plasma IL-1β and IL-6 levels are consistent and efficient predictors of outcome over time; pp. 1062–1073.
    1. Meduri G.U., Kohler G., Headley S., Tolley E., Stentz F., Postlethwaite A.J.C. Vol. 108. 1995. Inflammatory cytokines in the BAL of patients with ARDS: persistent elevation over time predicts poor outcome; pp. 1303–1314.
    1. McGonagle D., McDermott M.F. A proposed classification of the immunological diseases. PLoS Med. 2006;3
    1. Grateau G., Hentgen V., Stojanovic K.S., Jeru I., Amselem S., Steichen O. How should we approach classification of autoinflammatory diseases? Nat Rev Rheumatol. 2013;9:624–629.
    1. Grom A.A. Primary hemophagocytic lymphohistiocytosis and macrophage activation syndrome: the importance of timely clinical differentiation. J Pediatr. 2017;189:19–21. e1.
    1. Brisse E., Matthys P., Wouters C.H. Understanding the spectrum of haemophagocytic lymphohistiocytosis: update on diagnostic challenges and therapeutic options. Br J Haematol. 2016;174:175–187.
    1. Chen G., Wu D., Guo W., Cao Y., Huang D., Wang H. 2020. Clinical and immunologic features in severe and moderate forms of Coronavirus Disease 2019.
    1. Kopecky-Bromberg S.A., Martínez-Sobrido L., Frieman M., Baric R.A., Palese P. Severe acute respiratory syndrome coronavirus open reading frame (ORF) 3b, ORF 6, and nucleocapsid proteins function as interferon antagonists. J Virol. 2007;81:548–557.
    1. Cameron M.J., Kelvin A.A., Leon A.J., Cameron C.M., Ran L., Xu L. Lack of innate interferon responses during SARS coronavirus infection in a vaccination and reinfection ferret model. PLoS One. 2012;7 e45842-e45842.
    1. Falzarano D., de Wit E., Rasmussen A.L., Feldmann F., Okumura A., Scott D.P. Treatment with interferon-alpha2b and ribavirin improves outcome in MERS-CoV-infected rhesus macaques. Nat Med. 2013;19:1313–1317.
    1. Sainz B., Jr., Mossel E.C., Peters C., Garry R.F.J.V. Vol. 329. 2004. Interferon-beta and interferon-gamma synergistically inhibit the replication of severe acute respiratory syndrome-associated coronavirus (SARS-CoV) pp. 11–17.
    1. Zheng M., Gao Y., Wang G., Song G., Liu S., Sun D. 2020. Functional exhaustion of antiviral lymphocytes in COVID-19 patients; pp. 1–3.
    1. Zhang W., Du R.-H., Li B., Zheng X.-S., Yang X.-L., Hu B. Vol. 9. 2020. Molecular and serological investigation of 2019-nCoV infected patients: implication of multiple shedding routes; pp. 386–389.
    1. Chen W., Lan Y., Yuan X., Deng X., Li Y., Cai X. Vol. 9. 2020. Detectable 2019-nCoV viral RNA in blood is a strong indicator for the further clinical severity; pp. 469–473.
    1. Okabayashi T., Kariwa H., Yokota S., Iki S., Indoh T., Yokosawa N. Cytokine regulation in SARS coronavirus infection compared to other respiratory virus infections. J Med Virol. 2006;78:417–424.
    1. Mihara M., Hashizume M., Yoshida H., Suzuki M., Shiina M.J.C.S. Vol. 122. 2012. IL-6/IL-6 receptor system and its role in physiological and pathological conditions; pp. 143–159.
    1. Jones S.A., Richards P.J., Scheller J., Rose-John S.J.J.O.I., C. Research . Vol. 25. 2005. IL-6 transsignaling: the in vivo consequences; pp. 241–253.
    1. Le T.-T.T., Karmouty-Quintana H., Melicoff E., Le T.-T.T., Weng T., Chen N.-Y. Blockade of IL-6 <em>trans</em> Signaling attenuates pulmonary fibrosis. J Immunol. 2014;193:3755–3768.
    1. Scheller J., Ohnesorge N., Rose-John S. Interleukin-6 trans-signalling in chronic inflammation and cancer. Scand J Immunol. 2006;63:321–329.
    1. Garbers C., Rose-John S. Dissecting Interleukin-6 classic- and trans-signaling in inflammation and cancer. Methods Mol Biol (Clifton, NJ) 2018;1725:127–140.
    1. Velazquez-Salinas L., Verdugo-Rodriguez A., Rodriguez L.L., Borca M.V. Vol. 10. 2019. The role of interleukin 6 during viral infections; p. 1057.
    1. Ruan Q., Yang K., Wang W., Jiang L., Song J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020
    1. Baden L.R., Rubin E.J. Covid-19 — the search for effective therapy. New Engl J Med. 2020
    1. Bracaglia C., Caiello I., De Graaf K., D'Ario G., Guilhot F., Ferlin W. Interferon-gamma (IFNy) in macrophage activation syndrome (MAS) associated with systemic juvenile idiopathic arthritis (sJIA). High levels in patients and a role in a murine mas model. Pediatr Rheumatol Online J. 2014;12 O3-O3.
    1. Vallurupalli M., Berliner N. Emapalumab for the treatment of relapsed/refractory hemophagocytic lymphohistiocytosis. Blood. 2019;134:1783–1786.
    1. Dinarello C.A. Targeting interleukin 18 with interleukin 18 binding protein. Ann Rheum Dis. 2000;59(Suppl. 1):i17–i20.
    1. Russell C.D., Millar J.E., Baillie J.K. Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury. Lancet. 2020;395:473–475.
    1. Xu Z., Shi L., Wang Y., Zhang J., Huang L., Zhang C. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020
    1. Registry C.C.T. 2020. A multicenter, randomized controlled trial for the efficacy and safety of tocilizumab in the treatment of new coronavirus pneumonia (COVID-19)
    1. Gupta S., Weitzman S. Primary and secondary hemophagocytic lymphohistiocytosis: clinical features. Pathogen Ther. 2010;6:137–154.
    1. Le T.-T.T., Karmouty-Quintana H., Melicoff E., Le T.-T.T., Weng T., Chen N.-Y. Vol. 193. 2014. Blockade of IL-6 Trans signaling attenuates pulmonary fibrosis; pp. 3755–3768.
    1. Dosch S.F., Mahajan S.D., Collins A.R.J.V. Vol. 142. 2009. SARS coronavirus spike protein-induced innate immune response occurs via activation of the NF-κB pathway in human monocyte macrophages in vitro; pp. 19–27.
    1. Wang W., Ye L., Ye L., Li B., Gao B., Zeng Y. Vol. 128. 2007. Up-regulation of IL-6 and TNF-α induced by SARS-coronavirus spike protein in murine macrophages via NF-κB pathway; pp. 1–8.
    1. Channappanavar R., Fehr A.R., Vijay R., Mack M., Zhao J., Meyerholz D.K. Vol. 19. 2016. Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice; pp. 181–193.
    1. Dienz O., Rud J.G., Eaton S.M., Lanthier P.A., Burg E., Drew A. Vol. 5. 2012. Essential role of IL-6 in protection against H1N1 influenza virus by promoting neutrophil survival in the lung; pp. 258–266.
    1. Yang M.-L., Wang C.-T., Yang S.-J., Leu C.-H., Chen S.-H., Wu C.-L. Vol. 7. 2017. IL-6 ameliorates acute lung injury in influenza virus infection; pp. 1–11.
    1. Murphy E.A., Davis J.M., Brown A.S., Carmichael M.D., Ghaffar A., Mayer E.P.J.J. Effect of IL-6 deficiency on susceptibility to HSV-1 respiratory infection and intrinsic macrophage antiviral resistance. 2008;28:589–596.
    1. Bhargava R., Janssen W., Altmann C., Andrés-Hernando A., Okamura K., Vandivier R.W. Vol. 8. 2013. Intratracheal IL-6 protects against lung inflammation in direct, but not indirect, causes of acute lung injury in mice.
    1. Voiriot G., Razazi K., Amsellem V., Van Nhieu J.T., Abid S., Adnot S. Interleukin-6 displays lung anti-inflammatory properties and exerts protective hemodynamic effects in a double-hit murine acute lung injury. 2017;18:64.
    1. Ladel C.H., Blum C., Dreher A., Reifenberg K., Kopf M., S.J.I. Kaufmann and immunity . Vol. 65. 1997. Lethal tuberculosis in interleukin-6-deficient mutant mice; pp. 4843–4849.
    1. van der Poll T., Keogh C.V., Guirao X., Buurman W.A., Kopf M., Lowry S.F.J.J. Vol. 176. 1997. Interleukin-6 gene-deficient mice show impaired defense against pneumococcal pneumonia; pp. 439–444.
    1. Cenci E., Mencacci A., Casagrande A., Mosci P., Bistoni F., Romani L.J.T.J. Vol. 184. 2001. Impaired antifungal effector activity but not inflammatory cell recruitment in interleukin-6-deficient mice with invasive pulmonary aspergillosis; pp. 610–617.
    1. Kobayashi T., Tanaka K., Fujita T., Umezawa H., Amano H., Yoshioka K. Vol. 16. 2015. Bidirectional role of IL-6 signal in pathogenesis of lung fibrosis; p. 99.
    1. Ibrahim Y.F., Moussa R.A., Bayoumi A.M., Ahmed A.-S.F.J.I. Vol. 28. 2020. Tocilizumab attenuates acute lung and kidney injuries and improves survival in a rat model of sepsis via down-regulation of NF-κB/JNK: a possible role of P-glycoprotein; pp. 215–230.

Source: PubMed

3
Abonneren