18
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      COVID-19: more than a cytokine storm

      letter

      Read this article at

          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background In these first months of coronavirus disease-19 (COVID-19) pandemic, a mainstream pathogenetic hypothesis, likely stemming from early clinico-therapeutic observations, has been suggesting that severe COVID-19 may represent a sort of hyperimmune disorder, akin, in particular, to secondary hemophagocytic lymphohistiocytosis (sHLH) and macrophage activation syndrome (MAS) [1–3]. In this view, COVID-19-associated cytokine storm, with elevated plasma levels of IL-6, IL-1, and TNF-α, as well as ferritin and other inflammatory biomarkers, has been considered as a typical sign of sHLH/MAS, but the other “key feature” of COVID-19—the progressive lymphopenia with T cell exhaustion [4–6]—has largely been neglected. Of note, both CD4+ and CD8+ T lymphocytes were found to be remarkably decreased in severe cases (median 177.5 and 89.0 × 106/L, respectively), when compared to moderate ones (median 381.5 and 254.0 × 106/L, respectively), thus suggesting T cell lymphopenia may constitute a potential prognostic marker to be included in the monitoring of COVID-19 patients [4]. Frequencies of IFN-γ-producing CD4+ T cells (i.e., cytotoxic Th1 subset) tended to be lower in severe than in moderate illness (median 14.1% versus 22.8%, respectively), possibly indicating a progressive skew of the Th1/Th2 balance toward a tolerogenic response [4]. In addition, the percentages of both memory Th cells and regulatory T cells were found to decrease in severe cases [5]. Nonetheless, in patients with severe systemic hyper-inflammatory diseases driven by other viral infections, hemophagocytic syndrome can be expected as a rare but life-threatening event, and, indeed, sHLH has been recognized to occur in up to 4.3% of sepsis cases [1]. Hence, in those COVID-19 patients showing massive hyperinflammation, a clinical diagnosis of sHLH/MAS may be appropriate and deserves further investigation at the histological level. More recently, COVID-19 clinical syndrome and related immunopathogenesis have been compared with sepsis, recalling the need to target the underlying and shared impairment of protective T cell immunity, while suppressing the emergent cytokine storm [7–9]. In fact, severe COVID-19 has appeared as a peculiar clinicopathologic entity—yet poorly understood from a mechanistic viewpoint—which however, by definition, may represent a novel form of viral sepsis, being characterized by (a) T cell deficiencies, with early and progressive lymphopenia; (b) systemic hyperinflammation, with a peculiar time-course, often increasing at a late phase, when coagulopathy and fatal organ damage may eventually occur; and (c) COVID-19-associated coagulopathy, displaying some unique clinical and laboratory findings, compared with either disseminated intravascular coagulation or sepsis-induced coagulopathy [10]. Further investigations are required to shed light on the relationships between these clinic-immunologic features and organ failure, possibly paving the way to the treatment (or even prevention) of severe COVID-19, by modulation of host immune system with targeted immunotherapeutic drugs. During the last few years, cancer immunotherapy with immune checkpoint inhibitors (ICIs), such as anti-PD1/PD-L1 and anti-CTLA-4 monoclonal antibodies (e.g., nivolumab and ipilimumab, respectively), has allowed impressive restoration of T cell immunity against neoplastic cells, which commonly induce overexpression of PD-1/CTLA-4 ligands to foster T cell exhaustion/anergy and break anti-tumor immune surveillance. Intriguingly, several human viruses have been demonstrated to adopt such “cancer-like” immune-evasion strategies, mainly by upregulation of PD-L1 in infected cells, in order to hamper antiviral T cell responses and make a productive infection [11]. Recently, in the attempt to improve antiviral T cell immunity in COVID-19 patients, clinical trials have started to test such T cell activating treatments. Of note, an ongoing Spanish phase 2 study (NCT04335305) seems the first to evaluate the attractive strategy of combining anti-cytokine treatments with ICIs (namely, tocilizumab plus pembrolizumab). Alongside monoclonal antibodies activating T lymphocytes, it has also been suggested that the infusion of SARS-CoV-2-specific cytotoxic T lymphocytes, deriving from HLA-matched convalescent donors, could be explored as innovative cell therapy for COVID-19 [12]. Actually, to maximize potential benefits of different immunotherapeutic approaches against COVID-19, adequate patients’ selection is warranted, possibly performed on the basis of putative biomarkers and immune profiles predictive of response. In addition, by considering the typical disease course, often prolonged for several weeks, the optimal timing for these treatments should be defined. Thus, it seems conceivable that, during SARS-CoV-2 infection, especially in elderly patients and less frequently in young people, something can go wrong at the delicate interface between effective viral clearance and T cell tolerance. Indeed, COVID-19 may be characterized by different clinical pictures, ranging from almost asymptomatic/mild infections in children and young individuals to lethal “sepsis-like” illness with SARS, particularly in advanced age. What differs between these two distinct stages of life, with regard to the antiviral response toward SARS-CoV-2 infection? Generally, in young subjects and even more in children, T cell immunity is known to be more pronounced and active, especially in terms of lymphocyte counts and adequate antiviral responses, while aged individuals typically undergo a well-described decline in T cell functions, which correlates with higher susceptibility to life-threatening infections, autoimmunity, and cancer [13]. Susceptibility to SARS-CoV-2 infection, related to the different functions and proportions of CD27dull and CD27bright memory B cells, throughout life, has also recently been suggested [14]. In the fight against SARS-CoV-2 pandemic, a more comprehensive vision of COVID-19 immunopathogenesis and related clinical manifestations is warranted to reconcile COVID-19 hyper-inflammatory features—similarly observed in sepsis, sHLH/MAS, and cytokine release syndrome (CRS) [3] induced by chimeric antigen receptor (CAR) T cell therapy, as well as in Kaposi sarcoma herpesvirus-associated inflammatory cytokine syndrome (KICS) [15] occurring in immunocompromised patients—with a renewed pivotal role played by the impairment of antiviral T cell functions. In this perspective (Fig. 1), in parallel with targeted immunosuppressive strategies, an effective reversal of T cell impairment by immune-activating treatments should allow to improve viral clearance and promote a better disease control with faster resolution, probably more akin to what naturally occurs in children infected with SARS-CoV-2. Fig. 1 Working model for COVID-19 immunopathogenesis and related immunomodulatory treatments. Acronyms: SARS-CoV-2 severe acute respiratory syndrome-coronavirus-2, SARS severe acute respiratory syndrome, SIRS severe inflammatory response syndrome, sHLH secondary hemophagocytic lymphohistiocytosis, MAS macrophage activation syndrome, CRS cytokine release syndrome, KICS Kaposi sarcoma herpesvirus-associated inflammatory cytokine syndrome, COVID-19 coronavirus disease-19, DIC disseminated intravascular coagulation Conclusions SARS-COV-2 has arisen as a new pathogen frequently inducing sepsis-like manifestations in the host. Indeed, based on actual evidence showing hyperinflammation as well as T cell deficiencies and coagulation abnormalities, associated with life-threatening organ dysfunction, severe COVID-19 may be well consistent with a clinical diagnosis of viral sepsis, rather than with a mere hyper-inflammatory disease. This conceptual framing may help to improve clinical management of severe COVID-19 patients, by providing a rationale for the development of novel balanced immunomodulatory approaches, combining both suppressive and activating immunotherapies.

          Related collections

          Most cited references13

          • Record: found
          • Abstract: found
          • Article: found

          COVID-19: consider cytokine storm syndromes and immunosuppression

          As of March 12, 2020, coronavirus disease 2019 (COVID-19) has been confirmed in 125 048 people worldwide, carrying a mortality of approximately 3·7%, 1 compared with a mortality rate of less than 1% from influenza. There is an urgent need for effective treatment. Current focus has been on the development of novel therapeutics, including antivirals and vaccines. Accumulating evidence suggests that a subgroup of patients with severe COVID-19 might have a cytokine storm syndrome. We recommend identification and treatment of hyperinflammation using existing, approved therapies with proven safety profiles to address the immediate need to reduce the rising mortality. Current management of COVID-19 is supportive, and respiratory failure from acute respiratory distress syndrome (ARDS) is the leading cause of mortality. 2 Secondary haemophagocytic lymphohistiocytosis (sHLH) is an under-recognised, hyperinflammatory syndrome characterised by a fulminant and fatal hypercytokinaemia with multiorgan failure. In adults, sHLH is most commonly triggered by viral infections 3 and occurs in 3·7–4·3% of sepsis cases. 4 Cardinal features of sHLH include unremitting fever, cytopenias, and hyperferritinaemia; pulmonary involvement (including ARDS) occurs in approximately 50% of patients. 5 A cytokine profile resembling sHLH is associated with COVID-19 disease severity, characterised by increased interleukin (IL)-2, IL-7, granulocyte-colony stimulating factor, interferon-γ inducible protein 10, monocyte chemoattractant protein 1, macrophage inflammatory protein 1-α, and tumour necrosis factor-α. 6 Predictors of fatality from a recent retrospective, multicentre study of 150 confirmed COVID-19 cases in Wuhan, China, included elevated ferritin (mean 1297·6 ng/ml in non-survivors vs 614·0 ng/ml in survivors; p 39·4°C 49 Organomegaly None 0 Hepatomegaly or splenomegaly 23 Hepatomegaly and splenomegaly 38 Number of cytopenias * One lineage 0 Two lineages 24 Three lineages 34 Triglycerides (mmol/L) 4·0 mmol/L 64 Fibrinogen (g/L) >2·5 g/L 0 ≤2·5 g/L 30 Ferritin ng/ml 6000 ng/ml 50 Serum aspartate aminotransferase <30 IU/L 0 ≥30 IU/L 19 Haemophagocytosis on bone marrow aspirate No 0 Yes 35 Known immunosuppression † No 0 Yes 18 The Hscore 11 generates a probability for the presence of secondary HLH. HScores greater than 169 are 93% sensitive and 86% specific for HLH. Note that bone marrow haemophagocytosis is not mandatory for a diagnosis of HLH. HScores can be calculated using an online HScore calculator. 11 HLH=haemophagocytic lymphohistiocytosis. * Defined as either haemoglobin concentration of 9·2 g/dL or less (≤5·71 mmol/L), a white blood cell count of 5000 white blood cells per mm3 or less, or platelet count of 110 000 platelets per mm3 or less, or all of these criteria combined. † HIV positive or receiving longterm immunosuppressive therapy (ie, glucocorticoids, cyclosporine, azathioprine).
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Clinical and immunologic features in severe and moderate Coronavirus Disease 2019

            Journal of Clinical Investigation
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Dysregulation of immune response in patients with COVID-19 in Wuhan, China

              Abstract Background In December 2019, coronavirus disease 2019 (COVID-19) emerged in Wuhan and rapidly spread throughout China. Methods Demographic and clinical data of all confirmed cases with COVID-19 on admission at Tongji Hospital from January 10 to February 12, 2020, were collected and analyzed. The data of laboratory examinations, including peripheral lymphocyte subsets, were analyzed and compared between severe and non-severe patients. Results Of the 452 patients with COVID-19 recruited, 286 were diagnosed as severe infection. The median age was 58 years and 235 were male. The most common symptoms were fever, shortness of breath, expectoration, fatigue, dry cough and myalgia. Severe cases tend to have lower lymphocytes counts, higher leukocytes counts and neutrophil-lymphocyte-ratio (NLR), as well as lower percentages of monocytes, eosinophils, and basophils. Most of severe cases demonstrated elevated levels of infection-related biomarkers and inflammatory cytokines. The number of T cells significantly decreased, and more hampered in severe cases. Both helper T cells and suppressor T cells in patients with COVID-19 were below normal levels, and lower level of helper T cells in severe group. The percentage of naïve helper T cells increased and memory helper T cells decreased in severe cases. Patients with COVID-19 also have lower level of regulatory T cells, and more obviously damaged in severe cases. Conclusions The novel coronavirus might mainly act on lymphocytes, especially T lymphocytes. Surveillance of NLR and lymphocyte subsets is helpful in the early screening of critical illness, diagnosis and treatment of COVID-19.
                Bookmark

                Author and article information

                Contributors
                mario.luppi@unimore.it
                Journal
                Crit Care
                Critical Care
                BioMed Central (London )
                1364-8535
                1466-609X
                4 September 2020
                4 September 2020
                2020
                : 24
                : 549
                Affiliations
                [1 ]Department of Laboratory Medicine and Pathology, AUSL/AOU Policlinico, Modena, Italy
                [2 ]GRID grid.7548.e, ISNI 0000000121697570, Department of Medical and Surgical Sciences, , University of Modena and Reggio Emilia; Hematology Unit, AOU Policlinico, ; Via Del Pozzo 71, 41124 Modena, Italy
                [3 ]GRID grid.419425.f, ISNI 0000 0004 1760 3027, Pediatric Hematology/Oncology Unit, , IRCCS Policlinico San Matteo, ; Pavia, Italy
                Author information
                http://orcid.org/0000-0002-0373-1154
                Article
                3267
                10.1186/s13054-020-03267-w
                7472946
                32887652
                45915840-b9d9-4427-82e6-b8b42fad83c1
                © The Author(s) 2020

                Open AccessThis 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.

                History
                : 15 August 2020
                : 26 August 2020
                Categories
                Commentary
                Custom metadata
                © The Author(s) 2020

                Emergency medicine & Trauma
                Emergency medicine & Trauma

                Comments

                Comment on this article