An immune-based biomarker signature is associated with mortality in COVID-19 patients

Michael S Abers, Ottavia M Delmonte, Emily E Ricotta, Jonathan Fintzi, Danielle L Fink, Adriana A Almeida de Jesus, Kol A Zarember, Sara Alehashemi, Vasileios Oikonomou, Jigar V Desai, Scott W Canna, Bita Shakoory, Kerry Dobbs, Luisa Imberti, Alessandra Sottini, Eugenia Quiros-Roldan, Francesco Castelli, Camillo Rossi, Duilio Brugnoni, Andrea Biondi, Laura Rachele Bettini, Mariella D'Angio', Paolo Bonfanti, Riccardo Castagnoli, Daniela Montagna, Amelia Licari, Gian Luigi Marseglia, Emily F Gliniewicz, Elana Shaw, Dana E Kahle, Andre T Rastegar, Michael Stack, Katherine Myint-Hpu, Susan L Levinson, Mark J DiNubile, Daniel W Chertow, Peter D Burbelo, Jeffrey I Cohen, Katherine R Calvo, John S Tsang, NIAID COVID-19 Consortium, Helen C Su, John I Gallin, Douglas B Kuhns, Raphaela Goldbach-Mansky, Michail S Lionakis, Luigi D Notarangelo, Michael S Abers, Ottavia M Delmonte, Emily E Ricotta, Jonathan Fintzi, Danielle L Fink, Adriana A Almeida de Jesus, Kol A Zarember, Sara Alehashemi, Vasileios Oikonomou, Jigar V Desai, Scott W Canna, Bita Shakoory, Kerry Dobbs, Luisa Imberti, Alessandra Sottini, Eugenia Quiros-Roldan, Francesco Castelli, Camillo Rossi, Duilio Brugnoni, Andrea Biondi, Laura Rachele Bettini, Mariella D'Angio', Paolo Bonfanti, Riccardo Castagnoli, Daniela Montagna, Amelia Licari, Gian Luigi Marseglia, Emily F Gliniewicz, Elana Shaw, Dana E Kahle, Andre T Rastegar, Michael Stack, Katherine Myint-Hpu, Susan L Levinson, Mark J DiNubile, Daniel W Chertow, Peter D Burbelo, Jeffrey I Cohen, Katherine R Calvo, John S Tsang, NIAID COVID-19 Consortium, Helen C Su, John I Gallin, Douglas B Kuhns, Raphaela Goldbach-Mansky, Michail S Lionakis, Luigi D Notarangelo

Abstract

Immune and inflammatory responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) contribute to disease severity of coronavirus disease 2019 (COVID-19). However, the utility of specific immune-based biomarkers to predict clinical outcome remains elusive. Here, we analyzed levels of 66 soluble biomarkers in 175 Italian patients with COVID-19 ranging from mild/moderate to critical severity and assessed type I IFN-, type II IFN-, and NF-κB-dependent whole-blood transcriptional signatures. A broad inflammatory signature was observed, implicating activation of various immune and nonhematopoietic cell subsets. Discordance between IFN-α2a protein and IFNA2 transcript levels in blood suggests that type I IFNs during COVID-19 may be primarily produced by tissue-resident cells. Multivariable analysis of patients' first samples revealed 12 biomarkers (CCL2, IL-15, soluble ST2 [sST2], NGAL, sTNFRSF1A, ferritin, IL-6, S100A9, MMP-9, IL-2, sVEGFR1, IL-10) that when increased were independently associated with mortality. Multivariate analyses of longitudinal biomarker trajectories identified 8 of the aforementioned biomarkers (IL-15, IL-2, NGAL, CCL2, MMP-9, sTNFRSF1A, sST2, IL-10) and 2 additional biomarkers (lactoferrin, CXCL9) that were substantially associated with mortality when increased, while IL-1α was associated with mortality when decreased. Among these, sST2, sTNFRSF1A, IL-10, and IL-15 were consistently higher throughout the hospitalization in patients who died versus those who recovered, suggesting that these biomarkers may provide an early warning of eventual disease outcome.

Keywords: COVID-19; Chemokines; Cytokines; Immunology.

Conflict of interest statement

Conflict of interest: SLL and MJD are employees of and own stock in BioAegis Therapeutics, Inc, which is developing recombinant human plasma gelsolin for potential clinical use.

Figures

Figure 1. Biomarkers associated with activation of…
Figure 1. Biomarkers associated with activation of monocytes/macrophages and NF-κB signaling are markedly induced in COVID-19 patients.
(A) Shown are levels of soluble CD163 (sCD163), CCL2, ferritin, IL-15, CX3CL1, IL-12p70, IL-12p40, IL-6, and sTNFRSF1A in peripheral blood of COVID-19 patients with various severity groups (n = 94–119 depending on the biomarker) relative to healthy volunteers (HV; n = 45–60 depending on the biomarker). Ferritin concentrations were determined by clinical assays performed in Italian hospitals. The area shaded in gray reflects the normal range for HVs reported by the clinical laboratory. Groups were compared by Kruskal-Wallis test. When P < 0.05, pairwise comparisons were made using Dunn’s test with Benjamini-Hochberg adjustment for multiple comparisons. (B) Expression of 11 NF-κB–regulated genes was measured by NanoString and expressed as summary z scores in whole blood of COVID-19 patients (n = 29) and HVs (n = 22). Groups were compared by an unpaired Student’s t test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Figure 2. Neutrophil activation–associated biomarkers are increased…
Figure 2. Neutrophil activation–associated biomarkers are increased in COVID-19 patients with more severe disease.
Shown are levels of MPO, MMP-9, S100A9, NGAL, lactoferrin, IL-8, and IL-16 in peripheral blood of COVID-19 patients with various severity groups (n = 80–119 depending on the biomarker) relative to healthy volunteers (HV; n = 12–60 depending on the biomarker). Groups were compared by Kruskal-Wallis test. When P < 0.05, pairwise comparisons were made using Dunn’s test with Benjamini-Hochberg adjustment for multiple comparisons. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Figure 3. Th1-type immune response–associated biomarkers are…
Figure 3. Th1-type immune response–associated biomarkers are predominantly increased in patients with COVID-19 relative to Th2 and Th17 immune response–associated biomarkers, while sFASLG and sCD40LG are decreased.
(A) Shown are levels of IL-2, sFASLG, sCD40LG, CXCL9, IL-4, CCL22, IL-33, IL-17, and IL-10 in peripheral blood of COVID-19 patients with various severity groups (n = 94–119 depending on the biomarker) relative to healthy volunteers (HV; n = 34–60 depending on the biomarker). Groups were compared by Kruskal-Wallis test. When P < 0.05, pairwise comparisons were made using Dunn’s test with Benjamini-Hochberg adjustment for multiple comparisons. (B) Expression of 15 type II IFN–regulated (IFN-γ–regulated genes was measured by NanoString and expressed as summary z scores in whole blood of COVID-19 patients (n = 29) and HVs (n = 22). Groups were compared by an unpaired Student’s t test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Figure 4. Abnormal levels of biomarkers associated…
Figure 4. Abnormal levels of biomarkers associated with endothelial integrity and sepsis severity in COVID-19 patients.
Shown are levels of soluble VEGF receptor 1 (sVEGFR1), VEGF, sST2 LPS binding protein (LBP), receptor of advanced glycation end products (RAGE), and plasma gelsolin (pGSN) in peripheral blood of COVID-19 patients with various severity groups (n = 93–119) relative to healthy volunteers (HV; n = 14–60 depending on the biomarker). Groups were compared by Kruskal-Wallis test. When P < 0.05, pairwise comparisons were made using Dunn’s test with Benjamini-Hochberg adjustment for multiple comparisons. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Figure 5. Type I IFN mediators are…
Figure 5. Type I IFN mediators are increased in COVID-19 patients, but the transcriptional response of type I IFN genes in circulating immune cells is disproportionally low.
(AB) Shown are (A) IFN-α2a and (B) CXCL10 levels in peripheral blood of COVID-19 patients with various severity groups (n = 94–114 depending on the biomarker) relative to healthy volunteers (HV; n = 45–67 depending on the biomarker). Groups were compared by Kruskal-Wallis test. When P < 0.05, pairwise comparisons were made using Dunn’s test with Benjamini-Hochberg adjustment for multiple comparisons. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. (C) Expression of 28 type I IFN–induced genes was measured by NanoString and expressed as log10-transformed summary z scores. Shown is comparison of HVs (n = 22), COVID-19 patients (n = 84), and patients with the NLRP3 inflammasomopathy NOMID (n = 11); and the type I IFNopathies CANDLE (n = 9), SAVI (n = 9), and AGS (n = 7); the CANDLE mimic NEMO-NDAS (n = 9); and the IL-18opathy IL-18 PAP/MAS (n = 6). NOMID, neonatal onset multisystem inflammatory disease; CANDLE, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature; SAVI, STING-associated vasculopathy with onset in infancy; AGS, Aicardi-Goutières syndrome; NEMO-NDAS, NF-κB essential modulator-deleted exon 5 autoinflammatory syndrome; IL18 PAP/MAS, IL-18–mediated pulmonary alveolar proteinosis and macrophage activation syndrome. (D) Correlation of the transcript levels of IFNA2 in whole blood with blood levels of IFN-α2a in patients with COVID-19 (n = 22). (E) Correlation of the 28 type I IFN–induced gene score with transcript levels of IFNA2 in patients with COVID-19 (left panel) (n = 73) compared with the indicated type I IFNopathies (right panel) (n = 34).
Figure 6. A subset of immune-based biomarkers…
Figure 6. A subset of immune-based biomarkers is associated with mortality in COVID-19 patients in multivariable analyses.
Shown are forest plots and adjusted HRs (aHRs) of all 66 tested biomarkers and their association with mortality during COVID-19 by multivariable analysis, irrespective of when the first sample was collected relative to the hospital admission when adjusting for (left panel) the time of sample collection relative to hospital admission or (right panel) the time of sample collection relative to hospital admission with age, chronic kidney disease, and receipt of immunomodulatory medications. For biomarkers significantly associated with mortality (i.e., q < 0.025), aHR CIs are shown in red.
Figure 7. Association between the longitudinal trajectory…
Figure 7. Association between the longitudinal trajectory of biomarkers and the risk of death after COVID-19.
Shown are forest plots of the immune-based biomarkers (n = 66) whose longitudinal trajectories were significantly associated with increased patient mortality after controlling the FDR irrespective of when the first sample was collected relative to the hospital admission. aHR CIs for biomarkers significantly associated with mortality (i.e., q < 0.025) are shown in red when aHR > 1 and in blue when aHR < 1. aHR CIs for biomarkers with q > 0.025 are shown in black.
Figure 8. sTNFRSF1A, sST2, IL-10, and IL-15…
Figure 8. sTNFRSF1A, sST2, IL-10, and IL-15 may differentiate between survivors and patients who succumb to COVID-19 throughout the entire hospitalization.
Shown are loess-smoothed means with 95% CIs (shaded intervals) of sTNFRSF1A, sST2, IL-10, and IL-15 concentration throughout the hospitalization in patients with COVID-19 who survived or succumbed to the infection (n = 175). All biomarker concentrations are in pg/mL.

References

    1. John Hopkins Coronavirus Resource Center. COVID-19 Dashboard by the Center for Systems Science and Engineering (CSSE) at Johns Hopkins University (JHU). Updated November 30, 2020. Accessed November 30, 2020.
    1. Guan WJ, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382(18):1708–1720. doi: 10.1056/NEJMoa2002032.
    1. Chen N, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395(10223):507–513. doi: 10.1016/S0140-6736(20)30211-7.
    1. Huang C, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506. doi: 10.1016/S0140-6736(20)30183-5.
    1. Richardson S, et al. Presenting characteristics, comorbidities, and outcomes among 5700 patients hospitalized with COVID-19 in the New York City area. JAMA. 2020;323(20):2052–2059. doi: 10.1001/jama.2020.6775.
    1. Wu C, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med. 2020;180(7):934–943. doi: 10.1001/jamainternmed.2020.0994.
    1. Blanco-Melo D, et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell. 2020;181(5):1036–1045. doi: 10.1016/j.cell.2020.04.026.
    1. Merad M, Martin JC. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat Rev Immunol. 2020;20(6):355–362. doi: 10.1038/s41577-020-0331-4.
    1. Tay MZ, et al. The trinity of COVID-19: immunity, inflammation and intervention. Nat Rev Immunol. 2020;20(6):363–374. doi: 10.1038/s41577-020-0311-8.
    1. Hadjadj J, et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science. 2020;369(6504):718–724. doi: 10.1126/science.abc6027.
    1. Chen G, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130(5):2620–2629. doi: 10.1172/JCI137244.
    1. doi: 10.1101/2020.02.25.20025643. Gong J DH, et al. Correlation analysis between disease severity and inflammation-related parameters in patients with COVID-19 pneumonia [preprint]. Posted on medRxiv February 27, 2020.
    1. Zhou F, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395(10229):1054–1062. doi: 10.1016/S0140-6736(20)30566-3.
    1. Del Valle DM, et al. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat Med. 2020;26(10):1636–1643. doi: 10.1038/s41591-020-1051-9.
    1. Lucas C, et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature. 2020;584(7821):463–469. doi: 10.1038/s41586-020-2588-y.
    1. Arunachalam PS, et al. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans. Science. 2020;369(6508):1210–1220. doi: 10.1126/science.abc6261.
    1. Yang Y, et al. Plasma IP-10 and MCP-3 levels are highly associated with disease severity and predict the progression of COVID-19. J Allergy Clin Immunol. 2020;146(1):119–127. doi: 10.1016/j.jaci.2020.04.027.
    1. Mathew D, et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science. 2020;369(6508):eabc8511.
    1. Laing AG, et al. A dynamic COVID-19 immune signature includes associations with poor prognosis. Nat Med. 2020;26(10):1623–1635. doi: 10.1038/s41591-020-1038-6.
    1. Rosenbaum L. Facing Covid-19 in Italy — Ethics, logistics, and therapeutics on the epidemic’s front line. N Engl J Med. 2020;382(20):1873–1875. doi: 10.1056/NEJMp2005492.
    1. Wei P-F, ed. Diagnosis and treatment protocol for novel coronavirus pneumonia (trial version 7). Chin Med J (Engl). 2020;133(9):1087–1095.
    1. Elshazli RM, et al. Diagnostic and prognostic value of hematological and immunological markers in COVID-19 infection: a meta-analysis of 6320 patients. PLoS One. 2020;15(8):e0238160. doi: 10.1371/journal.pone.0238160.
    1. Tian W, et al. Predictors of mortality in hospitalized COVID-19 patients: a systematic review and meta-analysis. J Med Virol. 2020;92(10):1875–1883. doi: 10.1002/jmv.26050.
    1. Group RC, et al. Dexamethasone in hospitalized patients with Covid-19 — preliminary report [published online July 17, 2020]. N Engl J Med. .
    1. Laguna-Goya R, et al. IL-6-based mortality risk model for hospitalized patients with COVID-19. J Allergy Clin Immunol. 2020;146(4):799–807. doi: 10.1016/j.jaci.2020.07.009.
    1. Bedin AS, et al. Monocyte CD169 expression as a biomarker in the early diagnosis of COVID-19 [published online November 18, 2020]. J Infect Dis. .
    1. doi: 10.1101/2020.03.24.20042655. Zhang D GR, et al. COVID-19 infection induces readily detectable morphological and inflammation-related phenotypic changes in peripheral blood monocytes, the severity of which correlate with patient outcome [preprint]. Posted on medRxiv March 26, 2020.
    1. Giavridis T, et al. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med. 2018;24(6):731–738. doi: 10.1038/s41591-018-0041-7.
    1. Savic S, et al. Moving towards a systems-based classification of innate immune-mediated diseases. Nat Rev Rheumatol. 2020;16(4):222–237. doi: 10.1038/s41584-020-0377-5.
    1. Wilson JG, et al. Cytokine profile in plasma of severe COVID-19 does not differ from ARDS and sepsis. JCI Insight. 2020;5(17):140289.
    1. Shakoory B, et al. Interleukin-1 receptor blockade is associated with reduced mortality in sepsis patients with features of macrophage activation syndrome: reanalysis of a prior phase III trial. Crit Care Med. 2016;44(2):275–281. doi: 10.1097/CCM.0000000000001402.
    1. Dimopoulos G, et al. Favorable anakinra responses in severe Covid-19 patients with secondary hemophagocytic lymphohistiocytosis. Cell Host Microbe. 2020;28(1):117–123. doi: 10.1016/j.chom.2020.05.007.
    1. Huet T, et al. Anakinra for severe forms of COVID-19: a cohort study. Lancet Rheumatol. 2020;2(7):e393–e400. doi: 10.1016/S2665-9913(20)30164-8.
    1. Herold T, et al. Elevated levels of IL-6 and CRP predict the need for mechanical ventilation in COVID-19. J Allergy Clin Immunol. 2020;146(1):128–136. doi: 10.1016/j.jaci.2020.05.008.
    1. Mazzoni A, et al. Impaired immune cell cytotoxicity in severe COVID-19 is IL-6 dependent. J Clin Invest. 2020;130(9):4694–4703. doi: 10.1172/JCI138554.
    1. Vultaggio A, et al. Prompt predicting of early clinical deterioration of moderate-to-severe COVID-19 patients: usefulness of a combined score using IL-6 in a preliminary study. J Allergy Clin Immunol Pract. 2020;8(8):2575–2581. doi: 10.1016/j.jaip.2020.06.013.
    1. Lee JS, et al. Immunophenotyping of COVID-19 and influenza highlights the role of type I interferons in development of severe COVID-19. Sci Immunol. 2020;5(49):eabd1554. doi: 10.1126/sciimmunol.abd1554.
    1. doi: 10.1101/2020.06.13.20127605. Mann ER MM, et al. Longitudinal immune profiling reveals distinct features of COVID-19 pathogenesis [preprint]. Posted on medRxiv June 16, 2020.
    1. Perlin DS, et al. Levels of the TNF-related cytokine LIGHT Increase in hospitalized COVID-19 patients with cytokine release syndrome and ARDS. mSphere. 2020;5(4):e00699-20.
    1. Schulte-Schrepping J, et al. Severe COVID-19 is marked by a dysregulated myeloid cell compartment. Cell. 2020;182(6):1419–1440. doi: 10.1016/j.cell.2020.08.001.
    1. Silvin A, et al. Elevated calprotectin and abnormal myeloid cell subsets discriminate severe from mild COVID-19. Cell. 2020;182(6):1401–1408. doi: 10.1016/j.cell.2020.08.002.
    1. Skendros P, et al. Complement and tissue factor-enriched neutrophil extracellular traps are key drivers in COVID-19 immunothrombosis. J Clin Invest. 2020;130(11):6151–6157. doi: 10.1172/JCI141374.
    1. Wilk AJ, et al. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat Med. 2020;26(7):1070–1076. doi: 10.1038/s41591-020-0944-y.
    1. Legg JP, et al. Type 1 and type 2 cytokine imbalance in acute respiratory syncytial virus bronchiolitis. Am J Respir Crit Care Med. 2003;168(6):633–639. doi: 10.1164/rccm.200210-1148OC.
    1. Zhao Y, et al. Longitudinal COVID-19 profiling associates IL-1RA and IL-10 with disease severity and RANTES with mild disease. JCI Insight. 2020;5(13):e139834.
    1. Vestweber D. How leukocytes cross the vascular endothelium. Nat Rev Immunol. 2015;15(11):692–704. doi: 10.1038/nri3908.
    1. Ackermann M, et al. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in Covid-19. N Engl J Med. 2020;383(2):120–128. doi: 10.1056/NEJMoa2015432.
    1. Alves-Filho JC, et al. Interleukin-33 attenuates sepsis by enhancing neutrophil influx to the site of infection. Nat Med. 2010;16(6):708–712. doi: 10.1038/nm.2156.
    1. Chen KF, et al. Diagnostic accuracy of lipopolysaccharide-binding protein as biomarker for sepsis in adult patients: a systematic review and meta-analysis. PLoS One. 2016;11(4):e0153188. doi: 10.1371/journal.pone.0153188.
    1. Dubois C, et al. High plasma level of S100A8/S100A9 and S100A12 at admission indicates a higher risk of death in septic shock patients. Sci Rep. 2019;9(1):15660. doi: 10.1038/s41598-019-52184-8.
    1. Krychtiuk KA, et al. Predictive value of low interleukin-33 in critically ill patients. Cytokine. 2018;103:109–113. doi: 10.1016/j.cyto.2017.09.017.
    1. Pregernig A, et al. Prediction of mortality in adult patients with sepsis using six biomarkers: a systematic review and meta-analysis. Ann Intensive Care. 2019;9(1):125. doi: 10.1186/s13613-019-0600-1.
    1. Faustino LD, et al. Interleukin-33 activates regulatory T cells to suppress innate γδ T cell responses in the lung. Nat Immunol. 2020;21(11):1371–1383. doi: 10.1038/s41590-020-0785-3.
    1. Wang S, et al. S100A8/A9 in inflammation. Front Immunol. 2018;9:1298.
    1. Piktel E, et al. Plasma gelsolin: indicator of inflammation and its potential as a diagnostic tool and therapeutic target. Int J Mol Sci. 2018;19(9):2516. doi: 10.3390/ijms19092516.
    1. Ciancanelli MJ, et al. Infectious disease. life-threatening influenza and impaired interferon amplification in human IRF7 deficiency. Science. 2015;348(6233):448–453. doi: 10.1126/science.aaa1578.
    1. Lim HK, et al. Severe influenza pneumonitis in children with inherited TLR3 deficiency. J Exp Med. 2019;216(9):2038–2056. doi: 10.1084/jem.20181621.
    1. Hernandez N, et al. Inherited IFNAR1 deficiency in otherwise healthy patients with adverse reaction to measles and yellow fever live vaccines. J Exp Med. 2019;216(9):2057–2070. doi: 10.1084/jem.20182295.
    1. Hernandez N, et al. Life-threatening influenza pneumonitis in a child with inherited IRF9 deficiency. J Exp Med. 2018;215(10):2567–2585. doi: 10.1084/jem.20180628.
    1. Duncan CJ, et al. Human IFNAR2 deficiency: lessons for antiviral immunity. Sci Transl Med. 2015;7(307):307ra154. doi: 10.1126/scitranslmed.aac4227.
    1. Dupuis S, et al. Impaired response to interferon-alpha/beta and lethal viral disease in human STAT1 deficiency. Nat Genet. 2003;33(3):388–391. doi: 10.1038/ng1097.
    1. Hambleton S, et al. IRF8 mutations and human dendritic-cell immunodeficiency. N Engl J Med. 2011;365(2):127–138. doi: 10.1056/NEJMoa1100066.
    1. Pozzetto B, et al. Characteristics of autoantibodies to human interferon in a patient with varicella-zoster disease. J Infect Dis. 1984;150(5):707–713. doi: 10.1093/infdis/150.5.707.
    1. Walter JE, et al. Broad-spectrum antibodies against self-antigens and cytokines in RAG deficiency. J Clin Invest. 2015;125(11):4135–48. doi: 10.1172/JCI80477.
    1. Uggenti C, et al. Self-awareness: nucleic acid-driven inflammation and the type I interferonopathies. Annu Rev Immunol. 2019;37:247–267. doi: 10.1146/annurev-immunol-042718-041257.
    1. Channappanavar R, et al. Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host Microbe. 2016;19(2):181–193. doi: 10.1016/j.chom.2016.01.007.
    1. Kim H, et al. Development of a validated interferon score using NanoString technology. J Interferon Cytokine Res. 2018;38(4):171–185. doi: 10.1089/jir.2017.0127.
    1. Franco LM, et al. Immune regulation by glucocorticoids can be linked to cell type-dependent transcriptional responses. J Exp Med. 2019;216(2):384–406. doi: 10.1084/jem.20180595.
    1. Lionakis MS, Kontoyiannis DP. Glucocorticoids and invasive fungal infections. Lancet. 2003;362(9398):1828–1838. doi: 10.1016/S0140-6736(03)14904-5.
    1. Luo P, et al. Tocilizumab treatment in COVID-19: a single center experience. J Med Virol. 2020;92(7):814–818. doi: 10.1002/jmv.25801.
    1. Zain Mushtaq M, et al. Outcome of COVID-19 patients with use of tocilizumab: a single center experience. Int Immunopharmacol. 2020;88:106926.
    1. Arbeev KG, et al. Joint analyses of longitudinal and time-to-event data in research on aging: implications for predicting health and survival. Front Public Health. 2014;2:228.
    1. Ibrahim JG, et al. Basic concepts and methods for joint models of longitudinal and survival data. J Clin Oncol. 2010;28(16):2796–2801. doi: 10.1200/JCO.2009.25.0654.
    1. Zhou Z, et al. Heightened innate immune responses in the respiratory tract of COVID-19 patients. Cell Host Microbe. 2020;27(6):883–890. doi: 10.1016/j.chom.2020.04.017.
    1. Zhang Q, et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science. 2020;370(6515):eabd4570. doi: 10.1126/science.abd4570.
    1. Bastard P, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020;370(6515):eabd4585. doi: 10.1126/science.abd4585.
    1. Shi H, et al. Neutrophil calprotectin identifies severe pulmonary disease in COVID-19 [published online September 1, 2020]. J Leukoc Biol. .
    1. Morjaria S, et al. The effect of neutropenia and filgrastim (G-CSF) in cancer patients with COVID-19 infection [preprint]. Posted on medRxiv August 15, 2020.
    1. Grommes J, Soehnlein O. Contribution of neutrophils to acute lung injury. Mol Med. 2011;17(3–4):293–307.
    1. Leonard WJ, et al. The γc family of cytokines: basic biology to therapeutic ramifications. Immunity. 2019;50(4):832–850. doi: 10.1016/j.immuni.2019.03.028.
    1. Cassatella MA, McDonald PP. Interleukin-15 and its impact on neutrophil function. Curr Opin Hematol. 2000;7(3):174–177. doi: 10.1097/00062752-200005000-00008.
    1. Agouridakis P, et al. Association between increased levels of IL-2 and IL-15 and outcome in patients with early acute respiratory distress syndrome. Eur J Clin Invest. 2002;32(11):862–867. doi: 10.1046/j.1365-2362.2002.01081.x.
    1. Leahy TR, et al. Interleukin-15 is associated with disease severity in viral bronchiolitis. Eur Respir J. 2016;47(1):212–222. doi: 10.1183/13993003.00642-2015.
    1. Kandikattu HK, et al. IL-15 immunotherapy is a viable strategy for COVID-19. Cytokine Growth Factor Rev. 2020;54:24–31. doi: 10.1016/j.cytogfr.2020.06.008.
    1. Griesenauer B, Paczesny S. The ST2/IL-33 axis in immune cells during inflammatory diseases. Front Immunol. 2017;8:475.
    1. Hoogerwerf JJ, et al. Soluble ST2 plasma concentrations predict mortality in severe sepsis. Intensive Care Med. 2010;36(4):630–637. doi: 10.1007/s00134-010-1773-0.
    1. Watanabe M, et al. Soluble ST2 as a prognostic marker in community-acquired pneumonia. J Infect. 2015;70(5):474–482. doi: 10.1016/j.jinf.2015.02.004.
    1. Xia J, et al. Increased IL-33 expression in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol. 2015;308(7):L619–L627. doi: 10.1152/ajplung.00305.2014.
    1. Portugal CAA, et al. IL-33 and ST2 as predictors of disease severity in children with viral acute lower respiratory infection. Cytokine. 2020;127:154965.
    1. Giamarellos-Bourboulis EJ, et al. Complex immune dysregulation in COVID-19 patients with severe respiratory failure. Cell Host Microbe. 2020;27(6):992–1000. doi: 10.1016/j.chom.2020.04.009.
    1. Moratto D, et al. Flow cytometry identifies risk factors and dynamic changes in patients with COVID-19. J Clin Immunol. 2020;40(7):970–973. doi: 10.1007/s10875-020-00806-6.
    1. doi: 10.1101/2020.05.31.20112979. Neumann J PT, et al. An open resource for T cell phenotype changes in COVID-19 identifies IL-10-producing regulatory T cells as characteristic of severe cases [preprint]. Posted on medRxiv June 2, 2020.
    1. Chang J, et al. Negative regulation of MyD88-dependent signaling by IL-10 in dendritic cells. Proc Natl Acad Sci U S A. 2009;106(43):18327–18332. doi: 10.1073/pnas.0905815106.
    1. Chaudhry A, et al. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity. 2011;34(4):566–578. doi: 10.1016/j.immuni.2011.03.018.
    1. Bedoya F, et al. Viral antigen induces differentiation of Foxp3+ natural regulatory T cells in influenza virus-infected mice. J Immunol. 2013;190(12):6115–6125. doi: 10.4049/jimmunol.1203302.
    1. van der Sluijs KF, et al. IL-10 is an important mediator of the enhanced susceptibility to pneumococcal pneumonia after influenza infection. J Immunol. 2004;172(12):7603–9. doi: 10.4049/jimmunol.172.12.7603.
    1. Joyce DA, et al. Two inhibitors of pro-inflammatory cytokine release, interleukin-10 and interleukin-4, have contrasting effects on release of soluble p75 tumor necrosis factor receptor by cultured monocytes. Eur J Immunol. 1994;24(11):2699–2705. doi: 10.1002/eji.1830241119.
    1. Dickensheets HL, et al. Interleukin-10 upregulates tumor necrosis factor receptor type-II (p75) gene expression in endotoxin-stimulated human monocytes. Blood. 1997;90(10):4162–4171. doi: 10.1182/blood.V90.10.4162.
    1. Burbelo PD, et al. Sensitivity in detection of antibodies to nucleocapsid and spike proteins of severe acute respiratory syndrome coronavirus 2 in patients with coronavirus disease 2019. J Infect Dis. 2020;222(2):206–213. doi: 10.1093/infdis/jiaa273.
    1. R. Bayesian applied regression modeling via Stan. R package version 2211. Accessed November 25, 2020.
    1. doi: 10.5281/zenodo.1284334. Brilleman SL, et al. Joint longitudinal and time-to-event models via Stan. Paper presented at: StanCon 2018; January 10, 2018; Pacific Grove, California, USA. Accessed November 30, 2020.
    1. Storey JD, et al. The positive false discovery rate: a Bayesian interpretation and the q-value. Ann Statist. 2003;31(6):2013–2035. doi: 10.1214/aos/1074290335.
    1. R. qvalue: Q-value estimation for false discovery rate control. R package version 2200. Accessed November 25, 2020.
    1. van Buuren S, Groothuis-Oudshoorn K. mice: multivariate imputation by chained equations in R. J Stat Softw. 2011;45(3):1–67.

Source: PubMed

3
Předplatit