Cytokine levels associated with favorable clinical outcome in the CAPSID randomized trial of convalescent plasma in patients with severe COVID-19

Sixten Körper, Eva Vanessa Schrezenmeier, Hector Rincon-Arevalo, Beate Grüner, Daniel Zickler, Manfred Weiss, Thomas Wiesmann, Kai Zacharowski, Johannes Kalbhenn, Martin Bentz, Matthias M Dollinger, Gregor Paul, Philipp M Lepper, Lucas Ernst, Hinnerk Wulf, Sebastian Zinn, Thomas Appl, Bernd Jahrsdörfer, Markus Rojewski, Ramin Lotfi, Thomas Dörner, Bettina Jungwirth, Erhard Seifried, Daniel Fürst, Hubert Schrezenmeier, Sixten Körper, Eva Vanessa Schrezenmeier, Hector Rincon-Arevalo, Beate Grüner, Daniel Zickler, Manfred Weiss, Thomas Wiesmann, Kai Zacharowski, Johannes Kalbhenn, Martin Bentz, Matthias M Dollinger, Gregor Paul, Philipp M Lepper, Lucas Ernst, Hinnerk Wulf, Sebastian Zinn, Thomas Appl, Bernd Jahrsdörfer, Markus Rojewski, Ramin Lotfi, Thomas Dörner, Bettina Jungwirth, Erhard Seifried, Daniel Fürst, Hubert Schrezenmeier

Abstract

Objectives: To determine the profile of cytokines in patients with severe COVID-19 who were enrolled in a trial of COVID-19 convalescent plasma (CCP).

Methods: Patients were randomized to receive standard treatment and 3 CCP units or standard treatment alone (CAPSID trial, ClinicalTrials.gov NCT04433910). The primary outcome was a dichotomous composite outcome (survival and no longer severe COVID-19 on day 21). Time to clinical improvement was a key secondary endpoint. The concentrations of 27 cytokines were measured (baseline, day 7). We analyzed the change and the correlation between serum cytokine levels over time in different subgroups and the prediction of outcome in receiver operating characteristics (ROC) analyses and in multivariate models.

Results: The majority of cytokines showed significant changes from baseline to day 7. Some were strongly correlated amongst each other (at baseline the cluster IL-1ß, IL-2, IL-6, IL-8, G-CSF, MIP-1α, the cluster PDGF-BB, RANTES or the cluster IL-4, IL-17, Eotaxin, bFGF, TNF-α). The correlation matrix substantially changed from baseline to day 7. The heatmaps of the absolute values of the correlation matrix indicated an association of CCP treatment and clinical outcome with the cytokine pattern. Low levels of IP-10, IFN-γ, MCP-1 and IL-1ß on day 0 were predictive of treatment success in a ROC analysis. In multivariate models, low levels of IL-1ß, IFN-γ and MCP-1 on day 0 were significantly associated with both treatment success and shorter time to clinical improvement. Low levels of IP-10, IL-1RA, IL-6, MCP-1 and IFN-γ on day 7 and high levels of IL-9, PDGF and RANTES on day 7 were predictive of treatment success in ROC analyses. Low levels of IP-10, MCP-1 and high levels of RANTES, on day 7 were associated with both treatment success and shorter time to clinical improvement in multivariate models.

Conclusion: This analysis demonstrates a considerable dynamic of cytokines over time, which is influenced by both treatment and clinical course of COVID-19. Levels of IL-1ß and MCP-1 at baseline and MCP-1, IP-10 and RANTES on day 7 were associated with a favorable outcome across several endpoints. These cytokines should be included in future trials for further evaluation as predictive factors.

Keywords: COVID-19; chemokines; convalescent plasma; interleukins; predictive factors; randomized trial.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Körper, Schrezenmeier, Rincon-Arevalo, Grüner, Zickler, Weiss, Wiesmann, Zacharowski, Kalbhenn, Bentz, Dollinger, Paul, Lepper, Ernst, Wulf, Zinn, Appl, Jahrsdörfer, Rojewski, Lotfi, Dörner, Jungwirth, Seifried, Fürst and Schrezenmeier.

Figures

Figure 1
Figure 1
Dynamics of serum cytokine levels in the study cohort at baseline (day 0) and day 7 after randomization. Results are presented in pg/ml. Circles show individual measurements, the horizontal lines represent medians and IQR. Groups were compared by the Kruskal-Wallis-Test with Dunn´s post hoc test. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001, ns, not significant.
Figure 2
Figure 2
Heat map of the absolute values of the correlation matrix at baseline (day 0) (A) and on day 7 (B) of all patients. Clustered cytokines are indicated by the solid lines in the dendrograms and the clusters are indicated by the colors above and beside the graph. Non-clustered cytokines are shown by the dashed lines in the dendrogram. Clusters were formed by the unweighted group method. Cluster reports for the absolute values of the correlation matrix are presented in Table 2A.
Figure 3
Figure 3
Heat map of the absolute values of the correlation matrix on day 7 stratified by randomization group: control (A) and CCP (B). Clustered cytokines are indicated by the solid lines in the dendrograms and the clusters are indicated by the colors above and beside the graph. Non-clustered cytokines are shown by the dashed lines in the dendrogram. Clusters were formed by the unweighted group method. Cluster reports for the absolute values of the correlation matrix are presented in Table 2B.
Figure 4
Figure 4
Heat map of the absolute values of the correlation matrix on day 7 in an analysis including both control group and CCP group stratified by reaching the primary endpoint on day 21: failure (A) and success (B). Clustered cytokines are indicated by the solid lines in the dendrograms and the clusters are indicated by the colors above and beside the graph. Non-clustered cytokines are shown by the dashed lines in the dendrogram. Clusters were formed by the unweighted group method. Cluster reports for the absolute values of the correlation matrix are presented in Table 2B.
Figure 5
Figure 5
Receiver operating characteristics analysis of day 0 levels of IP-10, IFN-γ, IL-1ß and MCP-1 and primary endpoint (failure versus success on day 21). Low levels of these cytokines on day 0 indicate a positive condition, i.e. patients reached the primary endpoint.(A) All patients (irrespective of allocation to randomization group). Area under the curve (AUC) and p-values for AUC >0.5 were as follows: IP-10: AUC 0.74; p=0.0002; IFN-γ: AUC 0.67, p=0.013; MCP-1: AUC 0.64, p=0.03; IL-1ß: AUC 0.64, p=0.04).(B) Patients in the CCP group Area under the curve (AUC) and p-values for AUC >0.5 were as follows: IP-10: AUC 0.82; p<0.0001; IFN-γ: AUC 0.79, p=0.0002; MCP-1: AUC 0.76, p=0.002; IL-1ß: AUC 0.70, p=0.04). (C) Comparison of day 0 levels of IP-10, IFN-γ, MCP-1 and IL-1ß (from left to right) between patients not reaching the primary endpoint (brown symbols) or reaching the primary endpoint on day 21 (green symbols). Groups were compared by Mann-Whitney test. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001.
Figure 6
Figure 6
Receiver operating characteristics analysis of day 7 cytokine levels and primary endpoint (failure vs. success on day 21). (A) All patients (irrespective of allocation to randomization group). Low levels of IP-10, IL-1RA, MCP-1, IL-6 and IFN-γ on day 7 indicate treatment success. Area under the curve (AUC) and p-values for AUC >0.5 were as follows: IP-10: AUC 0.85; p<0.0001; IL-1RA: AUC 0.78, p<0.0001; IL-6: AUC 0.72, p=0.0012; MCP-1: AUC 0.71, p=0.0033; IFN-γ: AUC 0.70, p=0.0038). (B) Patients in the CCP group. Low levels of IP-10, IL-1RA, MCP-1 and IL-6 on day 7 indicate treatment success. AUC and p-values for AUC >0.5 were as follows: IP-10: AUC 0.82; p<0.0001; IL-1RA: AUC 0.89, p<0.0001; IL-6: AUC 0.71, p=0.0345; MCP-1: AUC 0.70, p=0.0499; IFN-γ: AUC 0.69, p=0.0516). (C) All patients (irrespective of allocation to randomization group). High levels of PDGF-BB, RANTES and IL-9 on day 7 indicate treatment success. AUC and p-values for AUC >0.5 were as follows: PDGF-BB: AUC 0.78; p<0.0001; RANTES: AUC 0.68, p=0.0080; IL-9: AUC 0.65, p=0.0260). (D) Patients in the CCP group. High levels of PDGF-BB, RANTES and IL-9 on day 7 indicate treatment success. AUC and p-values for AUC >0.5 were as follows: PDGF-BB: AUC 0.84; p<0.0001; RANTES: AUC 0.77, p=0.0012; IL-9: AUC 0.69, p=0.0279). (E-L) Bivariate comparison of day 7 levels of IP-10, IL-1RA, IL-6, MCP-1, IFN-γ, IL-9, PDGF-BB and RANTES between the patients not reaching the primary endpoint (brown symbols) or reaching the primary endpoint on day 21 (green symbols). Groups were compared by Mann-Whitney test. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001. ns, not significant.
Figure 7
Figure 7
Multivariate analyses of primary endpoint (survival and no longer meeting criteria for severe COVID-19 on day 21) including cytokine levels on day 0. The following variable were included in the model: Treatment group (control group, high titer plasma, low titer plasma), age (as continuous variable), gender (female (f)) and male (m), baseline WHO Severity Score (≤4 vs. > 4) (11) and the level of the respective cytokine on day 0 (≤ median (“low”) versus > median (“high”)). One cytokine each was included in the models: IL-1ß (A), IFN-γ (B), MCP-1 (C) or IP-10 (D).
Figure 8
Figure 8
Multivariate analyses of primary endpoint (survival and no longer meeting criteria for severe COVID-19 on day 21) including cytokine levels on day 7. The following variable were included in the model: Treatment group (control group, high titer plasma, low titer plasma), age (as continuous variable), gender, baseline WHO Severity Score (≤4 vs. > 4) (11) and the level of the respective cytokine on day 7 (≤ median (“low”) versus > median (“high”)). One cytokine each was included in the models: IL-1RA (A), IFN-γ (B), MCP-1 (C), IP-10 (D), IL-6 (E), RANTES (F), PDGF-BB (G) or IL-9 (H).
Figure 9
Figure 9
Multivariate analyses of the key secondary endpoint time to clinical improvement (≤ median vs. > median) including cytokine levels on day 7. The following variables were included in the model: Treatment group (control group, high titer plasma, low titer plasma), age (as continuous variable), gender, baseline WHO Severity Score (≤4 vs. > 4) (11) and the level of the respective cytokine on day 7 (≤ median (“low”) versus > median (“high”)). One cytokine each was included in the models: IL-1RA (A), IFN-γ (B), MCP-1 (C), IP-10 (D), IL-6 (E), RANTES (F), PDGF-BB (panel G) or IL-9 (panel H).
Figure 10
Figure 10
Multivariate analyses of the key secondary endpoint time to clinical improvement (≤ median vs. > median) including cytokine levels on day 0. The following variable were included in the model: Treatment group (control group, high titer plasma, low titer plasma), age (as continuous variable), gender, baseline WHO Severity Score (≤4 vs. > 4) (11) and the level of the respective cytokine on day 0 (≤ median (“low”) versus > median (“high”)). One cytokine each was included in the models: IL-1ß (A), IFN-γ (B), MCP-1 (C) or IP-10 (D).

References

    1. Ashrafzadeh-Kian S, Campbell MR, Jara Aguirre JC, Walsh J, Kumanovics A, Jenkinson G, et al. . Role of immune mediators in predicting hospitalization of SARS-CoV-2 positive patients. Cytokine (2022) 150:155790. doi: 10.1016/j.cyto.2021.155790
    1. Bandopadhyay P, D'Rozario R, Lahiri A, Sarif J, Ray Y, Paul SR, et al. . Nature and dimensions of systemic hyperinflammation and its attenuation by convalescent plasma in severe COVID-19. J Infect Dis (2021) 224:565–74. doi: 10.1093/infdis/jiab010
    1. Guo Y, Li T, Xia X, Su B, Li H, Feng Y, et al. . Different profiles of antibodies and cytokines were found between severe and moderate COVID-19 patients. Front Immunol (2021) 12:723585. doi: 10.3389/fimmu.2021.723585
    1. Hariharan A, Hakeem AR, Radhakrishnan S, Reddy MS, Rela M. The role and therapeutic potential of NF-kappa-B pathway in severe COVID-19 patients. Inflammopharmacology (2021) 29:91–100. doi: 10.1007/s10787-020-00773-9
    1. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. . Clinical features of patients infected with 2019 novel coronavirus in wuhan, China. Lancet (2020) 395:497–506. doi: 10.1016/S0140-6736(20)30183-5
    1. Kleymenov DA, Bykonia EN, Popova LI, Mazunina EP, Gushchin VA, Kolobukhina LV, et al. . A deep look into COVID-19 severity through dynamic changes in blood cytokine levels. Front Immunol (2021) 12:771609. doi: 10.3389/fimmu.2021.771609
    1. Kwon JS, Kim JY, Kim MC, Park SY, Kim BN, Bae S, et al. . Factors of severity in patients with COVID-19: Cytokine/Chemokine concentrations, viral load, and antibody responses. Am.J.Trop.Med Hyg (2020) 103:2412–8. doi: 10.4269/ajtmh.20-1110
    1. Pons MJ, Ymana B, Mayanga-Herrera A, Saenz Y, Alvarez-Erviti L, Tapia-Rojas S, et al. . Cytokine profiles associated with worse prognosis in a hospitalized Peruvian COVID-19 cohort. Front Immunol (2021) 12:700921. doi: 10.3389/fimmu.2021.700921
    1. Sunkara H, Dewan SMR. Coronavirus disease-2019: A review on the disease exacerbation via cytokine storm and concurrent management. Int Immunopharmacol (2021) 99:108049. doi: 10.1016/j.intimp.2021.108049
    1. Körper S, Weiss M, Zickler D, Wiesmann T, Zacharowski K, Corman VM, et al. . Results of the CAPSID randomized trial for high-dose convalescent plasma in patients with severe COVID-19. J Clin Invest (2021) 131:e152264. doi: 10.1172/JCI152264
    1. World Health Organization, WHO . R&D blueprint - novel coronavirus - COVID-19 therapeutic trial synopsis. (2020). Available at:
    1. Jahrsdorfer B, Kroschel J, Ludwig C, Corman VM, Schwarz T, Korper S, et al. . Independent side-by-Side validation and comparison of 4 serological platforms for SARS-CoV-2 antibody testing. J Infect.Dis (2021) 223:796–801. doi: 10.1093/infdis/jiaa656
    1. Kreye J, Reincke SM, Kornau HC, Sanchez-Sendin E, Corman VM, Liu H, et al. . A therapeutic non-self-reactive SARS-CoV-2 antibody protects from lung pathology in a COVID-19 hamster model. Cell (2020) 183:1058–69. doi: 10.1016/j.cell.2020.09.049
    1. Wölfel R, Corman VM, Guggemos W, Seilmaier M, Zange S, Müller MA. Virological assessment of hospitalized cases of coronavirus disease 2019. Nature (2020) 581:465–9. doi: 10.1038/s41586-020-2196-x
    1. Angioni R, Sanchez-Rodriguez R, Munari F, Bertoldi N, Arcidiacono D, Cavinato S, et al. . Age-severity matched cytokine profiling reveals specific signatures in covid-19 patients. Cell Death.Dis (2020) 11:957. doi: 10.1038/s41419-020-03151-z
    1. Chen Q, Yu B, Yang Y, Huang J, Liang Y, Zhou J, et al. . Immunological and inflammatory profiles during acute and convalescent phases of severe/ critically ill COVID-19 patients. Int Immunopharmacol (2021) 97:107685. doi: 10.1016/j.intimp.2021.107685
    1. Valle DMD, Kim-Schulze S, Huang HH, Beckmann ND, Nirenberg S, Wang B, et al. . An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat Med (2020) 26:1636–43. doi: 10.1038/s41591-020-1051-9
    1. Qin L, Li X, Shi J, Yu M, Wang K, Tao Y, et al. . Gendered effects on inflammation reaction and outcome of COVID-19 patients in wuhan. J.Med Virol (2020) 92:2684–92. doi: 10.1002/jmv.26137
    1. Varchetta S, Mele D, Oliviero B, Mantovani S, Ludovisi S, Cerino A, et al. . Unique immunological profile in patients with COVID-19. Cell Mol.Immunol (2021) 18:604–12. doi: 10.1038/s41423-020-00557-9
    1. Han H, Ma Q, Li C, Liu R, Zhao L, Wang W, et al. . Profiling serum cytokines in COVID-19 patients reveals IL-6 and IL-10 are disease severity predictors. Emerg Microbes Infect (2020) 9:1123–30. doi: 10.1080/22221751.2020.1770129
    1. Lucas C, Wong P, Klein J, Castro TBR, Silva J, Sundaram M, et al. . Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature (2020) 584:463–9. doi: 10.1038/s41586-020-2588-y
    1. Zhao Y, Qin L, Zhang P, Li K, Liang L, Sun J, et al. . Longitudinal COVID-19 profiling associates IL-1RA and IL-10 with disease severity and RANTES with mild disease. JCI Insight (2020) 5 :1-11. doi: 10.1172/jci.insight.139834
    1. Chang Y, Bai M, You Q. Associations between serum interleukins (IL-1beta, IL-2, IL-4, IL-6, IL-8, and IL-10) and disease severity of COVID-19: A systematic review and meta-analysis. Biomed Res Int (2022) 2022:2755246. doi: 10.1155/2022/2755246
    1. Hsu RJ, Yu WC, Peng GR, Ye CH, Hu S, Chong PCT, et al. . The role of cytokines and chemokines in severe acute respiratory syndrome coronavirus 2 infections. Front Immunol (2022) 13:832394. doi: 10.3389/fimmu.2022.832394
    1. Ozger HS, Karakus R, Kuscu EN, Bagriacik UE, Oruklu N, Yaman M, et al. . Serial measurement of cytokines strongly predict COVID-19 outcome. PLoS One (2021) 16:e0260623. doi: 10.1371/journal.pone.0260623
    1. Alshammary AF, Alsughayyir JM, Alharbi KK, Al-Sulaiman AM, Alshammary HF, Alshammary HF. T-Cell subsets and interleukin-10 levels are predictors of severity and mortality in COVID-19: A systematic review and meta-analysis. Front Med (Lausanne) (2022) 9:852749. doi: 10.3389/fmed.2022.852749
    1. Zeng Z, Yu H, Chen H, Qi W, Chen L, Chen G, et al. . Longitudinal changes of inflammatory parameters and their correlation with disease severity and outcomes in patients with COVID-19 from wuhan, China. Crit Care (2020) 24:525. doi: 10.1186/s13054-020-03255-0
    1. Herr C, Mang S, Mozafari B, Guenther K, Speer T, Seibert M, et al. . Distinct patterns of blood cytokines beyond a cytokine storm predict mortality in COVID-19. J Inflamm Res (2021) 14:4651–67. doi: 10.2147/JIR.S320685
    1. Chen IY, Moriyama M, Chang MF, Ichinohe T. Severe acute respiratory syndrome coronavirus viroporin 3a activates the NLRP3 inflammasome. Front Microbiol (2019) 10:50. doi: 10.3389/fmicb.2019.00050
    1. Ferreira AC, Soares VC, de Azevedo-Quintanilha IG, Dias SDSG, Fintelman-Rodrigues N, Sacramento CQ, et al. . SARS-CoV-2 engages inflammasome and pyroptosis in human primary monocytes. Cell Death.Discov (2021) 7:43. doi: 10.1038/s41420-021-00428-w
    1. Junqueira C, Crespo A, Ranjbar S, de Lacerda LB, Lewandrowski M, Ingber J, et al. . FcgammaR-mediated SARS-CoV-2 infection of monocytes activates inflammation. Nature (2022) 606:576–84. doi: 10.1038/s41586-022-04702-4
    1. Costa LS da, Outlioua A, Anginot A, Akarid K, Arnoult D. RNA Viruses promote activation of the NLRP3 inflammasome through cytopathogenic effect-induced potassium efflux. Cell Death.Dis (2019) 10:346. doi: 10.1038/s41419-019-1579-0
    1. Darif D, Hammi I, Kihel A, El IS, Guessous I,F, Akarid K. The pro-inflammatory cytokines in COVID-19 pathogenesis: What goes wrong? Microb Pathog (2021) 153:104799. doi: 10.1016/j.micpath.2021.104799
    1. Youn JY, Zhang Y, Wu Y, Cannesson M, Cai H. Therapeutic application of estrogen for COVID-19: Attenuation of SARS-CoV-2 spike protein and IL-6 stimulated, ACE2-dependent NOX2 activation, ROS production and MCP-1 upregulation in endothelial cells. Redox Biol (2021) 46:102099. doi: 10.1016/j.redox.2021.102099
    1. Sugrue JA, Bourke NM, O'Farrelly C. Type I interferon and the spectrum of susceptibility to viral infection and autoimmune disease: A shared genomic signature. Front Immunol (2021) 12:757249. doi: 10.3389/fimmu.2021.757249
    1. Prenzler S, Rudrawar S, Waespy M, Kelm S, Anoopkumar-Dukie S, Haselhorst T. The role of sialic acid-binding immunoglobulin-like-lectin-1 (siglec-1) in immunology and infectious disease. Int Rev.Immunol (2021), 1–26. doi: 10.1080/08830185.2021.1931171
    1. Rincon-Arevalo H, Aue A, Ritter J, Szelinski F, Khadzhynov D, Zickler D, et al. . Altered increase in STAT1 expression and phosphorylation in severe COVID-19. Eur J Immunol (2021) 52: 138–48. doi: 10.1101/2021.08.13.21262006
    1. Zhang Q, Bastard P, Liu Z, Le PJ, Moncada-Velez M, Chen J, et al. . Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science (2020) 370: 1–8. doi: 10.1126/science.abd4570
    1. Bastard P, Rosen LB, Zhang Q, Michailidis E, Hoffmann HH, Zhang Y, et al. . Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science (2020) 370: 1–17. doi: 10.1126/science.abd4585
    1. Schuhenn J, Meister TL, Todt D, Bracht T, Schork K, Billaud JN, et al. . Differential interferon-alpha subtype induced immune signatures are associated with suppression of SARS-CoV-2 infection. Proc Natl Acad Sci U S A (2022) 119: 1–12. doi: 10.1073/pnas.2111600119
    1. Giannella A, Riccetti S, Sinigaglia A, Piubelli C, Razzaboni E, Di BP, et al. . Circulating microRNA signatures associated with disease severity and outcome in COVID-19 patients. Front Immunol (2022) 13: 968991. doi: 10.3389/fimmu.2022.968991
    1. Kulasinghe A, Tan CW, Ribeiro Dos Santos Miggiolaro AF, Monkman J, SadeghiRad H, Bhuva DD, et al. . Profiling of lung SARS-CoV-2 and influenza virus infection dissects virus-specific host responses and gene signatures. Eur Respir J (2022) 59 :1–19. doi: 10.1183/13993003.01881-2021
    1. Liu Y, Wu Y, Liu B, Zhang Y, San D, Chen Y, et al. . Biomarkers and immune repertoire metrics identified by peripheral blood transcriptomic sequencing reveal the pathogenesis of COVID-19. Front Immunol (2021) 12:677025. doi: 10.3389/fimmu.2021.677025
    1. Persson J, Andersson B, van VS, Haks MC, Obudulu O, Torkzadeh S, et al. . Stratification of COVID-19 patients based on quantitative immune-related gene expression in whole blood. Mol Immunol (2022) 145:17–26. doi: 10.1016/j.molimm.2022.03.004
    1. Yekelchyk M, Madan E, Wilhelm J, Short KR, Palma AM, Liao L, et al. . Flower lose, a cell fitness marker, predicts COVID-19 prognosis. EMBO Mol.Med (2021) 13:e13714. doi: 10.15252/emmm.202013714
    1. Davidson M, Menon S, Chaimani A, Evrenoglou T, Ghosn L, Grana C, et al. . Interleukin-1 blocking agents for treating COVID-19. Cochrane.Database.Syst.Rev 1 (2022), CD015308. doi: 10.1002/14651858.CD015308
    1. Coperchini F, Chiovato L, Croce L, Magri F, Rotondi M. The cytokine storm in COVID-19: An overview of the involvement of the chemokine/chemokine-receptor system. Cytokine Growth Factor Rev (2020) 53: 25–32. doi: 10.1016/j.cytogfr.2020.05.003
    1. Teixeira PC, Dorneles GP, Santana Filho PC, da Silva IM, Schipper LL, Postiga IAL, et al. . Increased LPS levels coexist with systemic inflammation and result in monocyte activation in severe COVID-19 patients. Int Immunopharmacol (2021) 100: 108125. doi: 10.1016/j.intimp.2021.108125
    1. Avendano-Sola C, Ramos-Martinez A, Munez-Rubio E, Ruiz-Antoran B, Malo de MR, Torres F, et al. . A multicenter randomized open-label clinical trial for convalescent plasma in patients hospitalized with COVID-19 pneumonia. J Clin Invest (2021) 131 :1-11. doi: 10.1172/JCI152740
    1. Devos T, Van TQ, Compernolle V, Najdovski T, Romano M, Dauby N, et al. . Early high antibody titre convalescent plasma for hospitalised COVID-19 patients: DAWn-plasma. Eur Respir J (2022) 59 :1-2. doi: 10.1183/13993003.01724-2021
    1. O'Donnell MR, Grinsztejn B, Cummings MJ, Justman JE, Lamb MR, Eckhardt CM, et al. . A randomized double-blind controlled trial of convalescent plasma in adults with severe COVID-19. J Clin Invest (2021) 131 :1-10. doi: 10.1172/JCI150646
    1. RECOVERY Collaborative Group . Convalescent plasma in patients admitted to hospital with COVID-19 (RECOVERY): a randomised controlled, open-label, platform trial. Lancet (2021) 397:2049–59. doi: 10.1016/S0140-6736(21)00897-7
    1. Simonovich VA, Burgos Pratx LD, Scibona P, Beruto MV, Vallone MG, Vazquez C, et al. . A randomized trial of convalescent plasma in covid-19 severe pneumonia. N.Engl.J Med (2020) 384:619–29. doi: 10.1056/NEJMoa2031304

Source: PubMed

3
Se inscrever