Safety and long-term improvement of mesenchymal stromal cell infusion in critically COVID-19 patients: a randomized clinical trial

Carmen Lúcia Kuniyoshi Rebelatto, Alexandra Cristina Senegaglia, Claudio Luciano Franck, Debora Regina Daga, Patrícia Shigunov, Marco Augusto Stimamiglio, Daniela Boscaro Marsaro, Bruna Schaidt, Andressa Micosky, Ana Paula de Azambuja, Cleverson Alex Leitão, Ricardo Rasmussen Petterle, Valderez Ravaglio Jamur, Isadora May Vaz, Antônio Paulo Mallmann, Hipólito Carraro Junior, Eduardo Ditzel, Paulo Roberto Slud Brofman, Alejandro Correa, Carmen Lúcia Kuniyoshi Rebelatto, Alexandra Cristina Senegaglia, Claudio Luciano Franck, Debora Regina Daga, Patrícia Shigunov, Marco Augusto Stimamiglio, Daniela Boscaro Marsaro, Bruna Schaidt, Andressa Micosky, Ana Paula de Azambuja, Cleverson Alex Leitão, Ricardo Rasmussen Petterle, Valderez Ravaglio Jamur, Isadora May Vaz, Antônio Paulo Mallmann, Hipólito Carraro Junior, Eduardo Ditzel, Paulo Roberto Slud Brofman, Alejandro Correa

Abstract

Background: COVID-19 is a multisystem disease that presents acute and persistent symptoms, the postacute sequelae (PASC). Long-term symptoms may be due to consequences from organ or tissue injury caused by SARS-CoV-2, associated clotting or inflammatory processes during acute COVID-19. Various strategies are being chosen by clinicians to prevent severe cases of COVID-19; however, a single treatment would not be efficient in treating such a complex disease. Mesenchymal stromal cells (MSCs) are known for their immunomodulatory properties and regeneration ability; therefore, they are a promising tool for treating disorders involving immune dysregulation and extensive tissue damage, as is the case with COVID-19. This study aimed to assess the safety and explore the long-term efficacy of three intravenous doses of UC-MSCs (umbilical cord MSCs) as an adjunctive therapy in the recovery and postacute sequelae reduction caused by COVID-19. To our knowledge, this is one of the few reports that presents the longest follow-up after MSC treatment in COVID-19 patients.

Methods: This was a phase I/II, prospective, single-center, randomized, double-blind, placebo-controlled clinical trial. Seventeen patients diagnosed with COVID-19 who require intensive care surveillance and invasive mechanical ventilation-critically ill patients-were included. The patient infusion was three doses of 5 × 105 cells/kg UC-MSCs, with a dosing interval of 48 h (n = 11) or placebo (n = 6). The evaluations consisted of a clinical assessment, viral load, laboratory testing, including blood count, serologic, biochemical, cell subpopulation, cytokines and CT scan.

Results: The results revealed that in the UC-MSC group, there was a reduction in the levels of ferritin, IL-6 and MCP1-CCL2 on the fourteen day. In the second month, a decrease in the levels of reactive C-protein, D-dimer and neutrophils and an increase in the numbers of TCD3, TCD4 and NK lymphocytes were observed. A decrease in extension of lung damage was observed at the fourth month. The improvement in all these parameters was maintained until the end of patient follow-up.

Conclusions: UC-MSCs infusion is safe and can play an important role as an adjunctive therapy, both in the early stages, preventing severe complications and in the chronic phase with postacute sequelae reduction in critically ill COVID-19 patients. Trial registration Brazilian Registry of Clinical Trials (ReBEC), UTN code-U1111-1254-9819. Registered 31 October 2020-Retrospectively registered, https://ensaiosclinicos.gov.br/rg/RBR-3fz9yr.

Keywords: COVID-19; Cell therapy; Mesenchymal stromal cells; Postacute sequelae.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Flow chart for patient enrollment, intervention and follow-up
Fig. 2
Fig. 2
Characterization and quality control for UC-MSC. A Representative image of cell differentiation. (a, c, e) Control cells; (b) Cells differentiated into adipocytes characterized by the presence of lipidic vacuoles stained with Oil Red O; (d) Cells differentiated into osteoblasts characterized by the presence of calcium deposits stained with Alizarin Red S (red); (f) Presence of vacuoles around young chondrocytes and proteoglycan in the matrix. B Representative histograms of UC-MSC surface markers, cell viability and apoptosis/necrosis. The isotype control is shown as a red line histogram. C UC-MSC karyogram after cell expansion. Normal karyotype: 46, XX. D Representative histograms from the lymphocyte inhibition assay. MSCs were cultivated with PHA stimulated CD3 + lymphocytes labeled with CFSE. (a) CD3 + lymphocytes not labeled with CFSE; (b) CFSE-labeled CD3 + lymphocytes; (c) CD3 + lymphocytes labeled with CFSE and stimulated with PHA (1 µg/µL); (d) MSCs were cultivated with CD3 + lymphocytes labeled with CFSE 1:10; (e) MSCs were cultivated with CD3 + lymphocytes labeled with CFSE 1:5; (f) MSCs were cultivated with CD3 + lymphocytes labeled with CFSE in a 1:2 ratio
Fig. 3
Fig. 3
Viral load from day 1 (baseline) to 14 days after UC-MSC treatment. AD Graphs representing relative viral quantification. Viral load was determined based on the relative expression of the viral gene RdRP in relation to the human POLR2B gene (normalizer). Viral gene expression gradually decreased in patient samples from day 1 (baseline) to day 14 (A, B). C, D After linearizing the data by the natural logarithm (ln (x)) and obtaining the linear equation, the angular coefficient of the viral load line (slope) and the coefficient of determination (R2) were established. C UC-MSC and D Placebo. The angular coefficient (slope) and R2 of each patient are plotted in (E). Average of the angular coefficient of each group (F). Mean with SEM; Student's unpaired t test analysis. UC-MSC, umbilical cord mesenchymal stromal cell; ns, not significant
Fig. 4
Fig. 4
Coagulation parameters and inflammatory markers. Comparison between UC-MSC (red line) and placebo (blue line) groups over time. The bars show standard deviations (SD). *p ≤ 0.05 for comparison between groups (black asterisk). Statistical differences inside groups, longitudinal follow-up, were depicted in the graphs (UC-MSC—red line, placebo—blue line). *p ≤ 0.05 and **p ≤ 0.01 for longitudinal follow-up (red and blue asterisks). A D-dimer, B Platelets, C Neutrophils, D Ferritin and E C-Reactive Protein. UC-MSCs, umbilical cord mesenchymal stromal cells
Fig. 5
Fig. 5
Profile of plasma cytokines, chemokines and growth factors in the patients during different clinical stages. Comparison between groups (UC-MSCs vs Placebo) and evaluated time points (baseline, days 2, 4, 6, 14 and 2 and 4 months). A Heatmap jointly comparing all analytes evaluated over the treatment time, either with cell therapy (UC-MSCs group) or placebo. Graphs show the results of some analytes separately, comparing the UC-MSC (red line) and placebo (blue line) groups over time. The bars show standard deviations (SD), and the broken line is the trend line fitted to the data. B Granulocyte–Macrophage Colony-Stimulating Factor (GM‐CSF); C Interleukin 2 (IL-2); D Macrophage Inflammatory Protein 1-Alpha (MIP1a/CCL3); E Tumor Necrosis Factor (TNF); F Interleukin 6 (IL-6); G Interleukin 8 (IL-8); H Interleukin 10 (IL-10); I Monocyte chemoattractant protein-1 (MCP1-CCL2); J Interleukin 7 (IL-7). Abbreviations: UC-MSCs, Umbilical Cord Mesenchymal Stromal Cells. *p ≤ 0.04 and **p ≤ 0.02 for comparison between groups. Statistical differences inside groups, longitudinal follow-up, were not depicted in the graphs but indicated in the main text
Fig. 6
Fig. 6
Analysis of cell subpopulation. A Total lymphocytes, B CD3 T lymphocytes, C CD4 T lymphocytes, D Natural killer. Comparison between UC-MSC (red line) and placebo (blue line) groups over time. The bars show standard deviations (SD). *p ≤ 0.05 for comparison between groups (black asterisk). Statistical differences inside groups, longitudinal follow-up, were depicted in the graphs (UC-MSC—red line, placebo—blue line). *p ≤ 0.05 and **p ≤ 0.01 for longitudinal follow-up (red and blue asterisks). Flow cytometry strategy for lymphocyte analysis, total lympohocytes, CD3 T, CD8 T and CD4 T lymphocytes. E Lymphocyte gate FSC/SSC low; F Strong CD45 +  + ; G CD3 positive; H CD4/CD8 dotplot; I CD4 positive. UC-MSCs, umbilical cord mesenchymal stromal cells; SSC, side scatter; FSC, forward scatter
Fig. 7
Fig. 7
Cardiac and kidney function markers. Comparison between UC-MSC (red line) and placebo (blue line) groups over time. The bars show standard deviations (SD). *p ≤ 0.05 for comparison between groups (black asterisk). Statistical differences inside groups, longitudinal follow-up, were depicted in the graphs (UC-MSC—red line, placebo—blue line). *p ≤ 0.05 and **p ≤ 0.01 for longitudinal follow-up (red and blue asterisks). A Troponin I and B Creatinine. UC-MSCs, umbilical cord mesenchymal stromal cells
Fig. 8
Fig. 8
Representative images from chest CT at the level of the lower lobes in patients from the treatment and control groups. Red arrows point to ground-glass opacification, blue arrows to peripheral consolidations, and the yellow arrow to crazy-paving, all of which are typical features of COVID-19. It is possible to note the higher degree of clearance in patients from the treatment group (first line) compared to the control group (last line). UC-MSC, umbilical cord mesenchymal stromal cell

References

    1. Ahn DG, Shin HJ, Kim MH, Lee S, Kim HS, Myoung J, et al. Current status of epidemiology, diagnosis, therapeutics, and vaccines for novel coronavirus disease 2019 (COVID-19) J Microbiol Biotechnol. 2020;30:313–324.
    1. Jin Y, Yang H, Ji W, Wu W, Chen S, Zhang W, et al. Virology, epidemiology, pathogenesis, and control of COVID-19. Viruses. 2020;12:372.
    1. Khan M, Adil SF, Alkhathlan HZ, Tahir MN, Saif S, Khan M, et al. COVID-19: A global challenge with old history, epidemiology and progress so far. Molecules. 2020;26:39.
    1. World Health Organization. World Health Organization COVID-19 dashboard. Genebra: WHO. (2021). Accessed 13 Fev 2022.
    1. Pollard CA, Morran MP, Nestor-Kalinoski AL. The COVID-19 pandemic: a global health crisis. Physiol Genomics. 2020;52:549–557.
    1. Preskorn SH. COVID-19: why has the mortality rate declined? J Psychiatr Pract. 2020;26:394–399.
    1. Grasselli G, Greco M, Zanella A, Albano G, Antonelli M, Bellani G, et al. Risk factors associated with mortality among patients with COVID-19 in Intensive Care Units in Lombardy. Italy JAMA Intern Med. 2020;180:1345–1355.
    1. Secretaria da Saúde, Governo do Estado do Paraná. Boletim epidemiológico coronavírus 31-07-2021 (COVID-19). Curitiba: SESA. (2021). Accessed 14 Aug 2021.
    1. Secretaria da Saúde, Governo do Estado do Paraná. Boletim epidemiológico coronavírus 30-08-2021 (COVID-19). Curitiba: SESA. (2021). Accessed 14 Aug 2021.
    1. Armstrong RA, Kane AD, Cook TM. Outcomes from intensive care in patients with COVID-19: a systematic review and meta-analysis of observational studies. Anesthesia. 2020;75:1340–1349.
    1. de Andrade CLT, Pereira CCA, Martins M, Lima SML, Portela MC. COVID-19 hospitalizations in Brazil's Unified Health System (SUS) PLoS ONE. 2020;15:e0243126.
    1. Tomazini BM, Maia IS, Cavalcanti AB, Berwanger O, Rosa RG, Veiga VC, et al. Effect of dexamethasone on days alive and ventilator-free in patients with moderate or severe acute respiratory distress syndrome and COVID-19: The CoDEX Randomized Clinical Trial. JAMA. 2020;324:1307–1316.
    1. Sousa GJB, Garces TS, Cestari VRF, Florêncio RS, Moreira TMM, Pereira MLD. Mortality and survival of COVID-19. Epidemiol Infect. 2020;148:e123.
    1. Huertas A, Montani D, Savale L, Pichon J, Tu L, Parent F, et al. Endothelial cell dysfunction: a major player in SARS-CoV-2 infection (COVID-19)? Eur Respir J. 2020;56:2001634.
    1. Pons S, Fodil S, Azoulay E, Zafrani L. The vascular endothelium: the cornerstone of organ dysfunction in severe SARS-CoV-2 infection. Crit Care. 2020;24:353.
    1. Lopes-Pacheco M, Silva PL, Cruz FF, Battaglini D, Robba C, Pelosi P, et al. Pathogenesis of multiple organ injury in COVID-19 and potential therapeutic strategies. Front Physiol. 2021;12:593223.
    1. Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130:2620–2629.
    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.
    1. Proal AD, VanElzakker MB. Long COVID or post-acute sequelae of COVID-19 (PASC): an overview of biological factors that may contribute to persistent symptoms. Front Microbiol. 2021;12:698169.
    1. Del Rio C, Collins LF, Malani P. Long-term health consequences of COVID-19. JAMA. 2020;324:1723–1724.
    1. Adeloye D, Elneima O, Daines L, Poinasamy K, Quint JK, Walker S, et al. The long-term sequelae of COVID-19: an international consensus on research priorities for patients with pre existing and new-onset airways disease. Lancet Respir Med. 2021;9:1467–1478.
    1. Bell ML, Catalfamo CJ, Farland LV, Ernst KC, Jacobs ET, Klimentidis YC, et al. Postacute sequelae of COVID-19 in a nonhospitalized cohort: results from the Arizona CoVHORT. PLoS ONE. 2021;16:e0254347.
    1. Johnsen S, Sattler SM, Miskowiak KW, Kunalan K, Victor A, Pedersen L, et al. Descriptive analysis of long COVID sequelae identified in a multidisciplinary clinic serving hospitalised and non-hospitalised patients. ERJ Open Res. 2021;7:00205–2021.
    1. Logue JK, Franko NM, McCulloch DJ, McDonald D, Magedson A, Wolf CR, et al. Sequelae in adults at 6 months after COVID-19 infection. JAMA Netw Open. 2021;4:e210830.
    1. Li C, Zhao H, Wang B. Challenges for mesenchymal stem cell-based therapy for COVID-19. Drug Des Dev Ther. 2020;14:3995–4001.
    1. Zhang JY, Wang XM, Xing X, Xu Z, Zhang C, Song JW, et al. Single-cell landscape of immunological responses in patients with COVID-19. Nat Immunol. 2020;21:1107–1118.
    1. Tufan A, Avanoğlu Güler A, Matucci-Cerinic M. COVID-19, immune system response, hyperinflammation and repurposing antirheumatic drugs. Turk J Med Sci. 2020;50:620–632.
    1. Bamba C, Singh SP, Choudhury S. Can mesenchymal stem cell therapy be the interim management of COVID-19? Drug Discov Ther. 2020;14:139–142.
    1. Huang C, Huang L, Wang Y, Li X, Ren L, Gu X, et al. 6-month consequences of COVID-19 in patients discharged from hospital: a cohort study. Lancet. 2021;397:220–232.
    1. Prockop DJ, Oh JY. Mesenchymal stem/stromal cells (MSCs): role as guardians of inflammation. Mol Ther. 2012;20:14–20.
    1. Prockop DJ. The exciting prospects of new therapies with mesenchymal stromal cells. Cytotherapy. 2017;19:1–8.
    1. Sutton MT, Fletcher D, Ghosh SK, Weinberg A, van Heeckeren R, Kaur S, et al. Antimicrobial properties of mesenchymal stem cells: therapeutic potential for cystic fibrosis infection, and treatment. Stem Cells Int. 2016;2016:5303048.
    1. Alcayaga-miranda F, Cuenca J, Khoury M. Antimicrobial activity of mesenchymal stem cells: current status and new perspectives of antimicrobial peptide-based therapies. Front Immunol. 2017;8:339.
    1. Lopes-Pacheco M, Robba C, Rocco PR, Pelosi P. Current undesrtanding of the therapeutic benefits of mesenchymal stem cells in acute respiratory distress syndrome. Cell Biol Toxicol. 2020;36:83–102.
    1. Enes SR, Krasnodembskaya AD, English K, Dantos CC, Weiss DJ. Research Progress on strategies that can enhance the therapeutic benefits of mesenchymal stromal cells in respiratory diseases with a specific focus on acute respiratory distress syndrome and other inflammatory lung diseases. Front Pharmacol. 2021;12:647652.
    1. Akkoc T. COVID-19, and mesenchymal stem cell tretament: mystery or not. Adv Exp Med Biol. 2020;10:167–176.
    1. Pinky, Gupta S, Krishnakumar V, Sharma Y, Dinda AK, Mohanty S. Mesenchymal stem cell derived exosomes: a nano platform for therapeutics and drug delivery in combating COVID-19. Stem Cell Rev Rep. 2021;17:33–43.
    1. Kuba K, Imai Y, Rao S, Gao H, Guo F, Guan B, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat Med. 2005;11:875–879.
    1. Ge XY, Li JL, Yang XL, Chmura AA, Zhu G, Epstein JH, et al. Isolation and chracterization of a bat SARS-like coronavirus that uses the ACE2 receptor. Nature. 2013;503:535–538.
    1. Leng Z, Zhu R, Hou W, Feng Y, Yang Y, Han Q, et al. Transplantation of ACE2- mesenchymal stem cells improves the outcome of patients with COVID-19 pneumonia. Aging Dis. 2020;11:216–228.
    1. Stolzing A, Jones E, McGonagle D, Scutt A. Age-related changes in human bone marrow-derived mesenchymal stem cells: consequences for cell therapies. Mech Ageing Dev. 2008;129:163–173.
    1. Wagner W, Bork S, Horn P, Krunic D, Walenda T, Diehlmann A, et al. Aging and replicative senescence have related effects on human stem and progenitor cells. PLoS ONE. 2009;4:e5846.
    1. Fischer UM, Harting MT, Jimenez F, Monzon-Posadas WO, Xue H, Savitz SI, et al. Pulmonary passage is a major obstacle for intravenous stem cell delivery: the pulmonary. Stem Cells Dev. 2009;18:683–691.
    1. Li X, An G, Wang Y, Liang D, Zhu Z, Tian L. Targeted migration of bone marrow mesenchymal stem cells inhibits silica-induced pulmonary fibrosis in rats. Stem Cell Res Ther. 2018;9:335.
    1. Atluri S, Manocha V, Boddu N, Bhati S, Syed Z, Diwan S, et al. Safety and effectiveness of intravascular mesenchymal stem cells to treat organ failure and possible application in COVID-19 complications. Pain Phys. 2020;23:391–420.
    1. Feng Y, Huang J, Wu J, Xu Y, Chen B, Jiang L, et al. Safety and feasibility of umbilical cord mesenchymal stem cells in patients with COVID-19 pneumonia: a pilot study. Cell Prolif. 2020;53:e12947.
    1. Lanzoni G, Linetsky E, Correa D, Messinger Cayetano S, Alvarez RA, Kouroupis D. Umbilical cord mesenchymal stem cells for COVID-19 acute respiratory distress syndrome: a double-blind, phase 1/2a, randomized controlled trial. Stem Cells Transl Med. 2021;10:660–673.
    1. Meng F, Xu R, Wang S, Xu Z, Zhang C, Li Y, et al. Human umbilical cord-derived mesenchymal stem cell therapy in patients with COVID-19: a phase 1 clinical trial. Signal Transduct Target Ther. 2020;5:172.
    1. Shu L, Niu C, Li R, Huang T, Wang Y, Huang M, et al. Treatment of severe COVID-19 with human umbilical cord mesenchymal stem cells. Stem Cell Res Ther. 2020;11:361.
    1. Tang L, Jiang Y, Zhu M, Chen L, Zhou X, Zhou C, et al. Clinical study using mesenchymal stem cells for the treatment of patients with severe COVID-19. Front Med. 2020;14:664–673.
    1. Adas G, Cukurova Z, Yasar KK, Yilmaz R, Isiksacan N, Kasapoglu P, et al. The systematic effect of mesenchymal stem cell therapy in critical COVID-19 patients: a prospective double controlled trial. Cell Transplant. 2021;30:1–41.
    1. Dilogo IH, Aditianingsih D, Sugiarto A, Burhan E, Damayanti T, Sitompul PA, et al. Umbilical cord mesenchymal stromal cells as critical COVID-19 adjuvant therapy: a randomized controlled trial. Stem Cells Transl Med. 2021;10:1279–1287.
    1. Hashemian SR, Aliannejad R, Zarrabi M, Soleimani M, Vosough M, Hosseini SE, et al. Mesenchymal stem cells derived from perinatal tissues for treatment of critically ill COVID-19-induced ARDS patients: a case series. Stem Cell Res Ther. 2021;12:91.
    1. Saleh M, Vaezi AA, Aliannejad R, Sohrabpour AA, Kiaei SZF, Shadnoush M, et al. Cell therapy in patients with COVID-19 using Wharton's jelly mesenchymal stem cells: a phase 1 clinical trial. Stem Cell Res Ther. 2021;12:410.
    1. Shi L, Huang H, Lu X, Yan X, Jiang X, Xu R, et al. Effect of human umbilical cord-derived mesenchymal stem cells on lung damage in severe COVID-19 patients: a randomized, double-blind, placebo-controlled phase 2 trial. Signal Transduct Target Ther. 2021;6:58.
    1. Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8:420–422.
    1. Zhu R, Yan T, Feng Y, Liu Y, Cao H, Peng G, et al. Mesenchymal stem cell treatment improves outcome of COVID-19 patients via multiple immunomodulatory mechanisms. Cell Res. 2021;31:1244–1262.
    1. Shi L, Yuan X, Yao W, Wang S, Zhang C, Zhang B, et al. Human mesenchymal stem cells treatment for severe COVID-19: 1-year follow-up results of a randomized, double-blind, placebo-controlled trial. 2022; 75:103789.
    1. Rebelatto CK, Aguiar AM, Moretão MP, Senegaglia AC, Hansen P, Barchiki F, et al. Dissimilar differentiation of mesenchymal stem cells from bone marrow, umbilical cord blood, and adipose tissue. Exp Biol Med (Maywood) 2008;233:901–913.
    1. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–7.
    1. Borgonovo T, Vaz IM, Senegaglia AC, Rebelatto CLK, Brofman PRS. Genetic evaluation of mesenchymal stem cells by G-banded karyotyping in a Cell Technology Center. Rev Bras Hematol Hemoter. 2014;36:202–207.
    1. Corman VM, Landt O, Kaiser M, Molenkamp R, Meijer A, Chu DK, et al. Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT–PCR. Euro Surveill. 2020;25:23–30.
    1. Kalina T, Flores-Montero J, van der Velden VH, Martin-Ayuso M, Böttcher S, Ritgen M, et al. EuroFlow standardization of flow cytometer instrument settings and immunophenotyping protocols. Leukemia. 2012;26:1986–2010.
    1. Flores-Montero J, Grigore G, Fluxá R, Hernández J, Fernandez P, Almeida J, et al. EuroFlow lymphoid screening tube (LST) data base for automated identification of blood lymphocyte subsets. J Immunol Methods. 2019;475:112662.
    1. Li K, Wu J, Wu F, Guo D, Chen L, Fang Z, et al. The clinical and chest ct features associated with severe and critical COVID-19 pneumonia. Invest Radiol. 2020;55:327–331.
    1. Masarotto G, Varin C. Gaussian copula marginal regression. Electron J Stat. 2012;6:1517–1549.
    1. Bretz F, Hothorn T, Westfall P. Multiple comparisons using R. 1. New York: Chapman and Hall/CRC; 2010.
    1. Dessau RB, Pipper CB. R–en programpakke til statistisk databehandling og grafik ["R"-project for statistical computing] Ugeskr Laeger. 2008;170:328–330.
    1. Masarotto G, Varin C. Gaussian copula regression in R. J Stat Softw. 2017;77:1–26.
    1. Öztürk S, Elçin AE, Elçin YM. Mesenchymal stem cells for coronavirus (COVID-19)-induced pneumonia: revisiting the paracrine hypothesis with new hopes? Aging Dis. 2020;11:477–479.
    1. Galipeau J, Krampera M, Barrett J, Dazzi F, Deans RJ, DeBruijn J, et al. International Society for Cellular Therapy perspective on immune functional assays for mesenchymal stromal cells as potency release criterion for advanced phase clinical trials. Cytotherapy. 2016;18:151–159.
    1. Moll G, Geißler S, Catar R, Ignatowicz L, Hoogduijn MJ, Strunk D, et al. Cryopreserved or fresh mesenchymal stromal cells: only a matter of taste or key to unleash the full clinical potential of msc therapy? Adv Exp Med Biol. 2016;951:77–98.
    1. Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L, et al. Dexamethasone in hospitalized patients with COVID-19. N Engl J Med. 2021;384:693–704.
    1. Vargas A, Boivin R, Cano P, Murcia Y, Bazin I, Lavoie JP. Neutrophil extracellular traps are downregulated by glucocorticosteroids in lungs in an equine model of asthma. Respir Res. 2017;18:207.
    1. Sun Y, Chen C, Zhang X, Wang S, Zhu R, Zhou A, et al. Heparin improves alveolarization and vascular development in hyperoxia-induced bronchopulmonary dysplasia by inhibiting neutrophil extracellular traps. Biochem Biophys Res Commun. 2020;522:33–39.
    1. Wang T, Chen R, Liu C, Liang W, Guan W, Tang R, et al. Attention should be paid to venous thromboembolism prophylaxis in the management of COVID-19. Lancet Haematol. 2020;7:e362–e363.
    1. Sharma D, Zhao F. Updates on clinical trials evaluating the regenerative potential of allogenic mesenchymal stem cells in COVID-19. NPJ Regen Med. 2021;6:1–11.
    1. Guo Z, Chen Y, Luo X, He X, Zhang Y, Wang J. Administration of umbilical cord mesenchymal stem cells in patients with severe COVID-19 pneumonia. Crit Care. 2020;24:420.
    1. Sánchez-Guijo F, García-Arranz M, López-Parra M, Monedero P, Mata-Martínez C, Santos A, et al. Adipose-derived mesenchymal stromal cells for the treatment of patients with severe SARS-CoV-2 pneumonia requiring mechanical ventilation. A proof of concept study. EClinicalMedicine. 2020;25:100454.
    1. Ranzani OT, Bastos LSL, Gelli JGM, Marchesi JF, Baião F, Hamacher S, et al. Characterisation of the first 250,000 hospital admissions for COVID-19 in Brazil: a retrospective analysis of nationwide data. Lancet Respir Med. 2021;9:407–418.
    1. Patel SK, Velkoska E, Burrell LM. Emerging markers in cardiovascular disease: where does angiotensin-converting enzyme 2 fit in? Clin Exp Pharmacol Physiol. 2013;40:551–559.
    1. Bunders M, Altfeld M. Implications of sex differences in immunity for SARS-CoV-2 Pathogenesis and design of therapeutic interventions. Immunity. 2020;53:487–495.
    1. Li Q, Guan X, Wu P, Wang X, Zhou L, Tong Y, et al. early transmission dynamics in Wuhan, China, of novel coronavirus–infected pneumonia. N Engl J Med. 2020;382:1199–1207.
    1. Zhang JJ, Dong X, Cao YY, Yuan YD, Yang YB, Yan YQ, et al. Clinical characteristics of 140 patients infected with SARS-CoV-2 in Wuhan. China Allergy. 2020;75:1730–1741.
    1. Chan L, Chaudhary K, Saha A, Chauhan K, Vaid A, Baweja M, et al. Acute kidney injury in hospitalized patients with COVID-19. J Am Soc Nephrol. 2020;32:151–160.
    1. Robbins-Juarez SY, Qian L, King KL, Stevens JS, Husain SA, Radhakrishnan J, et al. Outcomes for patients with COVID-19 and acute kidney injury: a systematic review and meta-analysis. Kidney Int Rep. 2020;5:1149–1160.
    1. Kellum JA, van Till JWO, Mulligan G. Targeting acute kidney injury in COVID-19. Nephrol Dial Transplant. 2020;35:1652–1662.
    1. Arikan H, Ozturk S, Tokgoz B, Dursun B, Seyahi N, Trabulus S, et al. Characteristics and outcomes of acute kidney injury in hospitalized COVID-19 patients: a multicenter study by the Turkish society of nephrology. PLos ONE. 2021;16:e0256023.
    1. Ghonimi TAL, Alkad MM, Abuhelaiqa EA, Othman MM, Elgaali MA, Ibrahim RAM, et al. Mortality and associated risk factors for COVID-19 infection in dialysis patients in Qatar: a nationwide cohort study. PLoS ONE. 2021;16:e0254246.
    1. Costa RLD, Sória TC, Salles EF, Gerecht AV, Corvisier MF, Menezes MAM, et al. Acute kidney injury in patients with COVID-19 in a Brazilian ICU: incidence, predictors and in-hospital mortality. J Bras Nefrol. 2020;43:349–358.
    1. Lino K, Guimarães GMC, Alves LS, Oliveira AC, Faustino R, Fernandes CS, et al. Serum ferritin at admission in hospitalized COVID-19 patients as a predictor of mortality. Braz J Infect Dis. 2021;25:101569.
    1. Bermejo-Martin JF, González-Rivera M, Almansa R, Micheloud D, Tedim AP, Domínguez-Gil M, et al. Viral RNA load in plasma is associated with critical illness and a dysregulated host response in COVID-19. Crit Care. 2020;24:691.
    1. Middleton EA, He XY, Denorme F, Campbell RA, Ng D, Salvatore SP, et al. Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood. 2020;136:1169–1179.
    1. Leppkes M, Knopf J, Naschberger E, Lindemann A, Singh J, Herrmann I, et al. Vascular occlusion by neutrophil extracellular traps in COVID-19. EBioMedicine. 2020;58:102925.
    1. Liu Y, Du X, Chen J, Jin Y, Peng L, Wang HH, et al. Neutrophil-to-lymphocyte ratio as an independent risk factor for mortality in hospitalized patients with COVID-19. J Infect. 2020;81:e6–e12.
    1. Ackermann M, Anders HJ, Bilyy R, Bowlin G, Daniel C, Lorenza R, et al. Patients with COVID-19: in the dark-NETs of neutrophilis. Cell Death Differ. 2021;28:3125–3139.
    1. Soni M, Gopalakrishnan R, Vaishya R, Prabu P. D-dimer level is a useful predictor for mortality in patients with COVID-19: analysis of 483 cases. Diabetes Metab Syndr. 2020;14:2245–2249.
    1. Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062.
    1. Hayıroğlu Mİ, Çiçek V, Kılıç Ş, Çınar T. Mean serum D-dimer level to predict in-hospital mortality in COVID-19. Rev Assoc Med Bras. 2021;67:437–442.
    1. Henry BM, de Oliveira MHS, Benoit S, Plebani M, Lippi G. Hematologic, biochemical and immune biomarker abnormalities associated with severe illness and mortality in coronavirus disease 2019 (COVID-19): a meta-analysis. Clin Chem Lab Med. 2020;58:1021–1028.
    1. Sharifpour M, Rangaraju S, Liu M, Alabyad D, Nahab FB, Creel-Bulos CM, et al. C-Reactive protein as a prognostic indicator in hospitalized patients with COVID-19. PLoS ONE. 2020;15:e0242400.
    1. Payán-Pernía S, Gómez Pérez L, Remacha Sevilla ÁF, Sierra Gil J, Novelli CS. Absolute lymphocytes, ferritin, C-Reactive protein, and lactate dehydrogenase predict early invasive ventilation in patients with COVID-19. Lab Med. 2021;52:141–145.
    1. Kaftan AN, Hussain MK, Algenabi AA, Naser FH, Enaya MA. Predictive value of C-reactive protein, lactate dehydrogenase, ferritin and D-dimer levels in diagnosing COVID-19 patients: a retrospective study. Acta Inform Med. 2021;29:45–50.
    1. Fajgenbaum DC, June CH. Cytokine storm. N Engl J Med. 2020;383:2255–2273.
    1. Schulert GS, Grom AA. Pathogenesis of macrophage activation syndrome and potential for cytokine- directed therapies. Annu Rev Med. 2015;66:145–159.
    1. Brisse E, Wouters CH, Matthys P. Advances in the pathogenesis of primary and secondary hemophagocytic lymphohistiocytosis: differences and similarities. Br J Hematol. 2016;174:203–2017.
    1. Crayne CB, Albeituni S, Nichols KE, Cron RQ. The immunology of macrophage activation syndrome. Front Immunol. 2019;10:119.
    1. Liu F, Li L, Xu M, Wu J, Luo D, Zhu Y, et al. Prognostic value of interleukin-6, C-reactive protein, and procalcitonin in patients with COVID-19. J Clin Virol. 2020;127:104370.
    1. Ma A, Zhang L, Ye X, Chen J, Yu J, Zhuang L, et al. High levels of circulating IL-8 and soluble IL-2R are associated with prolonged illness in patients with severe COVID-19. Front Immunol. 2021;12:626235.
    1. Li L, Li J, Gao M, Fan H, Wang Y, Xu X, et al. Interleukin-8 as a biomarker for disease prognosis of coronavirus disease-2019 patients. Front Immunol. 2021;11:602395.
    1. Chen Y, Wang J, Liu C, Su L, Zhang D, Fan J, et al. IP-10 and MCP-1 as biomarkers associated with disease severity of COVID-19. Mol Med. 2020;26:97.
    1. Jamilloux Y, Henry T, Belot A, Viel S, Fauter M, El Jammal T, Walzer T, François B, Sève P. Should we stimulate or suppress immune responses in COVID-19? Cytokine and anti-cytokine interventions. Autoimmun Rev. 2020;19:102567.
    1. Nanjappa SG, Kim EH, Suresh M. Immunotherapeutic effects of IL-7 during a chronic viral infection in mice. Blood. 2011;117:5123–5132.
    1. Perales MA, Goldberg JD, Yuan J, Koehne G, Lechner L, Papadopoulos EB, et al. Recombinant human interleukin-7 (CYT107) promotes T-cell recovery after allogeneic stem cell transplantation. Blood. 2012;120:4882–4891.
    1. Diao B, Wang C, Tan Y, Chen X, Liu Y, Ning L, et al. Reduction and functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID-19) Front Immunol. 2020;11:827.
    1. Qin C, Zhou L, Hu Z, Zhang S, Yang S, Tao Y, et al. Dysregulation of immune response in patients with coronavirus 2019 (COVID-19) in Wuhan. China Clin Infect Dis. 2020;71:762–768.
    1. Wang F, Nie J, Wang H, Zhao Q, Xiong Y, Deng L, et al. Characteristics of peripheral lymphocyte subset alteration in COVID-19 pneumonia. J Infect Dis. 2020;221:1762–1769.
    1. Hadjadj J, Yatim N, Barnabei L, Corneau A, Boussier J, Smith N, et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science. 2020;369:718–724.
    1. Zheng M, Gao Y, Wang G, Song G, Liu S, Sun D, et al. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell Mol Immunol. 2020;17:533–535.
    1. Zheng HY, Zhang M, Yang CX, Zhang N, Wang XC, Yang XP, et al. Elevated exhaustion levels and reduced functional diversity of T cells in peripheral blood may predict severe progression in COVID-19 patients. Cell Mol Immunol. 2020;17:541–543.
    1. Chen J, Hu C, Chen L, Tang L, Zhu Y, Xu X, et al. Clinical study of mesenchymal stem cell treatment for acute respiratory distress syndrome induced by epidemic influenza A (H7N9) infection: a hint for COVID-19 treatment. Engineering. 2020;6:1153–1161.
    1. Liang B, Chen J, Li T, Wu H, Yang W, Li Y, et al. Clinical remission of a critically ill COVID-19 patient treated by human umbilical cord mesenchymal stem cells: a case report. Medicine (Baltimore) 2020;99:e21429.
    1. Herrera-Valdés R, Almaguer-López M, López-Marín L, Bacallao-Méndez R, Pérez-Oliva JF, Guerra-Bustillo G. COVID-19 and the kidneys: risk, damage and sequelae. MEDICC Rev. 2020;22:87–88.
    1. Hwang W, Cho MS, Oh JE, Lee JH, Jeong JC, Shin G-T, et al. Comparison of creatinine index and geriatric nutritional risk index for nutritional evaluation of patients with hemodialysis. Hemodial Int. 2018;22:507–514.
    1. Sterne JAC, Murthy S, Diaz JV, Slutsky AS, Villar J, Angus DC, et al. Association between administration of systemic corticosteroids and mortality among critically ill patients with COVID-19: a meta-analysis. JAMA. 2020;324:1330–1341.
    1. Sinclair KA, Yerkovich ST, Chen T, McQualter JL, Hopkins PM, Wells CA, et al. Mesenchymal stromal cells are readily recoverable from lung tissue, but not the alveolar space, in healthy humans. Stem Cells. 2016;34:2548–2558.
    1. Lerum TV, Aaløkken TM, Bronstad E, Aarli B, Ikdahl E, Lund KMA, et al. Dyspnoea, lung function and CT findings 3 months after hospital admission for COVID-19. Eur Respir J. 2021;57:2003448.
    1. Huang Y, Pinto MD, Borelli JL, Mehrabadi MA, Abrihim H, Dutt N, et al. COVID symptoms, symptom clusters, and predictors for becoming a long-hauler: looking for clarity in the haze of the pandemic. medRxiv. 2021. 10.1101/2021.03.03.21252086.

Source: PubMed

3
Abonnieren