3,251
Views
23
CrossRef citations to date
0
Altmetric
Review

Cytokine storm associated coagulation complications in COVID-19 patients: Pathogenesis and Management

, &
Pages 1397-1413 | Received 27 Jan 2021, Accepted 07 Apr 2021, Published online: 19 Apr 2021

References

  • Lu H, Stratton CW, Tang YW. Outbreak of pneumonia of unknown etiology in Wuhan, China: the mystery and the miracle. J Med Virol. 2020;92(4):401–402.
  • Zhu N, Zhang D, Wang W, et al. A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med. 2020;382(8):727–733.
  • Statement on the second meeting of the International Health Regulations (2005) Emergency Committee regarding the outbreak of novel coronavirus (2019-nCoV) https://www.who.int/news-room/detail/30-01-2020-statement-on-the-second-meeting-of-the-international-health-regulations-(2005)-emergency-committee-regarding-the-outbreak-of-novel-coronavirus-(2019-ncov). (Accessed 2020 May 27
  • WHO-audio-emergencies-coronavirus-press-conference-full-and-final. https://www.who.int/docs/default-source/coronaviruse/transcripts/who-audio-emergencies-coronavirus-press-conference-full-and-final-11mar2020.pdf?sfvrsn=cb432bb3_2 (accessed 2020 May 27
  • Weekly epidemiological update - 9 March 2021 https://www.who.int/publications/m/item/weekly%2depidemiological%2dupdate—10%2dmarch%2d2021 (accessed 2021 Mar 11
  • Weiss SR, Leibowitz JL. Coronavirus pathogenesis. Adv Virus Res. 2011;81:85–164.
  • Baviskar T, Raut D, Bhatt LK. Deciphering Vaccines for COVID-19: where do we stand today? Immunopharmacol. Immunotoxicol. 2021;43(1):8–21.
  • Su S, Wong G, Shi W, et al. Epidemiology, genetic recombination, and pathogenesis of coronaviruses. Trends Microbiol. 2016;24(6):490–502.
  • Cui J, Li F, Shi ZL. Origin and evolution of pathogenic coronaviruses. Nat Rev Microbiol. 2019;17(3):181–192.
  • Peiris JSM, Lai ST, Poon LLM, et al. Coronavirus as a possible cause of severe acute respiratory syndrome. Lancet. 2003;361(9366):1319–1325.
  • Kuiken T, Fouchier RAM, Schutten M, et al. Newly discovered coronavirus as the primary cause of severe acute respiratory syndrome. Lancet. 2003;362(9380):263–270.
  • Thompson BT, Chambers RC, Liu KD. Acute respiratory distress syndrome. N Engl J Med. 2017;377(6):562–572.
  • Drosten C, Günther S, Preiser W, et al. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med. 2003;348(20):1967–1976.
  • Ksiazek TG, Erdman D, Goldsmith CS, et al. A novel coronavirus associated with severe acute respiratory syndrome. N Engl J Med. 2003;348(20):1953–1966.
  • Zhong NS, Zheng BJ, Li YM, et al. Epidemiology and cause of severe acute respiratory syndrome (SARS) in guangdong, people’s republic of China, in february, 2003. Lancet. 2003;362(9393):1353–1358.
  • Zaki AM, Van Boheemen S, Bestebroer TM, et al. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med. 2012;367(19):1814–1820.
  • Wong G, Liu W, Liu Y, et al. MERS, SARS, and ebola: the role of super-spreaders in infectious disease. Cell Host Microbe. 2015;18(4):398–401.
  • Gheblawi M, Wang K, Viveiros A, et al. Angiotensin-converting enzyme 2: SARS-CoV-2 receptor and regulator of the renin-angiotensin system: celebrating the 20th anniversary of the discovery of ACE2. Circ Res. 2020;126(10):1456–1474.
  • Wan Y, Shang J, Graham R, et al. Receptor recognition by the novel coronavirus from wuhan: an analysis based on decade-long structural studies of SARS coronavirus. J Virol. 2020;94(7):e00127–20.
  • Zhou P, Lou YX, Wang XG, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579(7798):270–273.
  • Zhang W, Zhao Y, Zhang F, et al. The use of anti-inflammatory drugs in the treatment of people with severe coronavirus disease 2019 (COVID-19): the experience of clinical immunologists from China. Clin Immunol. 2020;214:108393.
  • Conti P, Ronconi G, Caraffa A, et al. Induction of pro-inflammatory cytokines (IL-1 and IL-6) and lung inflammation by Coronavirus-19 (COVI-19 or SARS-CoV-2): anti-inflammatory strategies. J Biol Regul Homeost Agents. 2020;34(2):327–331.
  • Li G, Fan Y, Lai Y, et al. Coronavirus infections and immune responses. J Med Virol. 2020;92(4):424–432.
  • Wu JT, Leung K, Leung GM. Nowcasting and forecasting the potential domestic and international spread of the 2019-nCoV outbreak originating in Wuhan, China: a modelling study. Lancet. 2020;395(10225):689–697.
  • Thevarajan I, Nguyen THO, Koutsakos M, et al. Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat Med. 2020;26(4):453–455.
  • Cao X. COVID-19: immunopathology and its implications for therapy. Nat Rev Immunol. 2020;20(5):269–270.
  • Van Kerkhove MD, Vandemaele KAH, Shinde V, et al. Risk factors for severe outcomes following 2009 influenza a (H1N1) infection: a global pooled analysis. PLoS Med. 2011;8(7):e1001053.
  • Yang J, Zheng Y, Gou X, et al. Prevalence of comorbidities and its effects in coronavirus disease 2019 patients: a systematic review and meta-analysis. Int J Infect Dis. 2020;94:91–95.
  • Rao S, Lau A, Hc S, Traits. Exploring diseases/traits and blood proteins causally related to expression of ACE2, the putative receptor of SARS-CoV-2: a mendelian randomization analysis highlights tentative relevance of diabetes-related. Diabetes Care. 2020;43(7):1416–1426.
  • Fernandez C, Rysä J, Almgren P, et al. Plasma levels of the proprotein convertase furin and incidence of diabetes and mortality. J Intern Med. 2018;284(4):377–387.
  • Shang J, Wan Y, Luo C, et al. Cell entry mechanisms of SARS-CoV-2. Proc Natl Acad Sci U S A. 2020;117(21):11727–11734.
  • Ma LY, Chen WW, Gao RL, et al. China cardiovascular diseases report 2018: an updated summary. J Geriatr Cardiol. 2020;17(1):1–8.
  • Fang L, Karakiulakis G, Roth M. Are patients with hypertension and diabetes mellitus at increased risk for COVID-19 infection?. Lancet Respir Med. 2020;8(4):e21.
  • Badawi A, Ryoo SG. Prevalence of comorbidities in the Middle East respiratory syndrome coronavirus (MERS-CoV): a systematic review and meta-analysis. Int J Infect Dis. 2016;49:129–133.
  • Chan JWM, Ng CK, Chan YH, et al. Short term outcome and risk factors for adverse clinical outcomes in adults with severe acute respiratory syndrome (SARS). Thorax. 2003;58(8):686–689.
  • Bonow RO, Fonarow GC, O’Gara PT, et al. association of coronavirus disease 2019 (COVID-19) with myocardial injury and mortality. JAMA Cardiol. 2020;5(7):751–753.
  • Yao X, Ye F, Zhang M, et al. In vitro antiviral activity and projection of optimized dosing design of hydroxychloroquine for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Clin Infect Dis. 2020;71(15):732–739.
  • Cui S, Chen S, Li X, et al. Prevalence of venous thromboembolism in patients with severe novel coronavirus pneumonia. J Thromb Haemost. 2020;18(6):1421–1424.
  • Klok FA, Kruip MJHA, Van Der Meer NJM, et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res. 2020;191:145–147.
  • Tang N, Bai H, Chen X, et al. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J Thromb Haemost. 2020;18(5):1094–1099.
  • Llitjos J, Leclerc M, Chochois C, et al. High incidence of venous thromboembolic events in anticoagulated severe COVID‐19 patients. J Thromb Haemost. 2020;18(7):1743–1746.
  • Cytokine Storm: MG. Is it the only major death factor in COVID-19 patients? Coagulation role. Med Hypotheses. 2020;142:109829.
  • Tang N, Li D, Wang X, et al. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost. 2020;18(4):844–847.
  • Weitz JI, Fredenburgh JC, Eikelboom JW. A test in context: d-dimer [internet]. J Am Coll Cardiol. 2017;70(19):2411–2420.
  • Phillippe HM. Overview of venous thromboembolism [Internet]. Am J Manag Care. 2017;23(20 Suppl):S376–82.
  • Bavishi C, Bonow RO, Trivedi V, et al. Acute myocardial injury in patients hospitalized with COVID-19 infection: a review. Prog Cardiovasc Dis. 2020;63(5):682–689.
  • Davidson S, Maini MK, Wack A. Disease-promoting effects of type i interferons in viral, bacterial, and coinfections. J Interf Cytokine Res. 2015;35(4):252–264.
  • Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nat Rev Immunol. 2013;13(12):875–887.
  • Channappanavar R, Fehr AR, Vijay 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.
  • Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506.
  • Marchingo JM, Sinclair LV, Howden AJ, et al. Quantitative analysis of how myc controls t cell proteomes and metabolic pathways during t cell activation. Elife. 2020;9:e53725.
  • Chen L, Liu HG, Liu W, et al. [Analysis of clinical features of 29 patients with 2019 novel coronavirus pneumonia]. Zhonghua Jie He He Hu Xi Za Zhi. 2020;43:E005.
  • Yang X, Yu Y, Xu J, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;8(5):475–481.
  • Ranieri VM, Rubenfeld GD, Thompson BT, et al. Acute respiratory distress syndrome: the Berlin definition. J Am Med Assoc. 2012;307:2526–2533.
  • Douda DN, Jackson R, Grasemann H, et al. Innate immune collectin surfactant protein D simultaneously binds both neutrophil extracellular traps and carbohydrate ligands and promotes bacterial trapping. J Immunol. 2011;187(4):1856–1865.
  • Parsons PE, Eisner MD, Thompson BT, et al. Lower tidal volume ventilation and plasma cytokine markers of inflammation in patients with acute lung injury. Crit Care Med. 2005;33(1):1–6.
  • Wang H, Ma S. The cytokine storm and factors determining the sequence and severity of organ dysfunction in multiple organ dysfunction syndrome. Am J Emerg Med. 2008;26(6):711–715.
  • Shimabukuro-Vornhagen A, Gödel P, Subklewe M, et al. Cytokine release syndrome. J Immunother Cancer. 2018;6(1):56.
  • Hunter CA, Jones SA. IL-6 as a keystone cytokine in health and disease. Nat Immunol. 2015;16(5):448–457.
  • Tanaka T, Narazaki M, Kishimoto T. Immunotherapeutic implications of IL-6 blockade for cytokine storm. Immunotherapy. 2016;8(8):959–970.
  • Pathan N, Hemingway CA, Alizadeh AA, et al. Role of interleukin 6 in myocardial dysfunction of meningococcal septic shock. Lancet. 2004;363(9404):203–209.
  • Hay KA, Hanafi LA, Li D, et al. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor–modified T-cell therapy. Blood. 2017;130(21):2295–2306.
  • Iwanaga S, Kawabata SI, Muta T. New types of clotting factors and defense molecules found in horseshoe crab hemolymph: their structures and functions. J Biochem. 1998;123(1):1–15.
  • Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity. Nat Rev Immunol. 2013;13(1):34–45.
  • Herter JM, Rossaint J, Zarbock A. Platelets in inflammation and immunity. J Thromb Haemost. 2014;12(11):1764–1775.
  • Maas C, Renne T. Coagulation factor XII in thrombosis and inflammation. Blood. 2018;131(17):1903–1909.
  • Conway EM. Reincarnation of ancient links between coagulation and complement. J Thromb Haemost. 2015;13:S121–32.
  • Burzynski LC, Humphry M, Pyrillou K, et al. The coagulation and immune systems are directly linked through the activation of interleukin-1α by thrombin. Immunity. 2019;50(4):1033–1042.
  • Thornton P, McColl BW, Greenhalgh A, et al. Platelet interleukin-1α drives cerebrovascular inflammation. Blood. 2010;115(17):3632–3639.
  • Bester J, Matshailwe C, Pretorius E. Simultaneous presence of hypercoagulation and increased clot lysis time due to IL-1β IL-6 and IL-8. Cytokine. 2018;110:237–242.
  • Brunn GJ, Saadi S, Platt JL. Constitutive repression of interleukin-1α in endothelial cells. Circ Res. 2008;102(7):823–830.
  • Tunjungputri RN, Li Y, De Groot PG, et al. The inter-relationship of platelets with interleukin-1β-mediated inflammation in humans. Thromb Haemost. 2018;118(12):2112–2125.
  • Cavalli G, Colafrancesco S, Emmi G, et al. Interleukin 1α: a comprehensive review on the role of IL-1α in the pathogenesis and treatment of autoimmune and inflammatory diseases. Autoimmun Rev. 2021;20:102763.
  • José RJ, Williams AE, Chambers RC. Proteinase-activated receptors in fibroproliferative lung disease. Thorax. 2014;69(2):190–192.
  • Feistritzer C, Riewald M. Endothelial barrier protection by activated protein C through PAR1-dependent sphingosine 1-phosphate receptor-1 crossactivation. Blood. 2005;105(8):3178–3184.
  • Günther A, Mosavi P, Heinemann S, et al. Alveolar fibrin formation caused by enhanced procoagulant and depressed fibrinolytic capacities in severe pneumonia: comparison with the acute respiratory distress syndrome. Am J Respir Crit Care Med. 2000;161(2):454–462.
  • Howell DCJ, Johns RH, Lasky JA, et al. Absence of proteinase-activated receptor-1 signaling affords protection from bleomycin-induced lung inflammation and fibrosis. Am J Pathol. 2005;166(5):1353–1365.
  • Mercer PF, Williams AE, Scotton CJ, et al. Proteinase-activated receptor-1, CCL2, and CCL7 regulate acute neutrophilic lung inflammation. Am J Respir Cell Mol Biol. 2014;50(1):144–157.
  • Chambers RC, Scotton CJ. Coagulation cascade proteinases in lung injury and fibrosis. Proc Am Thorac Soc. 2012;9(3):96–101.
  • Zhou F, Yu T, Du R, 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.
  • Smith SA, Mutch NJ, Baskar D, et al. Polyphosphate modulates blood coagulation and fibrinolysis. Proc Natl Acad Sci U S A. 2006;103(4):903–908.
  • Subramaniam S, Jurk K, Hobohm L, et al. Distinct contributions of complement factors to platelet activation and fibrin formation in venous thrombus development. Blood. 2017;129(16):2291–2302.
  • Noubouossie DF, Reeves BN, Strahl BD, et al. Neutrophils: back in the thrombosis spotlight. Blood. 2016;133(20):2186–2197.
  • Iba T, Levy JH. Inflammation and thrombosis: roles of neutrophils, platelets and endothelial cells and their interactions in thrombus formation during sepsis. J Thromb Haemost. 2018;16(2):231–241.
  • Rittirsch D, Flierl MA, Ward PA. Harmful molecular mechanisms in sepsis. Nat Rev Immunol. 2008;8(10):776–787.
  • Iba T, Levy JH. Derangement of the endothelial glycocalyx in sepsis. J Thromb Haemost. 2019;17(2):283–294.
  • Siddiqi HK, Mehra MR. COVID-19 illness in native and immunosuppressed states: a clinical–therapeutic staging proposal. J Heart Lung Transplant. 2020;39(5):405–407.
  • Cavalli G, Larcher A, Tomelleri A, et al. Interleukin-1 and interleukin-6 inhibition compared with standard management in patients with COVID-19 and hyperinflammation: a cohort study. Lancet Rheumatol. 2021 (online ahead of print).
  • Fu B, Xu X, Wei H. Why tocilizumab could be an effective treatment for severe COVID-19? J Transl Med. 2020;18(1):164.
  • Zhang C, Wu Z, Li JW, et al. Cytokine release syndrome in severe COVID-19: interleukin-6 receptor antagonist tocilizumab may be the key to reduce mortality. Int J Antimicrob Agents. 2020;55(5):105954.
  • Xu X, Han M, Li T, et al. Effective treatment of severe COVID-19 patients with tocilizumab. Proc Natl Acad Sci U S A. 2020;117(20):10970–10975.
  • Luo P, Liu Y, Qiu L, et al. Tocilizumab treatment in COVID-19: a single center experience. J Med Virol. 2020;92(7):814–818.
  • Fontana F, Alfano G, Mori G, et al. COVID-19 pneumonia in a kidney transplant recipient successfully treated with tocilizumab and hydroxychloroquine. Am J Transplant. 2020;20(7):1902–1906.
  • Mihai C, Dobrota R, Schröder M, et al. COVID-19 in a patient with systemic sclerosis treated with tocilizumab for SSc-ILD. Ann Rheum Dis. 2020;79(5):668–669.
  • Zhong J, Tang J, Ye C, et al. The immunology of COVID-19: is immune modulation an option for treatment? Lancet Rheumatol. 2020;2(7):e428–36.
  • Deisseroth A, Ko CW, Nie L, et al. FDA approval: siltuximab for the treatment of patients with multicentric castleman disease. Clin Cancer Res. 2015;21(5):950–954.
  • Palanques-Pastor T, López-Briz E, Poveda Andrés JL. Involvement of interleukin 6 in SARS-CoV-2 infection: siltuximab as a therapeutic option against COVID-19. Eur J Hosp Pharm. 2020;27(5):297–298.
  • Van Rhee F, Casper C, Voorhees PM, et al. Long-term safety of siltuximab in patients with idiopathic multicentric Castleman disease: a prespecified, open-label, extension analysis of two trials. Lancet Haematol. 2020;7(3):e209–17.
  • Richardson P, Griffin I, Tucker C, et al. Baricitinib as potential treatment for 2019-nCoV acute respiratory disease. Lancet. 2020;395(10223):e30–1.
  • Russell B, Moss C, George G, et al. Associations between immune-suppressive and stimulating drugs and novel COVID-19 - A systematic review of current evidence. Ecancermedicalscience. 2020;14:1022.
  • Schett G, Manger B, Simon D, et al. COVID-19 revisiting inflammatory pathways of arthritis. Nat Rev Rheumatol. 2020;16(8):465–470.
  • Dimopoulos G, De Mast Q, Markou N, et al. Favorable anakinra responses in severe covid-19 patients with secondary hemophagocytic lymphohistiocytosis. Cell Host Microbe. 2020;28:117–123.
  • Huet T, Beaussier H, Voisin O, et al. Anakinra for severe forms of COVID-19: a cohort study. Lancet Rheumatol. 2020;2(7):e393–400.
  • Navarro-Millán I, Sattui SE, Lakhanpal A, et al. Use of anakinra to prevent mechanical ventilation in severe COVID-19: a case series. Arthritis Rheumatol. 2020;72(12):1990–1997.
  • Cavalli G, De Luca G, Campochiaro C, et al. Interleukin-1 blockade with high-dose anakinra in patients with COVID-19, acute respiratory distress syndrome, and hyperinflammation: a retrospective cohort study. Lancet Rheumatol. 2020;2(6):e325–31.
  • Monteagudo LA, Boothby A, Gertner E. Continuous intravenous anakinra infusion to calm the cytokine storm in macrophage activation syndrome. ACR Open Rheumatol. 2020;2(5):276–282.
  • King A, Vail A, O’Leary C, et al. Anakinra in COVID-19: important considerations for clinical trials. Lancet Rheumatol. 2020;2(7):e379–81.
  • Aouba A, Baldolli A, Geffray L, et al. Targeting the inflammatory cascade with anakinra in moderate to severe COVID-19 pneumonia: case series. Ann Rheum Dis. 2020;79(10):1381–1382.
  • Riphagen S, Gomez X, Gonzalez-Martinez C, et al. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet. 2020;395(10237):1607–1608.
  • Ak C PM. Updates in thrombosis in pediatrics: where are we after 20 years? Hematol Am Soc Hematol Educ Program. 2012;439–43:2012.
  • Della Gatta AN, Rizzo R, Pilu G, et al. Coronavirus disease 2019 during pregnancy: a systematic review of reported cases. Am J Obstet Gynecol. 2020;223(1):36–41.
  • Nikolich-Zugich J, Knox KS, Rios CT, et al. SARS-CoV-2 and COVID-19 in older adults: what we may expect regarding pathogenesis, immune responses, and outcomes. GeroScience. 2020;42(2):505–514.
  • Fogarty H, Townsend L, Ni Cheallaigh C, et al. COVID19 coagulopathy in Caucasian patients. Br J Haematol. 2020; 189(6): 1044–9
  • Guan W, Ni Z, Hu Y, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382(18):1708–1720.
  • Wang YD, Zhang SP, Wei QZ, et al. COVID-19 complicated with DIC: 2 cases report and literatures review. Zhonghua Xue Ye Xue Za Zhi. 41, 245–247 (2020).
  • AlSubaie AM. Coagulopathies in novel coronavirus (SARS-CoV-2) pandemic: emerging evidence for hematologists. Saudi J Biol Sci. 2020;28(1):956–961.
  • Lippi G, Plebani M, Henry BM. Thrombocytopenia is associated with severe coronavirus disease 2019 (COVID-19) infections: a meta-analysis. Clin Chim Acta. 2020;506:145–148.
  • Glas GJ, Van Der Sluijs KF, Schultz MJ, et al. Bronchoalveolar hemostasis in lung injury and acute respiratory distress syndrome. J Thromb Haemost. 2013;11(1):17–25.
  • Yang X, Yang Q, Wang Y, et al. Thrombocytopenia and its association with mortality in patients with COVID‐19. J Thromb Haemost. 2020;18(6):1469–1472.
  • Machlus KR, Cardenas JC, Church FC, et al. Causal relationship between hyperfibrinogenemia, thrombosis, and resistance to thrombolysis in mice. Blood. 2011;117(18):4953–4963.
  • Dolhnikoff M, Duarte-Neto AN, De Almeida Monteiro RA, et al. Pathological evidence of pulmonary thrombotic phenomena in severe COVID-19. J Thromb Haemost. 2020;18(6):1517–1519.
  • Gattinoni L, Chiumello D, Caironi P, et al. COVID-19 pneumonia: different respiratory treatment for different phenotypes? Intensive Care Med. 2020;46(6):1099–1102.
  • Khandait H, Gandotra G, Sachdeva S, et al. COVID-19 and hematology—what do we know so far? SN Compr Clin Med. 2020;2(12):2631–2636. online ahead of print.
  • Asakura H, Ogawa H. COVID-19-associated coagulopathy and disseminated intravascular coagulation. Int J Hematol. 2021;113(1):45–57.
  • Zhang Y, Xiao M, Zhang S, et al. Coagulopathy and antiphospholipid antibodies in patients with covid-19. N Engl J Med. 2020;382(17):e38.
  • Thachil J, Tang N, Gando S, et al. ISTH interim guidance on recognition and management of coagulopathy in COVID‐19. J Thromb Haemost. 2020;18(5):1023–1026.
  • Bikdeli B, Madhavan MV, Jimenez D, et al. COVID-19 and thrombotic or thromboembolic disease: implications for prevention, antithrombotic therapy, and follow-up. J Am Coll Cardiol. 2020;75(23):2950–2973.
  • Karasu E, Nilsson B, Köhl J, et al. Targeting complement pathways in polytrauma- And sepsis-induced multiple-organ dysfunction. Front Immunol. 2019;10:543.
  • Licciardi F, Giani T, Baldini L, et al. COVID-19 and what pediatric rheumatologists should know: a review from a highly affected country. Pediatr Rheumatol. 2020;18(1):35.
  • Whyte CS, Morrow GB, Mitchell JL, et al. Fibrinolytic abnormalities in acute respiratory distress syndrome (ARDS) and versatility of thrombolytic drugs to treat COVID‐19. J Thromb Haemost. 2020;18(7):1548–1555.
  • Ranucci M, Ballotta A, Di Dedda U, et al. The procoagulant pattern of patients with COVID-19 acute respiratory distress syndrome. J Thromb Haemost. 2020;18(7):1747–1751.
  • Favresse J, Lippi G, Roy PM, et al. D-dimer: preanalytical, analytical, postanalytical variables, and clinical applications. Crit Rev Clin Lab Sci. 2018;55(8):548–577.
  • Wang D, Hu B, Hu C, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. J Am Med Assoc. 2020;323(11):1061–1069.
  • Huang I, Pranata R, Lim MA, et al. C-reactive protein, procalcitonin, D-dimer, and ferritin in severe coronavirus disease-2019: a meta-analysis. Ther Adv Respir Dis. 2020;14:1753466620937175.
  • Levi M, Thachil J, Iba T, et al. Coagulation abnormalities and thrombosis in patients with COVID-19. Lancet Haematol. 2020;7(6):e438–40.
  • Liu F, Li L, Da XM, et al. Prognostic value of interleukin-6, C-reactive protein, and procalcitonin in patients with COVID-19. J Clin Virol. 2020;127:104370.
  • Koozi H, Lengquist M, Frigyesi A. C-reactive protein as a prognostic factor in intensive care admissions for sepsis: a Swedish multicenter study. J Crit Care. 2020;56:73–79.
  • Solari F, Varacallo M. Low molecular weight heparin (LMWH). StatPearls. 2020.
  • Rotzinger DC, Beigelman-Aubry C, Von Garnier C, et al. Pulmonary embolism in patients with COVID-19: time to change the paradigm of computed tomography. Thromb Res. 2020;190:58–59.
  • Zakai NA, Mcclure LA. Racial differences in venous thromboembolism. J Thromb Haemost. 2011;9(10):1877–1882.
  • Stein PD, Kayali F, Olson RE, et al. Pulmonary thromboembolism in asians/pacific islanders in the United States: analysis of data from the national hospital discharge survey and the United States bureau of the census. Am J Med. 2004;116(7):435–442.
  • Middeldorp S, Coppens M, Van Haaps TF, et al. Incidence of venous thromboembolism in hospitalized patients with COVID‐19. J Thromb Haemost. 2020;18(8):1995–2002.
  • Helms J, Tacquard C, Severac F, et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med. 2020;46(6):1089–1098.
  • Poissy J, Goutay J, Caplan M, et al. Pulmonary Embolism in COVID-19 Patients: awareness of an Increased Prevalence. Circulation. 2020;142(2):184–186.
  • Wang TF, Milligan PE, Wong CA, et al. Efficacy and safety of high-dose thromboprophylaxis in morbidly obese inpatients. Thromb Haemost. 2013;111(1):88–93.
  • Pannucci CJ, Fleming KI, Holoyda K, et al. Enoxaparin 40 mg per day is inadequate for venous thromboembolism prophylaxis after thoracic surgical procedure. Ann Thorac Surg. 2018;106(2):404–411.
  • Young E. The anti-inflammatory effects of heparin and related compounds. Thromb Res. 2008;122(6):743–752.
  • Ozolina A, Sarkele M, Sabelnikovs O, et al. Activation of coagulation and fibrinolysis in acute respiratory distress syndrome: a prospective pilot study. Front Med. 2016;3:64.
  • Shukla D, Spear PG. Herpesviruses and heparan sulfate: an intimate relationship in aid of viral entry. J Clin Invest. 2001;108(4):503–510.
  • Vicenzi E, Canducci F, Pinna D, et al. Coronaviridae and SARS-associated coronavirus strain HSR1. Emerg Infect Dis. 2004;10(3):413–418.
  • Yamakawa K, Levy JH, Iba T. Recombinant human soluble thrombomodulin in patients with sepsis-associated coagulopathy (SCARLET): an updated meta-analysis. Crit Care. 2019;321(20):1993–2002.
  • Kienast J, Juers M, Wiedermann CJ, et al. Treatment effects of high-dose antithrombin without concomitant heparin in patients with severe sepsis with or without disseminated intravascular coagulation. J Thromb Haemost. 2006;4(1):90–97.
  • Iba T, Arakawa M, Ohchi Y, et al. Prediction of early death in patients with sepsis-associated coagulation disorder treated with antithrombin supplementation. Clin Appl Thromb. 2018;24(9_suppl):145S–9S.
  • Lin L, Lu L, Cao W, et al. Hypothesis for potential pathogenesis of SARS-CoV-2 infection–a review of immune changes in patients with viral pneumonia. Emerg Microbes Infect. 2020;9(1):727–732.
  • Iba T, Levy JH, Raj A, et al. Advance in the management of sepsis-induced coagulopathy and disseminated intravascular coagulation. J Clin Med. 2019;8(5):728.
  • Du L, Kao RY, Zhou Y, et al. Cleavage of spike protein of SARS coronavirus by protease factor Xa is associated with viral infectivity. Biochem Biophys Res Commun. 2007;359(1):174–179.
  • Kam Y-W, Okumura Y, Kido H, et al. Cleavage of the SARS coronavirus spike glycoprotein by airway proteases enhances virus entry into human bronchial epithelial cells in vitro. PLoS One. 2009;4(11):e7870.
  • Elfiky AA. Ribavirin, Remdesivir, Sofosbuvir, Galidesivir, and Tenofovir against SARS-CoV-2 RNA dependent RNA polymerase (RdRp): a molecular docking study. Life Sci. 2020;253:117592.
  • Zabetakis I, Lordan R, Norton C, et al. COVID-19: the inflammation link and the role of nutrition in potential mitigation. Nutrients. 2020;12(5):1466.
  • Zhang C, Huang S, Zheng F, et al. Controversial treatments: an updated understanding of the coronavirus disease 2019. J Med Virol. 2020;92(9):1441–1448.
  • Karim S, Islam A, Rafiq S, et al. The COVID-19 pandemic: disproportionate thrombotic tendency and management recommendations. Trop Med Infect Dis. 2021;6(1):26.
  • Smyth SS, Mcever RP, Weyrich AS, et al. Platelet functions beyond hemostasis. J Thromb Haemost. 2009;7(11):1759–1766.
  • Connors J, States U, Levy J, et al. COVID-19 and its implications for thrombosis and anticoagulation COVID-19 and its implications for thrombosis and anticoagulation. J Blood. 2020;2:1–21.
  • Nagge J, Crowther M, Hirsh J. Is impaired renal function a contraindication to the use of low-molecular-weight heparin? Arch Intern Med. 2002;162(22):2605–2609.
  • Hippensteel JA, LaRiviere WB, Colbert JF, et al. Heparin as a therapy for COVID-19: current evidence and future possibilities. Am J Physiol - Lung Cell Mol Physiol. 2020;319(2):L211–7.
  • Morrow DA, Braunwald E, Bonaca MP, et al. Vorapaxar in the secondary prevention of atherothrombotic events. N Engl J Med. 2012;366(15):1404–1413.
  • José R, Williams A, Sulikowski M, et al. Regulation of neutrophilic inflammation in lung injury induced by community-acquired pneumonia. Lancet. 2015;385:S52.
  • José RJ, Williams AE, Mercer PF, et al. Regulation of neutrophilic inflammation by proteinase-activated receptor 1 during bacterial pulmonary infection. J Immunol. 2015;194(12):6024–6034.
  • Jiang Y, Zhao G, Song N, et al. Blockade of the C5a–C5aR axis alleviates lung damage in hDPP4 -transgenic mice infected with MERS-CoV. Emerg Microbes Infect. 2018;7(1):1–12.
  • Reis ES, Berger N, Wang X, et al. Safety profile after prolonged C3 inhibition. Clin Immunol. 2018;197:96–106.
  • Vitiello A, La Porta R, D’Aiuto V, et al. Pharmacological approach for the reduction of inflammatory and prothrombotic hyperactive state in COVID-19 positive patients by acting on complement cascade. Hum Immunol. 2021;82(4):264–269. online ahead of print.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.