Defibrotide Therapy for SARS-CoV-2 ARDS

David Frame, Gianni B Scappaticci, Thomas M Braun, Mary Maliarik, Thomas H Sisson, Steven W Pipe, Daniel A Lawrence, Paul G Richardson, Michael Holinstat, Robert C Hyzy, Daniel R Kaul, Kevin S Gregg, Vibha N Lama, Gregory A Yanik, David Frame, Gianni B Scappaticci, Thomas M Braun, Mary Maliarik, Thomas H Sisson, Steven W Pipe, Daniel A Lawrence, Paul G Richardson, Michael Holinstat, Robert C Hyzy, Daniel R Kaul, Kevin S Gregg, Vibha N Lama, Gregory A Yanik

Abstract

Background: SARS-CoV-2-related ARDS is associated with endothelial dysfunction and profound dysregulation of the thrombotic-fibrinolytic pathway. Defibrotide is a polyanionic compound with fibrinolytic, antithrombotic, and antiinflammatory properties.

Research question: What is the safety and tolerability of defibrotide in patients with severe SARS-CoV-2 infections?

Study design and methods: We report a prospective, open-label, single-center safety trial of defibrotide for the management of SARS-CoV-2-related ARDS. Eligible participants were 18 years of age or older with clinical and radiographic signs of ARDS, no signs of active bleeding, a serum D-dimer of more than twice upper limit of normal, and positive polymerase chain reaction-based results for SARS-CoV-2. Defibrotide (6.25 mg/kg/dose IV q6h) was administered for a planned 7-day course, with serum D-dimer levels and respiratory function monitored daily during therapy.

Results: Twelve patients (median age, 63 years) were treated, with 10 patients receiving mechanical ventilation and 6 receiving vasopressor support at study entry. The median D-dimer was 3.25 μg/ml (range, 1.33-12.3) at study entry. The median duration of therapy was 7 days. No hemorrhagic or thrombotic complications occurred during therapy. No other adverse events attributable to defibrotide were noted. Four patients met the day 7 pulmonary response parameter, all four showing a decrease in serum D-dimer levels within the initial 72 h of defibrotide therapy. Three patients died of progressive pulmonary disease 11, 17, and 34 days after study entry. Nine patients (75%) remain alive 64 to 174 days after initiation of defibrotide. Day 30 all-cause mortality was 17% (95% CI, 0%-35%). All patients with a baseline Pao2 to Fio2 ratio of ≥ 125 mm Hg survived, whereas the three patients with a baseline Pao2 to Fio2 ratio of < 125 mm Hg died.

Interpretation: The use of defibrotide for management of SARS-CoV-2-related ARDS proved safe and tolerable. No hemorrhagic or thrombotic complications were reported during therapy, with promising outcomes in a patient population with a historically high mortality rate.

Trial registry: ClinicalTrials.gov; No.: NCT04530604; URL: www.

Clinicaltrials: gov.

Keywords: ARDS; COVID-19; SARS-CoV-2; defibrotide.

Copyright © 2022 American College of Chest Physicians. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
A, B, Chest radiographs from patient 6 obtained at study entry (A) and day 3 (B). At the time of study entry, the patient was mechanically ventilated, on 50% Fio2, with positive end-expiratory pressure of 10 cm H2O. The patient subsequently was extubated by day 4, and all supplemental oxygen support was removed by day 7.
Figure 2
Figure 2
Bar graph showing WHO ordinal scores obtained at baseline, day 7, and day 14 from study entry. The day 30 score was obtained 30 days after completion of study therapy. WHO = World Health Organization.
Figure 3
Figure 3
Graph showing overall survival from time of study entry. Shaded area denotes pointwise 95% CI.

References

    1. World Heath Organization Coronavirus (COVID-19) dashboard. World Heath Organization website.
    1. 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.
    1. Chen N., Zhou M., Dong X., 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.
    1. Bermejo-Martin J.F., Martín-Fernandez M., López-Mestanza C., et al. Shared features of endothelial dysfunction between sepsis and its preceding risk factors (aging and chronic disease) J Clin Med. 2018;7(11):400–415.
    1. Libby P., Luscher P. COVID-19 is, in the end, an endothelial disease. Eur Heart J. 2020;41(32):3038–3044.
    1. Jin Y., Ji W., Yang H., et al. Endothelial activation and dysfunction in COVID-19: from basic mechanisms to potential therapeutic approaches. Signal Transduct Target Ther. 2020;5(1):293.
    1. Klok F.A., Kruip M.J., van der Meer J.M., et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res. 2020;191:145–147.
    1. Llitjos J.F., Leclerc M., Chochois C., et al. High incidence of venous thromboembolic events in anticoagulated severe COVID-19 patients. J Thrombosis and Hemostas. 2020;18(7):1743–1746.
    1. Xu Z., Shi L., Wang Y., et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome 2020. Lancet Respir Med. 2020;8(4):420–422.
    1. Fox S.E., Akmatbekov A., Harbert J.L., et al. Pulmonary and cardiac pathology in African American patients with COVID-19: an autopsy series from New Orleans. Lancet Respir Med. 2020;8(7):681–686.
    1. The REMAP-CAP, ACTIV-4a, ATTACC Investigators, Ryan Zarychanski. Therapeutic anticoagulation in critically ill patients with Covid-19—preliminary report [published online ahead of print March 12, 2021]. medRxiv. doi: 10.1101/2021.03.10.21252749.
    1. Yin S., Huang M., Li D., et al. Difference of coagulation features between severe pneumonia induced by SARS-CoV2 and non-SARS-CoV2. J Thromb Thrombolysis. 2021;51(4):1107–1110.
    1. Townsend L., Fogarty H., Dyer A., et al. Prolonged elevation of D-dimer levels in convalescent COVID-19 patients is independent of the acute phase response. J Thromb Haemost. 2021;19(4):1064–1070.
    1. Hunt B., Retter A., McClintock C. Practical guidance for the prevention of thrombosis and management of coagulopathy and disseminated intravascular coagulation of patients infected with COVID-19. Thromb Hemost. March 25th, 2020
    1. 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.
    1. Richardson P.G., Riches M.L., Kernan N.A., et al. Phase 3 trial of defibrotide for the treatment of severe veno-occlusive disease and multi-organ failure. Blood. 2016;127(13):1656–1665.
    1. Kornblum N., Ayyanar K., Benimetskaya L., Richardson P., Iacobelli M., Stein C.A. Defibrotide, a polydisperse mixture of single-stranded phosphodiester oligonucleotides with lifesaving activity in severe hepatic veno-occlusive disease: clinical outcomes and potential mechanisms of action. Oligonucleotides. 2006;16:105–114.
    1. Richardson P.G., Carreras E., Iacobelli M., Nejadnik B. The use of defibrotide in blood and marrow transplantation. Blood Adv. 2018;2(12):1495–1509.
    1. Richardson P., Linden E., Revta C., Ho V. Use of defibrotide in the treatment and prevention of veno-occlusive disease. Expert Rev Hematol. 2009;2:365–376.
    1. Bianchi G., Barone D., Lanzarotti E., et al. Defibrotide, a single-stranded polydeoxyribonucleotide acting as an adenosine receptor agonist. Eur J Pharmacol. 1993;238:327–334.
    1. Palomo M., Diaz-Ricart M., Rovira M., et al. Defibrotide prevents the activation of macrovascular and microvascular endothelia caused by soluble factors released to blood by autologous hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2011;17(4):497–506.
    1. Richardson E., García-Bernal D., Calabretta E., et al. Defibrotide: potential for treating endothelial dysfunction related to viral and post-infectious syndromes. Expert Opin Ther Targets. 2021;25(6):423–433.
    1. Echart C.L., Graziadio B., Somaini S., et al. The fibrinolytic mechanism of defibrotide: effect of defibrotide on plasmin activity. Blood Coag Fibrinolysis. 2009;20(8):627–634.
    1. Calabretta E., Moraleda J.M., Iacobelli M., et al. COVID-19-induced endotheliitis: emerging evidence and possible therapeutic strategies. Br J Haematol. 2021;193(1):43–51.
    1. Palomo M., Mir E., Rovira M., et al. What is going on between defibrotide and endothelial cells? Snapshots reveal the hot spots of their romance. Blood. 2016;127(13):1719–1727.
    1. World Health Organization Clinical management for severe acute respiratory infection (SARI) when COVID-19 disease is suspected.
    1. Schulman S., Kearon C. Subcommittee on Control of Anticoagulation of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis. Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients. J Thromb Haemost. 2005;3(4):692–694.
    1. Xiao Y.J., Dong X., Yang H.Z., et al. Clinical features of 141 fatal cases of coronavirus disease in Jinyintan Hospital in Wuhan, China. Zhonghua Jie He He Hu Xi Za Zhi. 2021;44(4):354–359.
    1. COVID-ICU Group on behalf of the REVA Network and the COVID-ICU Investigators Clinical characteristics and day-90 outcomes of 4244 critically ill adults with COVID-19: a prospective cohort study. Intensive Care Medicine. 2021;47:60–73.
    1. Grasselli G., Zangrillo A., Zanella A., et al. Baseline characteristics and outcomes of 1591 patients infected with SARS-CoV-2 admitted to ICUs of the Lombardy Region, Italy. JAMA. 2020;323(16):1574–1581.
    1. Cummings M.J., Baldwin M.R., Abrams D., et al. Epidemiology, clinical course, and outcomes of critically ill adults with COVID-19 in New York City: a prospective cohort study. Lancet. 2020;395(10239):1763–1770.
    1. Xie J., Covassin N., Fan Z., et al. Association between hypoxemia and mortality in patients with COVID-19. Mayo Clin Proc. 2020;95(6):1138–1147.
    1. Tomazini B.N., Maia I.S., Cavalcanti A.B., 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(13):1–11.
    1. García-Bernal D., Richardson E., Vlodavsky I., et al. Endothelial dysfunction and its critical role in COVID-19-associated coagulopathy: defibrotide as an endothelium-protective, targeted therapy. EJHaem. 2021;2(3):680–681.
    1. Ruggeri A., Voza A., Liberatore C., et al. Proceedings of the American Society of Hematology; December 11-14. 2021. Use of defibrotide in patients with COVID-19 pneumonia: results of the Defi-VID19 phase 2 trial. Abstract 672.
    1. Gerosa C., Calvani A.B., Cornelli U., et al. A multicenter study of defibrotide in the prevention of deep venous thrombosis. Minerva Chir. 1989;44(10):1507–1516.
    1. Rizzi A., Radaelli F., Pisano M., et al. Prevention of deep venous thrombosis with defibrotide in chest surgery. Controlled multicenter study versus heparin. Minerva Med. 1987;78(11):745–750.
    1. Strouse C., Richardson P., Prentice G., et al. Defibrotide for treatment of severe veno-occlusive disease in pediatrics and adults: an exploratory analysis using data from the center for international blood and marrow transplant research. Biol Blood Marrow Transplant. 2016;22(7):1306–1312.
    1. Tomar B., Anders H.J., Desai J., Mulay S.R. Neutrophils and neutrophil extra-cellular traps drive necroinflammation in COVID-19. Cells. 2020;9(6):1383.
    1. Thierry A.R., Roch B. SARS-CoV2 may evade innate immune response, causing uncontrolled neutrophil extracellular traps formation and multi-organ failure. Clin Sci. 2020;134(12):1295–1300.
    1. Zou Y., Yalavarthi S., H Shi, et al. Neutrophil extracellular traps in COVID-19. CI Insight. 2020;5(11)
    1. Barnes B.J., Adrover J.M., Baxter-Stoltzfus A., et al. Targeting potential drivers of COVID-19: neutrophil extracellular traps. J Exp Med. 2020;217(6)
    1. Sawadogo S.A., Dighero-Kemp B., Ouedraogo D.D., et al. How NETosis could drive “post-COVID-19 syndrome” among survivors. Immunol Lett. 2020;228:35–37.
    1. Fousert E., Toes R., Desai J. Neutrophil extracellular traps (NETs) take the central stage in driving autoimmune responses. Cells. 2020;9(4):915.
    1. Hui S., Ghandi A.A., Smith S.A., et al. Endothelium-protective, histone-neutralizing properties of the polyanionic agent defibrotide. JCI Insight. 2021;6(17)
    1. Maccio A., Madeddu C., Caocci G., Oppi S., La Nasa G. Defibrotide in the COVID-19 coagulopathy: what is the timing? J Thromb Haemost. 2020;18(11):3113–3115.

Source: PubMed

3
S'abonner