Design and Rationale of a Randomized, Double-Blind, Placebo-Controlled, Phase 2/3 Study Evaluating Dociparstat in Acute Lung Injury Associated with Severe COVID-19

Joseph A Lasky, Jyotsna Fuloria, Marion E Morrison, Randall Lanier, Odin Naderer, Tom Brundage, Allen Melemed, Joseph A Lasky, Jyotsna Fuloria, Marion E Morrison, Randall Lanier, Odin Naderer, Tom Brundage, Allen Melemed

Abstract

Introduction: The COVID-19 global pandemic caused by the novel coronavirus, SARS-CoV-2, and the consequent morbidity and mortality attributable to progressive hypoxemia and subsequent respiratory failure threaten to overrun hospital critical care units globally. New agents that address the hyperinflammatory "cytokine storm" and hypercoagulable pathology seen in these patients may be a promising approach to treat patients, minimize hospital stays, and ensure hospital wards and critical care units are able to operate effectively. Dociparstat sodium (DSTAT) is a glycosaminoglycan derivative of heparin with robust anti-inflammatory properties, with the potential to address underlying causes of coagulation disorders with substantially reduced risk of bleeding compared to commercially available heparin.

Methods: This study is a randomized, double-blind, placebo-controlled, phase 2/3 trial to determine the safety and efficacy of DSTAT added to standard of care in hospitalized adults with COVID-19 who require supplemental oxygen. Phase 2 will enroll 12 participants in each of two dose-escalating cohorts to confirm the safety of DSTAT in this population. Following review of the data, an additional 50 participants will be enrolled. Contingent upon positive results, phase 3 will enroll approximately 450 participants randomized to DSTAT or placebo. The primary endpoint is the proportion of participants who survive and do not require mechanical ventilation through day 28.

Discussion: Advances in standard of care, recent emergency use authorizations, and positive data with dexamethasone have likely contributed to an increasing proportion of patients who are surviving without the need for mechanical ventilation. Therefore, examining the time to improvement in the NIAID score will be essential to provide a measure of drug effect on recovery. Analysis of additional endpoints, including supportive biomarkers (e.g., IL-6, HMGB1, soluble-RAGE, D-dimer), will be performed to further define the effect of DSTAT in patients with COVID-19 infection.

Trial registration: ClinicalTrials.gov identifier; NCT04389840, Registered 13 May 2020.

Keywords: Acute lung injury; COVID-19; Dociparstat; HMGB1; NETs; Neutrophil extracellular traps; P-selectin; Platelet factor 4.

Figures

Fig. 1
Fig. 1
Potential of DSTAT in COVID-19. Moderate-to-severe COVID-19 manifests with excessive inflammation, infiltration of activated immune cells into the lungs and coagulation disorders (e.g., clot formation) in the blood and tissues. DSTAT has the potential to prevent and mitigate these severe effects of SARS-CoV-2 infection by inhibiting several pathogenic pathways. (DSTAT, dociparstat sodium; HMGB1, high mobility group box protein 1; IL-6, interleukin-6; MCP-1, monocyte chemoattractant protein-1; NETs, neutrophil extracellular traps; PF4, platelet factor 4)
Fig. 2
Fig. 2
CMX-DS-004 study schematic. All participants receive randomized treatment for up to 7 days and are followed through day 28. The continuous infusion dose administered in phase 2, cohorts 2 and 3 will be confirmed after review of data by an independent Data Monitoring Committee (DMC) from prior cohort(s). The phase 3 continuous infusion dose will be the same as administered in phase 2, cohort 3

References

    1. Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA J Am Med Assoc Am Med Assoc. 2020;323:1061–1069. doi: 10.1001/jama.2020.1585.
    1. Wu F, Zhao S, Yu B, Chen YM, Wang W, Song ZG, et al. A new coronavirus associated with human respiratory disease in China. Nat Nat Res. 2020;579:265–269.
    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 Lancet Publ Group. 2020;8:420–422. doi: 10.1016/S2213-2600(20)30076-X.
    1. Tian S, Hu W, Niu L, Liu H, Xu H, Xiao SY. Pulmonary pathology of early-phase 2019 novel coronavirus (COVID-19) pneumonia in two patients with lung cancer. J Thorac Oncol. 2020;15:700–704. doi: 10.1016/j.jtho.2020.02.010.
    1. Levi M, Thachil J, Iba T, Levy JH. Coagulation abnormalities and thrombosis in patients with COVID-19. Lancet Haematol. 2020;7:e438–e440. doi: 10.1016/S2352-3026(20)30145-9.
    1. Tang N, Bai H, Chen X, Gong J, Li D, Sun Z. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J Thromb Haemost. 2020;18:1094–1099. doi: 10.1111/jth.14817.
    1. Paranjpe I, Fuster V, Lala A, Russak AJ, Glicksberg BS, Levin MA, et al. Association of treatment dose anticoagulation with in-hospital survival among hospitalized patients with COVID-19. J Am Coll Cardiol. 2020;76:122–124. doi: 10.1016/j.jacc.2020.05.001.
    1. Lauzier F, Arnold DM, Rabbat C, Heels-Ansdell D, Zarychanski R, Dodek P, et al. Risk factors and impact of major bleeding in critically ill patients receiving heparin thromboprophylaxis. Intensive Care Med. 2013;39:2135–2143. doi: 10.1007/s00134-013-3044-3.
    1. Rao NV, Argyle B, Xu X, Reynolds PR, Walenga JM, Prechel M, et al. Low anticoagulant heparin targets multiple sites of inflammation, suppresses heparin-induced thrombocytopenia, and inhibits interaction of RAGE with its ligands. Am J Physiol Cell Physiol. 2010;299:C97–110. doi: 10.1152/ajpcell.00009.2010.
    1. Chen L, Long X, Xu Q, Tan J, Wang G, Cao Y, et al. Elevated serum levels of S100A8/A9 and HMGB1 at hospital admission are correlated with inferior clinical outcomes in COVID-19 patients. Cell Mol Immunol. 2020;17:992–994. doi: 10.1038/s41423-020-0492-x.
    1. Kim S, Young Kim S, Pribis JP, Lotze M, Mollen KP, Shapiro R, et al. Signaling of high mobility group box 1 (HMGB1) through toll-like receptor 4 in macrophages requires CD14. Mol Med. 2013;19:88–98. doi: 10.2119/molmed.2012.00306.
    1. Herold T, Jurinovic V, Arnreich C, Lipworth BJ, Hellmuth JC, Von Bergwelt-Baildon M, Klein M, et al. Elevated levels of IL-6 and CRP predict the need for mechanical ventilation in COVID-19. J Allergy Clin Immunol. 2020;146(1):128–136.e4. doi: 10.1016/j.jaci.2020.05.008.
    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. Sharma L, Wu J, Patel V, Sitapara R, Rao NV, Kennedy TP, et al. Partially-desulfated heparin improves survival in Pseudomonas pneumonia by enhancing bacterial clearance and ameliorating lung injury. J Immunotoxicol. 2014;11:260–267. doi: 10.3109/1547691X.2013.839587.
    1. Griffin KL, Fischer BM, Kummarapurugu AB, Zheng S, Kennedy TP, Rao NV, et al. 2-O, 3-O-desulfated heparin inhibits neutrophil elastase-induced HMGB-1 secretion and airway inflammation. Am J Respir Cell Mol Biol. 2014;50:684–689. doi: 10.1165/rcmb.2013-0338RC.
    1. Zheng S, Kummarapurugu AB, Afosah DK, Sankaranarayanan NV, Boothello RS, Desai UR, et al. 2-O, 3-O desulfated heparin blocks high mobility group box 1 release by inhibition of p300 acetyltransferase activity. Am J Respir Cell Mol Biol. 2017;56:90–98. doi: 10.1165/rcmb.2016-0069OC.
    1. Fiuza C, Bustin M, Talwar S, Tropea M, Gerstenberger E, Shelhamer JH, et al. Inflammation-promoting activity of HMGB1 on human microvascular endothelial cells. Blood. 2003;101:2652–2660. doi: 10.1182/blood-2002-05-1300.
    1. Nativel B, Marimoutou M, Thon-Hon VG, Gunasekaran MK, Andries J, Stanislas G, et al. Soluble HMGB1 is a novel adipokine stimulating IL-6 secretion through RAGE receptor in SW872 preadipocyte cell line: contribution to chronic inflammation in fat tissue. Chen X, editor. PLoS ONE. 2013;8:e76039. doi: 10.1371/journal.pone.0076039.
    1. Middleton EA, He X-Y, Denorme F, Campbell RA, Ng D, Salvatore SP, et al. Neutrophil extracellular traps (NETs) contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood. 2020;136:1169–1179. doi: 10.1182/blood.2020007008.
    1. Barnes BJ, Adrover JM, Baxter-Stoltzfus A, Borczuk A, Cools-Lartigue J, Crawford JM, et al. Targeting potential drivers of COVID-19: Neutrophil extracellular traps. J Exp Med. 2020;217(6):e20200652. doi: 10.1084/jem.20200652.
    1. Zuo Y, Yalavarthi S, Shi H, Gockman K, Zuo M, Madison JA, et al. Neutrophil extracellular traps in COVID-19. JCI insight. 2020;5(11):e138999.
    1. Kowalska MA, Zhao G, Zhai L, David G, Marcus S, Krishnaswamy S, et al. Modulation of protein C activation by histones, platelet factor 4, and heparinoids: new insights into activated protein C formation. Arterioscler Thromb Vasc Biol. 2014;34:120–126. doi: 10.1161/ATVBAHA.113.302236.
    1. Manne BK, Denorme F, Middleton EA, Portier I, Rowley JW, Stubben CJ, et al. Platelet gene expression and function in COVID-19 patients. Blood. 2020;136(11):1317–1329. doi: 10.1182/blood.2020007214.
    1. Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, et al. Remdesivir for the treatment of Covid-19—preliminary report. N Engl J Med. 2020;383:992–994. doi: 10.1056/NEJMoa2007764.
    1. Harrill AH, Roach J, Fier I, Eaddy JS, Kurtz CL, Antoine DJ, et al. The effects of heparins on the liver: application of mechanistic serum biomarkers in a randomized study in healthy volunteers. Clin Pharmacol Ther. 2012;92:214–220. doi: 10.1038/clpt.2012.40.
    1. Joglekar MV, Quintana Diez PM, Marcus S, Qi R, Espinasse B, Wiesner MR, et al. Disruption of PF4/H multimolecular complex formation with a minimally anticoagulant heparin (ODSH) Thromb Haemost. 2012;107:717–725. doi: 10.1160/TH11-11-0795.

Source: PubMed

3
Abonnere