Study of alteplase for respiratory failure in severe acute respiratory syndrome coronavirus 2/COVID-19: Study design of the phase IIa STARS trial

Hunter B Moore, Christopher D Barrett, Ernest E Moore, Rashi Jhunjhunwala, Robert C McIntyre, Peter K Moore, Janice Wang, Negin Hajizadeh, Daniel S Talmor, Angela Sauaia, Michael B Yaffe, Hunter B Moore, Christopher D Barrett, Ernest E Moore, Rashi Jhunjhunwala, Robert C McIntyre, Peter K Moore, Janice Wang, Negin Hajizadeh, Daniel S Talmor, Angela Sauaia, Michael B Yaffe

Abstract

Background: The coronavirus disease 2019 (COVID-19) pandemic has caused a large surge of acute respiratory distress syndrome (ARDS). Prior phase I trials (non-COVID-19) demonstrated improvement in pulmonary function in patients ARDS using fibrinolytic therapy. A follow-up trial using the widely available tissue-type plasminogen activator (t-PA) alteplase is now needed to assess optimal dosing and safety in this critically ill patient population.

Objective: To describe the design and rationale of a phase IIa trial to evaluate the safety and efficacy of alteplase treatment for moderate/severe COVID-19-induced ARDS.

Patients/methods: A rapidly adaptive, pragmatic, open-label, randomized, controlled, phase IIa clinical trial will be conducted with 3 groups: intravenous alteplase 50 mg, intravenous alteplase 100 mg, and control (standard-of-care). Inclusion criteria are known/suspected COVID-19 infection with PaO2/FiO2 ratio <150 mm Hg for > 4 hours despite maximal mechanical ventilation management. Alteplase will be delivered through an initial bolus of 50 mg or 100 mg followed by heparin infusion for systemic anticoagulation, with alteplase redosing if there is a >20% PaO2/FiO2 improvement not sustained by 24 hours.

Results: The primary outcome is improvement in PaO2/FiO2 at 48 hours after randomization. Other outcomes include ventilator- and intensive care unit-free days, successful extubation (no reintubation ≤3 days after initial extubation), and mortality. Fifty eligible patients will be enrolled in a rapidly adaptive, modified stepped-wedge design with 4 looks at the data.

Conclusion: Findings will provide timely information on the safety, efficacy, and optimal dosing of t-PA to treat moderate/severe COVID-19-induced ARDS, which can be rapidly adapted to a phase III trial (NCT04357730; FDA IND 149634).

Keywords: COVID‐19; acute respiratory distress syndrome; clinical trial; coagulopathy; fibrinolysis shutdown; tissue‐type plasminogen activator.

© 2020 The Authors. Research and Practice in Thrombosis and Haemostasis published by Wiley Periodicals LLC on behalf of International Society on Thrombosis and Haemostasis.

Figures

FIGURE 1
FIGURE 1
Study design of the STARS trial

References

    1. Yao XH, Li TY, He ZC, Ping YF, Liu HW, Yu SC, et al. A pathological report of three COVID‐19 cases by minimally invasive autopsies. Zhonghua Bing Li Xue Za Zhi. 2020;49:E009.
    1. Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, 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):475–81.
    1. Wiedermann CJ. Anticoagulant therapy for septic coagulopathy and disseminated intravascular coagulation: where do KyberSept and SCARLET leave us? Acute Med Surg. 2020;7(1):e477.
    1. Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382(18):1708–20.
    1. Grasselli G, Pesenti A, Cecconi M. Critical care utilization for the COVID‐19 outbreak in Lombardy, Italy: early experience and forecast during an emergency response. JAMA. 2020;323(16):1545.
    1. Livingston E, Bucher K. Coronavirus disease 2019 (COVID‐19) in Italy. JAMA. 2020;323(14):1335.
    1. Grasselli G, Zangrillo A, Zanella A, Antonelli M, Cabrini L, Castelli 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.
    1. Bhatraju PK, Ghassemieh BJ, Nichols M, Kim R, Jerome KR, Nalla AK, et al. Covid‐19 in critically ill patients in the Seattle region ‐ case series. N Engl J Med. 2020;382(21):2012–22.
    1. Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med. 2020;180(7):934–43.
    1. Richardson S, Hirsch JS, Narasimhan M, Crawford JM, McGinn T, Davidson KW, et al. Presenting characteristics, comorbidities, and outcomes among 5700 patients hospitalized with COVID‐19 in the New York City area. JAMA. 2020;323(20):2052–9.
    1. Pan C, Chen L, Lu C, Zhang W, Xia JA, Sklar MC, et al. Lung Recruitability in SARS‐CoV‐2 associated acute respiratory distress syndrome: a single‐center, observational study. Am J Respir Crit Care Med. 2020;201(10):1294–7.
    1. Gattinoni L, Chiumello D, Caironi P, Busana M, Romitti F, Brazzi L, et al. COVID‐19 pneumonia: different respiratory treatments for different phenotypes? Intensive Care Med. 2020;46(6):1099–102.
    1. Paget J. Additional observations on obstructions of the pulmonary arteries. Med Chir Trans. 1845;28:353–71.
    1. Vesconi S, Rossi GP, Pesenti A, Fumagalli R, Gattinoni L. Pulmonary microthrombosis in severe adult respiratory distress syndrome. Crit Care Med. 1988;16(2):111–3.
    1. Mergoni M, Caberti P, Vergallo A, Pagliari S, Salvadori A, Gulli E. Vascular obstruction in patients with ARDS. A study with selective pulmonary angiography with a wedged catheter. Minerva Anestesiol. 1991;57(10):932–3.
    1. Greene R. Pulmonary vascular obstruction in the adult respiratory distress syndrome. J Thorac Imaging. 1986;1(3):31–8.
    1. Zapol WM, Jones R. Vascular components of ARDS. Clinical pulmonary hemodynamics and morphology. Am Rev Respir Dis. 1987;136(2):471–4.
    1. Hardaway RM 3rd, Mc KD. Disseminated intravascular coagulation: a cause of shock. Ann Surg. 1959;149(4):462–70.
    1. Cafferata HT, Aggeler PM, Robinson AJ, Blaisdell FW. Intravascular coagulation in the surgical patient: its significance and diagnosis. Am J Surg. 1969;118(2):281–91.
    1. Turpini R, Stefanini M. The nature and mechanism of the hemostatic breakdown in the course of experimental hemorrhagic shock. J Clin Invest. 1959;38(1, Part 1):53–65.
    1. Hardaway RM, Brune WH, Geever EF, Burns JW, Mock HP. Studies on the role of intravascular coagulation in irreversible hemorrhagic shock. Ann Surg. 1962;155:241–50.
    1. Hardaway RM, Drake DC. Prevention of “irreversible” hemorrhagic shock with fibrinolysin. Ann Surg. 1963;157:39–47.
    1. Hardaway RM. The significance of coagulative and thrombotic changes after haemorrhage and injury. J Clin Pathol. 1970;4 (suppl):110–20.
    1. Greene R, Lind S, Jantsch H, Wilson R, Lynch K, Jones R, et al. Pulmonary vascular obstruction in severe ARDS: angiographic alterations after i.v. fibrinolytic therapy. AJR Am J Roentgenol. 1987;148(3):501–8.
    1. Hardaway RM, Harke H, Tyroch AH, Williams CH, Vazquez Y, Krause GF. Treatment of severe acute respiratory distress syndrome: a final report on a phase I study. Am Surg. 2001;67(4):377–82.
    1. Etchill E, Sperry J, Zuckerbraun B, Alarcon L, Brown J, Schuster K, et al. The confusion continues: results from an American association for the surgery of trauma survey on massive transfusion practices among United States trauma centers. Transfusion. 2016;56(10):2478–86.
    1. Oxley TJ, Mocco J, Majidi S, Kellner CP, Shoirah H, Singh IP, et al. Large‐vessel stroke as a presenting feature of Covid‐19 in the young. N Engl J Med. 2020;382(20):e60.
    1. Esmon CT. Molecular events that control the protein C anticoagulant pathway. Thromb Haemost. 1993;70(1):29–35.
    1. World Medical Association Declaration of Helsinki . Recommendations guiding physicians in biomedical research involving human subjects. Cardiovasc Res. 1997;35(1):2–3.
    1. Konstantinides S, Geibel A, Heusel G, Heinrich F, Kasper W, Management S, et al. Heparin plus alteplase compared with heparin alone in patients with submassive pulmonary embolism. N Engl J Med. 2002;347(15):1143–50.
    1. Carless PA, Henry DA, Moxey AJ, O'Connell DL, Brown T, Fergusson DA. Cell salvage for minimising perioperative allogeneic blood transfusion. Cochrane Database Syst Rev. 2006;4 10.1002/14651858.CD001888.pub2
    1. Spanos A, Jhanji S, Vivian‐Smith A, Harris T, Pearse RM. Early microvascular changes in sepsis and severe sepsis. Shock. 2010;33(4):387–91.
    1. Driessen A, Schafer N, Albrecht V, Schenk M, Frohlich M, Sturmer EK, et al. Infrastructure and clinical practice for the detection and management of trauma‐associated haemorrhage and coagulopathy. Eur J Trauma Emerg Surg. 2015;41(4):413–20.
    1. Geddes AE, Burlew CC, Wagenaar AE, Biffl WL, Johnson JL, Pieracci FM, et al. Expanded screening criteria for blunt cerebrovascular injury: a bigger impact than anticipated. Am J Surg. 2016;212(6):1167–74.
    1. Dumville JC, Hahn S, Miles JNV, Torgerson DJ. The use of unequal randomisation ratios in clinical trials: a review. Contemp Clin Trials. 2006;27(1):1–12.
    1. Jitlal M, Khan I, Lee SM, Hackshaw A. Stopping clinical trials early for futility: retrospective analysis of several randomised clinical studies. Br J Cancer. 2012;107(6):910–7.
    1. Pieracci FM, Leasia K, Bauman Z, Eriksson EA, Lottenberg L, Majercik S, et al. A multicenter, prospective, controlled clinical trial of surgical stabilization of rib fractures in patients with severe, nonflail fracture patterns (Chest Wall Injury Society NONFLAIL). J Trauma Acute Care Surg. 2020;88(2):249–57.
    1. investigators G . An international randomized trial comparing four thrombolytic strategies for acute myocardial infarction. N Engl J Med. 1993;329(10):673–82.
    1. Lees KR, Emberson J, Blackwell L, Bluhmki E, Davis SM, Donnan GA, et al. Effects of alteplase for acute stroke on the distribution of functional outcomes: a pooled analysis of 9 trials. Stroke. 2016;47(9):2373–9.
    1. Gamble C, Krishan A, Stocken D, Lewis S, Juszczak E, Dore C, et al. Guidelines for the content of statistical analysis plans in clinical trials. JAMA. 2017;318(23):2337–43.
    1. Rothman KJ. No adjustments are needed for multiple comparisons. Epidemiology (Cambridge, Mass). 1990;1(1):43–6.
    1. Perneger TV. What’s wrong with Bonferroni adjustments. BMJ. 1998;316(7139):1236–8.
    1. Panigada M, Bottino N, Tagliabue P, Grasselli G, Novembrino C, Chantarangkul V, et al. Hypercoagulability of COVID‐19 patients in intensive care unit. a report of thromboelastography findings and other parameters of hemostasis. J Thromb Haemost. 2020;18(7):1738–42.
    1. Tang N, Li D, Wang X, Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost. 2020;18(4):844–7.
    1. Ranucci M, Ballotta A, Di Dedda U, Bayshnikova E, Dei Poli M, Resta M, et al. The procoagulant pattern of patients with COVID‐19 acute respiratory distress syndrome. J Thromb Haemost. 2020;18(7):1747–51.
    1. Klok FA, Kruip M, van der Meer NJM, Arbous MS, Gommers D, Kant KM, et al. Incidence of thrombotic complications in critically ill ICU patients with COVID‐19. Thromb Res. 2020;191:145–7.
    1. He XL, Liu Z, Xia SY. Vascular endothelial injuries and changes of blood coagulation and fibrinolysis indexes in patients with acute respiratory distress syndrome. Chin Med Sci. 2004;19(4):252–6.
    1. Hofstra JJ, Haitsma JJ, Juffermans NP, Levi M, Schultz MJ. The role of bronchoalveolar hemostasis in the pathogenesis of acute lung injury. Semin Thromb Hemost. 2008;34(5):475–84.
    1. Idell S. Coagulation, fibrinolysis, and fibrin deposition in acute lung injury. Crit Care Med. 2003;31(4 Suppl):S213–20.
    1. Idell S, Koenig KB, Fair DS, Martin TR, McLarty J, Maunder RJ. Serial abnormalities of fibrin turnover in evolving adult respiratory distress syndrome. Am J Physiol. 1991;261(4 Pt 1):L240–8.
    1. Olson TM, Driscoll DJ, Edwards WD, Puga FJ, Danielson GK. Pulmonary microthrombi. Caveat for successful modified Fontan operation. J Thorac Cardiovasc Surg. 1993;106(4):739–44.
    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(10229):1054–62.
    1. Spero JA, Lewis JH, Hasiba U. Disseminated intravascular coagulation. Findings in 346 patients. Thromb Haemost. 1980;43(1):28–33.
    1. Gando S, Levi M, Toh CH. Disseminated intravascular coagulation. Nat Rev Dis Primers. 2016;2:16037.
    1. Doshi SN, Marmur JD. Evolving role of tissue factor and its pathway inhibitor. Crit Care Med. 2002;30(5 Suppl):S241–S250.
    1. Sungurlu S, Kuppy J, Balk RA. Role of antithrombin iii and tissue factor pathway in the pathogenesis of sepsis. Crit Care Clin. 2020;36(2):255–65.
    1. Varga Z, Flammer AJ, Steiger P, Haberecker M, Andermatt R, Zinkernagel AS, et al. Endothelial cell infection and endotheliitis in COVID‐19. Lancet. 2020;395(10234):1417–8.
    1. Carty CL, Heagerty P, Heckbert SR, Jarvik GP, Lange LA, Cushman M, et al. Interaction between fibrinogen and IL‐6 genetic variants and associations with cardiovascular disease risk in the cardiovascular health study. Ann Hum Genet. 2010;74(1):1–10.
    1. Spiel AO, Mayr FB, Firbas C, Quehenberger P, Jilma B. Validation of rotation thrombelastography in a model of systemic activation of fibrinolysis and coagulation in humans. J Thromb Haemost. 2006;4(2):411–6.
    1. Biemond BJ, Levi M, Ten Cate H, Van der Poll T, Buller HR, Hack CE, et al. Plasminogen activator and plasminogen activator inhibitor I release during experimental endotoxaemia in chimpanzees: effect of interventions in the cytokine and coagulation cascades. Clin Sci (Lond). 1995;88(5):587–94.
    1. Levi M, ten Cate H, Bauer KA, van der Poll T, Edgington TS, Buller HR, et al. Inhibition of endotoxin‐induced activation of coagulation and fibrinolysis by pentoxifylline or by a monoclonal anti‐tissue factor antibody in chimpanzees. J Clin Invest. 1994;93(1):114–20.
    1. Renckens R, Weijer S, de Vos AF, Pater JM, Meijers JC, Hack CE, et al. Inhibition of plasmin activity by tranexamic acid does not influence inflammatory pathways during human endotoxemia. Arterioscler Thromb Vasc Biol. 2004;24(3):483–8.
    1. Wright FLVT, Moore EE, Moore HB, Wohlauer MV, Urban S, Nydam TL, et al. Fibrinolysis shutdown correlates to thromboembolic events in severe COVID‐19 infection. JACS. 2020;S1072‐7515(20):30400‐2.
    1. Ruhl H, Berens C, Winterhagen A, Muller J, Oldenburg J, Potzsch B. Label‐free kinetic studies of hemostasis‐related biomarkers including d‐dimer using autologous serum transfusion. PLoS ONE. 2015;10(12):e0145012.
    1. Chakrabarti R, Hocking ED, Fearnley GR. Reaction pattern to three stresses–electroplexy, surgery, and myocardial infarction–of fibrinolysis and plasma fibrinogen. J Clin Pathol. 1969;22(6):659–2.
    1. Moore HB, Moore EE, Neal MD, Sheppard FR, Kornblith LZ, Draxler DF, et al. Fibrinolysis shutdown in trauma: historical review and clinical implications. Anesth Analg. 2019;129(3):762–73.
    1. Moore HB, Moore EE, Gonzalez E, Chapman MP, Chin TL, Silliman CC, et al. Hyperfibrinolysis, physiologic fibrinolysis, and fibrinolysis shutdown: the spectrum of postinjury fibrinolysis and relevance to antifibrinolytic therapy. J Trauma Acute Care Surg. 2014;77(6):811–7.
    1. Moore HB, Moore EE, Liras IN, Gonzalez E, Harvin JA, Holcomb JB, et al. Acute fibrinolysis shutdown after injury occurs frequently and increases mortality: a multicenter evaluation of 2540 severely injured patients. J Am Coll Surg. 2016;222(4):347–55.
    1. Leeper CM, Neal MD, McKenna CJ, Gaines BA. Trending fibrinolytic dysregulation: fibrinolysis shutdown in the days after injury is associated with poor outcome in severely injured children. Ann Surg. 2017;266(3):508–15.
    1. Meizoso JP, Karcutskie CA, Ray JJ, Namias N, Schulman CI, Proctor KG. Persistent fibrinolysis shutdown is associated with increased mortality in severely injured trauma patients. J Am Coll Surg. 2017;224(4):575–82.
    1. Roberts DJ, Kalkwarf KJ, Moore HB, Cohen MJ, Fox EE, Wade CE, et al. Time course and outcomes associated with transient versus persistent fibrinolytic phenotypes after injury: a nested, prospective, multicenter cohort study. J Trauma Acute Care Surg. 2019;86(2):206–13.
    1. Gall LS, Vulliamy P, Gillespie S, Jones TF, Pierre RSJ, Breukers SE, et al. The S100A10 pathway mediates an occult hyperfibrinolytic subtype in trauma patients. Ann Surg. 2018;269(6):1184–91.
    1. Moore HB, Moore EE, Chapman MP, Hansen KC, Cohen MJ, Pieracci FM, et al. Does tranexamic acid improve clot strength in severely injured patients who have elevated fibrin degradation products and low fibrinolytic activity, measured by thrombelastography? J Am Coll Surg. 2019;229(1):92–101.
    1. Ji HL, Zhao R, Matalon S, Matthay MA. Elevated plasmin(ogen) as a common risk factor for COVID‐19 susceptibility. Physiol Rev. 2020;100(3):1065–75.
    1. Slutsky AS, Villar J. Early paralytic agents for ARDS? Yes, No, and sometimes. N Engl J Med. 2019;380(21):2061–3.
    1. Gebistorf F, Karam O, Wetterslev J, Afshari A. Inhaled nitric oxide for acute respiratory distress syndrome (ARDS) in children and adults. Cochrane Database Syst Rev. 2016;6 10.1002/14651858.CD002787.pub3
    1. Fuller BM, Mohr NM, Skrupky L, Fowler S, Kollef MH, Carpenter CR. The use of inhaled prostaglandins in patients with ARDS: a systematic review and meta‐analysis. Chest. 2015;147(6):1510–22.
    1. Fan E, Brodie D, Slutsky AS. Acute respiratory distress syndrome: advances in diagnosis and treatment. JAMA. 2018;319(7):698–710.
    1. Coccolini F, Sartelli M, Kluger Y, Pikoulis E, Karamagioli E, Moore EE, et al. COVID‐19 the showdown for mass casualty preparedness and management: the Cassandra syndrome. World J Emerg Surg. 2020;15(1):26.
    1. Adaptive designs for medical device clinical studies DRAFT GUIDANCE2015. [Accessed 2020 May 15] Available from
    1. Pallmann P, Bedding AW, Choodari‐Oskooei B, Dimairo M, Flight L, Hampson LV, et al. Adaptive designs in clinical trials: why use them, and how to run and report them. BMC Med. 2018;16(1):29.
    1. Hemming K, Haines TP, Chilton PJ, Girling AJ, Lilford RJ. The stepped wedge cluster randomised trial: rationale, design, analysis, and reporting. BMJ. BMJ. 2015;350:h391.
    1. Brown CA, Lilford RJ. The stepped wedge trial design: a systematic review. BMC Med Res Methodol. 2006;6(1):54.
    1. Group TGHS . The Gambia hepatitis intervention study. Can Res. 1987;47(21):5782–7.
    1. Nuzzo R. Scientific method: statistical errors. Nature. 2014;506(7487):150–2.
    1. Moore HB, Barrett CD, Moore EE, McIntyre RC, Moore PK, Talmor DS, et al. Is there a role for tissue plasminogen activator (tPA) as a novel treatment for refractory COVID‐19 associated acute respiratory distress syndrome (ARDS)? J Trauma Acute Care Surg. 2020;88(6):713–4.
    1. Wang J, Hajizadeh N, Moore EE, McIntyre RC, Moore PK, Veress LA, et al. Tissue plasminogen activator (tPA) treatment for COVID‐19 associated acute respiratory distress syndrome (ARDS): a case series. J Thromb Haemost. 202018(7):1752–5.

Source: PubMed

3
Abonneren