Inhaled Sargramostim (Recombinant Human Granulocyte-Macrophage Colony-Stimulating Factor) for COVID-19-Associated Acute Hypoxemia: Results of the Phase 2, Randomized, Open-Label Trial (iLeukPulm)

Robert Paine, Robert Chasse, E Scott Halstead, Jay Nfonoyim, David J Park, Timothy Byun, Bela Patel, Guido Molina-Pallete, Estelle S Harris, Fiona Garner, Lorinda Simms, Sanjeev Ahuja, John L McManus, Debasish F Roychowdhury, Robert Paine, Robert Chasse, E Scott Halstead, Jay Nfonoyim, David J Park, Timothy Byun, Bela Patel, Guido Molina-Pallete, Estelle S Harris, Fiona Garner, Lorinda Simms, Sanjeev Ahuja, John L McManus, Debasish F Roychowdhury

Abstract

Introduction: Granulocyte-macrophage colony-stimulating factor (GM-CSF), a protein produced in the lung, is essential for pulmonary host defense and alveolar integrity. Prior studies suggest potential benefits in several pulmonary conditions, including acute respiratory distress syndrome and viral infections. This trial evaluated the effect of the addition of inhaled sargramostim (yeast-derived, glycosylated recombinant human GM-CSF) to standard of care (SOC) on oxygenation and clinical outcomes in patients with COVID-19-associated acute hypoxemia.

Materials and methods: A randomized, controlled, open-label trial of hospitalized adults with COVID-19-associated hypoxemia (oxygen saturation <93% on ≥2 L/min oxygen supplementation and/or PaO2/FiO2 <350) randomized 2:1 to inhaled sargramostim (125 mcg twice daily for 5 days) plus SOC versus SOC alone. Institutional SOC before and during the study was not limited. Primary outcomes were change in the alveolar-arterial oxygen gradient (P(A-a)O2) by day 6 and the percentage of patients intubated within 14 days. Safety evaluations included treatment-emergent adverse events. Efficacy analyses were based on the modified intent-to-treat population, the subset of the intent-to-treat population that received ≥1 dose of any study treatment (sargramostim and/or SOC). An analysis of covariance approach was used to analyze changes in oxygenation measures. The intubation rate was analyzed using the chi-squared test. All analyses are considered descriptive. The study was institutional review board approved.

Results: In total, 122 patients were treated (sargramostim, n = 78; SOC, n = 44). The sargramostim arm experienced greater improvement in P(A-a)O2 by day 6 compared to SOC alone (least squares [LS] mean change from baseline [SE]: -102.3 [19.4] versus -30.5 [26.9] mmHg; LS mean difference: -71.7 [SE 33.2, 95% CI -137.7 to -5.8]; P = .033; n = 96). By day 14, 11.5% (9/78) of sargramostim and 15.9% (7/44) of SOC arms required intubation (P = .49). The 28-day mortality was 11.5% (9/78) and 13.6% (6/44) in the sargramostim and SOC arms, respectively (hazard ratio 0.85; P = .76). Treatment-emergent adverse events occurred in 67.9% (53/78) and 70.5% (31/44) on the sargramostim and SOC arms, respectively.

Conclusions: The addition of inhaled sargramostim to SOC improved P(A-a)O2, a measure of oxygenation, by day 6 in hospitalized patients with COVID-19-associated acute hypoxemia and was well tolerated. Inhaled sargramostim is delivered directly to the lung, minimizing systemic effects, and is simple to administer making it a feasible treatment option in patients in settings where other therapy routes may be difficult. Although proportionally lower rates of intubation and mortality were observed in sargramostim-treated patients, this study was insufficiently powered to demonstrate significant changes in these outcomes. However, the significant improvement in gas exchange with sargramostim shows this inhalational treatment enhances pulmonary efficiency in this severe respiratory illness. These data provide strong support for further evaluation of sargramostim in high-risk patients with COVID-19.

Conflict of interest statement

The institutions for R.P., R.C., E. Scott Halstead, J.N., D.J.P., T.B., B.P., G.M.-P., and Estelle S. Harris received research funding from Partner Therapeutics, Inc. F.G., J.L.M., and D.F.R. are employees of and have stock options in Partner Therapeutics, Inc. L.S. is a consultant to (d.b.a. Taylor Creek Consulting, Inc.) and has stock options for Partner Therapeutics, Inc. During this work, S.A. has been an employee of and consultant to Partner Therapeutics, Inc. and has stock options. Outside the current work, R.P. has received research grants from the US DVA and US National Heart, Lung, and Blood Institute and consulting fees from Partner Therapeutics, Inc.; R.C. serves as Vice President of the National Board for Respiratory Care who has supported travel to Board meets; E. Scott Halstead’s institution has received grants from the Eunice Kennedy Shriver National Institute of Child Health and Human Development; G.M.-P. was supported by Great Plain Health for meeting attendance/travel; and S.A. has received consulting fees from Partner Therapeutics, Inc.

© The Association of Military Surgeons of the United States 2022.

Figures

FIGURE 1.
FIGURE 1.
Eligibility, randomization, and follow-up of patients in the iLeukPulm study. Abbreviations: ABG = arterial blood gas; mITT = modified intention to treat; SOC = standard of care.

References

    1. Cornonavirus Resource Center : Telltale signs of a ‘Tripledemic’. Available at ; accessed October 20, 2022.
    1. Tzotzos SJ, Fischer B, Fischer H. et al.: Incidence of ARDS and outcomes in hospitalized patients with COVID-19: a global literature survey. Crit Care 2020; 24(1): 516.doi: 10.1186/s13054-020-03240-7.
    1. Armstrong RA, Kane AD, Kursumovic E. et al.: Mortality in patients admitted to intensive care with COVID-19: an updated systematic review and meta-analysis of observational studies. Anaesthesia 2021; 76(4): 537–48.doi: 10.1111/anae.15425.
    1. The Writing Committee for the COMEBAC Study Group : Four-month clinical status of a cohort of patients after hospitalization for COVID-19. JAMA 2021; 325(15): 1525–34.doi: 10.1001/jama.2021.3331.
    1. RECOVERY Collaborative Group : Dexamethasone in hospitalized patients with COVID-19. N Engl J Med 2021; 384(8): 693–704.doi: 10.1056/NEJMoa2021436.
    1. Weinreich DM, Sivapalasingam S, Norton T. et al.: REGN-COV2, a neutralizing antibody cocktail, in outpatients with COVID-19. N Engl J Med 2021; 384(3): 238–51.doi: 10.1056/NEJMoa2035002.
    1. Rando HM, Wellhausen N, Ghosh S. et al.: Identification and development of therapeutics for COVID-19. mSystems 2021; 6(6): e0023321.doi: 10.1128/mSystems.00233-21.
    1. Triggle CR, Bansal D, Ding H. et al.: A comprehensive review of viral characteristics, transmission, pathophysiology, immune response, and management of SARS-CoV-2 and COVID-19 as a basis for controlling the pandemic. Front Immunol 2021; 12: 631139.doi: 10.3389/fimmu.2021.631139.
    1. Siemieniuk RA, Bartoszko JJ, Ge L. et al.: Drug treatments for covid-19: living systematic review and network meta-analysis. BMJ 2020; 370: m2980.doi: 10.1136/bmj.m2980.
    1. Drożdżal S, Rosik J, Lechowicz K. et al.: An update on drugs with therapeutic potential for SARS-CoV-2 (COVID-19) treatment. Drug Resist Updat 2021; 59: 100794.doi: 10.1016/j.drup.2021.100794.
    1. US Food and Drug Administration : Emergency use authorization. Available at ; accessed October 20, 2022.
    1. Choi A, Koch M, Wu K. et al.: Safety and immunogenicity of SARS-CoV-2 variant mRNA vaccine boosters in healthy adults: an interim analysis. Nat Med 2021; 27(11): 2025–31.doi: 10.1038/s41591-021-01527-y.
    1. Ali I, Ali S, Iqbal S: COVID-19 vaccination: concerns about its accessibility, affordability, and acceptability. Front Med (Lausanne) 2021; 8: 647294.doi: 10.3389/fmed.2021.647294.
    1. Schneider C, Nobs SP, Kurrer M. et al.: Induction of the nuclear receptor PPAR-γ by the cytokine GM-CSF is critical for the differentiation of fetal monocytes into alveolar macrophages. Nat Immunol 2014; 15(11): 1026–37.doi: 10.1038/ni.3005.
    1. Trapnell BC, Whitsett JA, Nakata K: Pulmonary alveolar proteinosis. N Engl J Med 2003; 349(26) 2527–39.doi: 10.1056/NEJMra023226.
    1. Sever-Chroneos Z, Murthy A, Davis J. et al.: GM-CSF modulates pulmonary resistance to influenza A infection. Antiviral Res 2011; 92(2): 319–28doi: 10.1016/j.antiviral.2011.08.022.
    1. Wessendarp M, Watanabe-Chailland M, Liu S. et al.: Role of GM-CSF in regulating metabolism and mitochondrial functions critical to macrophage proliferation. Mitochondrion 2022; 62: 85–101.doi: 10.1016/j.mito.2021.10.009.
    1. Unkel B, Hoegner K, Clausen BE. et al.: Alveolar epithelial cells orchestrate DC function in murine viral pneumonia. J Clin Invest 2012; 122(10): 3652–64.doi: 10.1172/JCI62139.
    1. Lee E, Miller C, Ataya A. et al.: Opportunistic infection associated with elevated GM-CSF autoantibodies: a case series and review of the literature. Open Forum Infect Dis 2022; 9(5): ofac146.doi: 10.1093/ofid/ofac146.
    1. Wang EY, Mao T, Klein J. et al.: Diverse functional autoantibodies in patients with COVID-19. Nature 2021; 595(7866): 283–8.doi: 10.1038/s41586-021-03631-y.
    1. Halstead ES, Umstead TM, Davies ML. et al.: GM-CSF overexpression after influenza a virus infection prevents mortality and moderates M1-like airway monocyte/macrophage polarization. Respir Res 2018; 19(1): 3.doi: 10.1186/s12931-017-0708-5.
    1. Rosler B, Herold S: Lung epithelial GM-CSF improves host defense function and epithelial repair in influenza virus pneumonia—a new therapeutic strategy? Mol Cell Pediatr 2016; 3(1): 29.doi: 10.1186/s40348-016-0055-5.
    1. Herold S, Hoegner K, Vadasz I. et al.: Inhaled granulocyte/macrophage colony-stimulating factor as treatment of pneumonia-associated acute respiratory distress syndrome. Am J Respir Crit Care Med 2014; 189(5): 609–11.doi: 10.1164/rccm.201311-2041LE.
    1. Tazawa R, Ueda T, Abe M. et al.: Inhaled GM-CSF for pulmonary alveolar proteinosis. N Engl J Med 2019; 381(10): 923–32.doi: 10.1056/NEJMoa1816216.
    1. Tazawa R, Trapnell BC, Inoue Y. et al.: Inhaled granulocyte/macrophage-colony stimulating factor as therapy for pulmonary alveolar proteinosis. Am J Respir Crit Care Med 2010; 181(12): 1345–54.doi: 10.1164/rccm.200906-0978OC.
    1. Campo I, Mariani F, Paracchini E. et al.: Whole lung lavage followed by inhaled sargramostim as therapy of autoimmune pulmonary alveolar proteinosis. Am J Respir Crit Care Med 2016; 193: A6438.
    1. Paine R 3rd, Standiford TJ, Dechert RE. et al.: A randomized trial of recombinant human granulocyte-macrophage colony stimulating factor for patients with acute lung injury. Crit Care Med 2012; 40(1): 90–7.doi: 10.1097/CCM.0b013e31822d7bf0.
    1. Mathias B, Szpila BE, Moore FA. et al.: A review of GM-CSF therapy in sepsis. Medicine (Baltimore) 2015; 94(50): e2044.doi: 10.1097/MD.0000000000002044.
    1. Meisel C, Schefold JC, Pschowski R. et al.: Granulocyte-macrophage colony-stimulating factor to reverse sepsis-associated immunosuppression: a double-blind, randomized, placebo-controlled multicenter trial. Am J Respir Crit Care Med 2009; 180(7): 640–8.doi: 10.1164/rccm.200903-0363OC.
    1. Hall MW, Knatz NL, Vetterly C. et al.: Immunoparalysis and nosocomial infection in children with multiple organ dysfunction syndrome. Intensive Care Med 2011; 37(3): 525–32.doi: 10.1007/s00134-010-2088-x.
    1. LEUKINE® (sargramostim) for injection [Prescribing Information]. Lexington, MA, Partner Therapeutics, Inc., May2022.
    1. Mackey K, Ayers CK, Kondo KK. et al.: Racial and ethnic disparities in COVID-19-related infections, hospitalizations, and deaths: a systematic review. Ann Intern Med 2021; 174(3): 362–73.doi: 10.7326/M20-6306.
    1. Guidance for Industry and Food and Drug Administration Staff : Collection of race and ethnicity data in clinical trials. Available at ; accessed October 20, 2022.
    1. de Roos MP, Kilsdonk ID, Hekking PW. et al.: Chest computed tomography and alveolar-arterial oxygen gradient as rapid tools to diagnose and triage mildly symptomatic COVID-19 pneumonia patients. ERJ Open Res 2021; 7(1): 00737–2020.doi: 10.1183/23120541.00737-2020.
    1. Harris DE, Massie M: Role of alveolar-arterial gradient in partial pressure of oxygen and PaO2/fraction of inspired oxygen ratio measurements in assessment of pulmonary dysfunction. AANA J 2019; 87(3): 214–21.
    1. Arentz M, Yim E, Klaff L. et al.: Characteristics and outcomes of 21 critically ill patients with COVID-19 in Washington state. JAMA 2020; 323(16): 1612–4.doi: 10.1001/jama.2020.4326.
    1. 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): 1054–62.doi: 10.1016/S0140-6736(20)30566-3.
    1. Kim L, Garg S, O’Halloran A. et al.: Risk factors for intensive care unit admission and in-hospital mortality among hospitalized adults identified through the US Coronavirus Disease 2019 (COVID-19)-Associated Hospitalization Surveillance Network (COVID-NET). Clin Infect Dis 2021; 72(9): e206–e14.doi: 10.1093/cid/ciaa1012.
    1. Hall MW, Joshi I, Leal L. et al.: Immune immunomodulation in coronavirus disease 2019 (COVID-19): strategic considerations for personalized therapeutic intervention. Clin Infect Dis 2022; 74(1): 144–8.doi: 10.1093/cid/ciaa904.
    1. Tisoncik JR, Korth MJ, Simmons CP. et al.: Into the eye of the cytokine storm. Microbiol Mol Biol Rev 2012; 76(1): 16–32.doi: 10.1128/MMBR.05015-11.
    1. Huang FF, Barnes PF, Feng Y. et al.: GM-CSF in the lung protects against lethal influenza infection. Am J Respir Crit Care Med 2011; 184(2): 259–68.doi: 10.1164/rccm.201012-2036OC.
    1. Criner GJ, Lang FM, Gottlieb RL. et al.: Anti-granulocyte-macrophage colony-stimulating factor monoclonal antibody gimsilumab for COVID-19 pneumonia: a randomized, double-blind, placebo-controlled trial. Am J Respir Crit Care Med 2022; 205(11): 1290–9.doi: 10.1164/rccm.202108-1859OC.

Source: PubMed

3
購読する