Therapeutic blockade of granulocyte macrophage colony-stimulating factor in COVID-19-associated hyperinflammation: challenges and opportunities

Puja Mehta, Joanna C Porter, Jessica J Manson, John D Isaacs, Peter J M Openshaw, Iain B McInnes, Charlotte Summers, Rachel C Chambers, Puja Mehta, Joanna C Porter, Jessica J Manson, John D Isaacs, Peter J M Openshaw, Iain B McInnes, Charlotte Summers, Rachel C Chambers

Abstract

The COVID-19 pandemic is a global public health crisis, with considerable mortality and morbidity exerting pressure on health-care resources, including critical care. An excessive host inflammatory response in a subgroup of patients with severe COVID-19 might contribute to the development of acute respiratory distress syndrome (ARDS) and multiorgan failure. Timely therapeutic intervention with immunomodulation in patients with hyperinflammation could prevent disease progression to ARDS and obviate the need for invasive ventilation. Granulocyte macrophage colony-stimulating factor (GM-CSF) is an immunoregulatory cytokine with a pivotal role in initiation and perpetuation of inflammatory diseases. GM-CSF could link T-cell-driven acute pulmonary inflammation with an autocrine, self-amplifying cytokine loop leading to monocyte and macrophage activation. This axis has been targeted in cytokine storm syndromes and chronic inflammatory disorders. Here, we consider the scientific rationale for therapeutic targeting of GM-CSF in COVID-19-associated hyperinflammation. Since GM-CSF also has a key role in homoeostasis and host defence, we discuss potential risks associated with inhibition of GM-CSF in the context of viral infection and the challenges of doing clinical trials in this setting, highlighting in particular the need for a patient risk-stratification algorithm.

Trial registration: ClinicalTrials.gov NCT04326920.

Copyright © 2020 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Role of GM-CSF in homoeostasis, viral response, and inflammation GM-CSF has an important homoeostatic role in the maturation and function of alveolar macrophages, which clear and catabolise surfactant, and in host defence. In response to viral insults (eg, with SARS-CoV-2), alveolar type II epithelial cells secrete GM-CSF, improving the innate immune response of myeloid cells, particularly alveolar macrophages. In severe inflammatory states, GM-CSF production is upregulated by alveolar type II epithelial cells and monocyte-derived M1-like macrophages, thereby stimulating IL-6 production from CD14+ and CD16+ inflammatory monocytes, increasing Th1 and Th17 T cells and driving the recruitment and priming of neutrophils. The resulting autocrine, positive feedback loop of GM-CSF production further perpetuates the inflammatory milieu. GM-CSF=granulocyte macrophage colony-stimulating factor. IL=interleukin. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2. Th=T helper.
Figure 2
Figure 2
A window of opportunity in hyperinflammation for optimum treatment intervention Hyperinflammation can be initiated by an inciting trigger (eg, SARS-CoV-2 infection) and can progress from an early indolent state to a fulminant and fatal hypercytokinaemia. Withholding potentially lifesaving immunomodulatory treatment until a patient is intubated could result in a missed window of opportunity for optimum therapeutic intervention. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2.

References

    1. WHO Coronavirus disease 2019 (COVID-19): situation report—142. June 10, 2020.
    1. Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020;46:846–848.
    1. Siddiqi HK, Mehra MR. COVID-19 illness in native and immunosuppressed states: a clinical-therapeutic staging proposal. J Heart Lung Transplant. 2020;39:405–407.
    1. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet. 2020;395:1033–1034.
    1. Bloomberg FDA approves emergency IND use of Humanigen's lenzilumab for compassionate use in COVID-19 patients. April 2, 2020.
    1. Kiniksa Announces Early Evidence of Treatment Response with Mavrilimumab in 6 Patients with Severe COVID-19 Pneumonia and Hyperinflammation. April 1, 2020.
    1. Bloomberg FDA approves initiation of Humanigen's phase III study of lenzilumab in COVID-19 patients. April 15, 2020.
    1. McKee S. Izana initiates study assessing namilumab for COVID-19. April 6, 2020.
    1. Izana Bioscience's drug namilumab announced in major UK COVID-19 drugs trial. June 11, 2020.
    1. Roivant Sciences Roivant doses first patient in pivotal BREATHE clinical trial evaluating gimsilumab in COVID-19 patients for the prevention and treatment of acute respiratory distress syndrome. April 15, 2020.
    1. GlobeNewsWire I-Mab announces IND clearance from FDA for TJM2 to treat cytokine release syndrome (CRS) associated with severe coronavirus disease 19 (COVID-19) April 3, 2020.
    1. Zhou F, Yu T, Du R. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062.
    1. Calfee CS, Delucchi K, Parsons PE, Thompson BT, Ware LB, Matthay MA. Subphenotypes in acute respiratory distress syndrome: latent class analysis of data from two randomised controlled trials. Lancet Respir Med. 2014;2:611–620.
    1. Huang C, Wang Y, Li X. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506.
    1. Gattinoni L, Chiumello D, Rossi S. COVID-19 pneumonia: ARDS or not? Crit Care. 2020;24:154.
    1. Matthay MA, Aldrich JM, Gotts JE. Treatment for severe acute respiratory distress syndrome from COVID-19. Lancet Respir Med. 2020;8:433–434.
    1. Becher B, Tugues S, Greter M. GM-CSF: from growth factor to central mediator of tissue inflammation. Immunity. 2016;45:963–973.
    1. Hamilton JA. GM-CSF-dependent inflammatory pathways. Front Immunol. 2019;10
    1. Hamilton JA, Anderson GP. GM-CSF biology. Growth Factors. 2004;22:225–231.
    1. Rösler 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;3:29.
    1. Cakarova L, Marsh LM, Wilhelm J. Macrophage tumor necrosis factor-alpha induces epithelial expression of granulocyte-macrophage colony-stimulating factor: impact on alveolar epithelial repair. Am J Respir Crit Care Med. 2009;180:521–532.
    1. Steinwede K, Tempelhof O, Bolte K. Local delivery of GM-CSF protects mice from lethal pneumococcal pneumonia. J Immunol. 2011;187:5346–5356.
    1. Ballinger MN, Paine R, 3rd, Serezani CH. Role of granulocyte macrophage colony-stimulating factor during gram-negative lung infection with Pseudomonas aeruginosa. Am J Respir Cell Mol Biol. 2006;34:766–774.
    1. Unkel B, Hoegner K, Clausen BE. Alveolar epithelial cells orchestrate DC function in murine viral pneumonia. J Clin Invest. 2012;122:3652–3664.
    1. Matute-Bello G, Liles WC, Radella F., 2nd Modulation of neutrophil apoptosis by granulocyte colony-stimulating factor and granulocyte/macrophage colony-stimulating factor during the course of acute respiratory distress syndrome. Crit Care Med. 2000;28:1–7.
    1. Carter SJ, Tattersall RS, Ramanan AV. Macrophage activation syndrome in adults: recent advances in pathophysiology, diagnosis and treatment. Rheumatology. 2019;58:5–17.
    1. Mehta HM, Malandra M, Corey SJ. G-CSF and GM-CSF in neutropenia. J Immunol. 2015;195:1341–1349.
    1. Glasser L, Legolvan M, Horwitz HM. Florid histiocytic hemophagocytosis following therapy with long acting G-CSF (pegfilgrastim) Am J Hematol. 2007;82:753–757.
    1. Cornish AL, Campbell IK, McKenzie BS, Chatfield S, Wicks IP. G-CSF and GM-CSF as therapeutic targets in rheumatoid arthritis. Nat Rev Rheumatol. 2009;5:554–559.
    1. Quesnel B, Catteau B, Aznar V, Bauters F, Fenaux P. Successful treatment of juvenile rheumatoid arthritis associated haemophagocytic syndrome by cyclosporin A with transient exacerbation by conventional-dose G-CSF. Br J Haematol. 1997;97:508–510.
    1. Wang S, Degar BA, Zieske A, Shafi NQ, Rose MG. Hemophagocytosis exacerbated by G-CSF/GM-CSF treatment in a patient with myelodysplasia. Am J Hematol. 2004;77:391–396.
    1. Paydas S, Yetişir A, Mirili C. G-CSF related hemophagocytosis in a case with lymphoma. Clin Res Trials. 2018;4:1–2. doi: 10.15761/CRT.1000203.
    1. Padhi S, Varghese RGB, Ramdas A, Phansalkar MD, Sarangi R. Hemophagocytic lymphohistiocytosis: critical reappraisal of a potentially under-recognized condition. Front Med. 2013;7:492–498.
    1. Albeituni S, Verbist KC, Tedrick PE. Mechanisms of action of ruxolitinib in murine models of hemophagocytic lymphohistiocytosis. Blood. 2019;134:147–159.
    1. Burn TN, Weaver L, Rood JE. Genetic deficiency of interferon-γ reveals interferon-γ-independent manifestations of murine hemophagocytic lymphohistiocytosis. Arthritis Rheumatol. 2020;72:335–347.
    1. Yoshihara S, Li Y, Xia J, Danzl N, Sykes M, Yang YG. Posttransplant hemophagocytic lymphohistiocytosis driven by myeloid cytokines and vicious cycles of T-cell and macrophage activation in humanized mice. Front Immunol. 2019;10:186.
    1. Zinter MS, Hermiston ML. Calming the storm in HLH. Blood. 2019;134:103–104.
    1. Stiel L, Meziani F, Helms J. Neutrophil activation during septic shock. Shock. 2018;49:371–384.
    1. Potey PM, Rossi AG, Lucas CD, Dorward DA. Neutrophils in the initiation and resolution of acute pulmonary inflammation: understanding biological function and therapeutic potential. J Pathol. 2019;247:672–685.
    1. Vassallo A, Wood AJ, Subburayalu J, Summers C, Chilvers ER. The counter-intuitive role of the neutrophil in the acute respiratory distress syndrome. Br Med Bull. 2019;131:43–55.
    1. Zuo Y, Yalavarthi S, Shi H. Neutrophil extracellular traps in COVID-19. JCI Insight. 2020 doi: 10.1172/jci.insight.138999. published online April 24.
    1. Barnes BJ, Adrover JM, Baxter-Stoltzfus A. Targeting potential drivers of COVID-19: neutrophil extracellular traps. J Exp Med. 2020;217
    1. Kamp VM, Leentjens J, Pillay J. Modulation of granulocyte kinetics by GM-CSF/IFN-γ in a human LPS rechallenge model. J Leukoc Biol. 2013;94:513–520.
    1. Cowburn AS, Summers C, Dunmore BJ. Granulocyte/macrophage colony-stimulating factor causes a paradoxical increase in the BH3-only pro-apoptotic protein Bim in human neutrophils. Am J Respir Cell Mol Biol. 2011;44:879–887.
    1. Summers C, Singh NR, White JF. Pulmonary retention of primed neutrophils: a novel protective host response, which is impaired in the acute respiratory distress syndrome. Thorax. 2014;69:623–629.
    1. Juss JK, House D, Amour A. Acute respiratory distress syndrome neutrophils have a distinct phenotype and are resistant to phosphoinositide 3-kinase inhibition. Am J Respir Crit Care Med. 2016;194:961–973.
    1. De Alessandris S, Ferguson GJ, Dodd AJ. Neutrophil GM-CSF receptor dynamics in acute lung injury. J Leukoc Biol. 2019;105:1183–1194.
    1. Zhou Y, Fu B, Zheng X. Pathogenic T cells and inflammatory monocytes incite inflammatory storm in severe COVID-19 patients. Natl Sci Rev. 2020 doi: 10.1093/nsr/nwaa041. published online March 13.
    1. Neelapu SS, Locke FL, Bartlett NL. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377:2531–2544.
    1. Li Y, Li H, Fan R. Coronavirus infections in the central nervous system and respiratory tract show distinct features in hospitalized children. Intervirology. 2016;59:163–169.
    1. Sterner RM, Sakemura R, Cox MJ. GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood. 2019;133:697–709.
    1. Sterner RM, Cox MJ, Sakemura R, Kenderian SS. Using CRISPR/Cas9 to knock out GM-CSF in CAR-T cells. J Vis Exp. 2019 doi: 10.3791/59629. published online July 22.
    1. Lotfi N, Thome R, Rezaei N. Roles of GM-CSF in the pathogenesis of autoimmune diseases: an update. Front Immunol. 2019;10
    1. Mehta P, Cron RQ, Hartwell J, Manson JJ, Tattersall RS. Silencing the cytokine storm: the use of intravenous anakinra in haemophagocytic lymphohistocytosis or macrophage activation syndrome. Lancet Rheumatol. 2020;2:e358–e367.
    1. Ahmed A, Merrill SA, Alsawah F. Ruxolitinib in adult patients with secondary haemophagocytic lymphohistiocytosis: an open-label, single-centre, pilot trial. Lancet Haematol. 2019;6:e630–e637.
    1. Wu D, Yang XO. TH17 responses in cytokine storm of COVID-19: an emerging target of JAK2 inhibitor fedratinib. J Microbiol Immunol Infect. 2020;53:368–370.
    1. Dolhnikoff M, Duarte-Neto AN, de Almeida Monteiro RA. Pathological evidence of pulmonary thrombotic phenomena in severe COVID-19. J Thromb Haemost. 2020 doi: 10.1111/jth.14844. published online April 15.
    1. Herold S, Hoegner K, Vadász I. Inhaled granulocyte/macrophage colony-stimulating factor as treatment of pneumonia-associated acute respiratory distress syndrome. Am J Respir Crit Care Med. 2014;189:609–611.
    1. Paine R, 3rd, Standiford TJ, Dechert RE. A randomized trial of recombinant human granulocyte-macrophage colony stimulating factor for patients with acute lung injury. Crit Care Med. 2012;40:90–97.
    1. Meisel C, Schefold JC, Pschowski R. 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;180:640–648.
    1. Bhattacharya P, Budnick I, Singh M. Dual role of GM-CSF as a pro-inflammatory and a regulatory cytokine: implications for immune therapy. J Interferon Cytokine Res. 2015;35:585–599.
    1. Berg J, Zscheppang K, Fatykhova D. Tyk2 as a target for immune regulation in human viral/bacterial pneumonia. Eur Respir J. 2017;50
    1. Pinder EM, Rostron AJ, Hellyer TP. Randomised controlled trial of GM-CSF in critically ill patients with impaired neutrophil phagocytosis. Thorax. 2018;73:918–925.
    1. Ingraham NE, Lotfi-Emran S, Thielen BK. Immunomodulation in COVID-19. Lancet Respir Med. 2020;8:544–546.
    1. Arnaldez FI, O'Day SJ, Drake CG. The Society for Immunotherapy of Cancer perspective on regulation of interleukin-6 signaling in COVID-19-related systemic inflammatory response. J Immunother Cancer. 2020;8
    1. Calfee CS, Delucchi KL, Sinha P. Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised controlled trial. Lancet Respir Med. 2018;6:691–698.
    1. Shakoory B, Carcillo JA, Chatham WW. Interleukin-1 receptor blockade is associated with reduced mortality in sepsis patients with features of macrophage activation syndrome: reanalysis of a prior phase III trial. Crit Care Med. 2016;44:275–281.

Source: PubMed

3
Sottoscrivi