Safety and efficacy of multipotent adult progenitor cells in acute respiratory distress syndrome (MUST-ARDS): a multicentre, randomised, double-blind, placebo-controlled phase 1/2 trial

G Bellingan, F Jacono, J Bannard-Smith, D Brealey, N Meyer, D Thickett, D Young, A Bentley, B J McVerry, R G Wunderink, K C Doerschug, C Summers, M Rojas, A Ting, E D Jenkins, G Bellingan, F Jacono, J Bannard-Smith, D Brealey, N Meyer, D Thickett, D Young, A Bentley, B J McVerry, R G Wunderink, K C Doerschug, C Summers, M Rojas, A Ting, E D Jenkins

Abstract

Purpose: Bone marrow-derived, allogeneic, multipotent adult progenitor cells demonstrated safety and efficacy in preclinical models of acute respiratory distress syndrome (ARDS).

Methods: This phase 1/2 trial evaluated the safety and tolerability of intravenous multipotent adult progenitor cells in patients with moderate-to-severe ARDS in 12 UK and USA centres. Cohorts 1 and 2 were open-label, evaluating acute safety in three subjects receiving 300 or 900 million cells, respectively. Cohort 3 was a randomised, double-blind, placebo-controlled parallel trial infusing 900 million cells (n = 20) or placebo (n = 10) within 96 h of ARDS diagnosis. Primary outcomes were safety and tolerability. Secondary endpoints included clinical outcomes, quality of life (QoL) and plasma biomarkers.

Results: No allergic or serious adverse reactions were associated with cell therapy in any cohort. At baseline, the cohort 3 cell group had less severe hypoxia. For cohort 3, 28-day mortality was 25% for cell vs. 45% for placebo recipients. Median 28-day free from intensive care unit (ICU) and ventilator-free days in the cell vs. placebo group were 12.5 (IQR 0,18.5) vs. 4.5 (IQR 0,16.8) and 18.5 (IQR 0,22) vs. 6.5 (IQR 0,18.3), respectively. A prospectively defined severe ARDS subpopulation (PaO2/FiO2 < 150 mmHg (20 kPa); n = 16) showed similar trends in mortality, ICU-free days and ventilator-free days favouring cell therapy. Cell recipients showed greater recovery of QoL through Day 365.

Conclusions: Multipotent adult progenitor cells were safe and well tolerated in ARDS. The clinical outcomes warrant larger trials to evaluate the therapeutic efficacy and optimal patient population.

Trial registration: ClinicalTrials.gov NCT02611609.

Keywords: Acute respiratory distress syndrome (ARDS); Multipotent adult progenitor cells (MAPC); Stem cells.

Conflict of interest statement

Two of the authors, EJ and AT, were employees of Athersys and one, GB, received a travel grant from Athersys to two meetings. No other Conflict of interest are declared

© 2021. Crown.

Figures

Fig. 1
Fig. 1
Trial Design for Cohort 3
Fig. 2
Fig. 2
Ratio of Day 7 to baseline values of biomarker plasma concentrations. The ratio of the biomarker value at Day 7 compared to the baseline values are presented as medians with upper and lower quartiles. Biomarkers include: angiopoietin 1 (ANG1), angiopoietin 2 (ANG2), C-X-C Motif chemokine ligand 10 (CXCL10), Interferon (IFN) gamma, interleukin 1 beta (IL-1b), interleukin 1 receptor 2 (IL-1R2), interleukin 1 receptor antagonist (IL-1RA), interleukin 6 (IL-6), interleukin 8 (IL-8), interleukin 10 (IL-10), interleukin 12 (IL-12), keratinocyte growth factor (KGF), matrix metalloproteinase 2 (MMP-2), programmed cell death protein (PD-1), receptor for advanced glycation end-products (RAGE), regulated on activation, normal T cell expressed and secreted (RANTES), surfactant protein D (SP-D), tumour necrosis factor alpha (TNF alpha), soluble TNF receptor 1 (sTNFR1) and thrombospondin 1 (TSP-1). Green = patients receiving multipotent adult progenitor cells (n = 19); Blue = patients receiving placebo (n = 7)

References

    1. Matthay MA, Zemans RL, Zimmerman GA, et al. Acute respiratory distress syndrome. Nat Rev Dis Primers. 2019;5(1):18. doi: 10.1038/s41572-019-0069-0.
    1. Thompson BT, Chambers RC, Liu KD. Acute respiratory distress syndrome. N Engl J Med. 2017;377(19):1904–1905. doi: 10.1056/NEJMra1608077.
    1. Group RC. Horby P, Lim WS, et al. Dexamethasone in hospitalized patients with Covid-19—preliminary report. N Engl J Med. 2020 doi: 10.1056/NEJMoa2021436.
    1. Angus DC, Derde L, Al-Beidh F, et al. Effect of hydrocortisone on mortality and organ support in patients with severe COVID-19: the REMAP-CAP COVID-19 Corticosteroid Domain Randomized Clinical Trial. JAMA. 2020;324(13):1317–1329. doi: 10.1001/jama.2020.17022.
    1. The REMAP-CAP Investigators Interleukin-6 receptor antagonists in critically ill patients with Covid-19. N Engl J Med. 2021;384:1491–1502. doi: 10.1056/NEJMoa2100433.
    1. RECOVERY Collaborative Group Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. 2021;397:1637–1645. doi: 10.1016/S0140-6736(21)00676-0.
    1. Rosas IO, Diaz G, Gottlieb RL, et al. Tocilizumab and remdesivir in hospitalized patients with severe COVID-19 pneumonia: a randomized clinical trial. Intensive Care Med. 2021 doi: 10.1007/s00134-021-06507-x13.
    1. Menk M, Estenssoro E, Sahetya SK, et al. Current and evolving standards of care for patients with ARDS. Intensive Care Med. 2020;46:2157–2167. doi: 10.1007/s00134-020-06299-6.
    1. Gupta N, Su X, Popov B, Lee JW, Serikov V, Matthay MA. Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J Immunol. 2007;179(3):1855–1863. doi: 10.4049/jimmunol.179.3.1855.
    1. Matthay MA, Goolaerts A, Howard JP, Lee JW. Mesenchymal stem cells for acute lung injury: preclinical evidence. Crit Care Med. 2010;38(10 Suppl):S569–S573. doi: 10.1097/CCM.0b013e3181f1ff1d.
    1. Rojas M, Xu J, Woods CR, et al. Bone marrow-derived mesenchymal stem cells in repair of the injured lung. Am J Respir Cell Mol Biol. 2005;33(2):145–152. doi: 10.1165/rcmb.2004-0330OC.
    1. Moodley Y, Sturm M, Shaw K, et al. Human mesenchymal stem cells attenuate early damage in a ventilated pig model of acute lung injury. Stem cell Res. 2016;17(1):25–31. doi: 10.1016/j.scr.2016.05.005.
    1. Rojas M, Parker RE, Thorn N, et al. Infusion of freshly isolated autologous bone marrow derived mononuclear cells prevents endotoxin-induced lung injury in an ex-vivo perfused swine model. Stem Cell Res Ther. 2013;4(2):26. doi: 10.1186/scrt174.
    1. Horie S, Curley GF, Laffey JG. What’s new in cell therapies in ARDS? Intensive Care Med. 2016;42:779–782. doi: 10.1007/s00134-015-4140-3.
    1. Zheng G, Huang L, Tong H, et al. Treatment of acute respiratory distress syndrome with allogeneic adipose-derived mesenchymal stem cells: a randomized, placebo-controlled pilot study. Respir Res. 2014;15:39. doi: 10.1186/1465-9921-15-39.
    1. Wilson JG, Liu KD, Zhuo H, et al. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Respir Med. 2015;3(1):24–32. doi: 10.1016/S2213-2600(14)70291-7.
    1. Matthay MA, Calfee CS, Zhuo H, et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised phase 2a safety trial. Lancet Respir Med. 2019;7(2):154–162. doi: 10.1016/S2213-2600(18)30418-1.
    1. Simonson OE, Mougiakakos D, Heldring N, et al. In vivo effects of mesenchymal stromal cells in two patients with severe acute respiratory distress syndrome. Stem Cells Transl Med. 2015;4(10):1199–1213. doi: 10.5966/sctm.2015-0021.
    1. Lv H, Chen W, Xiang AP, et al. Mesenchymal stromal cells as a salvage treatment for confirmed acute respiratory distress syndrome: preliminary data from a single-arm study. Intensive Care Med. 2020;46:1944–1947. doi: 10.1007/s00134-020-06122-2.
    1. Hess DC, Wechsler LR, Clark WM, et al. Safety and efficacy of multipotent adult progenitor cells in acute ischaemic stroke (MASTERS): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol. 2017;16(5):360–368. doi: 10.1016/S1474-4422(17)30046-7.
    1. Maziarz RT, Devos T, Bachier CR, et al. Single and multiple dose MultiStem (multipotent adult progenitor cell) therapy prophylaxis of acute graft-versus-host disease in myeloablative allogeneic hematopoietic cell transplantation: a phase 1 trial. Biol Blood Marrow Transplant. 2015;21(4):720–728. doi: 10.1016/j.bbmt.2014.12.025.
    1. Penn MS, Ellis S, Gandhi S, et al. Adventitial delivery of an allogeneic bone marrow-derived adherent stem cell in acute myocardial infarction: phase I clinical study. Circ Res. 2012;110(2):304–311. doi: 10.1161/CIRCRESAHA.111.253427.
    1. Rojas M, Cardenes N, Kocyildirim E, et al. Human adult bone marrow-derived stem cells decrease severity of lipopolysaccharide-induced acute respiratory distress syndrome in sheep. Stem Cell Res Ther. 2014;5(2):42. doi: 10.1186/scrt430.
    1. Cardenes N, Aranda-Valderrama P, Carney JP, et al. Cell therapy for ARDS: efficacy of endobronchial versus intravenous administration and biodistribution of MAPCs in a large animal model. BMJ Open Respir Res. 2019;6(1):e000308. doi: 10.1136/bmjresp-2018-000308.
    1. Bellingan G, Jacono F, Bannard-Smith J, et al (2019) Primary analysis of a phase 1/2 study to assess MultiStem? Cell therapy, a regenerative advanced therapy medicinal product (ATMP), in acute respiratory distress syndrome (MUST-ARDS). In: B14. LATE BREAKING CLINICAL TRIALS. American Thoracic Society, pp A7353–A7353. 10.1164/ajrccm-conference.2019.199.1_MeetingAbstracts.A7353
    1. Ranieri VM, Rubenfeld GD, Thompson BT, et al. Acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012;307(23):2526–2533. doi: 10.1001/jama.2012.5669.
    1. Maiolo G, Collino F, Vasques F, et al. Reclassifying acute respiratory distress syndrome. Am J Respir Crit Care Med. 2018;197(12):1586–1595. doi: 10.1164/rccm.201709-1804OC.
    1. Fan E, Brodie D, Slutsky AS. Acute respiratory distress syndrome: advances in diagnosis and treatment. JAMA. 2018;319(7):698–710. doi: 10.1001/jama.2017.21907.
    1. McAuley DF, Cross LM, Hamid U, et al. Keratinocyte growth factor for the treatment of the acute respiratory distress syndrome (KARE): a randomised, double-blind, placebo-controlled phase 2 trial. Lancet Respir Med. 2017;5(6):484–491. doi: 10.1016/S2213-2600(17)30171-6.
    1. McAuley DF, Laffey JG, O'Kane CM, et al. Simvastatin in the acute respiratory distress syndrome. N Engl J Med. 2014;371(18):1695–1703. doi: 10.1056/NEJMoa1403285.
    1. Laffey JG, Matthay MA. Fifty years of research in ARDS. Cell-based therapy for acute respiratory distress syndrome. Biology and potential therapeutic value. Am J Respir Crit Care Med. 2017;196(3):266–273. doi: 10.1164/rccm.201701-0107CP.
    1. Auletta JJ, Cooke KR, Solchaga LA, Deans RJ, van’t Hof W. Regenerative stromal cell therapy in allogeneic hematopoietic stem cell transplantation: current impact and future directions. Biol Blood Marrow Transplant. 2010;16(7):891–906. doi: 10.1016/j.bbmt.2009.12.005.
    1. Wick KD, Leligdowicz A, Zhuo H, et al. Mesenchymal stromal cells reduce evidence of lung injury in patients with ARDS. JCI Insight. 2021;6(12):e148983. doi: 10.1172/jci.insight.148983.

Source: PubMed

3
구독하다