Repair of acute respiratory distress syndrome by stromal cell administration (REALIST) trial: A phase 1 trial

Ellen Gorman, Manu Shankar-Hari, Phil Hopkins, William S Tunnicliffe, Gavin D Perkins, Jonathan Silversides, Peter McGuigan, Anna Krasnodembskaya, Colette Jackson, Roisin Boyle, Jamie McFerran, Cliona McDowell, Christina Campbell, Margaret McFarland, Jon Smythe, Jacqui Thompson, Barry Williams, Gerard Curley, John G Laffey, Mike Clarke, Daniel F McAuley, Cecilia M O'Kane, Ellen Gorman, Manu Shankar-Hari, Phil Hopkins, William S Tunnicliffe, Gavin D Perkins, Jonathan Silversides, Peter McGuigan, Anna Krasnodembskaya, Colette Jackson, Roisin Boyle, Jamie McFerran, Cliona McDowell, Christina Campbell, Margaret McFarland, Jon Smythe, Jacqui Thompson, Barry Williams, Gerard Curley, John G Laffey, Mike Clarke, Daniel F McAuley, Cecilia M O'Kane

Abstract

Background: Mesenchymal stromal cells (MSCs) may be of benefit in acute respiratory distress syndrome (ARDS) due to immunomodulatory, reparative, and antimicrobial actions. ORBCEL-C is a population of CD362 enriched umbilical cord-derived MSCs. The REALIST phase 1 trial investigated the safety and feasibility of ORBCEL-C in patients with moderate to severe ARDS.

Methods: REALIST phase 1 was an open label, dose escalation trial in which cohorts of mechanically ventilated patients with moderate to severe ARDS received increasing doses (100, 200 or 400 × 106 cells) of a single intravenous infusion of ORBCEL-C in a 3 + 3 design. The primary safety outcome was the incidence of serious adverse events. Dose limiting toxicity was defined as a serious adverse reaction within seven days. Trial registration clinicaltrials.gov NCT03042143.

Findings: Nine patients were recruited between the 7th January 2019 and 14th January 2020. Study drug administration was well tolerated and no dose limiting toxicity was reported in any of the three cohorts. Eight adverse events were reported for four patients. Pyrexia within 24 h of study drug administration was reported in two patients as pre-specified adverse events. A further two adverse events (non-sustained ventricular tachycardia and deranged liver enzymes), were reported as adverse reactions. Four serious adverse events were reported (colonic perforation, gastric perforation, bradycardia and myocarditis) but none were deemed related to administration of ORBCEL-C. At day 28 no patients had died in cohort one (100 × 106), three patients had died in cohort two (200 × 106) and one patient had died in cohort three (400 × 106). Overall day 28 mortality was 44% (n = 4/9).

Interpretation: A single intravenous infusion of ORBCEL-C was well tolerated in patients with moderate to severe ARDS. No dose limiting toxicity was reported up to 400 × 106 cells.

Conflict of interest statement

EG receives funding by the Wellcome Trust Health Innovation Challenge Fund [reference 106939/Z/15/Z] for the described work. COK, DMcA, JL, JS & AK are investigators on the grant funding this work from Wellcome Trust Health Innovation Challenge Fund [reference 106939/Z/15/Z]. DMcA, COK & MC are investigators on a grant received from the Northern Ireland Health and Social Care Research and Development Division to fund an additional COVID-19 cohort during phase 2 of this study. JL reports consulting fees from Baxter Healthcare and GlaxoSmithKline, provision of medicolegal reports to the Irish Clinical Indemnity Scheme, participation in DSMB for other clinical trials and is named on a patent for InspireShield, a device to reduce COVID-19 spread. AK reports grants received from MRC UK (MR/R025096/1 and MR/S009426/1) and a HORIZON 2020 MSC individual fellowship. MSH is supported by a NIHR Clinician Scientist Fellowship (CS-2016–16–011). GDP is supported by the 10.13039/100005622Health Research (NIHR) Applied Research Collaboration (ARC) West Midlands and is a director of research for the Intensive Care Society. GC reports grants from Health Research Board (Ireland) Emerging Clinician Scientist Award and the United States Department of defense Discovery Award. DMcA reports grants from NIHR, Innovate UK, MRC and Novavax as an investigator in ARDS and COVID-19 studies. DMcA reports consultancy fees unrelated to this work from Bayer, GlaxoSmithKline, Boehringer Ingelheim, Novartis and Eli Lilly. DMcA reports payments from GlaxoSmithKline as an educational seminar speaker. DMcA is a member of the DSMB for Vir Biotechnology, Inc and Faron Pharmaceuticals. DMcA has a patent for a novel treatment for inflammatory disease. DMcA is a director of research for the Intensive Care Society and Director of the EME programme for MRC/NIHR. DMcA reports a spouse who has received consultancy fees from INSMED and from the California Institute for Regenerative unrelated to this clinical trial. COK has received consultancy fees from INSMED, unrelated to this work, and fees for participation in grant panels for the Californian Institute of Regenerative Medicine, unrelated to this clinical trial. COK reports a spouse who has received grants from NIHR, Innovate UK, MRC and Novavax. COK reports a spouse who has received consultancy fees unrelated to this work from Bayer, GlaxoSmithKline, Boehringer Ingelheim, Novartis and Eli Lilly. COK reports a spouse who has received payments from GlaxoSmithKline as an educational seminar speaker. COK reports a spouse who is a member of the DSMB for Vir Biotechnology, Inc and Faron Pharmaceuticals. COK reports a spouse who has a patent for a novel treatment for inflammatory disease. COK reports a spouse who is a director of research for the Intensive Care Society and Director of the EME programme for MRC/NIHR. All other authors report no declarations of interest. The views expressed in this publication are those of the authors and not necessarily those of the National Health Service, the National Institute for Health Research (NIHR), or the Department of Health and Social Care.

© 2021 The Author(s).

Figures

Fig. 1
Fig. 1
Consort diagram.
Fig. 2
Fig. 2
Pulmonary (Oxygenation Index, P/F ratio, Respiratory Compliance, Driving pressure) and non-pulmonary organ function (SOFA score) outcomes Mean (SD) are provided for each dose cohort (dose 1: 100 × 10 6; dose 2: 200 × 10 6; dose 3: 400 × 10 6) at day 0 (baseline), day 4, day 7 and day 14. (A) Oxygenation index (B) P/F Ratio (C) Respiratory Compliance (D) Driving Pressure (E) SOF A Score.
Fig. 3
Fig. 3
Plasma biological markers of systemic inflammatory response (IL-6, IL-8 and IL-18), markers of epithelial injury (Surfactant protein-D [SP-D]) and endothelial activation/injury (ICAM-1 and Angiopoietin 2 [Ang-2]). Mean (SD) are provided for each dose cohort (dose 1: 100 × 10 6; dose 2: 200 × 106; dose 3: 400 × 106) at day 0 (baseline), day 4, day 7 and day 14. A) IL-6 B) IL-8 C) IL-18 D) Ang-2 E) ICAM F) SP-D.

References

    1. Ranieri V.M., Rubenfeld G.D., Thompson B.T. Acute respiratory distress syndrome: the Berlin definition. JAMA. 2012;307(23):2526–2533.
    1. Herridge M.S., Tansey C.M., Matte A. Functional disability 5 years after acute respiratory distress syndrome. N Engl J Med. 2011;364(14):1293–1304.
    1. Bienvenu O.J., Friedman L.A., Colantuoni E. Psychiatric symptoms after acute respiratory distress syndrome: a 5-year longitudinal study. Intensive Care Med. 2018;44(1):38–47.
    1. Bellani G., Laffey J.G., Pham T. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA. 2016;315(8):788–800.
    1. Saha R., Assouline B., Mason G., Douiri A., Summers C., Shankar-Hari M. Impact of differences in acute respiratory distress syndrome randomised controlled trial inclusion and exclusion criteria: systematic review and meta-analysis. Br J Anaesth. 2021;127(1):85–101.
    1. MacSweeney R.M., McAuley D.F. Acute respiratory distress syndrome. Lancet. 2016;388(10058):2416–2430.
    1. Griffiths M.J.D., McAuley D.F., Perkins G.D. Guidelines on the management of acute respiratory distress syndrome. BMJ Open Resp Res. 2019;6(1)
    1. Shaw T.D., McAuley D.F., O'Kane C.M. Emerging drugs for treating the acute respiratory distress syndrome. Expert Opin Emerg Drugs. 2019;24(1):29–41.
    1. Horby P., Lim W.S., Emberson J.R. Dexamethasone in hospitalized patients with COVID-19. N Engl J Med. 2021;384(8):693–704.
    1. 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.
    1. Interleukin-6 receptor antagonists in critically Ill Patients with COVID-19. N Engl J Med. 2021;384(16):1491–1502.
    1. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. 2021;397(10285):1637–1645.
    1. Devaney J., Horie S., Masterson C. Human mesenchymal stromal cells decrease the severity of acute lung injury induced by E. coli in the rat. Thorax. 2015;70(7):625–635.
    1. Masterson C., Devaney J., Horie S. Syndecan-2-positive, bone marrow-derived human mesenchymal stromal cells attenuate bacterial-induced acute lung injury and enhance resolution of ventilator-induced lung injury in rats. Anesthesiology. 2018;129(3):502–516.
    1. Curley G.F., Jerkic M., Dixon S. Cryopreserved, Xeno-free human umbilical cord mesenchymal stromal cells reduce lung injury severity and bacterial burden in rodent escherichia coli-induced acute respiratory distress syndrome. Crit Care Med. 2017;45(2):e202–ee12.
    1. Horie S., Masterson C., Brady J. Umbilical cord-derived CD362(+) mesenchymal stromal cells for E. coli pneumonia: impact of dose regimen, passage, cryopreservation, and antibiotic therapy. Stem Cell Res Ther. 2020;11(1):116.
    1. Krasnodembskaya A., Song Y., Fang X. Antibacterial effect of human mesenchymal stem cells is mediated in part from secretion of the antimicrobial peptide LL-37. Stem Cells. 2010;28(12):2229–2238.
    1. Zhu H., Xiong Y., Xia Y. Therapeutic effects of human umbilical cord-derived mesenchymal stem cells in acute lung injury mice. Sci Rep. 2017;7:39889.
    1. Sung D.K., Chang Y.S., Sung S.I., Yoo H.S., Ahn S.Y., Park W.S. Antibacterial effect of mesenchymal stem cells against Escherichia coli is mediated by secretion of beta- defensin- 2 via toll- like receptor 4 signalling. Cell Microbiol. 2016;18(3):424–436.
    1. Gupta N., Krasnodembskaya A., Kapetanaki M. Mesenchymal stem cells enhance survival and bacterial clearance in murine Escherichia coli pneumonia. Thorax. 2012;67(6):533–539.
    1. Lee J.W., Fang X., Gupta N., Serikov V., Matthay M.A. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc Natl Acad Sci U S A. 2009;106(38):16357–16362.
    1. Fang X., Neyrinck A.P., Matthay M.A., Lee J.W. Allogeneic human mesenchymal stem cells restore epithelial protein permeability in cultured human alveolar type II cells by secretion of angiopoietin-1. J Biol Chem. 2010;285(34):26211–26222.
    1. Nemeth K., Leelahavanichkul A., Yuen P.S. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15(1):42–49.
    1. Jackson M.V., Morrison T.J., Doherty D.F. Mitochondrial transfer via tunneling nanotubes is an important mechanism by which mesenchymal stem cells enhance macrophage phagocytosis in the in vitro and in vivo models of ARDS. Stem Cells. 2016;34(8):2210–2223.
    1. Islam M.N., Das S.R., Emin M.T. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012;18(5):759–765.
    1. Morrison T.J., Jackson M.V., Cunningham E.K. Mesenchymal stromal cells modulate macrophages in clinically relevant lung injury models by extracellular vesicle mitochondrial transfer. Am J Respir Crit Care Med. 2017;196(10):1275–1286.
    1. Zhu Y.G., Feng X.M., Abbott J. Human mesenchymal stem cell microvesicles for treatment of Escherichia coli endotoxin-induced acute lung injury in mice. Stem Cells. 2014;32(1):116–125.
    1. Song Y., Dou H., Li X. Exosomal miR-146a contributes to the enhanced therapeutic efficacy of interleukin-1β-primed mesenchymal stem cells against sepsis. Stem Cells. 2017;35(5):1208–1221.
    1. Silva J.D., Su Y., Calfee C.S. MSC extracellular vesicles rescue mitochondrial dysfunction and improve barrier integrity in clinically relevant models of ARDS. Eur Respir J. 2020
    1. Fergie N., Todd N., McClements L., McAuley D., O'Kane C., Krasnodembskaya A. Hypercapnic acidosis induces mitochondrial dysfunction and impairs the ability of mesenchymal stem cells to promote distal lung epithelial repair. FASEB Journal. 2019;33(4):5585–5598.
    1. McIntyre L.A., Moher D., Fergusson D.A. Efficacy of mesenchymal stromal cell therapy for acute lung injury in preclinical animal models: a systematic review. PLoS ONE. 2016;11(1)
    1. Asmussen S., Ito H., Traber D.L. Human mesenchymal stem cells reduce the severity of acute lung injury in a sheep model of bacterial pneumonia. Thorax. 2014;69(9):819–825.
    1. Lee J.W., Fang X., Gupta N., Serikov V., Matthay M.A. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc Natl Acad Sci U S A. 2009;106(38):16357–16362.
    1. Lee J.W., Krasnodembskaya A., McKenna D.H., Song Y., Abbott J., Matthay M.A. Therapeutic effects of human mesenchymal stem cells in ex vivo human lungs injured with live bacteria. Am J Respir Crit Care Med. 2013;187(7):751–760.
    1. McAuley D.F., Curley G.F., Hamid U.I. Clinical grade allogeneic human mesenchymal stem cells restore alveolar fluid clearance in human lungs rejected for transplantation. Am J Physiol Lung Cell Mol Physiol. 2014;306(9):L809–L815.
    1. Zheng G., Huang L., Tong H. Treatment of acute respiratory distress syndrome with allogeneic adipose-derived mesenchymal stem cells: a randomized, placebo-controlled pilot study. Respir Res. 2014;15:39.
    1. Wilson J.G., Liu K.D., Zhuo H. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Resp Med. 2015;3(1):24–32.
    1. Matthay M.A., Calfee C.S., Zhuo H. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised phase 2a safety trial. Lancet Resp Med. 2019;7(2):154–162.
    1. Bellingan G, Jacono F, Bannard-Smith J. 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). B14 LATE BREAKING CLINICAL TRIALS: American Thoracic Society. Am J Respir Crit Care Med. 2019;199:A7353.
    1. Yip H.K., Fang W.F., Li Y.C. Human umbilical cord-derived mesenchymal stem cells for acute respiratory distress syndrome. Crit Care Med. 2020;48(5):e391–e399.
    1. Lv H., Chen W., Xiang A.P., Zhang Q., Yang Y., Yi H. 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(10):1944–1947.
    1. Chen J., Hu C., Chen L. Clinical study of mesenchymal stem cell treatment for acute respiratory distress syndrome induced by epidemic influenza a (H7N9) infection: a hint for COVID-19 treatment. Engineering (Beijing, China) 2020;6(10):1153–1161.
    1. Devarapu S.K., Junhui X., Darisipudi M., Rocanin Arjo A., Anders H.J. FP456CD362+ mesenchymal stem cell treatment of kidney disease in type 2 diabetic LEPR DB/DB mice. Nephrol Dial Transplant. 2015;30(suppl_3):iii223–iiiii4.
    1. Cook N., Hansen A.R., Siu L.L., Abdul Razak A.R. Early phase clinical trials to identify optimal dosing and safety. Mol Oncol. 2015;9(5):997–1007.
    1. Kollerup Madsen B., Hilscher M., Zetner D., Rosenberg J. Adverse reactions of dimethyl sulfoxide in humans: a systematic review. F1000 Res. 2018;7:1746.
    1. Seeley E., McAuley D.F., Eisner M., Miletin M., Matthay M.A., Kallet R.H. Predictors of mortality in acute lung injury during the era of lung protective ventilation. Thorax. 2008;63(11):994–998.
    1. Burt C., Cryer C., Fuggle S., Little A.M., Dyer P., HLA-A B. DR allele group frequencies in 7007 kidney transplant list patients in 27 UK centres. Int J Immunogenet. 2013;40(3):209–215.
    1. Thompson M., Mei S.H.J., Wolfe D. Cell therapy with intravascular administration of mesenchymal stromal cells continues to appear safe: an updated systematic review and meta-analysis. EClin Med. 2020;19
    1. Lv H., Chen W., Xiang A.P., Zhang Q., Yang Y., Yi H. 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(10):1944–1947.
    1. Jerkic M., Gagnon S., Rabani R. Human umbilical cord mesenchymal stromal cells attenuate systemic sepsis in part by enhancing peritoneal macrophage bacterial killing via heme oxygenase-1 induction in rats. Anesthesiology. 2020;132(1):140–154.
    1. Perlee D., van Vught L.A., Scicluna B.P. Intravenous infusion of human adipose mesenchymal stem cells modifies the host response to lipopolysaccharide in humans: a randomized, single-blind, parallel group, placebo controlled trial. Stem Cells. 2018;36(11):1778–1788.
    1. Ware L.B., Koyama T., Billheimer D.D. Prognostic and pathogenetic value of combining clinical and biochemical indices in patients with acute lung injury. Chest. 2010;137(2):288–296.
    1. Dolinay T., Kim Y.S., Howrylak J. Inflammasome-regulated cytokines are critical mediators of acute lung injury. Am J Respir Crit Care Med. 2012;185(11):1225–1234.
    1. Eisner M.D., Parsons P., Matthay M.A., Ware L., Greene K. Plasma surfactant protein levels and clinical outcomes in patients with acute lung injury. Thorax. 2003;58(11):983.
    1. Wada T., Jesmin S., Gando S. The role of angiogenic factors and their soluble receptors in acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) associated with critical illness. J. Inflamm. 2013;10(1):6.
    1. Calfee C.S., Eisner M.D., Parsons P.E. Soluble intercellular adhesion molecule-1 and clinical outcomes in patients with acute lung injury. Intensiv Care Med. 2009;35(2):248–257.
    1. Wick K.D., Leligdowicz A., Zhuo H., Ware L.B., Matthay M.A. Mesenchymal stromal cells reduce evidence of lung injury in patients with ARDS. JCI Insight. 2021;6(12)
    1. Le Blanc K., Tammik C., Rosendahl K., Zetterberg E., Ringden O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 2003;31(10):890–896.
    1. Skyler J.S., Fonseca V.A., Segal K.R., Rosenstock J. Allogeneic mesenchymal precursor cells in type 2 diabetes: a randomized, placebo-controlled, dose-escalation safety and tolerability pilot study. Diabetes Care. 2015;38(9):1742–1749.
    1. Hare J.M., Fishman J.E., Gerstenblith G. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA. 2012;308(22):2369–2379.
    1. Packham D.K., Fraser I.R., Kerr P.G., Segal K.R. Allogeneic mesenchymal precursor cells (MPC) in diabetic nephropathy: a randomized, placebo-controlled, dose escalation study. EBio Med. 2016;12:263–269.
    1. Barnhoorn M.C., Wasser M., Roelofs H. Long-term evaluation of allogeneic bone marrow-derived mesenchymal stromal cell therapy for crohn's disease perianal fistulas. J Crohns Colitis. 2020;14(1):64–70.
    1. García-Sancho J., Sánchez A., Vega A., Noriega D.C., Nocito M. Influence of HLA matching on the efficacy of allogeneic mesenchymal stromal cell therapies for osteoarthritis and degenerative disc disease. Transplant Direct. 2017;3(9):e205.
    1. Rowan K.M., Kerr J.H., Major E., McPherson K., Short A., Vessey M.P. Intensive care society's APACHE II study in Britain and Ireland–II: outcome comparisons of intensive care units after adjustment for case mix by the American APACHE II method. BMJ. 1993;307(6910):977–981.
    1. Simonson O.E., Ståhle E., Hansen T. Five-year follow-up after mesenchymal stromal cell–based treatment of severe acute respiratory distress syndrome. Am J Respir Crit Care Med. 2020;202(7):1051–1055.
    1. Gorman E., Shankar-Hari M., Hopkins P. Repair of acute respiratory distress syndrome by stromal cell administration in COVID-19 (REALIST-COVID-19): a structured summary of a study protocol for a randomised. Trials. 2020;21(1):462. controlled trial.

Source: PubMed

3
Subskrybuj