A nebulised antitumour necrosis factor receptor-1 domain antibody in patients at risk of postoperative lung injury: A randomised, placebo-controlled pilot study

James Ryan, Andrew I Bayliffe, Daniel F McAuley, Joyce Yeung, David R Thickett, Phillip A Howells, Ciara O'Donnell, Arlette M Vassallo, Tracey J Wright, Elizabeth McKie, Kelly Hardes, Charlotte Summers, Martin O Shields, William Powley, Robert Wilson, Aili L Lazaar, Andrew Fowler, Gavin D Perkins, James Ryan, Andrew I Bayliffe, Daniel F McAuley, Joyce Yeung, David R Thickett, Phillip A Howells, Ciara O'Donnell, Arlette M Vassallo, Tracey J Wright, Elizabeth McKie, Kelly Hardes, Charlotte Summers, Martin O Shields, William Powley, Robert Wilson, Aili L Lazaar, Andrew Fowler, Gavin D Perkins

Abstract

Background: Tumour necrosis factor receptor 1 (TNFR1) signalling mediates the cell death and inflammatory effects of TNF-α.

Objective: The current clinical trial investigated the effects of a nebulised TNFR1 antagonist (GSK2862277) on signs of lung injury in patients undergoing oesophagectomy.

Design: Randomised double-blind (sponsor unblind), placebo-controlled, parallel group study.

Setting: Eight secondary care centres, the United Kingdom between April 2015 and June 2017.

Patients: Thirty-three patients undergoing elective transthoracic oesophagectomy.

Interventions: Patients randomly received a single nebulised dose (26 mg) of GSK2862277 (n = 17) or placebo (n = 16), given 1 to 5 h before surgery; 14 and 16, respectively competed the study.

Main outcome measurements: Physiological and biochemical markers of lung injury, pharmacokinetic and safety endpoints were measured. The primary endpoint was the change from baseline in pulmonary vascular permeability index (PVPI) at completion of surgery, measured using single-indicator transpulmonary thermodilution. Adjusted point estimates and 95% credible intervals (analogous to conventional confidence intervals) were constructed for each treatment using Bayesian statistical models.

Results: The mean change (with 95% credible intervals) from baseline in PVPI on completion of surgery was 0.00 (-0.23, 0.39) in the placebo and 0.00 (-0.24, 0.37) in the GSK2862277 treatment groups. There were no significant treatment-related differences in PaO2/FiO2 or Sequential Organ Failure Assessment score. Levels of free soluble TNFR1, Macrophage Inflammatory Protein-1 alpha and total protein were significantly reduced in the bronchoalveolar lavage fluid of patients treated with GSK2862277 (posterior probability of decrease with GSK2862277 vs. placebo:≥0.977; equivalent to P < 0.05). The frequency of adverse events and serious adverse events were distributed evenly across the two treatment arms.

Conclusion: Pre-operative treatment with a single 26 mg inhaled dose of GSK2862277 did not result in significantly lower postoperative alveolar capillary leak or extra vascular lung water. Unexpectedly small increases in transpulmonary thermodilution-measured PVPI and extra vascular lung water index at completion of surgery suggest less postoperative lung injury than historically reported, which may have also compromised a clear assessment of efficacy in this trial. GSK2862277 was well tolerated, resulted in expected lung exposure and reduced biomarkers of lung permeability and inflammation.

Trial registration: clinicaltrials.gov: NCT02221037.

Figures

Fig. 1
Fig. 1
Study schematic. Dosing with nebulised GSK2862277 26 mg or placebo. BAL, bronchoalveolar lavage; OLV, one-lung ventilation; OP, operation. 1On completion of surgery, transpulmonary thermodilution measurement first then bronchoalveolar lavage were performed before tracheal extubation. 2Thermodilution performed if patient remained in ICU with patent indwelling PiCCO catheter.
Fig. 2
Fig. 2
Consort flow diagram. ∗1 subject subsequently rescreened and randomised. ∗∗1 Subject subsequently randomised. PVPI, pulmonary vascular permeability index.
Fig. 3
Fig. 3
Individual changes from baseline in pulmonary vascular permeability index immediately postsurgery. PVPI, pulmonary vascular permeability index.
Fig. 4
Fig. 4
Effect of GSK2862277 on inflammatory biomarkers (pg ml−1): (a) free soluble tumour necrosis factor receptor 1; (b) macrophage inflammatory protein-1 alpha; (c) macrophage inflammatory protein-1 beta; (d) IL-1β; (e) IL-8; (f) IL-10; (g) IL-6; (h) TNF-α. IL, interleukin; MIP1-α, macrophage inflammatory protein-1 alpha; MIP-1β, macrophage inflammatory protein-1 beta; sTNFR1, soluble tumour necrosis factor receptor 1. Boxplots display the median (solid horizontal line), the interquartile range (the box), geometric mean (solid symbol inside the box) and highest and lowest values (whiskers). Open symbols lying outside the whiskers denote outliers. Horizontal long dashed lines are the lower limit of quantification and horizontal short dash lines are the upper limit of quantification.
Fig. 5
Fig. 5
Effect of GSK2862277 on: (a) total protein; (b) total protein ratio. Boxplots display the median (solid horizontal line), the interquartile range (the box), geometric mean (solid symbol inside box) and highest and lowest values (whiskers). Open symbols lying outside the whiskers denote outliers. Horizontal long dashed lines are the lower limit of quantification and horizontal short dash lines are the upper limit of quantification.
Fig. 6
Fig. 6
Median plasma concentrations of GSK2862277 and individual patient bronchoalveolar lavage fluid-derived lung epithelial lining fluid concentrations.

References

    1. Ranieri VM, Rubenfeld GD, Thompson BT, et al. ARDS Definition Task Force. Acute respiratory distress syndrome: the Berlin definition. JAMA 2012; 307:2526–2533.
    1. Abbott TEF, Fowler AJ, Pelosi P, et al. StEP-COMPAC Group. A systematic review and consensus definitions for standardised end-points in perioperative medicine: pulmonary complications. Br J Anaesth 2018; 120:1066–1079.
    1. Perkins GD, Gates S, Park D, et al. BALTI-Prevention Collaborators. The beta agonist lung injury trial prevention. A randomized controlled trial. Am J Respir Crit Care Med 2014; 189:674–683.
    1. Proudfoot AG, McAuley DF, Griffiths MJD, et al. Human models of acute lung injury. Dis Model Mech 2011; 4:145–153.
    1. Shyamsundar M, McAuley DF, Shields MO, et al. Effect of simvastatin on physiological and biological outcomes in patients undergoing esophagectomy: a randomized placebo-controlled trial. Ann Surg 2014; 259:26–31.
    1. Qiu P, Cui X, Sun J, et al. Antitumor necrosis factor therapy is associated with improved survival in clinical sepsis trials: a meta-analysis. Crit Care Med 2013; 41:2419–2429.
    1. Tartaglia LA, Rothe M, Hu YF, et al. Tumor necrosis factor's cytotoxic activity is signaled by the p55 TNF receptor. Cell 1993; 7:213–216.
    1. Patel BV, Wilson MR, O’Dea KP, et al. TNF-induced death signaling triggers alveolar epithelial dysfunction in acute lung injury. J Immunol 2013; 190:4274–4282.
    1. Proudfoot A, Bayliffe A, O’Kane CM, et al. Novel antitumour necrosis factor receptor-1 (TNFR1) domain antibody prevents pulmonary inflammation in experimental acute lung injury. Thorax 2018; 73:723–730.
    1. Marchetti L, Klein M, Schlett K, et al. Tumor necrosis factor (TNF)-mediated neuroprotection against glutamate-induced excitotoxicity is enhanced by N-methyl-d-aspartate receptor activation. Essential role of a TNF receptor 2-mediated phosphatidylinositol 3-kinase-dependent NF-kappa B pathway. J Biol Chem 2004; 279:32869–32881.
    1. Bluml S, Binder N, Niederreiter B, et al. Analysis of TNFR2-mediated functions on osteoclast precursor cells. Ann Rheum Dis 2010; 69:A35–A36.
    1. Al-Lamki RS, Wang J, Vandenabeele P, et al. TNFR1- and TNFR2-mediated signalling pathways in human kidney are cell type-specific and differentially contribute to renal injury. FASEB J 2005; 19:1637–1645.
    1. Luo D, Luo Y, He Y, et al. Differential functions of tumor necrosis factor receptor 1 and 2 signaling in ischemia-mediated arteriogenesis and angiogenesis. Am J Pathol 2006; 169:1886–1898.
    1. Wilson MR, Goddard ME, O’Dea KP, et al. Differential roles of p55 and p75 tumor necrosis factor receptors on stretch-induced pulmonary edema in mice. Am J Physiol Lung Cell Mol Physiol 2007; 293:L60–L68.
    1. Ebach DR, Riehl TE, Stenson WF. Opposing effects of tumor necrosis factor receptor 1 and 2 in sepsis due to cecal ligation and puncture. Shock 2005; 23:311–318.
    1. Longhi L, Perego C, Ortolano F, et al. Tumor necrosis factor in traumatic brain injury: effects of genetic deletion of p55 or p75 receptor. J Cereb Blood Flow Metab 2013; 33:1182–1189.
    1. Cordy JC, Morley PJ, Wright TJ, et al. Specificity of human antivariable heavy (VH) chain autoantibodies and impact on the design and clinical testing of a VH domain antibody antagonist of tumour necrosis factor-α receptor 1. Clin Exp Immunol 2015; 182:139–148.
    1. Jozwiak M, Teboul JL, Monnet X. Extravascular lung water in critical care: recent advances and clinical applications. Ann Intens Care 2015; 5:38.
    1. Vincent JL, Moreno R, Takala J, et al. The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. Intensive Care Med 1996; 22:707–710.
    1. MedDRA The Medical Dictionary for Regulatory Activities. MedDRA SMQs. URL: [Accessed 30 Novemebr 2018].
    1. Howells P, Thickett D, Knox C, et al. The impact of the acute respiratory distress syndrome on outcome after oesophagectomy. Br J Anaesth 2016; 117:375–381.
    1. Parekh D, Dancer RCA, Scott A, et al. Vitamin D to prevent lung injury following esophagectomy – a randomized, placebo-controlled trial. Crit Care Med 2018; 46:e1128–e1135.
    1. Howells PA, Aldridge KA, Parekh D, et al. ARDS following oesophagectomy: a comparison of two trials. BMJ Open Respir Res 2017; 4:e000207.
    1. Serpa NA, Campos PP, Hemmes SN, et al. PROVE Network Investigators. Kinetics of plasma biomarkers of inflammation and lung injury in surgical patients with or without postoperative pulmonary complications. Eur J Anaesthesiol 2017; 34:229–238.
    1. Agrawal A, Zhuo H, Brady S, et al. Pathogenetic and predictive value of biomarkers in patients with ALI and lower severity of illness: results from two clinical trials. Am J Physiol Lung Cell Mol Physiol 2012; 303:L634–L639.
    1. Arozullah AM, Khuri SF, Henderson WG, et al. Participants in the National Veterans Affairs Surgical Quality Improvement Program. Development and validation of a multifactorial risk index for predicting postoperative pneumonia after major noncardiac surgery. Ann Intern Med 2001; 135:847–857.
    1. Torrance HD, Pearse RM, O’Dwyer MJ. Does major surgery induce immune suppression and increase the risk of postoperative infection? Curr Opin Anaesthesiol 2016; 29:376–383.
    1. Furst DE. The risk of infections with biologic therapies for rheumatoid arthritis. Semin Arthritis Rheum 2010; 39:327–346.

Source: PubMed

3
Se inscrever