Articles
The effect of intravenous interferon-beta-1a (FP-1201) on lung CD73 expression and on acute respiratory distress syndrome mortality: an open-label study

https://doi.org/10.1016/S2213-2600(13)70259-5Get rights and content

Summary

Background

Pulmonary vascular leakage occurs early in acute respiratory distress syndrome (ARDS). Mortality is high (35–45%), but no effective pharmacotherapy exists. Production of anti-inflammatory adenosine by ecto-5′-nucleotidase (CD73) helps maintain endothelial barrier function. We tested whether interferon-beta-1a (IFN-beta-1a), which increases CD73 synthesis, can reduce vascular leakage and mortality in patients with ARDS.

Methods

In ex-vivo studies, we first established that IFN-beta-1a induced CD73 up-regulation in cultured human lung tissue samples. We then tested the safety, tolerability, and efficacy of intravenous human recombinant IFN-beta-1a (FP-1201) in patients with ARDS in an open-label study (comprising dose-escalation and expansion phases). We recruited patients from eight intensive care units in the UK. Eligible patients were aged 18 years or older, had ARDS, and were being treated with assisted ventilation. We established an optimal tolerated dose (OTD) in the first, dose-escalation phase. Once established, we gave all subsequently enrolled patients the OTD of intravenous FP-1201 for 6 days. We assessed 28-day mortality (our primary endpoint) in all patients receiving the OTD versus 28-day mortality in a group of patients who did not receive treatment (this control group comprised patients in the study but who did not receive treatment because they were screened during the safety windows after dose escalation). This trial is registered with ClinicalTrials.gov, number NCT00789685, and the EU Clinical Trials Register EudraCT, number 2008-000140-13.

Findings

IFN-beta-1a increased the number of CD73-positive vessels in lung culture by four times on day 1 (p=0·04) and by 14·3 times by day 4 (p=0·004). For the clinical trial, between Feb 23, 2009, and April 7, 2011, we identified 150 patients, of whom 37 were enrolled into the trial and given treatment. The control group consisted of 59 patients who were recruited to take part in the study, but who did not receive treatment. Demographic characteristics and severity of illness did not differ between treatment and control groups. The optimal tolerated FP-1201 dose was 10 μg per day for 6 days. By day 28, 3 (8%) of 37 patients in the treatment cohort and 19 (32%) of 59 patients in the control cohort had died—thus, treatment with FP-1201 was associated with an 81% reduction in odds of 28-day mortality (odds ratio 0·19 [95% CI 0·03–0·72]; p=0·01).

Interpretation

FP-1201 up-regulates human lung CD73 expression, and is associated with a reduction in 28-day mortality in patients with ARDS. Our findings need to be substantiated in large, prospective randomised trials, but suggest that FP-1201 could be the first effective, mechanistically targeted, disease-specific pharmacotherapy for patients with ARDS.

Funding

Faron Pharmaceuticals, National Institute for Health Research University College Hospitals Biomedical Research Centre, the Finnish Academy, the Sigrid Juselius Foundation, and the Arvo and Inkeri Suominen Foundation.

Introduction

Acute respiratory distress syndrome (ARDS) is characterised by pulmonary inflammation, diffuse non-cardiogenic bilateral pulmonary oedema, and refractory hypoxaemia. The severity of ARDS is defined by the ratio between the partial pressure of arterial oxygen (PaO2), in kPa, divided by the inspired fraction of oxygen (FiO2; PaO2/FiO2 ratio): mild (39·9–26·7 kPA), moderate (26·6–13·4 kPa), and severe ARDS (≤13·3 kPa).1 Triggered by a variety of extra-pulmonary or pulmonary disease states, ARDS is often seen in patients in intensive care units, affecting nearly 200 000 critically ill patients a year in the USA alone.2 Associated fiscal, mortality, and morbidity burdens are substantial: health-care costs about US$5000 dollars per patient per day in intensive care,2, 3 and some 35–40% of patients with ARDS die.2 Survivors have impaired functional capacity and increased health-care costs extending up to 5 years or more after discharge.4 Treatment, however, remains supportive, and although paralysis with drugs such as cisatracurium might have a role in improving outcome,5 no mechanistically or disease-specific pharmacotherapies have been shown to improve outcome.6, 7

One key pathophysiological event in ARDS is an early increase in pulmonary capillary permeability, causing plasma exudation into the alveolar space and thus (potentially life-threatening) hypoxaemia. Ecto-5′-nucleotidase (cluster of differentiation 73, or CD73) is an enzyme expressed on vascular endothelium, epithelial cells, and a leucocyte subset. Through dephosphorylation of adenosine monophosphate), CD73 yields anti-inflammatory adenosine,8 which binds to its A2B receptor that is diversely expressed in tissues including the lung,9 mediating anti-inflammatory effects such as the prevention of vascular leakage and the inhibition of leucocyte recruitment into sites of inflammation.10, 11, 12 Such endogenous protective mechanisms are enhanced in the presence of hypoxia.13 Thus adenosine receptor knockout mice are especially vulnerable to acute lung injury and pulmonary vascular leakage,12, 14 and CD73 reduces pulmonary vascular leakage in acute lung injury in animal models.11

We have previously shown that interferon-beta (IFN-beta) decreases vascular permeability by upregulating CD73 expression and activity on cultured endothelial cells.15 Moreover, severity of experimental acute lung injury is greater in CD73-deficient mice than in wild-type mice, and can be alleviated with IFN-beta treatment in only wild-type mice, indicating that IFN-beta exerts its effect via CD73.15 Thus, findings from cell-culture and rodent studies suggest that up-regulation of CD73 by IFN-beta has the potential to ameliorate pulmonary vascular leakage and to improve outcome in ARDS. We assessed whether these findings were true in human beings—we sought to confirm that human IFN-beta could increase CD73 expression in human lung samples before doing a phase 1–2 clinical study.

Section snippets

Study design

A detailed description of this open-label, phase 1–2 study is given in the appendix. Briefly, for the ex-vivo study, we used lung tissue samples resected from patients from University Hospital, Turku (Finland) receiving surgery for lung cancer. With permission from the ethics authorities at University Hospital (Turku, Finland), samples were taken from a region of macroscopically and microscopically normal tissue distant to the site of the tumour itself, with patients giving verbal informed

Statistical analysis

We searched for between-cohort differences using Mann-Whitney U test for continuous variables (age, PaO2/FiO2, Acute Physiology And Chronic Health Evaluation II [APACHE-II] score, number of days alive) and χ2 or Fisher's exact test for categorical variables (sex, aetiology), with results presented as medians with IQR or percentages. We assessed changes over time using a random intercept model to account for within-participant measurement correlations. Biomarkers (MxA, neopterin, CD73,

Results

Practically all CD73 positivity was in blood vessels and lymphatic vessels (figure 1). Bronchial epithelial cells and infiltrated neutrophils were CD73 negative (figure 1). The response was both time-dependent and dose-dependent: 1000 IU/mL of IFN-beta-1a increased the number of CD73-positive vessels by four times on day 1 (p=0·04) and by 14·3 times on day 4 (0·004; figure 1).

For the clinical trial, we recruited patients between Feb 23, 2009, and April 7, 2011. 150 patients met all the

Discussion

Findings from our ex-vivo study suggest that IFN-beta-1a has a role in the upregulation of CD73. Findings from our in-vivo phase 1–2 trial suggest that intravenous administration of FP-1201 can decrease concentrations of inflammatory markers and improve respiratory function and survival. If substantiated in large prospective studies, these findings suggest that FP-1201 could be an effective pharmacotherapy for ARDS.

CD73 dephosphorylates adenosine monophosphate to yield adenosine,8 the

References (37)

  • JV Diaz et al.

    Therapeutic strategies for severe acute lung injury

    Crit Care Med

    (2010)
  • SP Colgan et al.

    Physiological roles for ecto-5'-nucleotidase (CD73)

    Purinergic Signal

    (2006)
  • RE Cagnina et al.

    Adenosine A2B receptors are highly expressed on murine type II alveolar epithelial cells

    Am J Physiol Lung Cell Mol Physiol

    (2009)
  • M Salmi et al.

    Cell-surface enzymes in control of leukocyte trafficking

    Nat Rev Immunol

    (2005)
  • J Reutershan et al.

    Adenosine and inflammation: CD39 and CD73 are critical mediators in LPS-induced PMN trafficking into the lungs

    FASEB J

    (2009)
  • T Eckle et al.

    A2B adenosine receptor signaling attenuates acute lung injury by enhancing alveolar fluid clearance in mice

    J Clin Invest

    (2008)
  • E Morschl et al.

    A3 adenosine receptor signaling influences pulmonary inflammation and fibrosis

    Am J Respir Cell Mol Biol

    (2008)
  • J Kiss et al.

    IFN-beta protects from vascular leakage via up-regulation of CD73

    Eur J Immunol

    (2007)
  • Cited by (124)

    View all citing articles on Scopus
    View full text