Prospective evaluation of the efficacy, safety, and optimal biomarker enrichment strategy for nangibotide, a TREM-1 inhibitor, in patients with septic shock (ASTONISH): a double-blind, randomised, controlled, phase 2b trial

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

  • Bruno François
  • Simon Lambden
  • Tom Fivez
  • Sebastien Gibot
  • Marc Derive
  • Jean-Marie Grouin
  • Margarita Salcedo-Magguilli
  • Jérémie Lemarié
  • Nicolas De Schryver
  • Ville Jalkanen
  • Tarik Hicheur
  • Jean-Jacques Garaud
  • Valérie Cuvier
  • Ricard Ferrer
  • Bestle, Morten Heiberg
  • Ville Pettilä
  • Jean-Paul Mira
  • Camille Bouisse
  • Emmanuelle Mercier
  • Joris Vermassen
  • Vincent Huberlant
  • Isabelle Vinatier
  • Nadia Anguel
  • Mitchell Levy
  • Pierre François Laterre
  • ASTONISH investigators

Background: Activation of the triggering receptor expressed on myeloid cells-1 (TREM-1) pathway is associated with septic shock outcomes. Data suggest that modulation of this pathway in patients with activated TREM-1 might improve survival. Soluble TREM-1 (sTREM-1), a potential mechanism-based biomarker, might facilitate enrichment of patient selection in clinical trials of nangibotide, a TREM-1 modulator. In this phase 2b trial, we aimed to confirm the hypothesis that TREM1 inhibition might improve outcomes in patients with septic shock. Methods: This double-blind, randomised, placebo-controlled, phase 2b trial assessed the efficacy and safety of two different doses of nangibotide compared with placebo, and aimed to identify the optimum treatment population, in patients across 42 hospitals with medical, surgical, or mixed intensive care units (ICUs) in seven countries. Non-COVID-19 patients (18–85 years) meeting the standard definition of septic shock, with documented or suspected infection (lung, abdominal, or urinary [in patients ≥65 years]), were eligible within 24 h of vasopressor initiation for the treatment of septic shock. Patients were randomly assigned in a 1:1:1 ratio to intravenous nangibotide 0·3 mg/kg per h (low-dose group), nangibotide 1·0 mg/kg per h (high-dose group), or matched placebo, using a computer-generated block randomisation scheme (block size 3). Patients and investigators were masked to treatment allocation. Patients were grouped according to sTREM-1 concentrations at baseline (established from sepsis observational studies and from phase 2a change to data) into high sTREM-1 (≥ 400 pg/mL). The primary outcome was the mean difference in total Sequential Organ Failure Assessment (SOFA) score from baseline to day 5 in the low-dose and high-dose groups compared with placebo, measured in the predefined high sTREM-1 (≥ 400 pg/mL) population and in the overall modified intention-to-treat population. Secondary endpoints included all-cause 28-day mortality, safety, pharmacokinetics, and evaluation of the relationship between TREM-1 activation and treatment response. This study is registered with EudraCT, 2018-004827-36, and Clinicaltrials.gov, NCT04055909. Findings: Between Nov 14, 2019, and April 11, 2022, of 402 patients screened, 355 were included in the main analysis (116 in the placebo group, 118 in the low-dose group, and 121 in the high-dose group). In the preliminary high sTREM-1 population (total 253 [71%] of 355; placebo 75 [65%] of 116; low-dose 90 [76%] of 118; high-dose 88 [73%] of 121), the mean difference in SOFA score from baseline to day 5 was 0·21 (95% CI −1·45 to 1·87, p=0·80) in the low-dose group and 1·39 (−0·28 to 3·06, p=0·104) in the high-dose group versus placebo. In the overall population, the difference in SOFA score from baseline to day 5 between the placebo group and low-dose group was 0·20 (−1·09 to 1·50; p=0·76),and between the placebo group and the high-dose group was 1·06 (−0·23 to 2·35, p=0·108). In the predefined high sTREM-1 cutoff population, 23 (31%) patients in the placebo group, 35 (39%) in the low-dose group, and 25 (28%) in the high-dose group had died by day 28. In the overall population, 29 (25%) patients in the placebo, 38 (32%) in the low-dose, and 30 (25%) in the high-dose group had died by day 28. The number of treatment-emergent adverse events (111 [96%] patients in the placebo group, 113 [96%] in the low-dose group, and 115 [95%] in the high-dose group) and serious treatment-emergent adverse events (28 [24%], 26 [22%], and 31 [26%]) was similar between all three groups. High-dose nangibotide led to a clinically relevant improvement in SOFA score (of two points or more) from baseline to day 5 over placebo in those with higher cutoff concentrations (≥532 pg/mL) of sTREM-1 at baseline. Low dose nangibotide displayed a similar pattern with lower magnitude of effect across all cutoff values. Interpretation: This trial did not achieve the primary outcome of improvement in SOFA score at the predefined sTREM-1 value. Future studies are needed to confirm the benefit of nangibotide at higher concentrations of TREM-1 activation. Funding: Inotrem.

OriginalsprogEngelsk
TidsskriftThe Lancet Respiratory Medicine
Vol/bind11
Udgave nummer10
Sider (fra-til)894-904
Antal sider11
ISSN2213-2600
DOI
StatusUdgivet - 2023

Bibliografisk note

Funding Information:
The authors acknowledge the support of the patients, their families, and study teams in delivering the ASTONISH trial. In addition, the data monitoring committee and data adjudication committee members are thanked for their rigorous review of the data throughout the trial process. The authors thank Sarah Legrand Demai for her technical support and medical writing assistance.

Publisher Copyright:
© 2023 Elsevier Ltd

ID: 386493350