Skip to main content
Erschienen in: Critical Care 1/2023

Open Access 01.12.2023 | Research

FX06 to rescue SARS-CoV-2-induced acute respiratory distress syndrome: a randomized clinical trial

verfasst von: Emmanuelle Guérin, Lisa Belin, Guillaume Franchineau, Loïc Le Guennec, David Hajage, Mamadou Hassimiou Diallo, Thomas Frapard, Lucie Le Fèvre, Charles-Edouard Luyt, Alain Combes, Stéphane Germain, Jan Hayon, Pierre Asfar, Nicolas Bréchot

Erschienen in: Critical Care | Ausgabe 1/2023

Abstract

Background

Vascular leakage is a major feature of acute respiratory distress syndrome (ARDS). We aimed to evaluate the efficacy of FX06, a drug under development that stabilizes interendothelial cell junctions, at reducing vascular leakage during SARS-CoV-2-induced ARDS.

Methods

This multicenter, double-blinded, randomized trial included adults with COVID-19-associated ARDS who had received invasive mechanical ventilation for < 5 days and were randomized to receive either intravenous FX06 (400 mg/d, for 5 days) or its vehicle as placebo. The primary endpoint was the lowering—from day 1 to day 7—of the transpulmonary thermodilution-derived extravascular lung-water index (EVLWi).

Results

Twenty-five patients were randomized to receive FX06 and 24 the placebo. Although EVLWi was elevated at baseline (median [IQR] 15.6 mL/kg [13.5; 18.5]), its declines from day 1 to day 7 were comparable for FX06 recipients and controls (respectively, − 1.9 [− 3.3; − 0.5] vs. − 0.8 [− 5.5; − 1.1] mL/kg; estimated effect − 0.8 [− 3.1; + 2.4], p = 0.51). Cardiac indexes, pulmonary vascular permeability indexes, and fluid balances were also comparable, as were PaO2/FiO2 ratios and durations of mechanical ventilation. Adverse event rates were similar for the 2 groups, although more FX06 recipients developed ventilator-associated pneumonia (16/25 (64%) vs. 6/24 (24%), p = 0.009).

Conclusions

In this unique-dosing–regimen study, FX06 did not lower SARS-CoV-2-induced pulmonary vascular leakage. Future investigations will need to evaluate its efficacy at earlier times during the disease or using other regimens.
Trial registration NCT04618042. Registered 5 November 2020.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s13054-023-04616-1.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ARDS
Acute respiratory distress syndrome
ECMO
Extracorporeal membrane oxygenation
EVLWi
Extravascular lung-water index
(s)VE-cadherin
(soluble) vascular endothelial cadherin
SARS-CoV-2
Severe acute respiratory syndrome–coronavirus-2
COVID-19
Coronavirus disease 2019

Introduction

Vascular leakage is a major feature of pathogen-induced acute respiratory distress syndrome (ARDS) [1]. Triggered by inflammation following endothelial and epithelial lesions, it is thought to play an important role in altering gas exchanges. Consequently, the extravascular lung-water index (EVLWi), a marker of pulmonary vascular leakage measured by transpulmonary thermodilution, is independently associated with ARDS patients’ outcomes [2, 3]. Because EVLWi is highly elevated during SARS-CoV-2-induced ARDS [4, 5], which causes high mortality [4, 6], controlling vascular leakage might be of major interest in managing this disease.
FX06, an innovative drug containing fibrin-derived peptide Bβ15-42, stabilizes vascular endothelial (VE)-cadherin–dependent interendothelial cell junctions [79]. It reduced capillary leakage in several animal models of lipopolysaccharide- or HCl-induced acute lung injury [9, 10] and prolonged survival in a murine model of dengue-virus infection [9]. In a phase II trial conducted on 234 patients suffering from ischemia–reperfusion injuries during acute coronary syndrome, FX06-treated patients had 58% smaller early necrotic core zones [11]. Importantly, adverse events were comparable between groups, indicating the drug’s high safety profile. FX06 was then used as salvage therapy for a patient with severe ARDS following Ebola-virus infection, with a temporal link between its injection and sharply decreased EVLWi [12]. More recently, FX06 (400 mg/d for 4–7 days) was given as compassionate therapy to 6 patients receiving extracorporeal membrane oxygenation (ECMO) for coronavirus disease 2019 (COVID-19) [13]; 4 experienced improvement and 2 died. No clear treatment-related adverse event occurred.
Taken together, those findings indicate that FX06 is well-tolerated by patients and is a potent regulator of vascular leakage during ARDS. We hypothesized that FX06 might limit pulmonary vascular hyperpermeability during ARDS induced by SARS-CoV-2 infection, thereby improving gas exchanges and patients’ outcomes.

Methods

Trial design

We conducted a multicenter, double-blinded, randomized trial. The independent ethics review board CPP Ouest VI, Brest, France, and the ANSM (Agence Nationale de Sécurité du Médicament et des Produits de Santé) approved the trial protocol (available in  Additional file 1). F4-Pharma Ges.m.b.H. (Vienna, Austria) provided FX06. An independent Data- and Safety-Monitoring Committee periodically reviewed safety outcomes, with recruitment interruptions planned after inclusions of 10 and 30 patients. Neither F4-Pharma nor trial sponsors participated in the trial design, data collection, analysis or interpretation, or the writing or submission of the manuscript. The study protocol was registered at ClinicalTrials.gov (NCT04618042).

Participants

To be eligible for inclusion, patients had to be ≥ 18 year old and receiving invasive mechanical ventilation for < 5 days for polymerase-chain reaction-confirmed SARS-CoV-2-induced ARDS, according to the Berlin definition [14]. Exclusion criteria were mechanical ventilation for > 4 days; participation in another interventional clinical trial; severe renal, hepatic or cardiac insufficiency, or in a moribund state at randomization (see Additional file 1); contraindication for vascular access implantation for transpulmonary thermodilution monitoring; chemotherapy, radiotherapy or immunotherapy for malignancy; pregnancy or lactation; any history of severe allergic drug reaction. Patients taking drugs interfering with inflammation were also excluded, unless the drug’s use during COVID-19 was stated in the hospital center’s written policy.
According to the specifications of emergency consent, randomization was possible without a close relative’s or surrogate’s consent, but informed consent by the patient or patient’s relatives was obtained for all patients.

Treatment allocation

Patients were randomly assigned to receive either FX06 or its vehicle (phosphate-buffered saline) as the placebo. The randomization list was computer-generated with a 1:1 ratio and undisclosed block sizes, stratified by center. Concealment of the study-group assignments used a centralized, secure, interactive, web-based response system (CleanWeb, Telemedicine Technologies S.A.S., Boulogne-Billancourt, France) accessible from each study center. All investigators, statisticians, and data analysts were blinded to arm assignments until the study and analysis were completed.

Interventions

Patients were randomized to receive intravenous FX06, 400 mg/d or the placebo for 5 days. Each dose was administered in two boluses separated by a 10-min interval. The dose regimen chosen was based on the results of previous studies, in animals and humans, that suggested safety and mechanistic engagement with this dosing (additional Methods in Additional file 2). The manufacturer provided each treatment in unrecognizable ready-to-use form (numbered and sealed therapeutic units containing 10 vials of active treatment or placebo solution), stocked in each intensive care unit (ICU) under the supervision of each facility’s pharmacy department.
Patients were monitored using transpulmonary thermodilution systems (EV1000/Volume View, Edwards Lifesciences, Irvine, CA, USA, or PiCCO2, Pulsion Medical Systems, Feldkirchen, Germany), with thermistor-tipped catheters introduced in a femoral artery and an internal jugular vein [24]. Extravascular lung water and other thermodilution-derived parameters were averaged from three injections of cold physiological saline solution, in supine position, and indexed to the patient’s predicted body weight. Thermodilution measurements were taken before treatment administration during the first 5 days post-inclusion, with a measurement repeated 3 h post-administration on day 2, to detect a possible short-time effect of the drug. For patients receiving venovenous (VV)-ECMO, measurements were taken during a transient diminution of ECMO blood flow to < 2 L/min. Preliminary study results showed that thermodilution parameters were not affected by ECMO blood flow under that level (see Additional file 2: Table S1 and Fig. S1).
For ARDS management, investigators were asked to follow the most recent recommendations from the French Society of Intensive Care Medicine (https://​www.​srlf.​org/​rfe-srlf-prise-en-charge-du-syndrome-de-detresse-respiratoire-aigue-sdra-de-ladulte-a-la-phase-initiale/​). Specific treatments targeting COVID-19 were discouraged, unless the drug’s use during COVID-19 was stated in the center’s written policy.
Serum interleukin (IL)-6, IL-10, and soluble (s)VE-cadherin were quantified with DuoSet Elisa kits (R&D systems, Minneapolis, MN, USA).

Outcomes

The primary endpoint was the EVLWi change, assessed by transpulmonary thermodilution, between day 1 and day 7. Secondary endpoints included the evolution of daily EVLWi, cardiac index, global end-diastolic volume index, and pulmonary vascular permeability index measured by transpulmonary thermodilution for 7 days; daily fluid balance; serum albumin; systolic, diastolic, and mean blood pressures; and heart rate for 7 days; partial oxygen pressure/fraction of inspired oxygen (PaO2/FiO2) ratio and Sequential Organ-Failure Assessment (SOFA) score over 15 days; rate of rescue with VV-ECMO; durations of invasive mechanical ventilation, vasopressor support, and renal replacement therapy over 30 days; Weinberg Radiological Severity score over 30 days [15]; survival at 30 and 60 days; nature and frequency of adverse events. Kinetics of serum d-dimers and C-reactive protein over 7 days were extracted from medical charts afterward. Serum IL-6, IL-10, and sVE-cadherin measurements on days 1 and 7 in available biological samples were added as post hoc measurements.

Statistical analyses

Assuming a baseline (inclusion) mean EVLWi of 13 mL/kg and standard deviation (SD) of 5 mL/kg [16], and a 30% EVLWi decrease in FX06-treated patients compared to controls on day 7 [9, 10, 12], for 80% power and an overall 5% two-sided α-risk, the required sample size was 25 patients/group.
Baseline characteristics are reported as number (%) for categorical variables and median [interquartile range, IQR] for continuous variables. Efficacy endpoints were analyzed according to intention-to-treat principles. Safety endpoints were analyzed for all patients who received at least one assigned-treatment dose.
Missing primary endpoints were replaced by imputation values for patients who died or whose conditions no longer warranted the transpulmonary thermodilution system before 7 days; the last thermodilution value was retained for the primary analyses. Primary endpoints were compared between groups using an adjusted analysis of covariance (ANCOVA) of EVLWi at randomization. Results are expressed in terms of adjusted mean change with 95% confidence interval (CI).
Three sensitivity analyses were computed: complete case analysis, worst-case analysis or using a different statistical method (Mann–Whitney U test). Prespecified subgroup analyses were conducted according to VV-ECMO or EVLWi > 10 mL/kg at inclusion.
Qualitative and quantitative secondary outcome measures were compared between groups using, respectively, Pearson’s Chi-square tests and t tests, or Mann–Whitney U tests. Overall survival was estimated with the Kaplan–Meier method. Longitudinal quantitative endpoints were compared using linear-mixed models with a random effect for subjects. This model was fitted to fixed effect by an interaction between treatment arm and time (since the date of randomization), with the slope parameter estimating the difference between groups. A restricted likelihood maximization-estimation method was used. The p-values associated with the fixed effects were calculated using the analysis of variance (ANOVA) function with Kenward–Roger approximation for calculating the number of degrees of freedom.
Analyses were computed with a 2-sided α risk of 5%. All analyses were performed using R software (R Foundation for Statistical Computing, Vienna, Austria), version 4.0.3.

Results

Forty-nine patients were randomized from November 2020 to April 2021 and retained for analysis (Fig. 1). Their main characteristics are reported in Table 1. They were very severely ill at baseline, with median PaO2/FiO2 ratio at 104, static pulmonary compliance of < 30 mL/cmH2O, and more than one-third of them were on VV-ECMO. One-third were receiving vasopressors. Specific therapies targeting COVID-19 were marginal, except corticosteroids, given to all participants.
Table 1
Characteristics of patients with COVID-19-associated ARDs at trial inclusion, according to assigned treatment arm
Characteristic
FX06 (n = 25)
Placebo (n = 24)
Age, y
59 [53; 67]
59 [53; 68]
Sex
 Male
19 (76)
16 (67)
 Female
6 (24)
8 (33)
Pre-existing condition
 Diabetes
9 (36)
6 (25)
 Hypertension
11 (44)
14 (58)
 Coronary artery disease
4 (16)
2 (8)
 Severe cardiovascular disease*
0
0
 Chronic respiratory disease
1 (4)
2 (8)
 Kidney disease†
1 (4)
0
 Chronic liver disease
0
0
 Immunodeficiency
0
0
 Cancer
0
3 (12)
Body mass index, kg/m2
32 [28; 35]
30 [27; 36]
Simplified Acute Physiology Score II
58 [41; 65]
56 [38; 66]
Days to inclusion
 From symptom onset
14 [10; 18]
12 [9; 14]
 From first positive PCR
12 [10–16]
9 [7–14]
 From oxygen therapy onset
5 [1–7]
3 [2–5]
 From invasive mechanical ventilation onset
2 [2; 4]
2 [2; 3]
COVID-19 therapy use
 Corticosteroids
25 (100)
24 (100)
 Remdesivir
0
0
 Immunomodulator (tocilizumab or sarilumab)
3 (12)
3 (12)
 Other
0
0
Respiratory support prior to intubation
 High flow oxygen device
24 (96)
19 (79)
 High flow oxygen device duration, days
2.5 [0.25–5]
2 [1–2.75]
 Non-invasive mechanical ventilation (NIV)
8 (32)
6 (25)
 NIV duration, days
2 [1–3]
1.5 [0–4.5]
Respiratory status
 Tidal volume, mL/kg of PBW
3.7 [2.7; 4.7]
4.4 [3.0; 4.8]
 Plateau pressure, cmH2O
26 [24; 28]
25 [24; 27]
 Positive end-expiratory pressure, cmH2O
12 [12; 15]
12 [10; 14]
 Cstrs‡, mL/cmH2O
26.2 [18.7; 29.4]
27.8 [21.1; 38.7]
 FiO2
100 [60; 100]
100 [64; 100]
 VV-ECMO
10 (40)
7 (29)
Arterial blood gases
 PaO2, mmHg
68 [60; 92]
82 [70; 96]
 PaO2/FiO2§, mmHg
102 [62; 145]
110 [73; 187]
 PaCO2, mmHg
42 [39; 47]
42 [39; 44]
 Arterial pH
7.43 [7.40; 7.46]
7.40 [7.39; 7.46]
Vasopressor use
9 (36)
9 (37)
Serum creatinine, μmol/L
66 [51; 86]
66 [46; 97]
C-reactive protein, mg/L
102 [58; 159]
120 [79; 215]
Fibrinogenemia, g/L
6.6 [5.8; 7.4]
7.0 [6.0; 8.4]
d-Dimers, mg/L
2650 [1029; 8348]
2625 [1335; 4872]
Values are expressed as n (%) or median [IQR 25th; 75th percentiles]. NIV non-invasive mechanical ventilation; PBW predicted body weight; VV-ECMO venovenous extracorporeal membrane oxygenation
*Defined as New York Heart Association class III or IV. †Determined from the most recent stable serum creatinine level prior to this hospital admission, except for patients on dialysis. Abnormal kidney function was defined as serum creatinine ≥ 130 μmol/L (≥ 1.5 mg/dL) for males or ≥ 100 μmol/L (≥ 1.1 mg/dL) for females not previously on dialysis. ‡Cstrs static compliance of the respiratory system; Cst is the tidal volume in mL divided by the driving pressure (plateau pressure—positive end-expiratory pressure) in cmH2O §For VV-ECMO patients, last ratio before ECMO implantation

Study drug

All but 5 patients received the complete treatment. One patient allocated to receive the placebo was accidentally included in another interventional study; the assigned treatment was interrupted after 3 days. One patient allocated to the FX06 arm stopped treatment after 3 days because of fungal co-infection. Two patients allocated to the placebo arm died on day 3 or day 4. Lastly, one patient’s treatment was accidentally withheld on day 4.

Primary outcome

The primary outcome of EVLWi change between days 1 and 7 did not differ between FX06-treated patients and controls (Fig. 2 and Table 2). EVLWi kinetics and their individual variations were also comparable between groups (Additional file 2: Figs. S2 and S3). Patients’s EVLWis were high at inclusion, comparable for the 2 groups, and remained elevated during the first 7 days. Eight patients—1 FX06 recipient and 7 controls—did not undergo transpulmonary thermodilution on day 7; their last available values were retained for the primary analysis: 3 had died (1 FX06 recipient and 2 controls), 3 had recovered sufficiently to allow removal of their thermodilution catheters, 1 had a catheter infection necessitating its removal, and 1 withdrew consent to participate. Excluding those patients did not affect the primary-analysis results.
Table 2
Primary and secondary outcomes according to assigned treatment arm
Characteristic
FX06
Placebo
Estimated effect*
(95% CI)
p-value
D1–D7 EVLWi variation†, mL/kg
− 1.9 [− 3.3; − 0.5]
− 0.8 [− 5.5; 1.1]
− 0.48 [− 3.2; 2.3]
0.72
EVLWi†, mL/kg
 D1
15.9 [13.7; 18.3]
15.5 [12.6; 18.6]
 
0.38
 D7
14.4 [12.3; 17.5]
12.4 [10.0; 17.2]
 
0.89
Survival
    
 D7
24/25 (96)
21/24 (88)
− 8% [− 10%; 29%]ʊ
0.35
 D30
21/25 (84)
17/24 (71)
− 13% [− 37%; 11%]ʊ
0.27
 D60
17/25 (68)
17/24 (71)
+ 3% [− 24%; 30%]ʊ
0.83
D30 respiratory support-free days‡
0 [0; 14]
9 [0; 19]
OR = 0.4 [0.1; 1.2]
/IRR = 1.1 [0.8; 1.5]
0.19
D30 vasopressor-free days§
18 [12; 29]
17 [0; 30]
1 [− 15; 20]
0.90
D30 RRT-free days¶
30 [23; 30]
30 [0; 30]
0 [− 12; 9]
0.49
D30 SOFA-free days||
0 [0; 0]
0 [0; 1]
OR = 0.4 [0.1; 1.5]
/IRR = 1.1 [0.3; 4.4]
0.41
Plasma d-dimer, ng/L
  
− 358 [− 1120; 634]ϕ
1.0
 D1
2650 [1029; 8348]
2625 [1335; 4872]
  
 D7
3190 [1691; 8360]
2270 [1339.5; 3544]
  
Serum C-reactive protein, mg/L
  
− 48 [− 118; 53]ϕ
0.90
 D1
102 [57.8; 159]
120 [79; 215]
  
 D7
124 [87; 185]
140 [79; 193]
  
Plasma IL-6**, pg/mL
  
29 [− 125; 115]ϕ
0.91
 D1
24.9 [15.5; 93.6]
23.4 [3.85; 42.1]
  
 D7
118.2 [41.8; 218.7]
25.2 [14.9; 153.3]
  
Plasma IL-10**, pg/mL
  
0 [− 188; 6]ϕ
0.93
 D1
0.2 [0; 34.9]
2.1 [0; 52.6]
  
 D7
8.1 [0; 25.7]
0.5 [0; 69.0]
  
Plasma sVE-cadherin**, ng/mL
  
38 [− 170; 195]ϕ
0.57
 D1
586.3 [540.1; 681.6]
473.5 [393.1; 570.9]
  
 D7
628.7 [569.4; 793.1]
676.6 [580.5; 692.1]
  
Values are expressed as n (%) or median [25th; 75th percentiles, IQR]. D day. *Between-group difference of the mean variation of extravascular lung-water index (EVLWi) from day 1 to day 7, adjusted to the initial EVLWi. †Extravascular lung-water indexed to the predicted body weight. ‡Number of days alive without invasive mechanical ventilation. The effect estimate from Hurdle model is odds ratio to have more than 0 days alive without invasive mechanical ventilation/incidence risk ratio and bootstrap 95% CI. §Number of days alive without vasopressors. The effect estimate was measured with a median difference between arms and bootstrap 95% CI. ¶Number of days alive without renal replacement therapy. The effect estimate was measured with a median difference between arms and bootstrap 95% CI. ||Number of days alive without organ failure, defined by one or more Sequential Organ-Failure sub-Scores ≥ 3. The effect estimate obtained from Hurdle model is incidence risk ratio to have more than 0 days alive without organ failure/incidence risk ratio and bootstrap 95% CI. **Available for 13 FX06 recipients and 11 controls. ʊthe effect estimate was measured with a difference of proportion between arms and its exact binomial 95% confidence interval. ϕThe effect estimate was measured as a median difference of change from baseline between arms and bootstrap 95% CI
Several sensitivity analyses were computed: analyzing the primary outcome for patients with ECMO vs without; most severely ill (EVLWi > 16 mL/kg) vs less severely ill; or indexing the dose received above vs below 4.2 mg/kg/d (median dose received). All failed to detect any significant FX06 effect on EVLWi (not shown). Interestingly, EVLWi measured before and 3 h after FX06 bolus injections was very similar (median variation − 0.02 [IQR − 0.57; 0.55], n = 22).

Secondary outcomes

The Pulmonary Vascular Permeability Index (PVPI) was also very high during the first 7 days and did not differ between groups (Additional file 2: Fig. S4). Cardiac index and global end-diastolic volume index (GEDVI) were also comparable (not shown). Daily fluid balance remained positive during the first week, with comparable levels for the 2 groups (Additional file 2: Fig. S5). Serum albumin, another marker of vascular leakage, was very low at inclusion (median 23 [IQR 19; 26] g/L; n = 48); it remained stable with no between-group differences during the first week.
PaO2/FiO2 remained very low during the first 15 days (Additional file 2: Fig. S6), and very few patients survived to be extubated on 30 days (Table 2). Interestingly, the Weinberg Radiological Severity score decreased less for FX06 recipients (estimated effect 0.13 [95% CI 0.07–0.18]; p < 0.001). However, for a post hoc analysis taking into account missing values and mortality as a competing risk by imputing the last score available for survivors and a score of 12 after the patient died, score kinetics did not differ between groups (Additional file 2: Fig. S7). Three patients (2 FX06 recipients and 1 control) received VV-ECMO rescue therapy after inclusion.
Although catecholamine-free days were comparable for the 2 groups (Table 2), FX06 recipients had significantly lower systolic and diastolic blood pressure values after day 4 (Additional file 2: Fig. S8). FX06 did not affect heart rate (not shown).
The baseline SOFA score, reflecting the extent of multiple organ failure, was high and remained stable for both groups for 15 days (Additional file 2: Fig. S9). Finally, survival was comparable for both groups, with 34/49 (69%) day-60 survivors (Table 2 and Additional file 2: Fig. S10). C-reactive protein and serum d-dimers, elevated at baseline, remained stable and comparable. Serum cytokine measurements were available for 24 patients. For both groups at inclusion, IL-6 was elevated and continued to rise during the first week, while IL-10 levels were also high and declined slightly during the first week. FX06 recipients had slightly higher baseline sVE-cadherin values that remained comparable to those of the placebo group thereafter.

Safety

Adverse event rates were comparable for the 2 groups (Table 3). Although overall secondary infections were not more common in FX06 recipients, they did develop more episodes of microbiologically confirmed ventilator-associated pneumonia [17].
Table 3
Serious adverse events (SAEs) reported according to assigned treatment arm
Serious adverse events
FX06
Placebo
p-value
Patients with at least one, n (%)
22/25 (88)
21/24 (88)
1.0
Number
94
72
0.36
SAE type*
   
 VV-ECMO rescue
2
1
0.59
 Infectious event
22
18
0.11
  Bacteriemia
4
9
0.11
  Ventilator-associated pneumonia, n (%)
16 (64%)
6 (24%)
0.009
  Septic shock
2
2
1.0
 Ischemic stroke
1
0
1.0
 Hemorrhagic stroke
0
2
0.23
 Cardiac rhythm disorder
0
2
0.23
 Cardiogenic shock
0
1
0.49
 Gastrointestinal ulcer
1
0
1.0
 Liver cytolysis/cholestasis
1
3
0.35
 Acute kidney injury
4
4
1.0
 Anemia
1
0
1.0
 Thrombocytopenia
0
2
1.0
 Bleeding episode
3
2
1.0
 Thrombosis
0
2
1.0
 Cutaneous reaction
1
0
1.0
 Rhabdomyolysis
0
1
0.49
Values are expressed as number unless stated otherwise. VV-ECMO Venovenous extracorporeal membrane oxygenation. *According to the Common Terminology Criteria for Adverse Events (CTCAE) v.4.0

Discussion

In this multicenter, double-blinded, randomized trial, FX06 did not alter the thermodilution-measured EVLWi evolution during SARS-CoV-2-induced ARDS. Other markers of pulmonary vascular leakage, e.g., patients’ functional outcomes reflecting pulmonary function and 60-day survival, were also not affected. Although elevated at baseline, circulating markers of inflammation and endothelial lesions were comparable for the 2 groups. Despite their similar rates of serious adverse events, FX06 was associated with higher rates of ventilator-associated pneumonia.
Inflammation-induced pulmonary vascular leakage is widely diffused during severe SARS-CoV-2 infection. Autopsies of COVID-19 patients revealed markedly elevated lung weights [18, 19] and disruption of interendothelial VE-cadherin-dependent junctions [20]. Furthermore, COVID-19 patients’ plasmas were able to trigger rapid and sustained enhanced permeability of human pulmonary microvascular endothelial cells (HPMVEC) cultured in vitro [21]. Our patients’ baseline EVLWi values were very high, confirming extensive vascular hyperpermeability. C-reactive protein and IL-6 levels further confirmed high levels of inflammation. VE-cadherin-dependent vascular leakage, the mechanism targeted by FX06, was thus likely involved in our patients’ pulmonary lesions.
However, several factors limit the interpretation of our results. SARS-CoV-2 is responsible for a particular form of ARDS, in which several mechanisms other than VE-cadherin-mediated endothelial barrier disruption could account for vascular leakage that might have masked the FX06 effect. Results of autopsy series previously showed disruption of endothelial tight junctions [20]. They also revealed severe endothelial cell injury, with images of cell apoptosis and membrane disruption [18, 22]. In vitro study observations confirmed the cytotoxicity of plasmas from SARS-CoV-2-infected patients on endothelial cell monolayers [23]. Importantly, all autopsy findings highlighted widespread microvascular thromboses in the lungs [18, 20, 22] that might be responsible for disseminated endothelial cell dysfunction, lysis, and death [24]. Thus, other ARDS forms not originating from COVID-19 might be more suitable for FX06 evaluation, as they may be associated with less pronounced non-inflammatory physiological processes implicated in vascular leakage.
Our population had other limitations hampering treatment evaluation. With a median of 13 days between first symptoms and treatment initiation, it is likely that we missed the initial exudative phase of ARDS. That supposition is supported by the elevated EVLWi in our population at baseline. Notably, pulmonary edema remains a dynamic balance between fluid extravasation and resorption. Likewise, VE-cadherin-dependent interendothelial cell-junction stability relies on a dynamic balance between its internalization and recycling to the membrane [25]. Thus, by stabilizing VE-cadherin homotypic interactions, FX06 might still have had a net effect on extravascular fluid balance, even at late stages of ARDS. Such a late effect was suggested by the first FX06 rescue therapy during ARDS in humans, which occurred after 11 days of Ebola-virus infection [12]. However, we cannot exclude a more powerful FX06 effect in preventing vascular leakage at earlier stages.
Better characterization of vascular leakage kinetics during ARDS in humans and drug evaluation earlier during disease evolution are needed to determine the optimal window for its administration during ARDS. FX06 assessment in other entities with inflammation-induced vascular leakage, that would allow its prompt administration earlier during the disease course, might also be highly contributive. In this setting, post-resuscitation syndrome, in which the drug was highly effective in pre-clinical studies [26], might be of particular interest. Moreover, with one-third of the patients on VV-ECMO, a median PaO2/FiO2 ratio ~ 100 mmHg, and median static compliance of 27 mL/cmH2O, our population might have been too severely ill to detect an effect. Evaluating FX06 in less severe forms of COVID-19 remains of interest. Lastly, all of our patients were receiving corticosteroids, as recommended after the RECOVERY trial [27], which might have partially dampened inflammation, thereby masking the FX06 therapeutic effect.
The dosing regimen used is also open to debate. At a median of 4.2 mg/kg/d, our FX06 dose was close to the optimal one described for animals [9, 28], and those used as rescue therapy in humans [12, 13]. Although indexing the dose received to patients’ weight did not change the primary endpoint herein, we cannot exclude that higher doses might have achieved a detectable effect. Likewise, the FX06 half-life in the plasma of healthy volunteers was short (11–17 min) [29], even though FX06 effects linked to VE-cadherin persisted for hours or days in animal models [9, 10]. No EVLWi change was detected shortly (3 h) after its bolus injections in our study, which does not exclude that dosing regimens with repeated or continuous injections or higher doses might be more effective. In this context, PK/PD analyses of FX06 administration to critically ill patients should be highly informative. Although initially planned in our study, that analysis could not be done for technical reasons, in the context of the pandemic. Taken together, our data highlight the need for careful reassessment of the FX06-dosing regimen in future studies on humans.
Beyond PK, evaluating FX06 biological activity is complex. Although thermodilution-measured EVLWi accurately reflects the level of pulmonary edema [30], the hypothesized effect of 30% reduction in 7 days in FX06-treated patients might have been too large. FX06 limited vascular leakage by 60–80% in animal models of ARDS [9, 10] and was associated with 52% EVLWi reduction in a patient treated for Ebola-induced ARDS [12]. However, the multiplicity of factors impacting this parameter in humans might suggest a more restrictive approach to its anticipated reduction in future studies.
Other techniques might also be more sensitive in detecting vascular leakage and serve as better surrogate markers for FX06 development. Among markers implicated in endothelial physiology, several have been associated with patients’ outcomes after various causes of critical illnesses [3133], including COVID-19 [3436]. Among them, angiopoietin-2 might have the strongest association with the level of vascular leakage [32, 33]. We used sVE-cadherin as a possible surrogate marker of VE-cadherin stabilization by FX06. Although samples were only available for 24 patients, its serum concentration on day 7 was unaffected by FX06. However, despite sVE-cadherin being a marker of endothelial barrier disruption [37], its accuracy in predicting vascular leakage and patients’ outcomes remains debated [34, 38]. Whether other markers of endothelial injury might have been more sensitive in detecting an FX06 effect in our study is unknown and is currently the focus of ongoing studies. Whether some might also help improve patient selection for future studies remains to be seen.
VV-ECMO, implanted in one-third of our patients, may have also interfered with the evaluation of an FX06 effect, with the device influencing drug PK. However, considering the low lipophilic properties of FX06, significant adsorption onto the membrane is unlikely. EVLWi evaluation itself is also affected by VV-ECMO blood flow [39, 40]. Our observations in a preliminary study conducted on 20 patients at the time of ECMO explantation (Additional file 2: Methods), combined with the results of another group in 7 patients [39], showed that EVLWi determination is minimally affected for blood flows of < 2 L/min, like those used herein. Excluding patients on ECMO from the analysis also did not modify the results. Theoretically, pulmonary vascular obstruction might also contribute to EVLWi underestimation. Our COVID-19 patients, particularly at risk for pulmonary embolism, were not routinely screened for it. Although pulmonary embolism events did not affect the results of thermodilution-derived EVWLis in a recent study on COVID-19 patients [4], we cannot exclude they might have interfered with our evaluations.
Our observations on FX06 effect on arterial pressure are noteworthy. After 4 days of treatment, FX06 recipients were indeed less hypertensive than patients given the placebo. Although that finding remains exploratory, it might indicate an FX06 effect on the vasculature to be investigated in future studies.
With comparable rates of serious adverse events reported for both trial groups, our results confirm the good safety profile of FX06. Although exploratory, the higher rates of ventilator-associated pneumonia episodes developing in FX06 recipients warrant attention. Fibrin fragment Bβ15-42 was shown to inhibit leukocyte transendothelial migration in vitro [7] and to dampen neutrophil recruitment in the lung in two models of lipopolysaccharide- or HCl-induced acute lung injury, indicating a risk of impaired bacterial clearance [10]. Importantly, incubation of FX06 with monocytes or alveolar macrophages did not impact their in vitro activation and capacity to release pro-inflammatory cytokines. Moreover, on the contrary, FX06 was shown to enhance bacterial clearance and ultimately survival in a model of secondary Pseudomonas aeruginosa infection [10]. Although this potential effect of FX06 needs to be better clarified, these findings indicate the need for close monitoring of secondary infections in future evaluations of it.

Conclusions

FX06 did not lower thermodilution-derived EVLWi during severe ARDS induced by SARS-CoV-2 infection. Whether other time-lines for its administration or other dosing regimens might be more efficient remains to be determined.

Acknowledgements

We are very grateful to Prof. Peter Radermacher, Prof. Jean Chastre, and Dr. Florence Caniou-Poitrine, who were responsible for the independent safety monitoring of the study. We also thank Dr Fanny Charbonnier-Beaupel and Dr Annick Tibi, for their constant help with pharmacology, technical, and organizational issues. We thank Edwards Lifesciences S.A., USA, for providing thermodilution systems.

Declarations

The independent ethics review board CPP Ouest VI, Brest, France, approved the trial protocol (CPP 1306-EudraCT 2020-002056-20/RIPH 20.06.15.56627). The trial was conducted in accordance with the ethical principles of the Declaration of Helsinki, the International Conference on Harmonisation–Good Clinical Practice (ICH-GCP) guideline, the Quality Management Standards for Drug Clinical Trials of the French Food and Drug Administration (ANSM, Agence Nationale de Sécurité du Médicament et des Produits de Santé). According to the specifications of emergency consent, randomization without a close relative or surrogate consent could be performed, but informed consent by the patient or patient’s relatives was obtained for all patients.
Not applicable.

Competing interests

Dr Bréchot is on the F4-Pharma advisory board, without any financial competing interest. He received a grant from the French Ministry of Health for another study evaluating FX06. He receives fees from Findimmune, outside the scope of this study. Dr Luyt reported receiving grants from French Ministry of Health during the conduct of the study; personal fees from Bayer Healthcare, Carmat, Faron, Merck Sharp & Dohme, ThermoFisher Brahms, and BioMérieux, outside the submitted work. Dr. Combes reports receiving grant support and lecture fees from Maquet and Baxter, and consulting fees from Hemovent.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
2.
Zurück zum Zitat Tagami T, Ong MEH. Extravascular lung water measurements in acute respiratory distress syndrome: why, how, and when? Curr Opin Crit Care. 2018;24:209–15.CrossRefPubMedPubMedCentral Tagami T, Ong MEH. Extravascular lung water measurements in acute respiratory distress syndrome: why, how, and when? Curr Opin Crit Care. 2018;24:209–15.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Jozwiak M, Silva S, Persichini R, Anguel N, Osman D, Richard C, et al. Extravascular lung water is an independent prognostic factor in patients with acute respiratory distress syndrome*. Crit Care Med. 2013;41:472–80.CrossRefPubMed Jozwiak M, Silva S, Persichini R, Anguel N, Osman D, Richard C, et al. Extravascular lung water is an independent prognostic factor in patients with acute respiratory distress syndrome*. Crit Care Med. 2013;41:472–80.CrossRefPubMed
4.
Zurück zum Zitat Shi R, Lai C, Teboul J-L, Dres M, Moretto F, De Vita N, et al. COVID-19 ARDS is characterized by higher extravascular lung water than non-COVID-19 ARDS: the PiCCOVID study. Crit Care Lond Engl. 2021;25:186.CrossRef Shi R, Lai C, Teboul J-L, Dres M, Moretto F, De Vita N, et al. COVID-19 ARDS is characterized by higher extravascular lung water than non-COVID-19 ARDS: the PiCCOVID study. Crit Care Lond Engl. 2021;25:186.CrossRef
5.
Zurück zum Zitat Rasch S, Schmidle P, Sancak S, Herner A, Huberle C, Schulz D, et al. Increased extravascular lung water index (EVLWI) reflects rapid non-cardiogenic oedema and mortality in COVID-19 associated ARDS. Sci Rep. 2021;11:11524.CrossRefPubMedPubMedCentral Rasch S, Schmidle P, Sancak S, Herner A, Huberle C, Schulz D, et al. Increased extravascular lung water index (EVLWI) reflects rapid non-cardiogenic oedema and mortality in COVID-19 associated ARDS. Sci Rep. 2021;11:11524.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Marconi VC, Ramanan AV, de Bono S, Kartman CE, Krishnan V, Liao R, et al. Efficacy and safety of baricitinib for the treatment of hospitalised adults with COVID-19 (COV-BARRIER): a randomised, double-blind, parallel-group, placebo-controlled phase 3 trial. Lancet Respir Med. 2021;9:1407–18.CrossRefPubMedPubMedCentral Marconi VC, Ramanan AV, de Bono S, Kartman CE, Krishnan V, Liao R, et al. Efficacy and safety of baricitinib for the treatment of hospitalised adults with COVID-19 (COV-BARRIER): a randomised, double-blind, parallel-group, placebo-controlled phase 3 trial. Lancet Respir Med. 2021;9:1407–18.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Yakovlev S, Gao Y, Cao C, Chen L, Strickland DK, Zhang L, et al. Interaction of fibrin with VE-cadherin and anti-inflammatory effect of fibrin-derived fragments. J Thromb Haemost JTH. 2011;9:1847–55.CrossRefPubMed Yakovlev S, Gao Y, Cao C, Chen L, Strickland DK, Zhang L, et al. Interaction of fibrin with VE-cadherin and anti-inflammatory effect of fibrin-derived fragments. J Thromb Haemost JTH. 2011;9:1847–55.CrossRefPubMed
8.
Zurück zum Zitat Bach TL, Barsigian C, Yaen CH, Martinez J. Endothelial cell VE-cadherin functions as a receptor for the beta15-42 sequence of fibrin. J Biol Chem. 1998;273:30719–28.CrossRefPubMed Bach TL, Barsigian C, Yaen CH, Martinez J. Endothelial cell VE-cadherin functions as a receptor for the beta15-42 sequence of fibrin. J Biol Chem. 1998;273:30719–28.CrossRefPubMed
9.
Zurück zum Zitat Gröger M, Pasteiner W, Ignatyev G, Matt U, Knapp S, Atrasheuskaya A, et al. Peptide Bbeta(15–42) preserves endothelial barrier function in shock. PLoS ONE. 2009;4: e5391.CrossRefPubMedPubMedCentral Gröger M, Pasteiner W, Ignatyev G, Matt U, Knapp S, Atrasheuskaya A, et al. Peptide Bbeta(15–42) preserves endothelial barrier function in shock. PLoS ONE. 2009;4: e5391.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Matt U, Warszawska JM, Bauer M, Dietl W, Mesteri I, Doninger B, et al. Bbeta(15–42) protects against acid-induced acute lung injury and secondary pseudomonas pneumonia in vivo. Am J Respir Crit Care Med. 2009;180:1208–17.CrossRefPubMed Matt U, Warszawska JM, Bauer M, Dietl W, Mesteri I, Doninger B, et al. Bbeta(15–42) protects against acid-induced acute lung injury and secondary pseudomonas pneumonia in vivo. Am J Respir Crit Care Med. 2009;180:1208–17.CrossRefPubMed
11.
Zurück zum Zitat Atar D, Petzelbauer P, Schwitter J, Huber K, Rensing B, Kasprzak JD, et al. Effect of intravenous FX06 as an adjunct to primary percutaneous coronary intervention for acute ST-segment elevation myocardial infarction results of the F.I.R.E. (Efficacy of FX06 in the Prevention of Myocardial Reperfusion Injury) trial. J Am Coll Cardiol. 2009;53:720–9. Atar D, Petzelbauer P, Schwitter J, Huber K, Rensing B, Kasprzak JD, et al. Effect of intravenous FX06 as an adjunct to primary percutaneous coronary intervention for acute ST-segment elevation myocardial infarction results of the F.I.R.E. (Efficacy of FX06 in the Prevention of Myocardial Reperfusion Injury) trial. J Am Coll Cardiol. 2009;53:720–9.
12.
Zurück zum Zitat Wolf T, Kann G, Becker S, Stephan C, Brodt H-R, de Leuw P, et al. Severe Ebola virus disease with vascular leakage and multiorgan failure: treatment of a patient in intensive care. Lancet Lond Engl. 2015;385:1428–35.CrossRef Wolf T, Kann G, Becker S, Stephan C, Brodt H-R, de Leuw P, et al. Severe Ebola virus disease with vascular leakage and multiorgan failure: treatment of a patient in intensive care. Lancet Lond Engl. 2015;385:1428–35.CrossRef
13.
Zurück zum Zitat Adam EH, Schmid B, Sonntagbauer M, Kranke P, Zacharowski K, Meybohm P. Fibrin-derived peptide Bβ15-42 (FX06) as salvage treatment in critically ill patients with COVID-19-associated acute respiratory distress syndrome. Crit Care. 2020;24:574.CrossRefPubMedPubMedCentral Adam EH, Schmid B, Sonntagbauer M, Kranke P, Zacharowski K, Meybohm P. Fibrin-derived peptide Bβ15-42 (FX06) as salvage treatment in critically ill patients with COVID-19-associated acute respiratory distress syndrome. Crit Care. 2020;24:574.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat ARDS Definition Task Force, Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, et al. Acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012;307:2526–33. ARDS Definition Task Force, Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, et al. Acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012;307:2526–33.
15.
Zurück zum Zitat Bréchot N, Demondion P, Santi F, Lebreton G, Pham T, Dalakidis A, et al. Intra-aortic balloon pump protects against hydrostatic pulmonary oedema during peripheral venoarterial-extracorporeal membrane oxygenation. Eur Heart J Acute Cardiovasc Care. 2018;7:62–9.CrossRefPubMed Bréchot N, Demondion P, Santi F, Lebreton G, Pham T, Dalakidis A, et al. Intra-aortic balloon pump protects against hydrostatic pulmonary oedema during peripheral venoarterial-extracorporeal membrane oxygenation. Eur Heart J Acute Cardiovasc Care. 2018;7:62–9.CrossRefPubMed
16.
Zurück zum Zitat Wang H, Cui N, Su L, Long Y, Wang X, Zhou X, et al. Prognostic value of extravascular lung water and its potential role in guiding fluid therapy in septic shock after initial resuscitation. J Crit Care. 2016;33:106–13.CrossRefPubMed Wang H, Cui N, Su L, Long Y, Wang X, Zhou X, et al. Prognostic value of extravascular lung water and its potential role in guiding fluid therapy in septic shock after initial resuscitation. J Crit Care. 2016;33:106–13.CrossRefPubMed
17.
Zurück zum Zitat Luyt C-E, Sahnoun T, Gautier M, Vidal P, Burrel S, Pineton de Chambrun M, et al. Ventilator-associated pneumonia in patients with SARS-CoV-2-associated acute respiratory distress syndrome requiring ECMO: a retrospective cohort study. Ann Intensive Care. 2020;10:158. Luyt C-E, Sahnoun T, Gautier M, Vidal P, Burrel S, Pineton de Chambrun M, et al. Ventilator-associated pneumonia in patients with SARS-CoV-2-associated acute respiratory distress syndrome requiring ECMO: a retrospective cohort study. Ann Intensive Care. 2020;10:158.
18.
Zurück zum Zitat Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T, Laenger F, et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N Engl J Med. 2020;383:120–8.CrossRefPubMedPubMedCentral Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T, Laenger F, et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N Engl J Med. 2020;383:120–8.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Wichmann D, Sperhake J-P, Lütgehetmann M, Steurer S, Edler C, Heinemann A, et al. Autopsy Findings and Venous Thromboembolism in Patients With COVID-19: A Prospective Cohort Study. Ann Intern Med. 2020;173:268–77.CrossRefPubMed Wichmann D, Sperhake J-P, Lütgehetmann M, Steurer S, Edler C, Heinemann A, et al. Autopsy Findings and Venous Thromboembolism in Patients With COVID-19: A Prospective Cohort Study. Ann Intern Med. 2020;173:268–77.CrossRefPubMed
20.
Zurück zum Zitat D’Agnillo F, Walters K-A, Xiao Y, Sheng Z-M, Scherler K, Park J, et al. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19. Sci Transl Med. 2021;13:eabj7790. D’Agnillo F, Walters K-A, Xiao Y, Sheng Z-M, Scherler K, Park J, et al. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19. Sci Transl Med. 2021;13:eabj7790.
21.
Zurück zum Zitat Michalick L, Weidenfeld S, Grimmer B, Fatykhova D, Solymosi PD, Behrens F, et al. Plasma mediators in patients with severe COVID-19 cause lung endothelial barrier failure. Eur Respir J. 2021;57:2002384.CrossRefPubMedPubMedCentral Michalick L, Weidenfeld S, Grimmer B, Fatykhova D, Solymosi PD, Behrens F, et al. Plasma mediators in patients with severe COVID-19 cause lung endothelial barrier failure. Eur Respir J. 2021;57:2002384.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Varga Z, Flammer AJ, Steiger P, Haberecker M, Andermatt R, Zinkernagel AS, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020;395:1417–8.CrossRefPubMedPubMedCentral Varga Z, Flammer AJ, Steiger P, Haberecker M, Andermatt R, Zinkernagel AS, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020;395:1417–8.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Rauch A, Dupont A, Goutay J, Caplan M, Staessens S, Moussa M, et al. Endotheliopathy Is Induced by Plasma From Critically Ill Patients and Associated With Organ Failure in Severe COVID-19. Circulation. 2020;142:1881–4.CrossRefPubMedPubMedCentral Rauch A, Dupont A, Goutay J, Caplan M, Staessens S, Moussa M, et al. Endotheliopathy Is Induced by Plasma From Critically Ill Patients and Associated With Organ Failure in Severe COVID-19. Circulation. 2020;142:1881–4.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Giannotta M, Trani M, Dejana E. VE-cadherin and endothelial adherens junctions: active guardians of vascular integrity. Dev Cell. 2013;26:441–54.CrossRefPubMed Giannotta M, Trani M, Dejana E. VE-cadherin and endothelial adherens junctions: active guardians of vascular integrity. Dev Cell. 2013;26:441–54.CrossRefPubMed
26.
Zurück zum Zitat Bergt S, Gruenewald M, Beltschany C, Grub A, Neumann T, Albrecht M, et al. The Fibrin-Derived Peptide Bβ15-42 (FX06) Ameliorates Vascular Leakage and Improves Survival and Neurocognitive Recovery: Implications From Two Animal Models of Cardiopulmonary Resuscitation. Crit Care Med. 2016;44:e988-995.CrossRefPubMed Bergt S, Gruenewald M, Beltschany C, Grub A, Neumann T, Albrecht M, et al. The Fibrin-Derived Peptide Bβ15-42 (FX06) Ameliorates Vascular Leakage and Improves Survival and Neurocognitive Recovery: Implications From Two Animal Models of Cardiopulmonary Resuscitation. Crit Care Med. 2016;44:e988-995.CrossRefPubMed
27.
Zurück zum Zitat RECOVERY Collaborative Group, Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, et al. Dexamethasone in Hospitalized Patients with Covid-19 - Preliminary Report. N Engl J Med. 2020; RECOVERY Collaborative Group, Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, et al. Dexamethasone in Hospitalized Patients with Covid-19 - Preliminary Report. N Engl J Med. 2020;
28.
Zurück zum Zitat Petzelbauer P, Zacharowski PA, Miyazaki Y, Friedl P, Wickenhauser G, Castellino FJ, et al. The fibrin-derived peptide Bbeta15-42 protects the myocardium against ischemia-reperfusion injury. Nat Med. 2005;11:298–304.CrossRefPubMed Petzelbauer P, Zacharowski PA, Miyazaki Y, Friedl P, Wickenhauser G, Castellino FJ, et al. The fibrin-derived peptide Bbeta15-42 protects the myocardium against ischemia-reperfusion injury. Nat Med. 2005;11:298–304.CrossRefPubMed
29.
Zurück zum Zitat Roesner JP, Petzelbauer P, Koch A, Mersmann J, Zacharowski PA, Boehm O, et al. The fibrin-derived peptide Bbeta15-42 is cardioprotective in a pig model of myocardial ischemia-reperfusion injury. Crit Care Med. 2007;35:1730–5.CrossRefPubMed Roesner JP, Petzelbauer P, Koch A, Mersmann J, Zacharowski PA, Boehm O, et al. The fibrin-derived peptide Bbeta15-42 is cardioprotective in a pig model of myocardial ischemia-reperfusion injury. Crit Care Med. 2007;35:1730–5.CrossRefPubMed
30.
Zurück zum Zitat Tagami T, Kushimoto S, Yamamoto Y, Atsumi T, Tosa R, Matsuda K, et al. Validation of extravascular lung water measurement by single transpulmonary thermodilution: human autopsy study. Crit Care. 2010;14:R162.CrossRefPubMedPubMedCentral Tagami T, Kushimoto S, Yamamoto Y, Atsumi T, Tosa R, Matsuda K, et al. Validation of extravascular lung water measurement by single transpulmonary thermodilution: human autopsy study. Crit Care. 2010;14:R162.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Wollborn J, Hassenzahl LO, Reker D, Staehle HF, Omlor AM, Baar W, et al. Diagnosing capillary leak in critically ill patients: development of an innovative scoring instrument for non-invasive detection. Ann Intensive Care. 2021;11:175.CrossRefPubMedPubMedCentral Wollborn J, Hassenzahl LO, Reker D, Staehle HF, Omlor AM, Baar W, et al. Diagnosing capillary leak in critically ill patients: development of an innovative scoring instrument for non-invasive detection. Ann Intensive Care. 2021;11:175.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat van de Weg CAM, Pannuti CS, van den Ham H-J, de Araújo ESA, Boas LSV, Felix AC, et al. Serum angiopoietin-2 and soluble VEGF receptor 2 are surrogate markers for plasma leakage in patients with acute dengue virus infection. J Clin Virol Off Publ Pan Am Soc Clin Virol. 2014;60:328–35.CrossRef van de Weg CAM, Pannuti CS, van den Ham H-J, de Araújo ESA, Boas LSV, Felix AC, et al. Serum angiopoietin-2 and soluble VEGF receptor 2 are surrogate markers for plasma leakage in patients with acute dengue virus infection. J Clin Virol Off Publ Pan Am Soc Clin Virol. 2014;60:328–35.CrossRef
33.
Zurück zum Zitat Clajus C, Lukasz A, David S, Hertel B, Lichtinghagen R, Parikh SM, et al. Angiopoietin-2 is a potential mediator of endothelial barrier dysfunction following cardiopulmonary bypass. Cytokine. 2012;60:352–9.CrossRefPubMedPubMedCentral Clajus C, Lukasz A, David S, Hertel B, Lichtinghagen R, Parikh SM, et al. Angiopoietin-2 is a potential mediator of endothelial barrier dysfunction following cardiopulmonary bypass. Cytokine. 2012;60:352–9.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Vassiliou AG, Keskinidou C, Jahaj E, Gallos P, Dimopoulou I, Kotanidou A, et al. ICU Admission Levels of Endothelial Biomarkers as Predictors of Mortality in Critically Ill COVID-19 Patients. Cells. 2021;10:186.CrossRefPubMedPubMedCentral Vassiliou AG, Keskinidou C, Jahaj E, Gallos P, Dimopoulou I, Kotanidou A, et al. ICU Admission Levels of Endothelial Biomarkers as Predictors of Mortality in Critically Ill COVID-19 Patients. Cells. 2021;10:186.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Spadaro S, Fogagnolo A, Campo G, Zucchetti O, Verri M, Ottaviani I, et al. Markers of endothelial and epithelial pulmonary injury in mechanically ventilated COVID-19 ICU patients. Crit Care. 2021;25:74.CrossRefPubMedPubMedCentral Spadaro S, Fogagnolo A, Campo G, Zucchetti O, Verri M, Ottaviani I, et al. Markers of endothelial and epithelial pulmonary injury in mechanically ventilated COVID-19 ICU patients. Crit Care. 2021;25:74.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Dupont A, Rauch A, Staessens S, Moussa M, Rosa M, Corseaux D, et al. Vascular Endothelial Damage in the Pathogenesis of Organ Injury in Severe COVID-19. Arterioscler Thromb Vasc Biol. 2021;41:1760–73.CrossRefPubMed Dupont A, Rauch A, Staessens S, Moussa M, Rosa M, Corseaux D, et al. Vascular Endothelial Damage in the Pathogenesis of Organ Injury in Severe COVID-19. Arterioscler Thromb Vasc Biol. 2021;41:1760–73.CrossRefPubMed
37.
Zurück zum Zitat Flemming S, Burkard N, Renschler M, Vielmuth F, Meir M, Schick MA, et al. Soluble VE-cadherin is involved in endothelial barrier breakdown in systemic inflammation and sepsis. Cardiovasc Res. 2015;107:32–44.CrossRefPubMed Flemming S, Burkard N, Renschler M, Vielmuth F, Meir M, Schick MA, et al. Soluble VE-cadherin is involved in endothelial barrier breakdown in systemic inflammation and sepsis. Cardiovasc Res. 2015;107:32–44.CrossRefPubMed
38.
Zurück zum Zitat Yu W-K, McNeil JB, Wickersham NE, Shaver CM, Bastarache JA, Ware LB. Vascular endothelial cadherin shedding is more severe in sepsis patients with severe acute kidney injury. Crit Care. 2019;23:18.CrossRefPubMedPubMedCentral Yu W-K, McNeil JB, Wickersham NE, Shaver CM, Bastarache JA, Ware LB. Vascular endothelial cadherin shedding is more severe in sepsis patients with severe acute kidney injury. Crit Care. 2019;23:18.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Loosen G, Conrad AM, Hagman M, Essert N, Thiel M, Luecke T, et al. Transpulmonary thermodilution in patients treated with veno-venous extracorporeal membrane oxygenation. Ann Intensive Care. 2021;11:101.CrossRefPubMedPubMedCentral Loosen G, Conrad AM, Hagman M, Essert N, Thiel M, Luecke T, et al. Transpulmonary thermodilution in patients treated with veno-venous extracorporeal membrane oxygenation. Ann Intensive Care. 2021;11:101.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Herner A, Lahmer T, Mayr U, Rasch S, Schneider J, Schmid RM, et al. Transpulmonary thermodilution before and during veno-venous extra-corporeal membrane oxygenation ECMO: an observational study on a potential loss of indicator into the extra-corporeal circuit. J Clin Monit Comput. 2020;34:923–36.CrossRefPubMed Herner A, Lahmer T, Mayr U, Rasch S, Schneider J, Schmid RM, et al. Transpulmonary thermodilution before and during veno-venous extra-corporeal membrane oxygenation ECMO: an observational study on a potential loss of indicator into the extra-corporeal circuit. J Clin Monit Comput. 2020;34:923–36.CrossRefPubMed
Metadaten
Titel
FX06 to rescue SARS-CoV-2-induced acute respiratory distress syndrome: a randomized clinical trial
verfasst von
Emmanuelle Guérin
Lisa Belin
Guillaume Franchineau
Loïc Le Guennec
David Hajage
Mamadou Hassimiou Diallo
Thomas Frapard
Lucie Le Fèvre
Charles-Edouard Luyt
Alain Combes
Stéphane Germain
Jan Hayon
Pierre Asfar
Nicolas Bréchot
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Critical Care / Ausgabe 1/2023
Elektronische ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-023-04616-1

Weitere Artikel der Ausgabe 1/2023

Critical Care 1/2023 Zur Ausgabe

Blutdrucksenkung schon im Rettungswagen bei akutem Schlaganfall?

31.05.2024 Apoplex Nachrichten

Der optimale Ansatz für die Blutdruckkontrolle bei Patientinnen und Patienten mit akutem Schlaganfall ist noch nicht gefunden. Ob sich eine frühzeitige Therapie der Hypertonie noch während des Transports in die Klinik lohnt, hat jetzt eine Studie aus China untersucht.

Ähnliche Überlebensraten nach Reanimation während des Transports bzw. vor Ort

29.05.2024 Reanimation im Kindesalter Nachrichten

Laut einer Studie aus den USA und Kanada scheint es bei der Reanimation von Kindern außerhalb einer Klinik keinen Unterschied für das Überleben zu machen, ob die Wiederbelebungsmaßnahmen während des Transports in die Klinik stattfinden oder vor Ort ausgeführt werden. Jedoch gibt es dabei einige Einschränkungen und eine wichtige Ausnahme.

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Hyponatriämie Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.

Häusliche Gewalt in der orthopädischen Notaufnahme oft nicht erkannt

28.05.2024 Häusliche Gewalt Nachrichten

In der Notaufnahme wird die Chance, Opfer von häuslicher Gewalt zu identifizieren, von Orthopäden und Orthopädinnen offenbar zu wenig genutzt. Darauf deuten die Ergebnisse einer Fragebogenstudie an der Sahlgrenska-Universität in Schweden hin.

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.