Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Research Article

Cirrhosis and Liver Failure

Granulocyte-colony stimulating factor (G-CSF) to treat


acute-on-chronic liver failure: A multicenter
randomized trial (GRAFT study)

Graphical abstract Authors

Cornelius Engelmann, Adam Herber,


Annegret Franke, ., Peter R. Galle,
Acute-on-chronic liver failure Granulocyte colony stimulating factor
Anett Schmiedeknecht, Thomas Berg

Coagulation failure
Correspondence
thomas.berg@medizin.uni-leipzig.
Brain failure de (T. Berg).

Lay summary
Liver failure Inefficacy Granulocyte-colony stimulating
factor was considered as a novel
treatment for acute-on-chronic
Cardiocirculatory
Respiratory failure
liver failure (ACLF). We performed
failure
the first randomized, multicenter,
Kidney failure controlled phase II trial, which
showed that G-CSF did not improve
survival or other clinical endpoints
in patients with ACLF. Therefore, G-
Highlights CSF should not be used to treat liver
 G-CSF was expected to be a novel therapy for acute-on-chronic disease outside clinical studies.
liver failure.
 In this first multicenter, randomized phase II trial, G-CSF did not
improve patient survival.

 G-CSF was unable to reduce the rate of complications and did not
improve organ function.

https://doi.org/10.1016/j.jhep.2021.07.033
© 2021 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved. J. Hepatol. 2021, 75, 1346–1354
Research Article
Cirrhosis and Liver Failure

Granulocyte-colony stimulating factor (G-CSF) to treat acute-on-


chronic liver failure: A multicenter randomized trial (GRAFT study)
Cornelius Engelmann1,2,3, Adam Herber1, Annegret Franke4, Tony Bruns5,6, Philipp Reuken6,
Ingolf Schiefke7, Alexander Zipprich8, Stefan Zeuzem9, Tobias Goeser10, Ali Canbay11,
Christoph Berg12, Jonel Trebicka9,13,14, Frank E. Uschner9,14, Johannes Chang14, Tobias Mueller2,
Niklas Aehling1, Moritz Schmelzle15, Katrin Splith15, Frank Lammert16, Christian M. Lange9,17,
Christoph Sarrazin9,18, Christian Trautwein5, Michael Manns19, Dieter Häussinger20,
Jan Pfeiffenberger21, Peter R. Galle22, Anett Schmiedeknecht4, Thomas Berg1,*
1
Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany; 2Department of Hepatology and
Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany; 3Berlin Institute of Health (BIH), Berlin, Germany; 4Clinical Trial
Centre (ZKS) Leipzig, Faculty of Medicine, University Leipzig, Leipzig, Germany; 5Department of Medicine III, Aachen University Hospital,
Aachen, Germany; 6Clinic for Internal Medicine IV, University Hospital Jena, Jena, Germany; 7Clinic for Gastroenterology, Hepatology and
Endocrinology, St. Georg Hospital, Leipzig, Germany; 8University Hospital for Internal Medicine 1, Martin-Luther-University Halle-Wittenberg,
Halle, Germany; 9Department of Internal Medicine I, University Hospital Frankfurt, Frankfurt, Germany; 10Clinic for Gastroenterology and
Hepatology, University Hospital Cologne, Cologne, Germany; 11Medical Department, University Hospital Ruhr-University Bochum, Bochum,
Germany; 12Department of Internal Medicine I, University Hospital Tuebingen, Tuebingen, Germany; 13European Foundation for the Study of
Chronic Liver Failure, Barcelona, Spain; 14Department of Internal Medicine I, University of Bonn, Bonn, Germany; 15Department of Surgery,
Campus Charité Mitte | Campus Virchow-Klinik, Charité - Universitätsmedizin Berlin, Berlin, Germany; 16Department of Medicine II, Saarland
University Medical Center, Homburg, Germany; 17Clinic for Gastroenterology and Hepatology, University Hospital Essen, Essen, Germany;
18
Medical Clinic 2, St. Josefs Hospital Wiesbaden, Wiesbaden, Germany; 19Department of Gastroenterology, Hepatology and Endocrinology,
Hannover Medical School, Hannover, Germany; 20Clinic and Policlinic of Gastroenterology, Hepatology and Infectious Disease, Heinrich Heine
University Duesseldorf, Düsseldorf, Germany; 21Department of Gastroenterology and Hepatology, University Hospital Heidelberg, Heidelberg,
Germany; 22Department of Internal Medicine, University Medical Center Mainz, Mainz, Germany

Background & Aims: Based on positive results from small single and HR 1.058; 95% CI 0.727–1.548; p = 0.768, respectively). G-CSF
center studies, granulocyte-colony stimulating factor (G-CSF) is did not improve liver function scores, the occurrence of in-
being widely used for the treatment of patients with acute-on- fections, or survival in subgroups of patients without infections,
chronic liver failure (ACLF). Herein, we aimed to evaluate the with alcohol-related ACLF, or with ACLF defined by the APASL
safety and efficacy of G-CSF in patients with ACLF. criteria. Sixty-one serious adverse events were reported in the G-
Methods: In this multicenter, prospective, controlled, open-label CSF+SMT group and 57 were reported in the SMT group. In total,
phase II study, 176 patients with ACLF (EASL-CLIF criteria) were 7 drug-related serious adverse reactions occurred in the G-CSF
randomized to receive G-CSF (5 lg/kg daily for the first 5 days group. The study was prematurely terminated due to futility
and every third day thereafter until day 26) plus standard after conditional power calculation.
medical therapy (SMT) (n = 88) or SMT alone. The primary effi- Conclusions: In contrast to previous findings, G-CSF had no
cacy endpoint was 90-day transplant-free survival analyzed by significant beneficial effect on patients with ACLF in this multi-
Cox regression modeling. The key secondary endpoints were center controlled trial, which suggests that it should not be used
overall and transplant-free survival after 360 days, the devel- as a standard treatment for ACLF.
opment of ACLF-related complications, and the course of liver ClinicalTrials.gov number: NCT02669680
function scores during the entire observation period. Lay summary: Granulocyte-colony stimulating factor was
Results: Patients treated with G-CSF had a 90-day transplant- considered as a novel treatment for acute-on-chronic liver failure
free survival rate of 34.1% compared to 37.5% in the SMT group (ACLF). We performed the first randomized, multicenter,
(hazard ratio [HR] 1.05; 95% CI 0.711–1.551; p = 0.805). controlled phase II trial, which showed that G-CSF did not
Transplant-free and overall survival at 360 days did not differ improve survival or other clinical endpoints in patients with
between the 2 arms (HR 0.998; 95% CI 0.697–1.430; p = 0.992 ACLF. Therefore, G-CSF should not be used to treat liver disease
outside clinical studies.
© 2022 The Authors. Published by Elsevier B.V. on behalf of European
Keywords: cirrhosis; inflammation; regeneration; stem cell mobilization; ACLF; Association for the Study of the Liver. This is an open access article
transplantation; organ failure.
Received 16 April 2021; received in revised form 8 July 2021; accepted 20 July 2021;
under the CC BY-NC-ND license (http://creativecommons.org/
available online 5 August 2021 licenses/by-nc-nd/4.0/).
* Corresponding author. Address: Division of Hepatology, Department of Medicine II,
Leipzig University Medical Center, Liebigstrasse 21, 04103 Leipzig, Germany; Introduction
Tel.: 00493419712330.
E-mail address: thomas.berg@medizin.uni-leipzig.de (T. Berg). Acute-on-chronic liver failure (ACLF) develops in more than
https://doi.org/10.1016/j.jhep.2021.07.033 30% of patients with acutely decompensated cirrhosis and is

Journal of Hepatology 2021 vol. 75 j 1346–1354


characterized by development of organ failures.1 Cirrhosis ac- syndrome, and vasopressor support) or white blood cell (WBC)
counts for about 1 million deaths worldwide and most of these count of >50 x 109/L were excluded (complete list of inclu-
patients die during an ACLF episode.2 Its overall 90-day mortality sion and exclusion criteria is included in the supplementary
is approximately 50%, which is substantially higher than the 14% information). All patients gave their written consent before
seen in patients with acute decompensation without organ fail- study inclusion.
ures.3 Liver transplantation is the only established treatment
approach,4,5 but due to scarcity of donor organs, costs and con- Intervention
traindications it is only accessible to a minority of patients Patients were randomized in a 1:1 ratio to receive either stan-
(approximately 5%).4 dard medical therapy (SMT) (SMT group) or recombinant G-CSF
Novel treatment approaches aim to target ACLF-specific (G-CSF, Ratiograstim®) plus SMT (G-CSF+SMT group). Patients
pathomechanisms, dominated by the co-existence of exagger- were centrally randomized with a Web-based system by using a
ated inflammatory responses on the one hand, and immune minimization algorithm with a factor for center and a probability
paralysis on the other, leading to mitochondrial dysfunction, cell of 0.2 for random allocation, to avoid potential loss of allocation
death and regenerative incapacity.6–9 Bone marrow-derived concealment within centers. Patients randomized to the G-
stem cells have been shown to be potent in modulating im- CSF+SMT group were scheduled to receive in total 12 doses of G-
mune functions and promoting regenerative capacities, thus CSF subcutaneously injected on a daily basis for the first 5 days
mitigating various types of liver injury in experimental and after randomization and every third day thereafter until day 26.
clinical studies.10–12 However, costs, high technical requirements The dosing scheme used corresponded to that published by Garg
and peri-interventional complications limit the general applica- et al.15 which was chosen based on its superior efficacy over a
bility of this approach. shorter treatment duration applied by Duan et al.16 Further
Granulocyte-colony stimulating factor (G-CSF) mobilizes he- randomized efficacy trials were not available at the time of trial
matopoietic stem and immune cells and represents an alterna- preparation. Injections were performed at 12 am (± 1 hour) on
tive for exogenous stem cell infusions.13,14 Several randomized every treatment day. Using pre-filled syringes provided by the
controlled trials in India and China using G-CSF in patients with supplier (30 Mio IU or 48 Mio IU), as part of the clinical routine,
ACLF reported improved survival and decreased rates of disease- G-CSF doses were guided by body weight using a cut-off value of
related complications, such as bacterial infections.15,16 Conse- 70 kg (< −70kg 30 Mio IU G-CSF, >70 kg 48 Mio IU G-CSF) (Fig. 1)
quently, G-CSF is being used as standard of care in many parts of (Supplementary CTAT Table).
the world. However, its role in treating ACLF is still under debate A placebo controlled and blinded design was not considered
as data supporting its efficacy have been tested in relatively feasible in the study population for 2 main reasons. First, pa-
small clinical trials outside of Europe and the US, and in a limited tients with ACLF can have pre-existing systemic inflammatory
number of centers. Therefore, the aim of our study was to responses and a high risk of infectious complications1; thus,
explore the efficacy of G-CSF in patients with ACLF in a large blinding could lead to misinterpretation of WBC count, as it will
multicenter, randomized and controlled trial. be unknown if an infection or G-CSF treatment causes the
response in WBC count. Alternative diagnostic approaches are
Patients and methods not reliable to detect infectious complications in cases of G-CSF-
Trial design related WBC increases.19 Second, according to the drug manu-
The GRAFT study was an investigator-initiated, multicenter, facturer’s instruction, G-CSF treatment was transiently stopped
randomized, controlled trial, which recruited patients with ACLF in cases where WBC rose to >70 x109/L and re-started at a WBC
in 18 German tertiary centers funded by the German research <50 x109/L, mainly because WBC exceeding 100 x109/L can be
foundation (DFG). The trial protocol (supplementary trial pro- accompanied by a high risk of symptomatic hyperviscosity syn-
tocol) was approved by the leading ethics committee (University drome.20 Any subsequent new ACLF episode, which developed
Leipzig) and by the German competent authority (BfArM). The after the qualifying ACLF episode for inclusion into the trial, was
trial design followed the CONSORT guidelines and complied with treated with SMT in both treatment arms.
the principals of the declaration of Helsinki from 1975 (Version
Sommerset West 1996),17 as well as all pertinent national laws
and the ICH guidelines for Good Clinical Practice (GCP) issued in G-CSF + SMT Prim. endpoint Long-term follow-up
June 1996 and CPMP/ICH/135/95 from September 1997. Enrolment
ACLF eligibility Rando Intervention: G-CSF 5 μg/kg s.c. day 1-5 once daily,
EF-CLIF criteria informed 1:1 day 6-26 every third day
(according to the body weight using a cut off value of 70 kg
consent
Study population (≤70 kg 30 Mio IU G-CSF, >70 kg 48 Mio IU G-CSF))

Patients with ACLF according to the criteria of the European SMT Prim. endpoint Long-term follow-up

Foundation for the Study of Chronic Liver Failure (EASL-CLIF)


consortium18 with underlying liver cirrhosis and aged >
−18 years Screening + randomisation Intervention Prim. endpoint Follow-up
48 hours 26 days 90 days 12 month
were eligible for the trial. ACLF was defined as acutely
decompensated cirrhosis (acute development or worsening of Baseline V1-V4 V5 V6 V7 V8

one of the following complications: ascites, hepatic encepha- [D1, D4, D7, D14] [D28] [D90] [D180] [D360]

lopathy, gastrointestinal hemorrhage, bacterial infection) Fig. 1. Study design. After enrolment patients with G-CSF were randomized to
combined with one or more organ failure(s) (definitions in receive either SMT or SMT+G-CSF for 26 days. The primary endpoint was
supplementary information).3 assessed after 90 days. Patients were followed up for in total 360 days. There
were 9 study visits (baseline, V1-V8) at which clinical data, adverse events and
Patients with prior non-curatively treated or active malig- blood samples were collected. ACLF, acute-on-chronic liver failure; D, day of
nancies, sickle cell disease, septic shock (defined by the study visit; G-CSF, granulocyte-colony stimulating factor; SMT, standard
presence of bacteriaemia, systemic inflammatory response medical therapy; V, study visit.

Journal of Hepatology 2021 vol. 75 j 1346–1354 1347


Research Article Cirrhosis and Liver Failure

Follow-up occurrence of disease-related complications in competing risk


After randomization patients were followed-up for 360 days or analyses (Fine and Gray proportional subdistribution hazard
until premature study termination defined as: regression model, R version 3.6.3). Additional analyses were
 withdrawal of informed consent (drop-out) performed in the per protocol (PP) population. These patients
 complete loss of contact (drop-out) fulfilled all inclusion and exclusion criteria and received at least
 death 75% of the intended G-CSF total dose, if medical reasons did not
result in premature end of treatment. Ninety-day transplant-free
During follow up visits V1-V8 clinical data and samples were survival analysis was also calculated post hoc in various sub-
collected as outlined in Fig. 1. populations. Detailed definitions are provided in the supple-
mentary information. Analyses were performed with SSPS
Endpoints software, version 25 (IBM inc.) and R (Supplementary CTAT
The primary endpoint was defined as transplant-free survival 90 Table). Safety issues were carefully monitored by standardized
days after inclusion into the study, with death and orthotopic SAE reporting and annual safety reports (according to legal re-
liver transplantation (OLT) counting as events. Patients were quirements), and presented to the DMSB. All results shown here
censored at the time point of last information still alive and were calculated from the intention-to-treat (ITT) population
without OLT. The key secondary endpoints were the overall and unless otherwise specified.
transplant-free survival after 360 days, the development of ACLF-
related complications, bacterial infections and the course of liver Results
function scores, such as the model of end-stage liver disease Patient and treatment characteristics
(MELD) score and Chronic Liver Failure-consortium (CLIF-C) or- After registering 180 patients in total, 176 were randomized be-
gan failure (OF) score, during the entire observation period. A tween March 2016 and April 2019 to receive either G-CSF with
complete list of endpoints and definitions is presented in the SMT or SMT alone (n = 88 G-CSF+SMT group, n = 88 SMT group)
supplementary information. We abstained from using the CLIF-C (Fig. 2). Table 1 summarizes the baseline characteristics for both
ACLF score18 for further analysis, because it includes WBC count groups, which did not differ significantly, indicating good
as part of the scoring system, making it susceptible to artificial randomization. All patients had underlying cirrhosis. The mean ±
WBC alteration by G-CSF-related cell mobilization. Safety was SD age was 54.4 ± 10.2 years in the G-CSF+SMT group and 57.1 ±
evaluated at each visit based on clinical data and physical ex- 9.6 years in the SMT group. The majority of patients enrolled
amination. Severe adverse events (SAEs) were reported within were male (56.8% G-CSF+SMT group, 69.3% SMT group) and had
24 hours after awareness and monitored until resolution. The an increased mean BMI >28 kg/m2. Most patients had ACLF grade
SAE reporting period started on day 0 and ended at day 28 1 or 2, with a mean number of organ failures of 1.7 ± 0.7 in the G-
except for newly developed malignancies. Annual safety analyses CSF+SMT group vs. 1.4 ± 0.6 in the SMT group. The mean baseline
were performed. Safety reports and data analyses were reviewed CLIF-C OF score and MELD score were 10.4 ± 1.9 and 24.4 ± 6.3 in
by the data monitoring and safety board (DMSB) as requested or the G-CSF+SMT group and 10.3 ± 2 and 24.5 ± 6.1 in the SMT
at least annually. group, respectively. Liver and kidney failures were the predom-
inant types of organ failure accounting for more than 60% of
Statistical analysis and sample size calculation cases in both groups. Alcohol abuse, bacterial infections and
Based on the CANONIC Study,3 we expected a transplant-free gastrointestinal bleeding were the most frequent precipitating
survival rate of 42% at day 90 with SMT. Two small single- events (Table 1). At baseline, 96.6% of patients in both groups had
center, randomized G-CSF trials in patients with ACLF reported ascites, and bacterial infections were present in 56.8% in the G-
significant effects at improving survival from 26% to 66% after 60 CSF+SMT group and 51.1% in the SMT group.
days15 and from 21% to 48% after 90 days.16 We aimed to detect In the G-CSF+SMT group, 83 patients (94.3%) received at least
an absolute difference of 20% (from 42% to 62%, HR 1.815) in 1 G-CSF injection whereas 5 patients remained untreated due to
transplant-survival at day 90. Assuming a 10% loss to follow-up, early death (n = 4) and other medical reasons (n = 1). The mean
we originally determined that 292 patients would be required to number of G-CSF injections was 8.6 ± 3.9 and the mean duration
have 90% power, at a 2-sided significance level of 5%. One interim of G-CSF treatment was 17.5 days ± 9.7. In total, 29 patients (33%)
analysis was scheduled once the primary endpoint was available received all 12 injections and 51 patients >
−9 injections. Therefore,
for 50% of patients, allowing for detection of marked superiority 80 patients in the G-CSF+SMT arm and 84 patients in the SMT
of G-CSF (of >
−33%) on the primary endpoint, with a power of at arm fulfilled criteria for the PP cohort. Patients treated with G-
least 80%. Multiplicity adjustment followed O’Brien,21 with CSF showed increasing WBC count during the treatment course,
nominal significance levels of p = 0.005 at interim analysis and whereas it remained unchanged in the SMT group (p
p = 0.048 at final analysis. All patients enrolled up to this point <0.001) (Fig. S1).
were followed up until regular end of observation or an
endpoint. Cox regression was used to estimate the treatment Patient outcomes
effect on hazard ratio (HR) scale with 2-sided 95% CIs provided in The primary endpoint, defined by death or OLT within 90 days
time-to-event endpoints. Two-sided chi-square or Fisher’s exact after study enrolment, was reached in 54 patients (61.4%) in the
tests were used to compare qualitative variables, as appropriate. G-CSF+SMT group vs. 51 patients (58.0%) in the SMT group from
Courses of prognostic scores (MELD; CLIF-C OF score) were the ITT cohort. Five patients from the G-CSF+SMT group and 9
analyzed by repeated-measures ANCOVA, imputing single in- from the SMT group received an OLT. The HR for the risk of death
between values by linear interpolation and adding the last or transplantation when treated with G-CSF was 1.05 (95% CI
value observed in cases of death, OLT or dropout. We considered 0.711–1.551; p = 0.805) (Fig. 3 and 4). The median transplant-free
OLT competing with death and death/OLT competing with survival was 35 days (95% CI 19.8–50.2) in the G-CSF+SMT group

1348 Journal of Hepatology 2021 vol. 75 j 1346–1354


Registered
n = 180

Screening failure n = 4
- n = 3 not fulfilling the inclusion criteria
[no ACLF acc. to EF-CLIF criteria]
- n = 1 no reason provided

Randomized
n = 176

SMT + G-CSF SMT


ITT n = 88 ITT n = 88

Drop-outs n = 10 Drop-outs n = 18
n = 7 until day 90 n = 11 until day 90
n = 3 after day 90 until day 360 n = 7 after day 90 until day 360
Types of drop-out Types of drop-out
n = 5 withdrawal of consent n = 14 withdrawal of consent
n = 3 lost to follow-up n = 4 lost to follow-up
n = 2 other reasons*

Outcome Day 90 Outcome Day 90


n = 27 alive in follow-up n = 26 alive in follow-up
n = 50 deaths without OLT n = 45 deaths without OLT
n = 4 OLT (n = 1 death after OLT) n = 6 OLT (n = 2 death after OLT)

Outcome Day 360 Outcome Day 360


n = 16 alive at end of observation n = 9 alive at end of observation
n = 57 deaths without OLT n = 52 deaths without OLT
n = 5 OLT (n = 1 death after OLT) n = 9 OLT (n = 3 death after OLT)

Fig. 2. CONSORT flow chart of patients. After registering 180 patients, 176 patients fulfilled the inclusion and exclusion criteria, were randomized (n = 88
patients in each arm) and analyzed according to the ITT analysis. Patients were followed up for 360 days. *Other reasons for dropouts were: treatment of bronchial
carcinoma n = 1 and medical decision n = 1; ACLF, acute-on-chronic liver failure; EF-CLIF, European Foundation for the Study of Chronic Liver Failure; ITT,
intention-to-treat (all patients within the full analysis set were treated as randomized); OLT, orthotopic liver transplantation; SMT, standard medical care.

and 34 days (95% CI 9.9–58.1) in the SMT group. Neither the transplant-free survival after 90-days and 0.0046 for overall
overall survival (HR 1.058; 95% CI 0.727–1.548; p = 0.768), nor survival, which led to the premature termination of patient
360-day transplant-free survival (HR 0.998; 95% CI 0.697–1.430; recruitment after consultation with the study’s DMSB.
p = 0.992) were statistically different between arms in the ITT
cohort. Similar results were observed in the PP analysis. The HR Secondary endpoints
for the primary endpoint was calculated as 1.034 (95% CI Fatalities occured predominently in patients with increasing
0.698–1.531; p = 0.869] (Fig. 4). For overall survival, the HR was MELD score during the treatment period until day 28 in both
1.059 (95% CI 0.724–1.548; p = 0.769) and for transplant-free groups (Fig. S5). The distribution of patients with increasing or
survival 0.998 (95% CI 0.694–1.436; p = 0.992) when treated decreasing MELD or CLIF-C OF score, did not differ between the 2
with G-CSF (Fig. 4). groups (ANCOVA analysis: significance between groups p = 0.884
During the study period most fatalities were caused by ACLF for MELD and p = 0.757 for CLIF-C OF score; Fig. 5 and S5).
or other liver-related complications in both arms (G-CSF+SMT: Patients who recovered from the first ACLF episode were
27/88 [30.7%]; SMT: 21/88 [22.7%]). Gastrointestinal bleeding followed up for development of subsequent ACLF episodes (ACLF
leading to death was more frequent in the SMT arm (G-CSF+SMT: recurrence). The risk of developing further ACLF episodes was
2/88 [2.3%]; SMT 8/88 [9.1%]). In total 8 deaths, 3 in the G- not different between the 2 groups (HR 1.94; 95% CI 0.83–4.53;
CSF+SMT arm and 5 in the SMT arm, were related to bacterial p = 0.13) (Fig. S4) although the number of patients with ACLF
infections (Table S2) recurrence randomized into the G-CSF+SMT group was slightly
As liver transplant and dropouts were competing with death, higher (65.2% vs. 34.8 %, p = 0.19) (Table S3). Groups of patients
we performed a competing risk analysis by calculating the cu- with or without ACLF recurrence were further characterized at
mulative incidence of death, which confirmed that G-CSF treat- baseline and at the time point of remission from the first ACLF
ment failed to show a significant benefit on survival in patients episode. Patients with ACLF recurrence more often had ACLF
with ACLF (HR 1.11; 95% CI 1.61–0.77) (Fig. S2). For futility grade 1 at baseline (87% vs. 55%, p = 0.019). At the time point of
analysis, we calculated a conditional power of 1-b=0.0266 for remission the same patients had higher values for MELD score

Journal of Hepatology 2021 vol. 75 j 1346–1354 1349


Research Article Cirrhosis and Liver Failure

Table 1. Baseline characteristics for the 2 study arms.


1.0
Parameter G-CSF+SMT SMT Treatment arm
group (n = 88) group (n = 88) G-CSF+SMT
SMT
0.8
General

Cumulative survival
Age (years) 54.4 ± 10.2 57.1 ± 9.6
Gender (w/m) 38 (43.2%)/50 27 (30.7%)/61 0.6
(56.8%) (69.3%)
BMI (kg/m2) 28.9 ± 4.8 28.8 ± 5.6 0.4
Mean arterial 79.9 ± 12 82.7 ± 13.2
pressure (mmHg) 0.2 HR 1.05 [95% CI 0.711; 1.551 (p = 0.805)]

Disease severity
ACLF grade* 0.0
Grade 1 39 (44.3%) 45 (51.1%) 0 15 30 45 60 75 90 105
Grade 2 37 (42.0%) 28 (31.8%) Time from baseline in days
Grade 3 12 (13.6%) 15 (17%) Patients at risk
Number of organ failures 1.7 ± 0.7 1.4 ± 0.6 G-CSF + SMT 88 61 42 35 29 28 27
CLIF-C OF score 10.4 ± 1.9 10.3 ± 2 SMT 88 54 43 35 31 28 26
MELD score 24.4 ± 6.3 24.5 ± 6.1
CLIF-C ACLF score 51.9 ± 8.7 51.2 ± 7.4 Fig. 3. 90-day transplant-free survival in ITT cohort. Out of 176 patients with
Organ failures ACLF, 88 were randomized to receive G-CSF+SMT (G-CSF+SMT group) or SMT
Liver failure 58 (65.9%) 56 (63.6%) only (SMT group). Death or liver transplantation were considered as events in a
Kidney failure 58 (65.9%) 57 (64.8%) Cox Regression analysis calculating HR for G-CSF therapy. ACLF, acute-on-
Brain failure 5 (5.7%) 5 (5.7%) chronic liver failure; G-CSF, granulocyte-colony stimulating factor; HR, haz-
Circulatory failure 8 (9.1%) 6 (6.8%) ard ratio; ITT, intention-to-treat; SMT, standard medical therapy.
Coagulation failure 22 (25%) 15 (17%)
Respiratory failure 2 (2.3%) 6 (6.8%)
Precipitating events# Bacterial infections occurred in 71 (80.8%) patients in the G-
Alcohol related 49 (55.7%) 46 (52.3%)
CSF+SMT group and 69 (78.4%) in the SMT group (p = 0.709). Of
Bacterial infection 36 (40.9%) 33 (37.5%)
Bleeding 13 (14.8%) 7 (8%)
those, 50 out of 88 (56.6%) in the G-CSF+SMT arm and 45 out of
Others 7 (8%) 8 (9.1%) 88 (51.1%) had an infection at baseline and 31 out of 88 (35.2%)
Unknown 16 (18.2%) 18 (20.5%) and 32 out of 88 (36.4%) in the respective treatment arms
Complications of cirrhosis developed an infection during the observational period. Urinary
Ascites 85 (96.6%) 85 (96.6%) tract infections, spontaneous bacterial peritonitis, pneumonia
Hepatorenal syndrome 57 (64.8%) 58 (65.9%) and blood stream infections were the most common sites of
Hepatic encephalopathy 55 (62.5%) 61 (69.3%)
infection during the study. There was a predominance of spon-
Bacterial infection at baseline 50 (56.8%) 45 (51.1%)
taneous bacterial peritonitis in the G-CSF+SMT arm and blood
Laboratory values
stream infections in the SMT arm. There were no major differ-
Bilirubin (mg/dl) 18.0 ± 12.0 18.9 ± 14.5
ALT (U/L) 76.0 ± 144.7 81.7 ± 167.0 ences in the rate of infections across the different ACLF grades in
AST (U/L) 139.0 ± 217.0 124.1 ± 136.5 comparison between both arms (Table S4). There were 6 fungal
GGT (U/L) 195.2 ± 287.5 165.0 ± 224.0 infections in each cohort with Candida being the most frequent
ALP (U/L) 150.7 ± 80.6 148.8 ± 116.4 causative pathogen (6 out of 12). Two patients (1 in each group)
Creatinine (mg/dl) 2.4 ± 1.6 2.4 ± 1.5
had a fungal peritonitis (Table S5).
Urea (mg/dl) 101.5 ± 60.9 115.3 ± 61.3
INR 2.2 ± 0.8 2.1 ± 1.0
There was no difference between both groups with respect to
Albumin (g/dl) 3.0 ± 0.7 3.0 ± 0.7 the development of other complications, such as gastrointestinal
C-reactive protein (mg/dl) 39.9 ± 29.2 41.3 ± 39.6 bleeding, hepatic encephalopathy, and hepatorenal syndrome
Procalcitonin (mg/L) 1.7 ± 2.1 1.4 ± 1.4 during the observation period (Table 2).
WBC count (109/L) 14.8 ± 12.4 11.2 ± 7.1
ACLF, acute-on-chronic liver failure; ALP, alkaline phosphatase; ALT, alanine amino-
transferase; AST, aspartate aminotransferase; CLIF-C, Chronic Liver Failure-con- Post hoc sensitivity analyses
sortium; G-CSF, granulocyte-colony stimulating factor; GGT, gamma Sensitivity analyses focused on subgroups of patients with
glutamyltransferase; INR, international normalized ratio; MELD, model for end-stage
liver disease; OF, organ failure; SMT, standard medical therapy.
different disease severities (Fig. 4A) (ACLF grades and types of
#
Precipitating events counted individually in patients with more than one precipi- organ failures), on subgroups of patients who were included in
tating event. previous G-CSF trials (Fig. 4B)15,22 (ACLF defined according to the
*These values represent the number of patients in every respective ACLF grade.
APASL criteria, ACLF due to an alcohol-related precipitating
Values were calculated based on individual parameters. However, patients were
included based on ACLF grade allocations provided by the study centers. Based on event) and on subgroups with a higher likelihood of treatment
back calculations, 2 patients in the SMT group and 1 patient in the G-CSF+SMT group response (such as patients without infections, 7-day survivors
did not fulfil the criteria for ACLF. Percentages shown here were calculated against and patients fulfilling criteria for best cases). In none of these
the overall number of patients included in the trial.
cohorts did G-CSF show a significant effect on transplant-free
survival after 90 days (Table S1).
(17.8 vs. 13.1, p = 0.002), bilirubin (13.5 vs. 7, p = 0.024) and C- Multivariate Cox regression analysis identified risk factors for
reactive protein (38.9 vs. 27.2; p = 0.004) and more often had mortality such as age (HR 1.043; 95% CI 1.019–1.066; p <0.001),
high-grade ascites (60.9% vs. 22.5%, p = 0.002) (Table S3). the ACLF severity grade at baseline (HR 2.249; 95% CI

1350 Journal of Hepatology 2021 vol. 75 j 1346–1354


A Kidney failure 0.724 (0.453; 1.156), p = 0.176

Liver failure 1.013 (0.634; 1.617), p = 0.958

ACLF grade 3 1.162 (0.511; 2.640), p = 0.720

ACLF grade 2 1.037 (0.544; 1.979), p = 0.911

ACLF grade 1 1.005 (0.547; 1.846), p = 0.911

Overall cohort (per protocol) 1.034 (0.698; 1.531), p = 0.869

Overall cohort (intention to treat) 1.050 (0.711; 1.551), p = 0.805

0.1 1 10

B
Best case 1.138 (0.417; 3.1), p = 0.801

ACLF-APASL criteria 1.233 (0.762; 1.963), p = 0.404

Alcohol related ACLF 1.047 (0.594; 1.843), p = 0.875

7-day survivors 0.901 (0.583; 1.39), p = 0.64

No infections 1.055 (0.521; 2.137), p = 0.883

Infection 1.415 (0.777; 2.575), p = 0.254

Overall cohort (per protocol) 1.034 (0.698; 1.531), p = 0.869

Overall cohort (intention to treat) 1.050 (0.711; 1.551), p = 0.805

0.1 1 10

Fig. 4. Forrest plot for risk of all sub-populations analyzed. (A) Cox regression analyses calculating the hazard ratio (HR) for G-CSF therapy in subgroups with
different ACLF severity grades (ACLF grade 1 n = 83, ACLF grade 2 n = 64, ACLF grade 3 n = 27) and organ failures (liver failure n = 114, kidney failure n = 115) were
performed to evaluate the impact of disease severity on treatment efficacy. G-CSF was unable to improve the 90-day transplant-free survival in any of the severity
subgroups. (B) Cox regression analyses adjusted for ACLF grades were performed to calculate the hazard ratio of death or transplant until 90 days of observation
in additional subgroups. Along with the ITT cohort and per protocol sets of patients, analyses were performed in a variety of subgroups of patients with pre-
defined clinical conditions of potentially different prognosis. Best case (n = 65) – patients who survived regular end of treatment without transplant for at least 14
days and received at least 75% of intended dose if applicable; ACLF-APASL criteria (n = 114) – patients who fulfilled the APASL criteria for ACLF; alcohol-related
ACLF (n = 95) – patients who had an alcohol-related precipitating event; 7-day survivors (n = 155) – patients who survived randomization for a minimum of 7
days; no infection (n = 73) – patients without detectable infection at inclusion; infection (n = 69) – patients with a bacterial infection as a precipitating event.
ACLF, acute-on-chronic liver failure; APASL, Asian Pacific Association for the Study of the Liver; G-CSF, granulocyte-colony stimulating factor; ITT, intention-
to-treat.

1.703–2.970; p <0.001) and the WBC count at baseline (HR 1.044; CSF-related adverse reactions were reported in 18 patients and 7
95% CI 1.02–1.066; p <0.001) whereas treatment with G-CSF, the serious adverse reactions were reported in 4 patients (Table S6).
presence of an alcohol-related precipitating event or bacterial The study treatment was paused in 3 out of 4 patients with
infection at baseline (precipitating events) did not impact on serious adverse reactions, one of which was related to a WBC
transplant-free survival (Table 3). count above the threshold of 70 x109/L. In total, G-CSF therapy
had to be transiently stopped due to a WBC increase in 8 pa-
Safety characteristics tients (9.1%).
Overall, 403 adverse events were reported from 80 patients in
the G-CSF+SMT group and 354 from 78 patients in the SMT Discussion
group. Sixty-one SAEs were reported in 54 patients from the G- This is the first large multicenter study evaluating G-CSF as a
CSF+SMT group, as well as 57 SAEs in 47 patients from the SMT potential novel therapy for patients with bona fide ACLF. Despite
group, including 37 and 36 deaths during the SAE reporting G-CSF having immune-modulatory and pro-regenerative capac-
period, respectively. ACLF was the most frequent cause of death ities,23 our study confirmed the overall dismal prognosis of ACLF
in both groups (n = 17/37 [46%] and 16/36 [44%], respectively, p = and failed to demonstrate superiority of G-CSF over SMT. Hence,
1.0), followed by hemorrhages (n = 6 [16%] and 8 [22%], respec- the data provided here are in strong disagreement with 2 single-
tively, p = 0.564), and infection-related deaths (n = 4 [11%] and center trials that showed a marked improvement in survival and
n = 5 [14%], respectively, p = 0.736). In the G-CSF+SMT group, G- reduced infectious complications after G-CSF therapy13,14; thus,

Journal of Hepatology 2021 vol. 75 j 1346–1354 1351


Research Article Cirrhosis and Liver Failure

trials, G-CSF reduced the number of infectious complications,


A DeltaOF-Score - Overall cohort
improved immune dysfunction by increasing the number of
10 G-CSF+SMT
dendritic cells whilst decreasing interferon-producing T-cells
DeltaOF-Score between baseline

SMT

p = 0.757 and enhanced the homing of CD34+ hematopoietic cells, which


5 are believed to promote regenerative capacities.14,15,24 However,
in our trial, G-CSF was neither able to reduce the number of
and day 28

infections or any other complication nor improve severity scores,


0
such as the MELD score and CLIF-C score significantly. This is in
line with recently published data in children with ACLF,25 and
-5 supports other studies that did not find any beneficial effect of G-
CSF.24,25 Notably, some pre-clinical studies have even shown a
worsening of liver injury upon G-CSF use in ACLF.26
-10 There are several reasons that may explain the discrepancies
B DeltaMELD - Overall cohort
in G-CSF treatment outcome observed in our and previous
20 single-center trials.15,16 A recent meta-analysis summarized all
G-CSF+SMT
SMT G-CSF studies which enrolled patients with severe alcoholic
DeltaMELD between baseline

10 p = 0.884 hepatitis and underlined the huge heterogeneity among studies


and pointed towards a conflict between Asian and European
results,26 suggesting that a selection bias might play a role.
and day 28

0
However, when we stratified our patients according to criteria
-10
that were used in the Asian studies, most importantly patients
with an alcohol-related precipitating event and ACLF according
-20
to the APASL criteria, we were not able to demonstrate superi-
ority of the G-CSF approach compared to SMT. In this context, it
has to be mentioned that the definition of an alcohol-related
-30
precipitating event was at investigators discretion and not part
Fig. 5. Development of CLIF-C OF score during the treatment course. The of the study protocol. It has to be assumed that this cohort
distribution of patients with increasing and decreasing (A) CLIF-C OF score and comprised predominantly patients with alcoholic hepatitis.
(B) MELD score (delta between baseline and day 28) was not different when A recent review has indicated already that disease severities
comparing the G-CSF (G-CSF+SMT; blue bars) arm and the control arm (SMT;
grey bars) (between groups effect: DeltaOF-score p = 0.757, DeltaMELD score
and patient selections were comparable among Asian and Eu-
p = 0.884). Difference between groups over time was calculated by ANCOVA. ropean studies.12 Furthermore, G-CSF’s inefficacy was confirmed
Missing values due to death or transplant were imputed by individuals’ last across the different ACLF severity grades and types of organ
value. ACLF, acute-on-chronic liver failure; CLIF-C, Chronic Liver Failure- failures. Overall, these results indicate that selection criteria and
consortium; G-CSF, granulocyte-colony stimulating factor; MELD, model for
the study design do not fully explain differences in efficacy data
end-stage liver disease; OF, organ failure; SMT, standard medical therapy.
among previous studies and ours.
It might be tempting to say that the high number of infections
our data do not support the use of G-CSF as a potential standard in our study has driven an overwhelming inflammatory
therapy for patients with ACLF.23 We showed that 90-day response, which was aggravated by G-CSF thereby inhibiting its
transplant-free survival (HR 1.05, p = 0.805) and overall sur- beneficial effect. And indeed, patients with ACLF recurrence after
vival (HR 1.058, p = 0.768) were unaffected by G-CSF adminis- recovery from the first ACLF episode had higher C-reactive pro-
tration, and this observation was also confirmed in the subgroup tein values and tend to be treated more often with G-CSF. The
of patients treated strictly according to the protocol (PP) with an discrepancy between a high number of overall infections
HR of 1.034 (p = 0.869) for 90-day transplant-free survival and of developing during the study and low number of infection-related
1.059 (p = 0.769) for overall survival. In smaller single-center deaths is intriguing but might be related to the fact that only

Table 2. Development of cirrhosis-related complications during the observational period.*


Complication Treatment group (G-CSF+SMT) Control group (SMT) p value
ACLF remission observed1 33/872 (37.9%) 30/862 (34.9%) 0.685
ACLF re-episode3 15/87 (17.2%) 8/862 (9.3%) 0.134
Ascites 87 (98.9%) 87 (98.9%) 1.0004
Hepatic encephalopathy 64 (72.7%) 67 (76.1%) 0.6044
Hepatorenal syndrome 71 (80.7%) 69 (78.4%) 0.7094
Bacterial infections 71 (80.8%) 69 (78.4%) 0.7094
Variceal bleeding 7 (8%) 7 (8%) 1.0004
ACLF, acute-on-chronic liver failure; G-CSF, recombinant granulocyte colony stimulating factor; SMT, standard medical therapy.
*Groups were balanced regarding history of complications.
1
Remission before an endpoint-related/competing event or end of individual observational period.
2
Only patients with ACLF grade of > −1 at baseline were included.
3
ACLF re-episodes were considered only in case of a previous ACLF remission.
4
Group comparisons were performed by Chi-Square test.
5
Competing risk analysis by cumulative incidence function and hazard ratio.

1352 Journal of Hepatology 2021 vol. 75 j 1346–1354


Table 3. Multivariate Cox regression analysis for 90-day transplant-free survival.
Multivariate analysis
Baseline factor Hazard ratio 95% CI p value
Treatment arm (risk for G-CSF+SMT) 0.946 0.635–1.408 0.785
Age (years) 1.043 1.019–1.066 <0.001
ACLF grade 2.249 1.703–2.970 <0.001
Precipitating event bacterial infection 1.023 0.938–1.117 0.604
Precipitating event alcohol related 1.041 0.943–1.149 0.426
WBC count (109/L) 1.044 1.022–1.066 <0.001
ACLF, acute-on-chronic liver failure; G-CSF, recombinant granulocyte colony stimulating factor; SMT, standard medical therapy; WBC, white blood cell.

primary causes of death were documented by investigators and collection of acute precipitating events, which we considered as
data on a potential link between cause of death and underlying important determinants of the disease course.
infection was not systematically collected. This multicenter randomized study confirms the dismal
Stimulating the immune response may be an effect also prognosis of patients with ACLF and failed to demonstrate any
related to G-CSF per se independent of infections as shown by beneficial effect of treating this condition with G-CSF mono-
several pre-clinical studies. The rational for treating ACLF with therapy. G-CSF was associated with drug-related serious adverse
GCSF comes from animal models showing pro-regenerative and reactions and should not be used outside clinical trials for the
anti-inflammatory effects,27,28 and the earlier Asian studies treatment of ACLF.
seemed to confirm these findings in humans.15,16 It has to be
emphasized that G-CSF may act like a double-edged sword. Toll- Abbreviations
like receptor 4 (TLR4)-activation was observed to be a major ACLF, acute-on-chronic liver failure; AE, adverse events; APASL,
driver of disease progression in ACLF.29,30 As shown in septic Asian Pacific Association for the Study of the Liver; CLIF-C,
liver injury models, G-CSF might enhance sensitivity to endo- Chronic Liver Failure-consortium; DMSB, data monitoring and
toxins and increase hepatocyte uptake of lipopolysaccharide safety board; EASL-CLIF, European Foundation for the Study of
through the lipopolysaccharide-binding protein/TLR4 axis, Chronic Liver Failure; G-CSF, granulocyte-colony stimulating
thereby aggravating tissue injury, as shown by HMGB1 trans- factor; HR, hazard ratio; ITT, intention-to-treat; MELD, model of
location and mortality in rat models of septic liver injury.31,32 G- end-stage liver disease score; OF, organ failure; OLT, orthotopic
CSF also activates neutrophils that carry TLR433 and migrate into liver transplantation; PP, per protocol; SAEs, severe adverse
injured tissues, potentially causing additional cellular damage.34 events; SMT, standard medical therapy; TLR4, Toll-like receptor
Therefore, the effect of G-CSF, either beneficial or detrimental, 4; WBC, white blood cell.
might depend on the disease stage. In our trial, the number of
possibly drug-related SAEs and the trend that patients with ACLF Financial support
recurrence were more often treated with G-CSF points towards a The German Research Foundation (DFG) – EN 1100/1-1, funded
potential clinical risk when using G-CSF, although these data the study. Cornelius Engelmann is participant in the BIH-Charité
were purely descriptive. The comparison with previous trials is Clinician Scientist Program funded by the Charité – Uni-
difficult as WBC-related or drug-related severe complications versitätsmedizin Berlin and the Berlin Institute of Health. Moritz
were not reported.15,16 Schmelzle and Katrin Splith received grants for ancillary studies
The study presented here has several limitations. Firstly, pa- funded by the German Research Foundation (DFG) (SCHM2661/
tient recruitment had to be prematurely terminated due to fu- 3-1, SCHM2661/3-2). Jonel Trebicka is supported by grants from
tility after performance of conditional power analyses (in a the German Research Foundation (DFG) (SFB TRR57 to P18),
variety of sub-populations) in parallel to the planned interim European Union’s Horizon 2020 Research and Innovation Pro-
analysis. A calculated conditional power of 0.0266 for the pri- gramme (Galaxy, No. 668031 and MICROB-PREDICT, No. 825694),
mary endpoint and <0.005 for overall survival made it unethical and Societal Challenges - Health, Demographic Change and
to continue the publicly funded trial although there was no Wellbeing (No. 731875), and Cellex Foundation (PREDICT).
safety concern. We have also not provided any data about the
mode of action of G-CSF in ACLF. We did not specifically address
Conflict of interest
potential ethnic differences which could be a potential issue.
CE: has on-going research collaboration with Merz Pharmaceu-
However, so far there is no evidence to suggest that G-CSF itself
tical and Novartis. He has received speaker fees from Novartis,
acts differently or that the ethnic background of patients in-
Gilead and Merz Pharmaceuticals. TB: Receipt of honoraria or
fluences ACLF’s phenotype beyond the different diagnostic
consultation fees or participation in a company sponsored
criteria applied in Asia and Europe. Open labeled trials are al-
speaker’s bureau: Abbvie, Alexion, Bayer, Gilead, Eisai, Falk
ways at risk of bias. However, given the high number of positive
Foundation, Intercept, Ipsen, Janssen, MSD/Merck, Novartis, and
previous study results one would expect an overestimation of a
Sequana Medical. Receipt of grants/research supports: Abbvie,
therapeutic effect rather than failing to proof the benefit of G-
BMS, Gilead, MSD/Merck, Humedics, Intercept, Merz, Sequana
CSF. We therefore consider a bias of the effects associated with
Medical. MS: Receipt of honoraria or consultation fees or
the study design to be very unlikely. Unfortunately, we were not
participation in a company sponsored speaker’s bureau: Merck
collecting data on the etiology of the underlying chronic liver
Serono GmbH, Bayer AG, ERBE Elektromedizin GmbH, Amgen
disease. That was related to the fact that our focus was on a clear
Inc., Johnson&Johnson Medical GmbH, ERBE Elektromedizin
diagnosis of ACLF according to the EASL-CLIF criteria and
GmbH, Takeda Pharmaceutical Limited, Olympus K.K., Medtronic

Journal of Hepatology 2021 vol. 75 j 1346–1354 1353


Research Article Cirrhosis and Liver Failure

GmbH, Intuitive Surgical Inc.. JT: has received speaking and/or vein embolization enhances functional hepatic reserves after extended
consulting fees from Gore, Bayer, Alexion, MSD, Gilead, Intercept, right hepatectomy: a retrospective single-center study. Ann Surg
2012;255:79–85.
Norgine, Grifols, Versantis, and Martin Pharmaceutical. All other [12] Engelmann C, Martino VD, Kerbert AJC, Weil-Verhoeven D, Aehling NF,
authors declared no conflict of interest. Herber A, et al. The current status of granulocyte-colony stimulating
Please refer to the accompanying ICMJE disclosure forms for factor to treat acute-on-chronic liver failure. Semin Liver Dis 2021.
further details. [13] Demetri GD, Griffin JD. Granulocyte colony-stimulating factor and its re-
ceptor. Blood 1991;78:2791–2808.
[14] Engelmann C, Splith K, Berg T, Schmelzle M. Effects of granulocyte-
Authors’ contributions colony stimulating factor (G-CSF) on stem cell mobilization in patients
CE, TB, MS, KS, AF and AS were substantially involved the with liver failure. Eur J Intern Med 2016;36:e37–e39.
conception and design of the project. CE, TB and AF were [15] Garg V, Garg H, Khan A, Trehanpati N, Kumar A, Sharma BC, et al. Gran-
involved in data analysis and interpretation. All authors were ulocyte colony-stimulating factor mobilizes CD34(+) cells and improves
survival of patients with acute-on-chronic liver failure. Gastroenterology
involved in data acquisition, revised the manuscript critically, 2012;142:505–512 e501.
approved the final version to be published and agreed on the [16] Duan XZ, Liu FF, Tong JJ, Yang HZ, Chen J, Liu XY, et al. Granulocyte-colony
accuracy and integrity of data presented here. stimulating factor therapy improves survival in patients with hepatitis B
virus-associated acute-on-chronic liver failure. World J Gastroenterol
2013;19:1104–1110.
Data availability statement
[17] Shephard DA. The 1975 declaration of Helsinki and consent. Can Med
Data are available upon request from the corresponding author. Assoc J 1976;115:1191–1192.
[18] Jalan R, Saliba F, Pavesi M, Amoros A, Moreau R, Gines P, et al. Devel-
opment and validation of a prognostic score to predict mortality in pa-
Acknowledgement tients with acute-on-chronic liver failure. J Hepatol 2014;61:1038–1047.
We would like to thank Laura A. Kehoe for her excellent English [19] Pieri G, Agarwal B, Burroughs AK. C-reactive protein and bacterial infec-
language editing. We also like to acknowledge Prof. Reiner Wiest, tion in cirrhosis. Ann Gastroenterol 2014;27:113–120.
Prof. Tilmann Sauerbruch and Prof. Walter Lehmacher for their [20] Malkan UY, Ozcebe OI. Leukapheresis do not improve early death rates in
acute myeloid leukemia patients with hyperleukocytosis. Transfus Apher
deliberate and critical contribution as members of the DMSB.
Sci 2017;56:880–882.
[21] O’Brien PC, Fleming TR. A multiple testing procedure for clinical trials.
Supplementary data Biometrics 1979;35:549–556.
Supplementary data to this article can be found online at https:// [22] Singh V, Sharma AK, Narasimhan RL, Bhalla A, Sharma N, Sharma R.
Granulocyte colony-stimulating factor in severe alcoholic hepatitis: a
doi.org/10.1016/j.jhep.2021.07.033.
randomized pilot study. Am J Gastroenterol 2014;109:1417–1423.
[23] Sarin SK, Kedarisetty CK, Abbas Z, Amarapurkar D, Bihari C, Chan AC, et al.
References Acute-on-chronic liver failure: consensus recommendations of the asian
Author names in bold designate shared co-first authorship pacific association for the study of the liver (APASL) 2014. Hepatol Int
2014;8:453–471.
[1] Arroyo V, Moreau R, Jalan R. Acute-on-Chronic liver failure. N Engl J Med [24] Khanam A, Trehanpati N, Garg V, Kumar C, Garg H, Sharma BC, et al.
2020;382:2137–2145. Altered frequencies of dendritic cells and IFN-gamma-secreting T cells
[2] Pimpin L, Cortez-Pinto H, Negro F, Corbould E, Lazarus JV, Webber L, et al. with granulocyte colony-stimulating factor (G-CSF) therapy in acute-on-
Burden of liver disease in Europe: epidemiology and analysis of risk fac- chronic liver failure. Liver Int 2014;34:505–513.
tors to identify prevention policies. J Hepatol 2018;69:718–735. [25] Sharma S, Lal SB, Sachdeva M, Bhatia A, Varma N. Role of granulocyte
[3] Moreau R, Jalan R, Gines P, Pavesi M, Angeli P, Cordoba J, et al. Acute-on- colony stimulating factor on the short-term outcome of children with
chronic liver failure is a distinct syndrome that develops in patients with acute on chronic liver failure. J Clin Exp Hepatol 2020;10:201–210.
acute decompensation of cirrhosis. Gastroenterology 2013;144:1426-1437, [26] Marot A, Singal AK, Moreno C, Deltenre P. Granulocyte colony-stimulating
1437 e1421-1429. factor for alcoholic hepatitis: a systematic review and meta-analysis of
[4] Artru F, Louvet A, Ruiz I, Levesque E, Labreuche J, Ursic-Bedoya J, et al. randomised controlled trials. JHEP Rep 2020;2:100139.
Liver transplantation in the most severely ill cirrhotic patients: a multi- [27] Piscaglia AC, Shupe TD, Oh SH, Gasbarrini A, Petersen BE. Granulocyte-
center study in acute-on-chronic liver failure grade 3. J Hepatol colony stimulating factor promotes liver repair and induces oval cell
2017;67:708–715. migration and proliferation in rats. Gastroenterology 2007;133:619–631.
[5] Engelmann C, Thomsen KL, Zakeri N, Sheikh M, Agarwal B, Jalan R, et al. [28] Theocharis SE, Margeli AP, Kittas CN. Effect of granulocyte colony-
Validation of CLIF-C ACLF score to define a threshold for futility of stimulating-factor administration on tissue regeneration due to
intensive care support for patients with acute-on-chronic liver failure. Crit thioacetamide-induced liver injury in rats. Dig Dis Sci 1999;44:1990–1996.
Care 2018;22:254. [29] Engelmann C, Sheikh M, Sharma S, Kondo T, Loeffler-Wirth H, Zheng YB,
[6] Albillos A, Lario M, Alvarez-Mon M. Cirrhosis-associated immune et al. Toll-like receptor 4 is a therapeutic target for prevention and
dysfunction: distinctive features and clinical relevance. J Hepatol treatment of liver failure. J Hepatol 2020;73:102–112.
2014;61:1385–1396. [30] Engelmann C, Adebayo D, Oria M, De Chiara F, Novelli S, Habtesion A,
[7] Moreau R, Claria J, Aguilar F, Fenaille F, Lozano JJ, Junot C, et al. Blood et al. Recombinant alkaline phosphatase prevents acute on chronic liver
metabolomics uncovers inflammation-associated mitochondrial dysfunc- failure. Sci Rep 2020;10:389.
tion as a potential mechanism underlying ACLF. J Hepatol 2020;72:688–701. [31] Fang H, Jin H, Hua C, Liu A, Song Z, Chen X, et al. The LPS responsiveness in
[8] Engelmann C, Becker C, Boldt A, Herta T, Boehlig A, Splith K, et al. Ascites’ BN and LEW rats and its severity are modulated by the liver. J Immunol
neutrophil function is significantly impaired in patients with decom- Res 2018;2018:6328713.
pensated cirrhosis but can be restored by autologous plasma incubation. [32] Fang H, Liu A, Sun J, Kitz A, Dirsch O, Dahmen U. Granulocyte colony
Sci Rep 2016;6:37926. stimulating factor induces lipopolysaccharide (LPS) sensitization via
[9] Engelmann C, Claria J, Szabo G, Bosch J, Bernardi M. Pathophysiology of upregulation of LPS binding protein in rat. PloS One 2013;8:e56654.
decompensated cirrhosis: portal hypertension, circulatory dysfunction, [33] Acorci-Valerio MJ, Bordon-Graciani AP, Dias-Melicio LA, de Assis Golim M,
inflammation, metabolism and mitochondrial dysfunction. J Hepatol Nakaira-Takahagi E, de Campos Soares AM. Role of TLR2 and TLR4 in
2021;75(Suppl 1):S49–S66. human neutrophil functions against Paracoccidioides brasiliensis. Scand J
[10] Li N, Zhang L, Li H, Fang B. Human CD34+ cells mobilized by granulocyte Immunol 2010;71:99–108.
colony-stimulating factor ameliorate radiation-induced liver damage in [34] Fiuza C, Salcedo M, Clemente G, Tellado JM. Granulocyte colony-
mice. Stem Cell Res Ther 2010;1:22. stimulating factor improves deficient in vitro neutrophil trans-
[11] am Esch JS, Schmelzle M, Furst G, Robson SC, Krieg A, Duhme C, et al. endothelial migration in patients with advanced liver disease. Clin Diagn
Infusion of CD133+ bone marrow-derived stem cells after selective portal Lab Immunol 2002;9:433–439.

1354 Journal of Hepatology 2021 vol. 75 j 1346–1354

You might also like