PreprintPDF Available

Phase 3 Trial of Recombinant Human Alkaline Phosphatase for Patients with Sepsis-Associated Acute Kidney Injury (REVIVAL)

Authors:

Abstract

Purpose: Ilofotase alfa is a human recombinant alkaline phosphatase with reno-protective effects that showed improved survival and reduced MAKE90 in sepsis-associated acute kidney injury (SA-AKI) patients. ‘REVIVAL’, was aphase 3 trial, conducted to confirm its efficacy and safety. Methods: In this international double-blinded randomized-controlled trial, SA-AKI patients were enrolled <72 hours on vasopressor and <24 hours of AKI. The primary endpoint was 28-day all-cause mortality. The key secondary endpoint was Major Adverse Kidney Events up to day 90 (MAKE90). Results: 650 patients were treated and analyzed for safety; and 649 for efficacy data (ilofotase alfa n=330; placebo n=319). The observed mortality rates in the ilofotase alfa and placebo groups were 27.9% and 27.9% (nominal one-sided p-value of 0.50) at 28 days, and 33.9% and 34.8% (p=0.41) at 90 days. The trial was stopped for futility on the primary endpoint. The observed proportion of patients with MAKE90 was 56.7% in the ilofotase alfa group vs. 64.6% in the placebo group (p=0.02), mainly due to the number of patients who received renal replacement therapy (28.2% vs. 36.4%). There was evidence of heterogeneity of treatment effect with a marked reduction in MAKE90 events in patients with pre-existent impaired renal function randomized to ilofotase alfa (p=0.024). Adverse events were reported in 67.9% and 75.0% patients in the ilofotase and placebo group. Conclusion: Among critically ill patients with SA-AKI, ilofotase alfa did not improve day 28 survival. There may however be reno-protective properties, especially among patients with pre-existing renal disease. No safety concerns were identified. Trial registration and date of registration: ClinicalTrials.gov number NCT04411472, May-28-2020
Page 1/21
Phase 3 Trial of Recombinant Human Alkaline
Phosphatase for Patients with Sepsis-Associated
Acute Kidney Injury (REVIVAL)
Peter Pickkers ( Peter.Pickkers@radboudumc.nl )
University Medical Centre St Radboud Nijmegen https://orcid.org/0000-0002-1104-4303
Derek Angus
University of Pittsburgh School of Medicine
Kristie Bass
: D&A Pharma
Rinaldo Bellomo
Austin Hospital
Erik van den Berg
: D&A Pharma
Juliane Bernholz
: D&A Pharma
Morten H Bestle
Copenhagen University Hospital Glostrup: Rigshospitalet Glostrup
Kent Doi
University of Tokyo: Tokyo Daigaku
Christopher Doig
University of Calgary Cumming School of Medicine
Ricard Ferrer
Vall d'Hebron Research Institute: Vall d'Hebron Institut de Recerca
Bruno Francois
INSERM
Henrik Gammelager
Aarhus Universitetshospital
Ulf Goettrup
Zealand University Hospital Koge: Sjaellands Universitetshospital Koge
Eric Hoste
Ghent University: Universiteit Gent
Susanne Iversen
Slagelse Hospital: Slagelse Sygehus
Michael Joannidis
Page 2/21
Medical University of Innsbruck: Medizinische Universitat Innsbruck
John Kellum
Spectral Solutions
Kathleen Liu
University of California San Francisco Department of Medicine
Melanie Meersch
University Hospital Munster: Universitatsklinikum Munster
Ravindra Mehta
UC San Diego: University of California San Diego
Scott Millington
University of Ottawa
Patrick Murray
University College Dublin
Alistair Nichol
University College Dublin
Marlies Ostermann
Guy's and Saint Thomas' Hospitals NHS Trust: Guy's and St Thomas' NHS Foundation Trust
Ville Pettila
University of Helsinki: Helsingin Yliopisto
Christopher Solling
Viborg Regional Hospital: Regionshospitalet Viborg
Matthias Winkel
: D&A Pharma
Paul Young
Wellington Hospital Library: University of Otago Wellington
Alexander Zarbock
University Hospital Munster: Universitatsklinikum Munster
Research Article
Keywords: Sepsis, Acute Kidney Injury, Chronic Kidney Disease, MAKE90
Posted Date: August 2nd, 2023
DOI: https://doi.org/10.21203/rs.3.rs-3210421/v1
License: This work is licensed under a Creative Commons Attribution 4.0 International License. 
Read Full License
Page 3/21
Abstract
Purpose: Ilofotase alfa is a human recombinant alkaline phosphatase with reno-protective effects that
showed improved survival and reduced MAKE90 in sepsis-associated acute kidney injury (SA-AKI)
patients. REVIVAL, was aphase 3 trial, conducted to conrm its ecacy and safety.
Methods: In this international double-blinded randomized-controlled trial, SA-AKI patients were enrolled
<72 hours on vasopressor and <24 hours of AKI. The primary endpoint was 28-day all-cause mortality.
The key secondary endpoint was Major Adverse Kidney Events up to day 90 (MAKE90).
Results: 650 patients were treated and analyzed for safety; and 649 for ecacy data (ilofotase alfa
n=330; placebo n=319). The observed mortality rates in the ilofotase alfa and placebo groups were 27.9%
and 27.9% (nominal one-sided p-value of 0.50) at 28 days, and 33.9% and 34.8% (p=0.41) at 90 days. The
trial was stopped for futility on the primary endpoint. The observed proportion of patients with MAKE90
was 56.7% in the ilofotase alfa group vs. 64.6% in the placebo group (p=0.02), mainly due to the number
of patients who received renal replacement therapy (28.2% vs. 36.4%). There was evidence of
heterogeneity of treatment effect with a marked reduction in MAKE90 events in patients with pre-existent
impaired renal function randomized to ilofotase alfa (p=0.024). Adverse events were reported in 67.9%
and 75.0% patients in the ilofotase and placebo group.
Conclusion: Among critically ill patients with SA-AKI, ilofotase alfa did not improve day 28 survival. There
may however be reno-protective properties, especially among patients with pre-existing renal disease. No
safety concerns were identied.
Trial registration and date of registration: ClinicalTrials.gov number NCT04411472, May-28-2020
Take-home message
Sepsis-associated acute kidney injury in patients admitted to an intensive care unit is associated with
signicant morbidity and mortality. There is currently no pharmaceutical treatment. Although we found
no evidence that ilofotase alfa improved survival, its use reduced major adverse kidney events (mortality,
new onset, RRT, >25% reduction in eGFR, or rehospitalization) up to 90 days. The reduction in such events
was most marked in patient with pre-existent renal impairment.
140-character Tweet
Ilofotase alfa did not reduce mortality but showed a consistent pattern of reno-protection in patients with
sepsis-associated AKI.
Introduction
Sepsis is the leading cause of acute kidney injury (AKI) in critically ill patients as a consequence of
inammatory, direct nephrotoxic, and ischemic processes[1–4]. Development of AKI during sepsis (SA-
Page 4/21
AKI) is independently associated with increased morbidity and mortality[5, 6]. Patients with SA-AKI are at
risk of developing chronic kidney disease (CKD) resulting in a considerable burden for patients and
society[7]. Conversely, underlying CKD markedly increases the risk of AKI and the risk increases
proportionally with severity of CKD[8]. Pre-existent CKD complicated by AKI is common in critically ill
patients and associated with delayed kidney function recovery and increased risk of rehospitalization and
development of end stage renal disease[9].
There are no pharmacological therapies approved for the treatment of SA-AKI. Management consists of
only secondary prevention and supportive care strategies, such as uids and renal replacement therapy
RRT[5, 10]. Alkaline phosphatase (ALP) is an endogenous detoxifying enzyme that plays a signicant role
in host defense and innate immunity, particularly acting as an endogenous anti-inammatory protein[11,
12]. For example, removal of one of two phosphate groups abolishes the biological activity of endotoxin;
the dephosphorylated endotoxin acts as a toll-like receptor 4 (TLR4) antagonist[13]. Furthermore, ALP
dephosphorylates extracellular adenosine triphosphate (ATP), that is pro-inammatory, resulting in
generation of adenosine, which has anti-inammatory and tissue protective effects. In particular, the
kidney is negatively affected by increased levels of ATP while adenosine has reno-protective effects[14].
Hence, dephosphorylation attenuates the inammatory response and exerts tissue-protective
properties[15]. In animal models of sepsis, ALP administration dampens inammation and reduces
mortality[12, 16], but also protects against ischemia-reperfusion injury[17–19].
To augment therapeutic ecacy, human recombinant ALP, named ilofotase alfa, was developed,
consisting of an intestinal ALP sequence (highest biological activity) and a crown domain corresponding
with placental ALP sequence (to enhance stability)[20]. Dephosphorylation properties of ilofotase alfa
were conrmed[20] and considered to reduce systemic and local inammation and attenuate organ
damage[14]. Two small phase 2 studies with bovine ALP[15, 21] demonstrated reduced urinary excretion
of tubular injury markers and more pronounced improvement of endogenous creatinine clearance. In
addition to renal protective effects, a survival benet was observed in a large phase 2 trial[22]. The
REVIVAL study aimed to conrm the effect of ilofotase alfa on 28-day all-cause mortality in critically ill
patients with SA-AKI; major adverse kidney events by 90 days (MAKE90) was the key secondary endpoint.
Materials and methods
Ethics and dissemination
This trial was conducted in accordance with the protocol and consensus ethical principles of
international guidelines including the Declaration of Helsinki, Council for International Organizations of
Medical Sciences (CIOMS) International Ethical Guidelines, and International Conference on
Harmonization (ICH) Good Clinical Practice (GCP) Guidelines. The protocol, the single substantial
protocol amendment and other relevant documents were reviewed and approved by the Institutional
Research Board (IRB) / Institutional Ethics Committee (IEC) in the relevant centers prior to being used in
the trial.
Page 5/21
Trial design
This was a phase 3, multi-center, randomized, double-blind, placebo-controlled, 2-arm parallel-group-
sequential design trial in which patients with SA-AKI were randomly assigned in a 1:1 ratio to ilofotase
alfa or matching placebo in Europe, North America, Australia, New Zealand, and Japan.
Patients, randomization, and study medication
The population studied were adult patients with sepsis and recent onset AKI requiring vasopressor
support. The complete inclusion and exclusion criteria are described in supplemental Table1. Full details
on population and study design (including the protocol amendment) and conduct were previously
published[23]. Briey, three patient cohorts were dened: i) patients with SA-AKI with a pre-AKI reference
estimated glomerular ltration rate (eGFR)  45 mL/min/1.73 m2 and no proven or suspected COVID-19
at time of randomization (‘main trial population’); ii) patients with a pre-AKI reference eGFR  25 and < 45
mL/min/1.73 m2 (‘moderate-to-severe CKD population or mCKD’) with sepsis and AKI and no proven or
suspected COVID-19 at time of randomization; iii) patients with COVID-19-induced sepsis and AKI at time
of randomization (‘COVID-19 population’). The ‘all combined population’ includes all three cohorts. The
randomization schedule was stratied by site and modied Sequential Organ Failure Assessment
(mSOFA), excluding the neurological component of SOFA. An independent statistician generated a
permuted block randomization schedule for an interactive voice/web response system, which linked
sequential patient randomization numbers to treatment codes.
Study medication was administered, within 24 hours if sepsis was present prior to AKI or within 48 hours
if AKI was present when sepsis was diagnosed, at 1.6mg (1,000 U) per kg of patient body weight up to
120 kg, with a xed dose of 192mg in patients > 120 kg[23]. Patients received study medication as a 1-
hour infusion once daily for 3 consecutive days. All personnel involved in this study were blinded to
treatment assignment and clinicians were not allowed to measure serum ALP concentrations until day
14[24]. The trial drug was provided in addition to usual care as outlined in the Surviving Sepsis Campaign
guidelines[25] and KDIGO guidelines[5]. Initiation and termination of RRT was based on conventional
criteria[26–28]. A Data Management Committee (DMC) was installed and provided with stopping rules
based on predened threshold for futility or early success.
Primary endpoint
The primary ecacy endpoint was 28-day all-cause mortality.
Secondary endpoints
Renal endpoints
Key secondary endpoint was Major Adverse Kidney Events by day 90 (MAKE90) and its components. Two
denitions of MAKE90 were used. ‘MAKE-on-day-90’ was based on mortality through day 90 or an eGFR
drop of > 25% at day 28
and
day 90 compared to pre-AKI value, or need for RRT
at
day 90; in comparison
Page 6/21
and in accordance with existing data in the literature[29], ‘MAKE-through-day-90’ included any RRT events
through
day 28 or RRT status
at
day 90 or mortality through day 90, or an eGFR drop of > 25% at day 90,
or rehospitalization[30]. Furthermore, to examine the impact of prior renal function on outcome, we
determined the effect of pre-AKI eGFR on MAKE90.
Other secondary endpoints
Additional secondary endpoints were days alive and free of mechanical ventilation (MV), vasopressors or
inotropes; days alive and discharged from Intensive Care Unit (ICU) through day 28; and time to death
through day 90.
Safety Endpoints
Safety endpoints and adverse events (AEs) were monitored until study day 28 (inclusive). All deaths were
recorded up to study day 180.
Data analyses and statistics
Analysis of the primary ecacy endpoint was conducted according to protocol and Statistical Analysis
Plan (SAP) based on the mITT population, dened as all patients in whom drug administration was
started and analyzed according to treatment allocation. A supplementary SAP was established prior to
unblinding of the data during the rst interim analysis for the exploratory analysis of the all combined
population[23]. Safety analyses were conducted in the safety population as dened in the protocol.
For categorical and binary demographic and baseline characteristics, absolute and relative frequencies
are presented; and for continuous variables, the median and inter quartile ranges (IQR) are presented.
For the endpoints 28-day all-cause mortality, 90-day all-cause mortality, ‘MAKE on day 90’ and “MAKE-
through day 90’, observed proportions are presented along with the difference in proportions (i.e. ilofotase
alfa minus placebo), the 95% condence interval for the difference in proportions and the one-sided
nominal p-value. Because the study was stopped early due to futility, estimated (difference in)
proportions and the 95% condence interval and one-sided nominal p-values using the Kaplan-Meier
method are also provided[31]. In addition, for each treatment group, the effect of pre-AKI eGFR on the
predicted probability of a ‘MAKE through day 90’ outcome was investigated using a logistic regression
model which included treatment group, pre-AKI eGFR, and an interaction term for treatment group by pre-
AKI eGFR.
Safety analyses were conducted in the safety population as dened in the protocol. For patients with AEs,
relative and absolute frequencies are presented, and the distribution of the number of patients with
(serious) AEs has been compared using the chi-squared test.
SAS (SAS software version 9.4; SAS Institute Inc., Cary, NC, U.S.A.) was used for all statistical analyses. A
one-sided p-value < 0.025 was considered to indicate statistical signicance. In case the primary was not
Page 7/21
met, further statistical testing was considered explorative and therefore no correction for multiple testing
was performed.
Results
Patient recruitment and demographic characteristics
From November 2020 to July 2022, 649 patients (main [n=567] CKD [n=49] and COVID-19 [n=33] cohorts)
were enrolled and treated in 107 sites in North America, Europe, Japan, and Oceania (Figure 1) and
included in the analyses. Demographic characteristics are presented in Table 1 (demographics per cohort
in Supplemental Table 2). Patient demographics were comparable between the ilofotase alfa and
placebo groups in all three populations and comparable across analysis populations.
Interim analysis
At the time of the rst interim analysis (based on 411 patients in the main trial population) all-cause 28-
day mortality was 61/208 (29.3%) and 52/203 (25.7%) for the ilofotase alfa and placebo groups,
respectively. The recommendation from the DMC was to stop the REVIVAL trial early based on futility
according to the pre-dened thresholds for predictive probability of success.
Mortality
For the combined population (including main, CKD, COVID-19 cohorts), there was no difference in
mortality between the ilofotase alfa (92 of 330 patients) and the placebo (89 of 319 patients) groups at
day 28 (27.9% vs 27.9%, respectively). The difference in proportions was -0.02% with a 95% condence
interval of [-6.9%; 6.9%] and a nominal one-sided p-value of 0.50. Day 90 mortality was 112 of 330
patients (33.9%) vs 111 of 319 patients (34.8%). The difference in proportions was -0.86% with a 95%
condence interval of [-8.2%; 6.5%] and a nominal one-sided p-value of 0.41). The Kaplan-Meier analysis
had similar results with nominal one-sided p-values of 0.48 and 0.34 for day 28 and day 90 mortality.
Mortality data per cohort is presented in Supplementary Table 3.
Renal endpoints
There was no difference in observed proportions of patients with a “MAKE on day 90” event between
ilofotase alfa 36.4% (120 of 330) and the placebo 40.1% (128 of 319) groups (difference -3.8% 95%CI
[-11.2%, 3.7%] and a nominal one-sided p-value of 0.16. The estimated proportion of patients with a
‘MAKE-on-day-90’ event was 37.4% versus 42.8% for ilofotase alfa and placebo (difference -5.4%, 95%CI
[-13.2%, 2.4%], with a nominal one-sided p-value of 0.09).
The difference in observed proportions of patients with a ‘MAKE-through-day-90’ event between ilofotase
alfa 56.7% (187 of 330) and the placebo 64.6% (206 of 319) groups was 7.9, 95%CI [-15.4%, -0.4%] and a
nominal one sided p-value of 0.02. The estimated proportion of patients with a ‘MAKE-through-day-90’
event was also lower in the ilofotase alfa group compared to the placebo group (57.2% versus 64.7%,
Page 8/21
respectively (difference -7.4%, 95%CI [-15.2%, 0.4%], with a nominal one-sided p-value of 0.03 (Figure 2).
This effect was predominately driven by the difference in receipt of RRT between the ilofotase alfa and
placebo groups (28.2% vs 36.4%), see Supplementary Table 4.
There was evidence of an interaction between renal function prior to the SA-AKI episode (pre-AKI eGFR)
and the incidence of the ‘MAKE-through-day-90’ event suggesting that the therapeutic ecacy of ilofotase
alfa was more pronounced in patients with a lower pre-AKI eGFR, i.e., more severe pre-existent CKD
(Figure 3).Ilofotase alfa showed a benet over placebo in the probability of a ‘MAKE-through-day-90’
event for patients with pre-AKI eGFR below approximately 90 mL/min/1.73 m2. For each 10 mL/min/1.73
m2 decrease in pre-AKI eGFR, the predicted probability of a ‘MAKE-through-day-90’ event on placebo was
increased by 4% from an average at predicted probability of 0.57 for patients who received ilofotase alfa
(nominal one-sided p-value of 0.024).
Other secondary endpoints
There were no notable differences in the use of other types of organ support, as days alive and free of
non-renal organ support were similar between treatment groups. Also, ICU-LOS was similar, as number of
days alive and discharged from the ICU through day 28were similar between treatment groups
(Supplemental Table 5).
Safety parameters
Reported AEs are summarized by occurrence, percentage of occurrence, severity, seriousness, outcome,
and relation to treatment in the combined patient population (Table 2). A lower number of patients
experienced any AEs in the ilofotase alfa group compared to the placebo group [224/330 (67.9%) in the
ilofotase alfa group and 240/320 (75.0%) in the placebo group with a nominal two-sided p-value based
on the chi-squared test of 0.045]. Overall, the incidence of
serious
AEs was similar between the two
treatment groups. There were no relevant differences observed on SOC (System Organ Class Level)
(Supplemental Table 6).
Discussion
Given the observation that AKI complicates sepsis and patients with AKI have far worse short- and long-
term outcomes, there is high interest in strategies that reduce the incidence or ameliorate the course of
AKI, with the goal of reducing the overall burden on patients. In this phase 3 multi-center, international
double-blind RCT ilofotase alfa did not decrease 28-day all-cause mortality. However, there was evidence
to suggest ilofotase alfa reduced MAKE90 events, mainly driven by lowering the need for RRT in these
patients. Ilofotase alfa was well tolerated, and no safety issues emerged.
The previously demonstrated survival benet of ilofotase alfa[22] was not conrmed in the current trial
and several reasons for this discrepancy with the previous trial in the observed effect of ilofotase alfa on
survival can be put forward. First, the drug might not improve survival and the effect observed in the
Page 9/21
previous phase 2 STOP-AKI trial was a false-positive type 1 error. Second, slight changes in eligibility
criteria between both trials could be responsible. For example, in the REVIVAL trial, patients who already
had AKI when they presented with sepsis were eligible, in contrast to the STOP AKI trial. It is plausible that
the duration of AKI was longer in these patients, possibly limiting the therapeutic ecacy of ilofotase
alfa. It is also plausible that other differences in phenotype between the STOP-AKI and REVIVAL patients
existed that may have been of relevance, e.g. due to an overall change in the ICU population, because of
the COVID-19 pandemic and general heterogeneity in the sepsis population. Indeed, we found evidence of
heterogeneity of treatment effect with a more marked reduction in MAKE90 events in patients with pre-
existent impaired renal function. Third, REVIVAL was conducted in 107 sites worldwide vs 55 sites in
STOP-AKI, which increases generalizability but may have also negatively inuenced therapeutic ecacy.
Although the trial was stopped early because of the lack of an apparent survival benet, ilofotase alfa did
demonstrate reno-protective effects, which is consistent with previous trial results. Earlier clinical data
showed that bovine ALP attenuated urinary excretion of tubular injury marker Glutathione S-transferase
A1 (GSTA1-1), improved creatinine clearance and reduced the need for RRT[15]. Subsequently, a human
recombinant Alkaline Phosphatase (ilofotase alfa) was developed and tested in a phase 2 trial in 301
patients with SA-AKI. Although the primary endpoint in this trial, improvement of endogenous creatinine
clearance (ECC) in 7 days, was not met, longer-term ECC over the 28 day study period was signicantly
better, and in addition, a survival benet and reduction in MAKE-90 events were observed with ilofotase
alfa compared to placebo[22]. In REVIVAL, MAKE90 was markedly reduced in the ilofotase alfa treatment
arm, mainly driven by lower requirement for RRT. Need for RRT is an important component of the
composite MAKE90 endpoint especially due to the strong association with poor outcomes in patients
with sepsis[32–35]. Furthermore, it is recognized that patients with pre-existent CKD are more likely to
suffer from AKI when they develop sepsis and have a higher risk of not recovering renal function[8, 36]. It
is therefore not surprising that patients with a pre-existent lower eGFR, i.e. reduced or no renal reserve, are
more likely to meet a MAKE90 criterion following sepsis and that the reno-protective effect observed with
ilofotase alfa in REVIVAL was more pronounced in these patients. Increased ecacy in patients with pre-
existent CKD has been observed with other interventions as well[8, 27]. As pre-existent renal function was
not collected in the previous (recombinant) alkaline phosphatase trials, the interplay between pre-existent
renal function and therapeutic ecacy of ilofotase alfa is a novel nding that emerged from post-hoc
analysis, and should be interpreted as a hypothesis generating nding that needs to be conrmed.
Strengths of this study include its generalizability, as it was conducted in multiple countries worldwide.
Second, the group sequential trial design allowed for the results to be reported in case the trial was
terminated prematurely for futility. The latter also relates to two limitations. First, in case the primary
endpoint was not met, further statistical testing was considered explorative and therefore no correction
for multiple testing was performed; and second, analyses of combined cohorts were reported to explore
trends based on all enrolled patients. The number of patients enrolled in the COVID-19 and CKD cohorts
were small, and depicted in supplemental les for transparency. Further randomized controlled trials
would be necessary to assess the reno-protective effects of ilofotase alfa.
Page 10/21
Conclusions
The REVIVAL trial was stopped for futility due to lack of evidence of reduced 28-day mortality with
ilofotase alfa treatment in critically ill patients with SA-AKI. Despite this, our ndings were consistent with
preclinical studies and phase 2 trials suggesting reno-protective effects of ilofotase alfa. In particular, we
observed that ilofotase alfa reduced MAKE90, mainly driven by an attenuated need for RRT. The reduction
in MAKE90 with ilofotase alfa appeared most pronounced in patients with a lower eGFR prior to the SA-
AKI episode. A prospective randomized controlled trial is warranted to conrm the benecial renal effects
of ilofotase alfa in patients with SA-AKI and pre-existing CKD.
Declarations
Acknowledgments
PI’s of all enrolling sites and part of the REVIVAL investigators are depicted in supplemental le 1. We
thank all participating patients, their families for their support, as well as study coordinators and research
nurses of all participating sites in delivering the REVIVAL trial. The members of the data monitoring
committee are thanked for their rigorous review of the data throughout the trial process. The authors
thank Danielle Forkink (AM-Pharma, Clinical Trial Coordinator), Mariam Hamed (Phastar Programming
Consultant), Annelies Legters (AM-Pharma, Sr. Director, Global Clinical Trials) and Sharon Richards
(Phastar Biostatistician Consultant) for their support. 
Funding
This work was supported by AM-Pharma. The role of the sponsor in the design of the study was to
coordinate and facilitate processes, where the scientic input was provided by the members of the
protocol committee, steering committee, and specic input by external experts in data management and
statistics. The sponsor contracted an external contract research organization to operationally conduct the
study at the study sites. The contract research organization was responsible for setting up the technical
systems, data collection, quality control, pharmacovigilance, statistics, and further overall management
of the study, under coordination and supervision of the sponsor. The statistical analysis plan was
prepared by the contract research organization with input by principal investigator, sponsor, and external
experts in statistics. The analyses were performed by external contract research organizations. Data were
interpreted by the members of the steering committee, and, in a later phase, all coauthors and external
experts, coordinated by the sponsor, could provide input. The principal investigator was responsible for
preparation of the manuscript. All coauthors reviewed, made adjustments, and approved the manuscript.
The decision to submit the manuscript was made by the principal investigator and other coauthors.
Conict of interests statements:
Peter Pickkers has received travel reimbursements and consulting fees from AM-Pharma in relation to his
role as PI for REVIVAL, and consulting fees from Adrenomed, EBI Paion, Sphingotec, and 4Teen4 outside
Page 11/21
the submitted work.
Derek C. Angus has received consulting fees from AM-Pharma.
Kristie Bass, AM-Pharma BV, The Netherlands.
Rinaldo Bellomo has received consulting fees and research support from AM-Pharma, Baxter, Paion,
Viatris, Jafron Biomedical, and CSL Behring.
Erik van den Berg, AM-Pharma BV, The Netherlands.
Juliane Bernholz, AM-Pharma BV, TheNetherlands.
Morten H. Bestle has received consulting fees from AM-Pharma in relation to his role for REVIVAL and
has conducted contract research for Inotrem outside of the submitted work.
Kent Doi has received consulting fees from AM-Pharma.
Chistopher J. Doig reports no conicts of interest.
Ricard Ferrer hasreceived consulting fees from AM-Pharma.
Bruno Francois has received consulting fees from AM-Pharma as a member of the REVIVAL steering
committee, and consulting fees from Inotrem, Aridis and Enlivex outside the submitted work.
Henrik Gammelagerreports funding from various companies in the form of research grants to (and
administered by) Aarhus University or Aarhus University Hospital. H.G. has received support for attending
meetings by Baxter A/S.
Ulf Goettrup-Pedersen reports no conicts of interest.
Eric Hoste has received a travel grant from AM-Pharma.
Susanne Iversen reports no conicts of interest.
Michael Joannidis has received honoraria or research support from Baxter Healthcare Corp, AM-Pharma,
CLS Behring, Fresenius, Takeda, Sano and Novartis.
John A. Kellum discloses fees paid by AM-Pharma in relation to his role as national PI for REVIVAL and is
currently a full-time employee of Spectral Medical.
Kathleen Liu has been a member of the REVIVAL Steering Committee for AM Pharma. She has been a
consultant/member of the DSMB for Seastar, Novartis, BOA Medical, Baxter, and Biomerieux, and she
holds stock in Amgen.
Melanie Meersch has received lecture fees from Baxter and Fresenius Medical Care.
Page 12/21
Ravindra Mehta reports honoraria for consulting fromBaxter, Biomerieux, Mallinckrodt, GE Healthcare,
Sano, Abiomed, NovaBiomed, Renasym and advisory board reimbursements from AM Pharma, Renibus,
Alexion, Novartis and Guard.
Scott Millington reports no conicts of interest.
Patrick T. Murray has received consulting fees from AM-Pharma (for Clinical Trial Steering Committee
activities), Novartis, Renibus Therapeutics, and Alexion.
Alistair Nichol reports an unrestricted grant from Baxter to support the renal sub study of the TAME trial.
Marlies Ostermann has received speaker honoraria from Fresenius Medical, Baxter and Biomerieux; her
institution received research funding from Baxter, Fresenius Medical, Biomerieux and LaJolla Pharma. 
Christoffer Solling reports no conicts of interest.
Pettila Ville reports no conicts of interest.
Matthias Winkel, AM-Pharma BV, The Netherlands.
Paul J Young has received consulting fees from AM Pharma and from Baxter Healthcare Pty.
Alexander Zarbock has received consulting fees from Astute-Biomerieux, Baxter, Bayer, Novartis, Guard
Therapeutics, AM Pharma, Paion, Renibus, Fresenius, research funding from Astute-Biomerieux,
Fresenius, Baxter, and speakers fees from Astute-Biomerieux, Fresenius, Baxter.
References
1. Wen X, Peng Z, Kellum JA, (2011) Pathogenesis of acute kidney injury: effects of remote tissue
damage on the kidney. Contrib Nephrol 174: 129-137
2. Gomez H, Ince C, De Backer D, Pickkers P, Payen D, Hotchkiss J, Kellum JA, (2014) A unied theory of
sepsis-induced acute kidney injury: inammation, microcirculatory dysfunction, bioenergetics, and
the tubular cell adaptation to injury. Shock 41: 3-11
3. Verma SK, Molitoris BA, (2015) Renal endothelial injury and microvascular dysfunction in acute
kidney injury. Semin Nephrol 35: 96-107
4. Zarbock A, Nadim MK, Pickkers P, Gomez H, Bell S, Joannidis M, Kashani K, Koyner JL, Pannu N,
Meersch M, Reis T, Rimmelé T, Bagshaw SM, Bellomo R, Cantaluppi V, Deep A, De Rosa S, Perez-
Fernandez X, Husain-Syed F, Kane-Gill SL, Kelly Y, Mehta RL, Murray PT, Ostermann M, Prowle J, Ricci
Z, See EJ, Schneider A, Soranno DE, Tolwani A, Villa G, Ronco C, Forni LG, (2023) Sepsis-associated
acute kidney injury: consensus report of the 28th Acute Disease Quality Initiative workgroup. Nat Rev
Nephrol 19: 401-417
Page 13/21
5. Kellum JA, Lameire N, (2013) Diagnosis, evaluation, and management of acute kidney injury: a
KDIGO summary (Part 1). Crit Care 17: 204
. Kellum JA, Chawla LS, Keener C, Singbartl K, Palevsky PM, Pike FL, Yealy DM, Huang DT, Angus DC,
(2016) The Effects of Alternative Resuscitation Strategies on Acute Kidney Injury in Patients with
Septic Shock. Am J Respir Crit Care Med 193: 281-287
7. Chawla LS, Eggers PW, Star RA, Kimmel PL, (2014) Acute kidney injury and chronic kidney disease as
interconnected syndromes. N Engl J Med 371: 58-66
. Khosla N, Soroko SB, Chertow GM, Himmelfarb J, Ikizler TA, Paganini E, Mehta RL, (2009) Preexisting
chronic kidney disease: a potential for improved outcomes from acute kidney injury. Clin J Am Soc
Nephrol 4: 1914-1919
9. Abdalrahim MS, Khalil AA, Alramly M, Alshlool KN, Abed MA, Moser DK, (2020) Pre-existing chronic
kidney disease and acute kidney injury among critically ill patients. Heart Lung 49: 626-629
10. Kellum JA, Prowle JR, (2018) Paradigms of acute kidney injury in the intensive care setting. Nat Rev
Nephrol 14: 217-230
11. Verweij WR, Bentala H, Huizinga-van der Vlag A, Miek van Loenen-Weemaes A, Kooi K, Meijer DK,
Poelstra K, (2004) Protection against an Escherichia coli-induced sepsis by alkaline phosphatase in
mice. Shock 22: 174-179
12. Su F, Brands R, Wang Z, Verdant C, Bruhn A, Cai Y, Raaben W, Wulferink M, Vincent JL, (2006)
Benecial effects of alkaline phosphatase in septic shock. Crit Care Med 34: 2182-2187
13. Wy CA, Goto M, Young RI, Myers TF, Muraskas J, (2000) Prophylactic treatment of endotoxic shock
with monophosphoryl lipid A in newborn rats. Biol Neonate 77: 191-195
14. Peters E, Geraci S, Heemskerk S, Wilmer MJ, Bilos A, Kraenzlin B, Gretz N, Pickkers P, Masereeuw R,
(2015) Alkaline phosphatase protects against renal inammation through dephosphorylation of
lipopolysaccharide and adenosine triphosphate. Br J Pharmacol 172: 4932-4945
15. Peters E, van Elsas A, Heemskerk S, Jonk L, van der Hoeven J, Arend J, Masereeuw R, Pickkers P,
(2013) Alkaline phosphatase as a treatment of sepsis-associated acute kidney injury. J Pharmacol
Exp Ther 344: 2-7
1. Beumer C, Wulferink M, Raaben W, Fiechter D, Brands R, Seinen W, (2003) Calf intestinal alkaline
phosphatase, a novel therapeutic drug for lipopolysaccharide (LPS)-mediated diseases, attenuates
LPS toxicity in mice and piglets. J Pharmacol Exp Ther 307: 737-744
17. Rosin DL, Hall JP, Zheng S, Huang L, Campos-Bilderback S, Sandoval R, Bree A, Beaumont K, Miller E,
Larsen J, Hariri G, Kaila N, Encarnacion IM, Gale JD, van Elsas A, Molitoris BA, Okusa MD, (2022)
Human Recombinant Alkaline Phosphatase (Ilofotase Alfa) Protects Against Kidney Ischemia-
Reperfusion Injury in Mice and Rats Through Adenosine Receptors. Front Med (Lausanne) 9: 931293
1. Peters E, Ergin B, Kandil A, Gurel-Gurevin E, van Elsas A, Masereeuw R, Pickkers P, Ince C, (2016)
Effects of a human recombinant alkaline phosphatase on renal hemodynamics, oxygenation and
inammation in two models of acute kidney injury. Toxicol Appl Pharmacol 313: 88-96
Page 14/21
19. Davidson JA, Khailova L, Treece A, Robison J, Soranno DE, Jaggers J, Ing RJ, Lawson S, Lujan SO,
(2019) Alkaline Phosphatase Treatment of Acute Kidney Injury in an Infant Piglet Model of
Cardiopulmonary Bypass with Deep Hypothermic Circulatory Arrest. Sci Rep 9: 14175
20. Kiffer-Moreira T, Sheen CR, Gasque KC, Bolean M, Ciancaglini P, van Elsas A, Hoylaerts MF, Millán JL,
(2014) Catalytic signature of a heat-stable, chimeric human alkaline phosphatase with therapeutic
potential. PLoS One 9: e89374
21. Heemskerk S, Masereeuw R, Moesker O, Bouw MP, van der Hoeven JG, Peters WH, Russel FG,
Pickkers P, (2009) Alkaline phosphatase treatment improves renal function in severe sepsis or septic
shock patients. Crit Care Med 37: 417-423, e411
22. Pickkers P, Mehta RL, Murray PT, Joannidis M, Molitoris BA, Kellum JA, Bachler M, Hoste EAJ, Hoiting
O, Krell K, Ostermann M, Rozendaal W, Valkonen M, Brealey D, Beishuizen A, Meziani F, Murugan R, de
Geus H, Payen D, van den Berg E, Arend J, (2018) Effect of Human Recombinant Alkaline
Phosphatase on 7-Day Creatinine Clearance in Patients With Sepsis-Associated Acute Kidney Injury:
A Randomized Clinical Trial. Jama 320: 1998-2009
23. Pickkers P, Angus DC, Arend J, Bellomo R, van den Berg E, Bernholz J, Bestle M, Broglio K, Carlsen J,
Doig CJ, Ferrer R, Joannidis M, Francois B, Doi K, Kellum JA, Laterre PF, Liu K, Mehta RL, Murray PT,
Ostermann M, Pettilä V, Richards S, Young P, Zarbock A, Kjølbye AL, (2023) Study protocol of a
randomised, double-blind, placebo-controlled, two-arm parallel-group, multi-centre phase 3 pivotal
trial to investigate the ecacy and safety of recombinant human alkaline phosphatase for treatment
of patients with sepsis-associated acute kidney injury. BMJ Open 13: e065613
24. Pickkers P, Snellen F, Rogiers P, Bakker J, Jorens P, Meulenbelt J, Spapen H, Tulleken JE, Lins R,
Ramael S, Bulitta M, van der Hoeven JG, (2009) Clinical pharmacology of exogenously administered
alkaline phosphatase. Eur J Clin Pharmacol 65: 393-402
25. Levy MM, Evans LE, Rhodes A, (2018) The Surviving Sepsis Campaign Bundle: 2018 update.
Intensive Care Med 44: 925-928
2. Naorungroj T, Neto AS, Wang A, Gallagher M, Bellomo R, (2022) Renal outcomes according to renal
replacement therapy modality and treatment protocol in the ATN and RENAL trials. Crit Care 26: 269
27. Bagshaw SM, Neto AS, Smith O, Weir M, Qiu H, Du B, Wang AY, Gallagher M, Bellomo R, Wald R,
(2022) Impact of renal-replacement therapy strategies on outcomes for patients with chronic kidney
disease: a secondary analysis of the STARRT-AKI trial. Intensive Care Med 48: 1736-1750
2. Ronco C, Bellomo R, Kellum JA, (2019) Acute kidney injury. Lancet 394: 1949-1964
29. Billings FTt, Shaw AD, (2014) Clinical trial endpoints in acute kidney injury. Nephron Clin Pract 127:
89-93
30. Rewa O, Bagshaw SM, (2014) Acute kidney injury-epidemiology, outcomes and economics. Nat Rev
Nephrol 10: 193-207
31. Zee J, Xie SX, (2018) The Kaplan-Meier Method for Estimating and Comparing Proportions in a
Randomized Controlled Trial with Dropouts. Biostat Epidemiol 2: 23-33
Page 15/21
32. Joannidis M, Druml W, Forni LG, Groeneveld ABJ, Honore PM, Hoste E, Ostermann M, Oudemans-van
Straaten HM, Schetz M, (2017) Prevention of acute kidney injury and protection of renal function in
the intensive care unit: update 2017 : Expert opinion of the Working Group on Prevention, AKI section,
European Society of Intensive Care Medicine. Intensive Care Med 43: 730-749
33. Forni LG, Darmon M, Ostermann M, Oudemans-van Straaten HM, Pettilä V, Prowle JR, Schetz M,
Joannidis M, (2017) Renal recovery after acute kidney injury. Intensive Care Med 43: 855-866
34. Liu C, Peng Z, Dong Y, Li Z, Song X, Liu X, Andrijasevic NM, Gajic O, Albright RC, Jr., Kashani KB,
(2021) Continuous Renal Replacement Therapy Liberation and Outcomes of Critically Ill Patients
With Acute Kidney Injury. Mayo Clin Proc 96: 2757-2767
35. Li Y, Li H, Zhang D, (2019) Timing of continuous renal replacement therapy in patients with septic
AKI: A systematic review and meta-analysis. Medicine (Baltimore) 98: e16800
3. Ostermann M, Chang R, (2008) Correlation between the AKI classication and outcome. Crit Care 12:
R144
Tables
Table 1:Demographic and baseline characteristics
Page 16/21
Combined Patient population
Ilofotase alfa
(N=330) Placebo
(N=319)
Sex, n (%) Female 124 (37.6 %) 113 (35.4 %)
Male 206 (62.4 %) 206 (64.6 %)
Age, in years 70.0 (62.0,76.0) 70.0 (61.0,76.0)
Weight in kg 84.0 (72.0,100.0) 82.0 (70.0,100.0)
Height in cm 172.0
(164.0,178.0) 170.0
(165.0,178.0)
Body Mass Index in kg/m2 28.9 (25.0,33.2) 27.6 (24.6,34.0)
Mechanical Ventilation Status,
n(%) Off 84 (25.5 %) 74 (23.2 %)
On 246 (74.5 %) 245 (76.8 %)
AKI stage at randomization, n(%) 0 or 1 144 (43.6 %) 129 (40.4 %)
2 93 (28.2 %) 105 (32.9 %)
3 93 (28.2 %) 84 (26.3 %)
Missing  0  1 (0.3 %)
Baseline eGFR 31.2 (21.2,43.3) 30.6 (20.4,42.0)
Pre-AKI eGFR 74.0 (60.1,88.3) 73.9 (59.8,89.0)
mSOFA  9.0 (7.0,10.0)  9.0 (8.0,11.0)
CRP 260.5
(141.2,341.2) 235.0
(155.7,321.3)
Lactate  2.2 (1.5,3.6)  2.4 (1.5,3.7)
PaO2/FiO2 ratio: 225.8
(146.3,304.2) 191.7
(134.0,292.5)
Infection Status Proven 156 (47.3 %) 160 (50.2 %)
Suspected 174 (52.7 %) 159 (49.8 %)
Infection Site: Abdominal 120 (36.4 %) 115 (36.1 %)
CNS   7 (2.1 %)   3 (0.9 %)
Other 32 (9.7 %) 34 (10.7 %)
Pulmonary 97 (29.4 %) 95 (29.8 %)
Page 17/21
Combined Patient population
Ilofotase alfa
(N=330) Placebo
(N=319)
Skin or soft-
tissue 24 (7.3 %) 30 (9.4 %)
Unknown 15 (4.5 %) 18 (5.6 %)
Urinary tract 35 (10.6 %) 24 (7.5 %)
Pathogen Proven/Suspected Proven 132 (40.0 %) 126 (39.5 %)
Suspected 192 (58.2 %) 190 (59.6 %)
Missing  6 (1.8 %)  3 (0.9 %)
Type of pathogen: Viral 28 (8.5 %) 18 (5.6 %)
Bacterial 251 (76.1 %) 239 (74.9 %)
 Gram Positive 77 (23.3 %) 67 (21.0 %)
 Gram Negative 92 (27.9 %) 82 (25.7 %)
 Mixed 49 (14.8 %) 53 (16.6 %)
 Missing 33 (10.0 %) 37 (11.6 %)
Missing 51 (15.5 %) 62 (19.4 %)
Note:
- Continuous variable: Median (IQR)
- Patients have been analyzed according to treatment received
Table 2. Overall summary of number of patients with adverse events in the combined patient population
Page 18/21
Treatment Group
Ilofotase alfa
(N=330)
n (%)
Placebo
(N=320)
n (%)
P-value*
Adverse events 224 ( 67.9%) 240 ( 75.0%) 0.0446
Serious adverse events 143 ( 43.3%) 141 ( 44.1%) 0.8514
Fatal adverse events 99 ( 30.0%) 98 ( 30.6%)
Serious non-fatal adverse events 64 ( 19.4%) 62 ( 19.4%)
Drug-related adverse events 30 ( 9.1%) 32 ( 10.0%)
Serious drug-related adverse events 16 ( 4.8%) 16 ( 5.0%)
Adverse events leading to withdrawal of trial drug 9 ( 2.7%) 6 ( 1.9%)
Serious adverse events leading to withdrawal of trial drug 8 ( 2.4%) 5 ( 1.6%)
Adverseeventsbyseverity**
Mild 29 ( 8.8%) 31 ( 9.7%)
Note:
- Patients have been analyzed according to treatment received
* Nominal p-value based on chi-square test
Figures
Page 19/21
Figure 1
Flow chart of the patients participating in the REVIVAL trial.
Page 20/21
Figure 2
Time to rst “MAKE through day 90” event
Proportions of patients without "MAKE through day90”: Ilofotase alfa 42.77%, Placebo 35.33%.
Proportion of patients with “MAKE through day 90” event at Day 90: Ilofotase alfa 57.23%, Placebo
64.67%
The difference (Ilofotase alfa-Placebo) in proportion of patients with “MAKE through day 90” event at Day
90 is -7.44% with a 95% condence interval of (-15.23%, 0.36%).
The nominal one-sided p-value based on the Wald statistic for the difference in proportions is 0.030705
Patients who withdrew prior to day 90 (inclusive) without a “MAKE through day 90” event are censored at
their date of withdrawal.
Patients who withdrew/completed the trial after day 90 were censored at day 90.
Patients who died or were rehospitalized after day 90 are censored at day 90.
Patient with a “Make through day 90” event based on receiving RRT at their Day 90 visit , or a >25% drop
in eGFR at their Day90 visit, are counted as having a “MAKE through day 90” event at day 90.
Page 21/21
Figure 3
Predicted Probability of a “MAKE though day 90” event with 95% Condence Limits
Predicted probabilities are taken from the results of the logistic regression model which included “MAKE
through day 90” as the outcome of interest, and
treatment, pre-AKI reference eGFR, and pre-AKI reference eGFR by treatment interaction (p=0.0235).
Patients who did not meet the criteria for having a conrmed “MAKE through day 90” event are assumed
to not have a “MAKE through day 90” event in the logistic regression model.
Supplementary Files
This is a list of supplementary les associated with this preprint. Click to download.
CONSORTchecklist.docx
Supplle1REVIVALSiteprincipleinvestigators.xlsx
SupplementalTableandFiguresREVIVAL.docx
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Introduction: Sepsis, the leading cause of acute kidney injury (AKI), is associated with a high morbidity and mortality. Alkaline phosphatase (ALP) is an endogenous detoxifying enzyme. A recombinant human ALP compound, ilofotase alfa, showed no safety or tolerability concerns in a phase 2 trial. Renal function improvement over 28 days was significantly greater in the ilofotase alfa group. Moreover, a significant relative reduction in 28-day all-cause mortality of >40% was observed. A follow-up trial has been designed to confirm these findings. Methods and analysis: This is a phase 3, global, multi-centre, randomised, double-blind, placebo-controlled, sequential design trial in which patients are randomly assigned to either placebo or 1.6 mg/kg ilofotase alfa. Randomisation is stratified by baseline modified Sequential Organ Failure Assessment (mSOFA) score and trial site. The primary objective is to confirm the survival benefit with ilofotase alfa by demonstrating a reduction in 28-day all-cause mortality in patients with sepsis-associated AKI requiring vasopressors. A maximum of 1400 patients will be enrolled at ∼120 sites in Europe, North America, Japan, Australia and New Zealand. Up to four interim analyses will take place. Based on predefined decision rules, the trial may be stopped early for futility or for effectiveness. In addition, patients with COVID-19 disease and patients with 'moderate to severe' chronic kidney disease are analysed as 2 separate cohorts of 100 patients each. An independent Data Monitoring Committee evaluates safety data at prespecified intervals throughout the trial. Ethics and dissemination: The trial is approved by relevant institutional review boards/independent ethics committees and is conducted in accordance with the ethical principles of the Declaration of Helsinki, guidelines of Good Clinical Practice, Code of Federal Regulations and all other applicable regulations. Results of this study will determine the potential of ilofotase alfa to reduce mortality in critically ill patients with sepsis-associated AKI and will be published in a peer-reviewed scientific journal. Trial registration number: EudraCT CT Number 2019-0046265-24. US IND Number 117 605 Pre-results. Clinicaltrials: gov number: NCT04411472.
Article
Full-text available
PurposeTo assess whether pre-existing chronic kidney disease (CKD) modified the relationship between the strategy for renal-replacement theraphy (RRT) initiation and clinical outcomes in the STARRT-AKI trial.Methods This was a secondary analysis of a multi-national randomized trial. We included patients who had documented pre-existing estimated glomerular filtration rate (eGFR) data prior to hospitalization, and we defined CKD as an eGFR ≤ 59 mL/min/1.73 m2. The primary outcome was all-cause mortality at 90 days. Secondary outcomes included RRT dependence and RRT-free days at 90 days. We used logistic and linear regression and interaction testing to explore the effect of RRT initiation strategy on outcomes by CKD status.ResultsWe studied 1121 patients who had pre-hospital measures of kidney function. Of these, 432 patients (38.5%) had CKD. The median (IQR) baseline serum creatinine was 130 (114–160) and 76 (64–90) µmol/L for those with and without CKD, respectively. Patients with CKD were older and more likely to have cardiovascular comorbidities and diabetes mellitus. Patients with CKD had higher 90-day mortality (47% vs. 40%, p < 0.001) compared to those without CKD, though this was not significant after covariate adjustment (adjusted odds ratio [aOR], 1.05; 95% CI, 0.79–1.41). Patients with CKD were more likely to remain RRT dependent at 90 days (14% vs. 8%; aOR, 1.89; 95% CI, 1.05–3.43). CKD status did not modify the effect of RRT initiation strategy on 90-day mortality. Among patients with CKD, allocation to the accelerated strategy conferred more than threefold greater odds of RRT dependence at 90 days (aOR 3.18; 95% CI, 1.41–7.91) compared with the standard strategy, whereas RRT initiation strategy had no effect on this outcome among those without CKD (aOR 0.71; 95% CI, 0.34–1.47, p value for interaction, 0.009).Conclusion In this secondary analysis of the STARRT-AKI trial, an accelerated strategy of RRT initiation conferred a higher risk of 90-day RRT dependence among patients with pre-existing CKD; however, no effect was observed in the absence of CKD.
Article
Full-text available
Background In critically ill patients with acute kidney injury, renal replacement therapy (RRT) modality and treatment protocols may affect kidney recovery. This study explored whether RRT modality and treatment protocol affected RRT dependence in the ‘Randomized Evaluation of Normal versus Augmented Level of RRT’ and the ‘Acute Renal Failure Trial Network’ (ATN) trials. Methods Primary outcome was 28-day RRT dependence. Secondary outcomes included RRT dependence among survivors and in different SOFA-based treatment protocol groups. We used the Fine-Gray competing-risk model sub-distribution hazard ratio (SHR) to assess the primary outcome. Analyses were adjusted for confounders. Results Of 2542 patients, 2175 (85.5%) received continuous RRT (CRRT) and 367 (14.4%) received intermittent hemodialysis (IHD) as first RRT modality. CRRT-first patients had greater illness severity. After adjustment, there was no between-group difference in 28-day RRT dependence (SHR, 0.96 [95% CI 0.84–1.10]; p = 0.570) or hospital mortality (odds ratio [OR], 1.14 [95% CI 0.86–1.52]; p = 0.361) However, among survivors, CRRT-first was associated with decreased 28-day RRT dependence (OR, 0.54 [95% CI 0.37–0.80]; p = 0.002) and more RRT-free days (common OR: 1.38 [95% CI 1.11–1.71]). Moreover, among CRRT-first patient, the ATN treatment protocol was associated with fewer RRT-free days, greater mortality, and a fourfold increase in RRT dependence at day 28. Conclusions There was no difference in RRT dependence at day 28 between IHD and CRRT. However, among survivors and after adjustment, both IHD-first and the ATN treatment protocol were strongly associated with greater risk of RRT dependence at 28 days after randomization. Trial registration NCT00221013 registered September 22, 2005, and NCT00076219 registered January 19, 2004.
Article
Full-text available
Acute kidney injury (AKI) is associated with prolonged hospitalization and mortality following infant cardiac surgery, but therapeutic options are limited. Alkaline phosphatase (AP) infusion reduced AKI in phase 2 sepsis trials but has not been evaluated for cardiac surgery-induced AKI. We developed a porcine model of infant cardiopulmonary bypass (CPB) with deep hypothermic circulatory arrest (DHCA) to investigate post-CPB/DHCA AKI, measure serum/renal tissue AP activity with escalating doses of AP infusion, and provide preliminary assessment of AP infusion for prevention of AKI. Infant pigs underwent CPB with DHCA followed by survival for 4 h. Groups were treated with escalating doses of bovine intestinal AP (1, 5, or 25U/kg/hr). Anesthesia controls were mechanically ventilated for 7 h without CPB. CPB/DHCA animals demonstrated histologic and biomarker evidence of AKI as well as decreased serum and renal tissue AP compared to anesthesia controls. Only high dose AP infusion significantly increased serum or renal tissue AP activity. Preliminary efficacy evaluation demonstrated a trend towards decreased AKI in the high dose AP group. The results of this dose-finding study indicate that AP infusion at the dose of 25U/kg/hr corrects serum and tissue AP deficiency and may prevent AKI in this piglet model of infant CPB/DHCA.
Article
Background The impact of pre-existing chronic kidney disease (CKD) and acute kidney injury (AKI) on health outcomes in critically ill patients is unclear. Yet, CKD complicated by AKI in critically ill patients is common. Objectives : To compare risk of death within one-month of admission in critically ill patients with and without pre-existing CKD who developed AKI. Methods A multicenter retrospective comparative study using medical records review was conducted. Study participants consisted of 826 adult patients who received mechanical ventilation for at least 6 h in the critical care units from January 2012 to December 2017. Assessment of kidney function was established by serum creatinine. Severity and staging of AKI were defined using RIFLE criteria: Risk, Injury, Failure, Loss and End stage of renal disease. Chronic kidney disease was defined as eGFR > 60 ml/mg/1.73 m² on admission. Results Pre-existing CKD was present in 55% of patients and 7% had AKI within 7 days of admission. The overall mortality rate among these patients was 87.3%. The mortality rate was highest in patients with CKD (70.1%) followed by that of patients without pre-existing CKD but with AKI (20.7%) and that of patients with pre-existing CKD (7.1%) and AKI. Risks associated with mortality were APACHE II score (1.03; 95% CI 1.02–1.05;(P<0.001) and AKI (1.68; 95% CI 1.12–2.5;P<0.01) in patients with pre-existing CKD. Only APACHI-II (1.03; 95% CI 1.0–1.1; p < 0.001) was predictive of death in patients without pre-existing CKD. Conclusion : Pre-existing comorbid CKD increases risks of death among critically ill patients compared to patients without CKD and regardless of whether they develop AKI or not. Early identification of CKD and recognition of the risk for mortality among these patients may result in earlier intervention that could reduce mortality.
Article
Importance Sepsis-associated acute kidney injury (AKI) adversely affects long-term kidney outcomes and survival. Administration of the detoxifying enzyme alkaline phosphatase may improve kidney function and survival. Objective To determine the optimal therapeutic dose, effect on kidney function, and adverse effects of a human recombinant alkaline phosphatase in patients who are critically ill with sepsis-associated AKI. Design, Setting, and Participants The STOP-AKI trial was an international (53 recruiting sites), randomized, double-blind, placebo-controlled, dose-finding, adaptive phase 2a/2b study in 301 adult patients admitted to the intensive care unit with a diagnosis of sepsis and AKI. Patients were enrolled between December 2014 and May 2017, and follow-up was conducted for 90 days. The final date of follow-up was August 14, 2017. Interventions In the intention-to-treat analysis, in part 1 of the trial, patients were randomized to receive recombinant alkaline phosphatase in a dosage of 0.4 mg/kg (n = 31), 0.8 mg/kg (n = 32), or 1.6 mg/kg (n = 29) or placebo (n = 30), once daily for 3 days, to establish the optimal dose. The optimal dose was identified as 1.6 mg/kg based on modeling approaches and adverse events. In part 2, 1.6 mg/kg (n = 82) was compared with placebo (n = 86). Main Outcomes and Measures The primary end point was the time-corrected area under the curve of the endogenous creatinine clearance for days 1 through 7, divided by 7 to provide a mean daily creatinine clearance (AUC1-7 ECC). Incidence of fatal and nonfatal (serious) adverse events ([S]AEs) was also determined. Results Overall, 301 patients were enrolled (men, 70.7%; median age, 67 years [interquartile range {IQR}, 59-73]). From day 1 to day 7, median ECC increased from 26.0 mL/min (IQR, 8.8 to 59.5) to 65.4 mL/min (IQR, 26.7 to 115.4) in the recombinant alkaline phosphatase 1.6-mg/kg group vs from 35.9 mL/min (IQR, 12.2 to 82.9) to 61.9 mL/min (IQR, 22.7 to 115.2) in the placebo group (absolute difference, 9.5 mL/min [95% CI, −23.9 to 25.5]; P = .47). Fatal adverse events occurred in 26.3% of patients in the 0.4-mg/kg recombinant alkaline phosphatase group; 17.1% in the 0.8-mg/kg group, 17.4% in the 1.6-mg/kg group, and 29.5% in the placebo group. Rates of nonfatal SAEs were 21.0% for the 0.4-mg/kg recombinant alkaline phosphatase group, 14.3% for the 0.8-mg/kg group, 25.7% for the 1.6-mg/kg group, and 20.5% for the placebo group. Conclusions and Relevance Among patients who were critically ill with sepsis-associated acute kidney injury, human recombinant alkaline phosphatase compared with placebo did not significantly improve short-term kidney function. Further research is necessary to assess other clinical outcomes. Trial Registration ClinicalTrials.gov Identifier: NCT02182440
Article
Previous iterations of the sepsis bundle were introduced as a means of providing education and improvement related to sepsis management. The literature supports the use of sepsis bundles for improving outcomes in patients with sepsis and septic shock. This new sepsis "hour-1 bundle," based on the 2016 guidelines, should be introduced to emergency department, floor, and ICU staff as the next iteration of ever-improving tools in the care of patients with sepsis and septic shock as we all work to lessen the global burden of sepsis.
Article
Acute kidney injury (AKI) is not a single disease but a loose collection of syndromes characterized by an abrupt decrease in glomerular filtration rate Broad classifications of AKI — according to the dominant aetiology (such as sepsis or nephrotoxicity) as opposed to pseudo-anatomical categories (for example, prerenal or intrarenal) — demonstrate more consistent relationships with pathophysiology and can improve therapeutic approaches Broad aetiology-based classification may still be insufficient, as heterogeneity could exist at the molecular level; however, whether this heterogeneity leads to differences in response to therapy is not yet known Protein biomarkers and other in vitro diagnostics are likely to have a role in both characterizing clinical phenotypes of AKI and aiding the discovery of new endophenotypes, with the ultimate goal of matching subsets of patients to effective therapeutic approaches
Article
We propose a method for estimating and comparing proportions of study participants who reached an event of interest during a randomized controlled trial. Standard methods for estimating this proportion include the intent-to-treat method, which counts the number who reached the event of interest divided by the total number of participants, and the completers-only method, which counts the number who reached the event only among those who completed the entire study. When participants drop out of the study early, however, these methods will either be biased or inefficient. We propose to use the Kaplan–Meier method from survival analysis to estimate the proportion of interest in this non-survival setting. We show through extensive simulation studies that the Kaplan–Meier method has less bias and is more efficient than the standard methods. We demonstrate the performance of all methods for estimating proportions in one sample and for comparing proportions across two samples. Finally, we apply the proposed method to a data-set for estimating and comparing proportions of patients who achieved treatment response during a Parkinson's disease trial for the treatment of impulse control disorders.
Article
Two small clinical trials indicated that administration of bovine intestinal alkaline phosphatase (AP) improves renal function in critically ill patients with sepsis-associated acute kidney injury (AKI), for which the mechanism of action is not completely understood. Here, we investigated the effects of a newly developed human recombinant AP (recAP) on renal oxygenation and hemodynamics and prevention of kidney damage and inflammation in two in vivo AKI models. To induce AKI, male Wistar rats (n = 18) were subjected to renal ischemia (30 min) and reperfusion (I/R), or sham-operated. In a second model, rats (n = 18) received a 30 min infusion of lipopolysaccharide (LPS; 2.5 mg/kg), or saline, and fluid resuscitation. In both models, recAP (1000 U/kg) was administered intravenously (15 min before reperfusion, or 90 min after LPS). Following recAP treatment, I/R-induced changes in renal blood flow, renal vascular resistance and oxygen delivery at early, and cortical microvascular oxygen tension at late reperfusion were no longer significantly affected. RecAP did not influence I/R-induced effects on mean arterial pressure. During endotoxemia, recAP treatment did not modulate the LPS-induced changes in systemic hemodynamics and renal oxygenation. In both models, recAP did exert a clear renal protective anti-inflammatory effect, demonstrated by attenuated immunostaining of inflammatory, tubular injury and pro-apoptosis markers. Whether this renal protective effect is sufficient to improve outcome of patients suffering from sepsis-associated AKI is being investigated in a large clinical trial.