Bagshaw 2017

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Journal of Critical Care 42 (2017) 138–146

Contents lists available at ScienceDirect

Journal of Critical Care

journal homepage: www.jccjournal.org

Renal

The effect of low-dose furosemide in critically ill patients with early


acute kidney injury: A pilot randomized blinded controlled trial
(the SPARK study)
Sean M. Bagshaw, MD, MSc a,⁎, R.T. Noel Gibney, MD a, Peter Kruger, MD, PhD b, Imran Hassan, MSc c,
Finlay A. McAlister, MD, MSc d, Rinaldo Bellomo, MD, PhD e
a
Department of Critical Care Medicine, Faculty of Medicine and Dentistry, University of Alberta, 2-124E Clinical Science Building, 8440-112 Street, Edmonton, Alberta T6G 2B7, Canada
b
Department of Intensive Care Medicine, Princess Alexandra Hospital, Ipswich Rd, Woolloongabba, and School of Medicine, University of Queensland, Queensland 4012, Australia
c
Epidemiology Coordinating and Research Centre (EPICORE), Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, 362 Heritage Medical Research Centre, Edmonton,
Alberta T6G 2S2, Canada
d
Division of General Internal Medicine, Faculty of Medicine and Dentistry, University of Alberta, 5-112 Clinical Sciences Building, 8440-112 Street, Edmonton, Alberta T6G 2B7, Canada
e
Department of Intensive Care, Austin Hospital, Studley Rd, Heidelberg, Victoria 3084, Australia

a r t i c l e i n f o a b s t r a c t

Keywords: Purpose: Furosemide is commonly prescribed in acute kidney injury (AKI). Prior studies have found conflicting
Acute kidney injury findings on whether furosemide modifies the course and outcome of AKI.
Furosemide Methods: Pilot multi-center randomized blinded placebo-controlled trial in adult patients with AKI admitted to
Urine output
three intensive care units. Participants were randomly allocated to furosemide bolus and infusion or 0.9% saline
Renal replacement therapy
Recovery
placebo. Primary endpoint was worsening AKI, defined by the RIFLE criteria. Secondary endpoints were kidney
Mortality recovery, renal replacement therapy (RRT) and adverse events.
Randomized Results: The trial was terminated after enrollment of 73 participants (37 to furosemide and 36 to placebo). Mean
Placebo (SD) age was 61.7 (14.3), 79.5% were medical admissions, mean (SD) APACHE II score was 26.6 (7.8), 90.4% re-
ceived mechanical ventilation and 61.6% received vasoactives. Groups were similar at baseline. No differences
were found in the proportion with worsening AKI (43.2% vs. 37.1%, p = 0.6), kidney recovery (29.7% vs. 42.9%,
p = 0.3), or RRT (27.0% s. 28.6%, p = 0.8). Adverse events, mostly electrolyte abnormalities, were more common
in furosemide-treated patients (p b 0.001). Protocol deviations were common, due often to supplementary furo-
semide.
Conclusions: In this pilot trial, furosemide did not reduce the rate of worsening AKI, improve recovery or reduce
RRT; however, was associated with greater electrolyte abnormalities.
Trial registration: ClinicalTrials.gov Identifier: NCT00978354 registered September 9, 2014.
© 2017 Elsevier Inc. All rights reserved.

1. Introduction repair and recovery for AKI, few, if any, have proven effective to favor-
ably modify the course or outcome once AKI is established [1-3]. How-
Acute kidney injury (AKI) is a frequent and increasingly encountered ever, there remain important knowledge gaps in our understanding of
complication of critical illness, with an incremental risk for morbidity the pathophysiology, case-mix identification, and response to interven-
and chronic kidney disease (CKD) [1]. Despite investigation of numer- tions commonly used in AKI [4].
ous preventative and therapeutic interventions to prevent or facilitate Loop diuretics, most commonly furosemide [5-7], are received by an
estimated 60–70% of patients with AKI in intensive care unit (ICU) set-
tings, in spite of a contradictory evidence-base on their effectiveness
and safety [8-10].
Furosemide acts at the medullary thick ascending loop of Henle to
⁎ Corresponding author.
inhibit the Na+/K+/2Cl− pump on the luminal cell membrane surface.
E-mail addresses: bagshaw@ualberta.ca, @drseanbagshaw (S.M. Bagshaw),
ngibney@ualberta.ca (R.T.N. Gibney), Peter.Kruger@health.qld.gov.au (P. Kruger),
Experimental data and small clinical studies have shown AKI increases
ihassan@ualberta.ca (I. Hassan), fmcalist@ualberta.ca (F.A. McAlister), kidney metabolic demand and oxidative stress [11], and this may be at-
rinaldo.bellomo@austin.org.au (R. Bellomo). tenuated by furosemide [12-14]. Furosemide reduces renal tubular

http://dx.doi.org/10.1016/j.jcrc.2017.07.030
0883-9441/© 2017 Elsevier Inc. All rights reserved.
S.M. Bagshaw et al. / Journal of Critical Care 42 (2017) 138–146 139

oxygen demand by impairing tubular Na+ reabsorption [14], and may 2.2. Randomization and blinding
attenuate ischemia/reperfusion-induced apoptosis and associated gene
transcription in AKI [15,16]. In contrary, recent data have suggested Eligible participants were randomly allocated in a 1:1 ratio to furose-
bolus furosemide aimed at augmenting urine output may increase kid- mide (bolus followed by continuous infusion) or placebo-control (0.9%
ney oxidative stress [17]. saline). Computerized random blocks of variable size (4–8) were strat-
Small clinical studies have further suggested that furosemide may ified within each site and by the presence of sepsis. Randomization was
attenuate the severity or shorten the duration of AKI, and perhaps performed by an unblinded clinical trial pharmacist using a web-based
delay or ameliorate need for renal replacement therapy (RRT); howev- application through the Epidemiology Coordinating and Research Cen-
er, findings have been inconsistent and improvements in survival or tre (EPICORE) at the University of Alberta (available at: https://www.
renal recovery have yet to be confirmed [8-10,18-22]. epicore.ualberta.ca/home/). This was concealed from study
There has recently been recognition of the need for greater invest- investigators.
ment in strategies to improve outcomes in AKI from randomized trials
[4,23,24]. Arguably, scientific focus should also aim to improve our un- 2.3. Procedures
derstanding of common therapies received by patients with AKI, such
as furosemide [17]. The furosemide administration protocol was adapted from a prior
Accordingly, we performed a pilot randomized trial in critically ill trial [26]. Each furosemide infusion bag contained 2000 mg of furose-
patients with mild AKI to compare the efficacy and safety of continuous mide in 500 mL of 0.9% saline for a final concentration of 4 mg/mL
infusions of furosemide or placebo titrated to urine output on the inci- (Item S1). To ensure blinding of investigators and treating clinicians,
dence of worsening AKI. We further aimed to address the feasibility of study infusion bags for furosemide and placebo appeared identical and
such an approach, including gaining understanding of clinician equi- were identified by a 4-digit coded identifier.
poise for use of furosemide in critically ill patients with AKI. Each participant received a weight-based loading dose of 0.4 mg/kg
followed by a continuous infusion commenced at a dose of
0.05 mg/kg/h. Estimated ideal body weight (IBW), based on the formula
2. Materials and methods described by Devine [27], was used to determine the urine output target
(Tables S2–S4). The study infusion was titrated to achieve and maintain
Details of the trial protocol were published and registered a target urine output in the range of 1.0–2.0 mL/kg/h. The maximum in-
(ClinicalTrials.gov Identifier: NCT00978354 registered September 14, fusion rate was 0.40 mg/kg/h. Urine output was assessed hourly. The
2009) [25]. The Human Research Ethics Committee at each site treatment algorithm for titration of the continuous infusion is shown
reviewed and approved the study prior to commencement (University in the Table S5.
of Alberta HREB file #Pro00002230). Written informed consent was ob- The study infusion was continued for a minimum of 24 h and
tained from all participants or their legal representatives. discontinued if any one of the following events occurred: 1) kidney re-
covery; 2) decision to start RRT; 3) death; 4) adverse reaction attributed
to study intervention; 5) ICU discharge; or 6) a total of 7-days of study
2.1. Study design, setting and participants drug had been administered.
All other aspects of patient management within the parameters
We performed a pilot randomized, blinded, placebo-controlled trial outlined (i.e., methods of fluid resuscitation, choice of fluids, vasoactive
at three tertiary university-affiliated hospital intensive care units therapy, choice of vasoactive therapy) were left to the discretion of the
(ICU): General Systems Intensive Care Unit, University of Alberta Hospi- treating ICU team. No additional interventions were performed.
tal (Edmonton, Canada), Intensive Care Unit, Austin Hospital (Mel-
bourne, Australia) and Intensive Care Unit, Princess Alexandra 2.4. Operational definitions
Hospital (Brisbane, Australia).
We screened patients admitted to a study ICU for eligibility. Inclusion Acute kidney injury (AKI) – AKI was defined and classified according
Criteria (all patients were required to fulfill the following criteria): 1) to the RIFLE criteria (acronym indicating Risk of renal dysfunction; Inju-
evidence of early AKI (RIFLE category – RISK); 2) peripheral or central ry to the kidney; Failure of kidney function; Loss of kidney function; and
intravenous catheter and urinary catheter; 3) ≥2 criteria for the system- End-stage kidney disease) [28]. The presence of early AKI was defined by
ic inflammatory response syndrome (SIRS) within 24 h of screening; a minimum of RIFLE category – RISK. Renal replacement therapy (RRT) –
and 4) achieved immediate resuscitation goals, based on the judgment RRT was defined as any form of extracorporeal kidney support provided
of the most responsible treating physician and including one or more to patients with documented AKI. By protocol and in order to minimize
of the following: fluid resuscitation and/or vasoactive therapy to the potential bias of clinician discretion on when to initiate RRT, at least
achieve mean arterial pressure (MAP) ≥65 mm Hg, central venous pres- one conventional indications for RRT was required to be fulfilled prior to
sure (CVP) ≥8 cm H2O, central venous oxygen saturation (ScvO2) ≥70% starting RRT (Item S1). Renal recovery – For the trial, renal recovery was
(if measured) and/or cardiac index (CI) ≥ 2.5 L/min/1.73 m2 (if mea- defined as the return of serum creatinine to within 10% of baseline levels
sured). Exclusion criteria included any of: 1) age b 18 years; 2) con- and a spontaneous urine output ≥1.0 mL/kg/h for a minimum of 24 h
firmed or suspected pregnancy (verified by serum [β-HCG] pregnancy and not receiving RRT. Systemic inflammatory response syndrome (SIRS)
test if indicated); 3) suspected or confirmed obstructive etiology for – To fulfill SIRS, any 2 or more of the SIRS criteria were required to be
AKI; 4) ≥stage 4 chronic kidney disease (CKD) (defined by an estimated fulfilled (see Item S1) [29]. Sepsis – The clinical syndrome of sepsis
glomerular filtration rate (eGFR) b30 mL/min/1.73 m2), end stage kid- was defined by the presence of suspected or confirmed infection and
ney disease (ESKD) receiving maintenance dialysis or kidney transplan- the presence of ≥2 SIRS criteria.
tation; 5) recent RRT during ICU or index hospitalization; 6) recovering
AKI, defined as a ≥ 25% or ≥ 44.2 μmol/L decline from peak increase in 2.5. Endpoints
serum creatinine; 7) acute pulmonary edema mandating urgent furose-
mide administration or RRT initiation or patient was already receiving a The primary endpoint was worsening AKI, defined as progression
continuous furosemide infusion; 8) moribund status with expected from RIFLE category – RISK to a more severe category of AKI (i.e.,
death within 24 h or significant limitations of medical therapy RIFLE categories – INJURY or FAILURE or receipt of RRT) in the 7 days fol-
(LOMT); 9) suspected or known allergy to furosemide; and 10) prior lowing randomization. Pre-specified secondary endpoints compared
enrolment. differences in cumulative fluid balance, serum electrolytes (i.e.,
140 S.M. Bagshaw et al. / Journal of Critical Care 42 (2017) 138–146

potassium, magnesium), acid-base status (i.e., pH, serum bicarbonate), recruitment and funding limitations. The termination was supported
rates of RRT initiation, rates of renal recovery, and hospital mortality be- by the Data Safety Monitoring Committee. These disruptions included:
tween furosemide and placebo groups, respectively. 1) secondment of research personnel to bedside clinical duties during
pandemic influenza A(H1N1) in October 2009 (6 months); and 2) a
2.6. Statistical analysis North American shortage of furosemide due to generic drug maker
Sandoz Canada suspending production in October 2011, along with in-
Based on observational data, we conservatively estimated that 60% ability to import furosemide and restricted access imposed by Alberta
of critically ill patients with AKI would worsen and progress [30]. We es- Health Services (12 months).
timated that furosemide would contribute to a 20% absolute reduction No patients withdrew consent. One patient allocated to the placebo
in the proportion who progress from RIFLE class – RISK. This required arm did not receive the study intervention. The primary endpoint was
a sample size of 214 patients to provide 80% power (alpha 0.05) [25]. available for all 72 participants and included in the final intention-to-
Statistical analysis was performed using the principle of intention- treat analysis.
to-treat. Normally or near normally distributed variables are reported
as means with standard deviations (SD) and compared using the t-
3.1. Baseline characteristics
test. Continuous data that were far from normality were reported as
medians with inter-quartile ranges (IQR) and compared using the
The mean (SD) age of enrolled participants was 61.7 (14.3), 16
Mann Whitney U test. Hodge-Lehmann estimate of location shift with
(21.9%) were female, mean (SD) APACHE II score was 26.6 (7.8), mean
its confidence interval was reported along with the Mann Whitney U
(SD) sequential organ failure assessment (SOFA) score was 10.8 (3.7),
test's p-value. Categorical data, including the primary endpoint, and
58 (79.5%) were medical admissions, 66 (90.4%) were receiving me-
need for RRT, renal recovery and hospital mortality, were reported as
chanical ventilation and 45 (61.6%) were receiving vasoactive therapy,
proportions and compared using Chi-square test. Fisher's Exact test
respectively.
was used when small expected frequencies invalidates the Chi-square
Following randomization, baseline characteristics, acute physiology,
test. Multi-variable logistic regression was used to control for confound-
laboratory parameters and treatment intensity were comparable be-
ing due to imbalances in baseline characteristics after randomization.
tween furosemide and placebo groups (Tables 1 and 2). Participants al-
located to furosemide had more exposure to vancomycin (67.6% vs.
2.7. Role of the funding source
38.9%, p = 0.01).
The funder had no role in the design, data collection, data analysis,
interpretation or in the writing of the manuscript. The corresponding 3.2. Processes of care
author (SMB) and biostatistician (IH) had full access to the data.
In the 72 h prior to randomization, 30 patients (41.0%) had been ex-
3. Results posed to furosemide (38.9% for furosemide vs. 48.5% for placebo, p =
0.5). Urine output in the 6 h preceding randomization was similar be-
Between September 1, 2009 and June 2, 2014, we enrolled 73 partic- tween the furosemide (210 mL [160–505]) and placebo (235 mL
ipants from 3 sites: 37 were randomly allocated to receive furosemide [154–443]) groups (p = 0.9). In total, 69.4% had an episode of oliguria
and 36 to placebo (Fig. 1). (≤0.5 mL/kg/h × 6 h) in the 72 h prior to randomization (77.8% for furo-
The trial was terminated early due to feasibility of achieving recruit- semide vs. 61.1% for placebo, p = 0.3). Cumulative fluid balance in the
ment following two unavoidable enrollment disruptions to trial 24 h prior to randomization was a median (IQR) 2741 mL (1136–

Fig. 1. Trial profile (consort diagram).


S.M. Bagshaw et al. / Journal of Critical Care 42 (2017) 138–146 141

Table 1 Table 2
Baseline clinical characteristics by treatment arm. Pre-randomization baseline physiological and laboratory values by treatment arm.

Variable Furosemide Placebo Variable Furosemide Placebo


(n = 37) (n = 36) (n = 37) (n = 36)

Age (years), (median, IQR) 61.9 (49.5–69.6) 65.5 (54.9–71.2) Serum creatinine (mg/dL) (median, IQR) 1.69 (1.07–2.79) 1.88 (1.13–2.63)
Male sex (n, %) 30 (81.1) 27 (75.0) (n = 37) (n = 36)
Weight (kg), (median, IQR) 82.0 (70.0–100.0) 83.7 (72.3–95.3) Urine output (mL) 210 (160–505) 235 (154–443)
No. of comorbid diseases (median, IQR) 3 (2–4) 3.5 (2–5) (6 h pre-randomization) (median, IQR) (n = 37) (n = 36)
Susceptibilities for AKI Fluid balance (6 h pre-randomization) 709 (434–954) 505 (248–988)
Diabetes mellitus (n, %) 7 (19.4) 10 (28.6) (median, IQR) (n = 37) (n = 35)
Chronic liver disease (n, %) 6 (16.7) 5 (14.3) Cumulative fluid balance 6363 (3325–9496) 5906 (2760–8979)
Heart failure (n, %) 4 (11.1) 2 (5.7) (from ICU admission) (median, IQR) (n = 37) (n = 35)
Chronic kidney disease (n, %) 0 (0) 2 (5.7) Blood urea nitrogen (mg/dL) 34.2 (17.9–52.4) 34.2 (25.4–45.7)
Hospital exposures for AKI (median, IQR) (n = 36) (n = 36)
ACEi/ARB (n, %) 10 (27.8) 11 (31.4) Serum Sodium (mmol/L) (median, IQR) 141 (136–143) 139 (134–142)
NSAID (n, %) 5 (13.5) 10 (27.8) (n = 36) (n = 36)
Radiocontrast media (n, %) 12 (33.3) 12 (34.3) Serum Potassium (mmol/L) 3.8 (3.6–4.4) 4.1 (3.6–4.8)
Aminoglycoside/amphotericin (n, %) 4 (11.1) 4 (11.1) (median, IQR) (n = 36) (n = 36)
Vancomycin (n, %) 25 (67.6) 4 (11.1) Serum Chloride (mmol/L) (median, IQR) 110 (102−113) 107 (103−111)
Rhabdomyolysis (CK N 5000 U/L) (n, %) 0 (0) 2 (5.6) (n = 36) (n = 36)
Primary diagnostic category Serum Bicarbonate (mEq/L) 22.7 (20.3–24.1) 22.8 (19.9–25.1)
Cardiovascular (n, %) 4 (10.8) 4 (11.1) (median, IQR) (n = 36) (n = 36)
Respiratory (n, %) 10 (27.0) 13 (36.1) Serum Magnesium (mmol/L) 0.99 (0.81–1.15) 1.01 (0.82–1.17)
Gastrointestinal (n, %) 1 (2.7) 5 (13.9) (median, IQR) (n = 32) (n = 35)
Neurological (n, %) 6 (16.2) 1 (2.8) Serum pH (median, IQR) 7.40 (7.36–7.44) 7.40 (7.34–7.46)
Sepsis (n, %) 12 (32.4) 6 (16.7) (n = 35) (n = 35)
Trauma (n, %) 3 (8.1) 3 (8.3) Base Excess (median, IQR) −1.0 (−4.0–1.0) −1.4 (−6.0–2.0)
Metabolic/poisoning (n, %) 0 3 (8.3) (n = 35) (n = 34)
Hematologic (n, %) 1 (2.7) 0 Serum hemoglobin (lowest) (g/L) 96.0 (90.0–111.0) 105.0 (89.0–120.0)
Other (n, %) 0 1 (2.8) (median, IQR) (n = 36) (n = 36)
Admission type Serum platelets (lowest) (109/mL) 132.0 (73.0–215.0) 146.0 (88.0–227.0)
Medical (n, %) 31 (83.8) 27 (75.0) (median, IQR) (n = 35) (n = 35)
Emergency surgical (n, %) 5 (13.5) 4 (11.1) White cell count (highest) (109/mL) 12.5 (7.7–17.0) 12.8 (8.5–18.0)
Elective surgical (n, %) 1 (2.7) 5 (13.9) (median, IQR) (n = 36) (n = 36)
APACHE II score (mean, SD) 25.6 (7.4) 27.7 (8.1) Serum bilirubin (highest) (mg/dL) 1.55 (0.82–2.92) 1.32 (0.76–2.22)
SOFA score (mean, SD) 10.9 (3.5) 10.7 (4.0) (median, IQR) (n = 34) (n = 36)
Mechanical ventilation (n, %) 33 (89.2) 33 (91.7) Serum lactate (highest) (mmol/L) 2.5 (1.4–3.8) 2.60 (1.65–4.60)
Vasopressors/inotropes (n, %) 23 (62.2) 22 (62.9) (median, IQR) (n = 35) (n = 32)
FiO2 (highest) (%) (median, IQR) 0.65 (0.45–1.00) 0.70 (0.60–0.80)
Abbreviations: IQR = intraquartile range; AKI = acute kidney injury; ACEi = ACE inhibi-
(n = 34) (n = 33)
tor; ARB = angiotensinogen receptor blocker; NSAID = non-steroidal anti-inflammatory
PaO2 (lowest) (mm Hg) (median, IQR) 75.0 (65.0–86.0) 71.0 (57.0–81.0)
drug; APACHE = Acute Physiology and Chronic Health Evaluation; SOFA = Sequential
(n = 35) (n = 34)
Organ Failure Assessment.
PaCO2 (highest) (mm Hg) (median, IQR) 44.0 (37.0–49.0) 44.0 (39.0–49.0)
(n = 34) (n = 34)

4512) and 2002 mL (755–4479) in the furosemide and placebo groups


(p = 0.5), respectively. 3.3. Primary and secondary endpoints
The median (IQR) duration of study treatment was 2 days (2–3) in
the furosemide group and 3 days (2–5) in the placebo group (p = There was no statistically significant difference in the primary end-
0.2). Among those allocated to furosemide, the median (IQR) total point of worsening AKI between furosemide and placebo groups (16
dose of furosemide was 1.65 mg/kg (0.7–4.8) and 3.2 mg/kg (1.4– [43.2%] vs. 13 [37.1%]; OR 1.52, 95% CI, 0.61–3.83, p = 0.48) (Table 3).
16.1) in the first 24 h and over the study period, respectively. The There was also no difference in worsening AKI in the subgroup with sep-
urine output in the first 6 h (745 mL [369–1360] vs. 210 mL [149– sis (14 [43.8%] vs. 11 [37.9%]; OR 1.28; 95% CI 0.50–3.30). In multivari-
425], p b 0.001) and first 24 h (3006 mL [1475–5035] vs. 1165 [758– able analysis, adjusting for APACHE II score (OR 1.14; 95% CI, 0.42–
2100], p = 0.002) after randomization was significantly greater in the 3.00), vancomycin exposure (OR 1.08; 0.39–2.98), pre-randomization
furosemide compared with placebo group. exposure to furosemide (OR 1.39; 95% CI, 0.52–3.76) or pre-randomiza-
There were no significant differences in organ failure scores, hemo- tion oliguria (OR 1.18; 95% CI, 0.45–3.15, p = 0.7) made no difference.
dynamic profile, or fluid balance between groups during the study inter- No statistically significant differences were observed across second-
vention (Fig. 2). While there were no significant differences in serum ary endpoints, including kidney recovery, RRT utilization, mortality or a
creatinine and potassium levels during the study intervention, those al- composite of RRT and/or mortality (Table 3). While there was potential-
located to furosemide showed higher serum sodium and bicarbonate ly a clinically meaningful difference in cumulative fluid balance
levels (Fig. 3). (−1081 mL [95% CI, −2697 to 467]), this was not statistically signifi-
Similar proportions of patients in furosemide and placebo groups cant. There were no significant between group differences in maximal
received potassium supplementation in the first 24 h (46.0% vs. 48.6%, changes in electrolytes and acid-base status (Table 4).
p = 1.0) and 48 h (62.5% vs. 60.0%, p = 1.0) after randomization; how-
ever, the furosemide group received greater a cumulative dose over the
first 48 h (124 mmol [60–206] vs. 80 mmol [31 − 110], p = 0.006). 3.4. Adverse events and protocol deviations
There was no difference in the proportion receiving magnesium
supplementation (24 h: 18.9% vs. 17.1%, p = 1.0; 48 h: 12.5% vs. There was no difference in the proportion of patients with adverse
20.0%, p = 0.5) or in the cumulative dose in the first 48 h (5 g [3-5] events (p = 0.6); however, the incidence rate was higher in those re-
vs. 5 g [2-8], p = 0.5). ceiving furosemide (p b 0.001) (Table 5). In the furosemide arm, 25
142 S.M. Bagshaw et al. / Journal of Critical Care 42 (2017) 138–146

Fig. 2. Changes in daily organ failure, hemodynamic parameters and fluid balance stratified by study intervention. A) SOFA score; B) mean arterial pressure; C) central venous pressure; and
D) daily fluid balance (Error bars show 95% CI for mean differences between groups on each day of study intervention).

(67.6%) participants had 111 adverse events reported (incidence rate however, we found no differences between furosemide and placebo-
4.4 events/patient), of which 2 (5.4%) contributed to study intervention treated groups. A substantial number of adverse events occurred, pre-
discontinuation. In the placebo arm, 22 (61.1%) participants had 57 ad- dominantly attributable to minor electrolyte imbalances, with rates
verse events reported (incidence rate 2.6 events/patient), of which 1 higher among those receiving furosemide. Similarly, protocol deviations
(2.8%) contributed to study intervention discontinuation. The majority frequently occurred in both groups. These were largely due to non-ad-
of adverse events were related to occurrence of electrolyte abnormali- herence to the study algorithm and extra (i.e., non-protocol) furose-
ties in both groups. Protocol deviations were common; however, not mide use.
significantly different between groups (p = 0.8). The majority were re-
lated to deviation in the treatment algorithm. Compared with the furo-
semide arm, those allocated to placebo were more likely to receive 4.1. Context with prior literature
supplementary diuretic therapy (10.8% vs. 30.6% vs. p = 0.046).
The necessity for additional clinical trials specifically focused on
unraveling the optimal use, efficacy and safety of furosemide in AKI
4. Discussion stems from an apparent disconnect in the widespread utilization of fu-
rosemide in practice juxtaposed with evidence suggestive of no clinical
There is a pathophysiological mechanistic basis for giving low-dose benefit and potential harm [5,31-33]. Early clinical trials of furosemide
furosemide in early AKI to reduce kidney metabolic work [14-16] and use in AKI were small, confounded by co-interventions (i.e., mannitol,
we hypothesized that it may mitigate worsening AKI and facilitate ear- dopamine), characterized by late administration (i.e., long durations of
lier recovery of kidney function. We tested this hypothesis in a multi- oliguria or already receiving RRT), and by use of large bolus furosemide
center pilot randomized trial of critically ill patients with early AKI com- doses with no specific titration of therapy to physiologic endpoints such
paring continuous furosemide infusion to placebo-control on the occur- as urine output [8]. Such data lack generalizability to AKI and current fu-
rence of worsening AKI [25]. rosemide practices in ICU settings.
We found worsening AKI was common in this cohort of critically ill Observational studies detailing the risk of furosemide for develop-
patients with AKI, occurring in 39.7%, but with no difference between ment of AKI or poor outcome associated with its use are almost certainly
the furosemide and placebo-control groups (43.2% vs. 37.1%, p = 0.6), susceptible to confounding by indication [5,33]. In these studies, the risk
respectively. We did not find any clinically important differences in of death or non-recovery was predominantly borne by patients with
key secondary endpoints including kidney recovery, duration of AKI, more severe oliguric AKI who did not show a robust response to initial
or mortality. RRT use was also frequent, initiated in 27.4% of patients; diuretic challenge [5,33]. It is likely many of these patients had delay
S.M. Bagshaw et al. / Journal of Critical Care 42 (2017) 138–146 143

Fig. 3. Changes to daily biochemical profile stratified by study intervention. A) in serum creatinine; B) serum sodium; C) serum potassium; and D) serum bicarbonate (Error bars show 95%
CI for mean differences between groups on each day of study intervention).

to timely start of RRT or had unmeasured differences in probability of Indeed, in a cohort of acute lung injured patients with concomitant
being offered RRT. Consistent with the findings of our pilot trial, addi- AKI, after adjusting for fluid balance, there was no ceiling dose of furose-
tional observational studies have not shown significant differences in mide above which mortality was shown to increase [34].
outcomes associated with furosemide exposure and have failed to con- While our pilot trial was clearly underpowered and we did not show
firm any specific incremental risk with furosemide use in AKI [6,7,34]. furosemide reduced the severity or shortened the duration of AKI, furo-
semide may still have an important role for managing fluid balance and
to counterbalance the obligatory fluid intake and optimal delivery of nu-
trition routinely given to critically ill AKI patients [35,36]. Moreover,
Table 3
Summary of study outcomes.
loop diuretics such as furosemide are a common strategy used by clini-
cians to avert starting RRT among those with severe AKI [37,38]. How-
Variable Furosemide Placebo OR/difference p-Value ever, it remains uncertain whether furosemide exposure in this
(n = 37) (n = 36) (95% CI)
context portends risk or benefit for kidney recovery or mortality.
Worsened AKI (n, %)a 16 (43.2) 13 (37.1) – 0.6
Cumulative fluid balance 877 (−515 2407 (−522 −1081 0.2
(mL) (med [IQR]) to 2920) to 4383) (−2697 to
467) 4.2. Implications for clinicians and research
Kidney recovery (n, %)b 11 (29.7) 15 (42.9) – 0.3
Time to recovery (h) 48.6 54.6 4 (−33.5 to 0.7 Despite survey data suggesting clinicians had sufficient equipoise to
(median, IQR) (42.9–82.4) (43.5–82.7) 19.4) enroll AKI patients in such a trial, we found this protocol was challeng-
Received RRT (n, %) 10 (27.0) 10 (28.6) – 0.9
ing to implement [39,40]. Moreover, our data suggested a continuous
ICU death (n, %) 3 (8.1) 6 (17.1) – 0.3
Hospital death (n, %) 3 (8.1) 5 (14.3) – 0.5 furosemide infusion is not likely to prevent worsening AKI; however,
Mortality at 90-days (n, 8 (21.6) 11 (30.5) – 0.4 it may still represent an important approach to maintaining fluid ho-
%) meostasis. Clinicians appeared concerned about patient fluid status with
Received RRT or death at 17 (46.0) 19 (54.3) – 0.5
numerous occurrences of non-protocol administration of furosemide
90-days (n, %)
[35]. We believe these observations align with recent data showing di-
a
Worsening RIFLE category (primary endpoint) - defined as any worsening in RIFLE uretic therapy did not confer incremental risk of death or non-recovery
category and/or receipt of RRT.
b
Renal recovery – the operational definition of renal recovery will be the return of se-
in selected critically ill patients when aimed at mitigating fluid accumula-
rum creatinine to within 10% of baseline levels and a spontaneous urine output tion and complications of fluid overload [6,34]. Furosemide use in ICU set-
≥1.0 mL/kg/h for a minimum of 24 h independent of RRT. tings is predominantly triggered in response to fluid accumulation in the
144 S.M. Bagshaw et al. / Journal of Critical Care 42 (2017) 138–146

Table 4
Summary of physiological/laboratory process outcomes.

Variablea Furosemide Placebo Difference p-Value


(n = 37) (n = 36) (95% CI)

Largest Δ serum potassium (mean, SD) 0.59 (0.68) 0.62 (0.65) −0.03 (−0.34 to 0.29) 0.9
(n = 36) (n = 35)
Largest Δ serum magnesium (mean, SD) 0.13 (0.20) 0.07 (0.28) 0.06 (−0.06 to 0.18) 0.3
(n = 32) (n = 34)
Largest Δ serum pH (mean, SD) 0.03 (0.06) 0.06 (0.07) −0.02 (−0.06 to 0.01) 0.2
(n = 35) (n = 33)
Largest Δ serum bicarbonate (mean, SD) 0.35 (3.29) 0.30 (2.90) 0.05 (−1.42 to 1.52) 0.9
(n = 36) (n = 35)
a
Largest Δ referenced from baseline value at randomization to lowest observed value.

context of secondary organ injury (i.e., acute lung injury) rather than iso- type I error and has suboptimal statistical power. In addition, we expe-
lated AKI per se [40]. More recently, the urine output response to a bolus rienced a high rate of protocol deviations, highlighting the challenges
dose of furosemide (i.e., furosemide stress test [FST]) has shown utility as with a trial of furosemide administration in critically ill patients. Finally,
a functional dynamic measure to predict AKI severity [41,42]. In critically while allocation was concealed and the intervention blinded, it is plau-
ill patients with AKI, non-response to a furosemide challenge may repre- sible that unblinding may have occurred in selected patients with brisk
sent a logical prompt for support escalation and starting RRT [36,43]. Fu- urine output responses to receiving furosemide; however, our primary
ture research should focus on the ideal interventional strategies for fluid outcome was objectively measured and analysts were blinded to
balance management incorporating the use loop diuretics among critical- group assignment.
ly ill patients with AKI [36].
5. Conclusions
4.3. Strengths and limitations
In summary, in this pilot study, furosemide infusion in critically ill
Our study had a number of strengths, including being multi-centric, patients with early AKI did not reduce the incidence of worsening AKI
randomized, placebo-controlled, obtaining complete ascertainment of or favorably impact any secondary endpoints including kidney recovery,
primary endpoints and evaluating a biologically plausible and titratable duration of AKI, cumulative fluid balance, RRT use or mortality. In the
approach to furosemide administration in critically ill patients with AKI ICU setting, further research should aim to evaluate the ideal contribu-
[25]. However, our trial also has noteworthy limitations. Our trial was tion for furosemide in the routine management of critically ill patients
small and was prematurely terminated, and as such, is predisposed to with AKI, such as maintaining fluid balance homeostasis.

Table 5
Summary of adverse events and protocol deviations/violations.

Variable Furosemide Placebo


(n = 37) (n = 36)

No. events No. patients No. patients discontinued No. events No. patients No. patients discontinued
(n = 111) (n = 25) (n = 2) (n = 57) (n = 22) (n = 1)

Adverse eventsa
Drug reaction 1 1 1 1 1 1
Tinnitus 0 0 0 1 1 0
Arrhythmia – SVT 4 3 0 5 4 0
Arrhythmia – VT/VF 2 2 0 0 0 0
Cardiopulmonary arrest 0 0 0 0 0 0
Other 0 0 0 1 2 1
Electrolyte abnormalities 104b 22c 1 49b 14c 0
Serum sodium ≥ 150 mmol/L 53 9 1 1 1 0
Serum potassium b 3.0 mmol/L 7 4 0 2 2 0
Serum magnesium b 0.7 mmol/L 2 2 0 6 4 0
Serum bicarbonate ≥ 30 or pH ≥ 7.5 62 18 1 40 10 0

Variable Furosemide Placebo


(n = 37) (n = 36)

No. events No. patients No. patients discontinued No. events No. patients No. patients discontinued
(n = 171) (n = 28) (n = 7) (n = 201) (n = 29) (n = 3)

Protocol deviation/violationa
Did not receive study intervention 0 0 0 1 1 1
Supplementary diuretic therapy given 12 4 0 76 11 0
Study intervention interrupted 4 4 0 7 6 1
Study intervention discontinued 7 7 7 2 2 0
Study algorithm deviation 123 22 0 103 18 1
Other 25 11 0 12 6 0
a
Numbers in columns of adverse events and protocol deviations/violations are not mutually exclusive.
b
More than one electrolyte abnormality was reported with each discrete adverse event episode.
c
More than one adverse event was reported per patient.
S.M. Bagshaw et al. / Journal of Critical Care 42 (2017) 138–146 145

Abbreviations Nephrology. FAM was supported by a Senior Health Scholar Award from
AKI acute kidney injury Alberta Innovates – Health Solutions and the Capital Health/University
CKD chronic kidney disease of Alberta Chair in Cardiovascular Outcomes Research. We thank Dr.
CVP central venous pressure Ross Tsuyuki (Director) and the staff at EPICORE for facilitation data co-
ESKD end-stage kidney disease ordination and management. We thank Dr. Andrew Shaw (chair), Dr.
IBW ideal body weight Patrick Murray and Dr. Paul Miles for serving on the Data Safety Moni-
ICU intensive care unit toring Committee.
IQR intraquartile range The following are the participating centers, investigators and coordi-
LOMT limitation of medical therapy nators for the SPARK study: University of Alberta Hospital (Edmonton):
MAP mean arterial pressure Sean M. Bagshaw, R.T. Noel Gibney, Finlay A. McAlister, Samantha Tay-
RIFLE risk, injury, failure, loss, end-stage lor, Nadia Baig; Kristen Reid; Austin Hospital (Melbourne): Rinaldo
RRT renal replacement therapy Bellomo, Glenn Eastwood; Princess Alexandra Hospital (Brisbane): Peter
ScvO2 central venous oxygen saturation Kruger; Jason Meyer.
SD standard deviation
SIRS systemic inflammatory response syndrome Appendix A. Supplementary data
SOFA sequential organ failure assessment
Supplementary data to this article can be found online at http://dx.
Ethics approval and consent to participate doi.org/10.1016/j.jcrc.2017.07.030.

The Human Research Ethics Committee at each site reviewed and References
approved the study prior to commencement (University of Alberta
HREB file # Pro00002230). Written informed consent was obtained [1] Rewa O, Bagshaw SM. Acute kidney injury-epidemiology, outcomes and economics.
Nat Rev Nephrol 2014;10(4):193–207.
from all participants or their legal representatives. [2] Abel RM, Beck Jr CH, Abbott WM, Ryan Jr JA, Barnett GO, Fischer JE. Improved surviv-
al from acute renal failure after treatment with intravenous essential L-amino acids
Consent for publication and glucose. Results of a prospective, double-blind study. N Engl J Med 1973;
288(14):695–9.
[3] Lameire NH, Bagga A, Cruz D, De Maeseneer J, Endre Z, Kellum JA, et al. Acute kidney
Not applicable. injury: an increasing global concern. Lancet 2013;382(9887):170–9.
[4] Palevsky PM, Molitoris BA, Okusa MD, Levin A, Waikar SS, Wald R, et al. Design of
clinical trials in acute kidney injury: report from an NIDDK workshop on trial meth-
Availability of data and material
odology. Clin J Am Soc Nephrol 2012;7(5):844–50.
[5] Mehta RL, Pascual MT, Soroko S, Chertow GM, Group PS. Diuretics, mortality, and
The datasets used and/or analyzed during the current study are nonrecovery of renal function in acute renal failure. JAMA 2002;288(20):2547–53.
available from the corresponding author on reasonable request. [6] Teixeira C, Garzotto F, Piccinni P, Brienza N, Iannuzzi M, Gramaticopolo S, et al. Fluid
balance and urine volume are independent predictors of mortality in acute kidney
injury. Crit Care 2013;17(1):R14.
Competing interests [7] Uchino S, Doig GS, Bellomo R, Morimatsu H, Morgera S, Schetz M, et al. Diuretics and
mortality in acute renal failure. Crit Care Med 2004;32(8):1669–77.
[8] Bagshaw SM, Delaney A, Haase M, Ghali WA, Bellomo R. Loop diuretics in the man-
SMB reports having received honoraria and has consulted for Baxter agement of acute renal failure: a systematic review and meta-analysis. Crit Care
Healthcare Corp. Resusc 2007;9(1):60–8.
[9] Ho KM, Sheridan DJ. Meta-analysis of frusemide to prevent or treat acute renal fail-
ure. BMJ 2006;333(7565):420.
Funding [10] Sampath S, Moran JL, Graham PL, Rockliff S, Bersten AD, Abrams KR. The efficacy of
loop diuretics in acute renal failure: assessment using Bayesian evidence synthesis
This work was supported by a grant from the Alberta Innovates – techniques. Crit Care Med 2007;35(11):2516–24.
[11] Redfors B, Bragadottir G, Sellgren J, Sward K, Ricksten SE. Acute renal failure is NOT
Health Solutions (Grant File No. 200700566) and Intensivist Group in
an “acute renal success”—a clinical study on the renal oxygen supply/demand rela-
the Department of Critical Care Medicine, Faculty of Medicine and Den- tionship in acute kidney injury. Crit Care Med 2010;38(8):1695–701.
tistry, University of Alberta. The funding agency had no role in the de- [12] De Torrente A, Miller PD, Cronin RE, Paulsin PE, Erickson AL, Schrier RW. Effects of
furosemide and acetylcholine in norepinephrine-induced acute renal failure. Am J
sign or conduct of the study, in the collection, management, analysis
Physiol 1978;235(2):F131–136.
or interpretation of the data, or in the preparation, review or approval [13] Kramer HJ, Schuurmann J, Wassermann C, Dusing R. Prostaglandin-independent
of the manuscript. protection by furosemide from oliguric ischemic renal failure in conscious rats. Kid-
ney Int 1980;17(4):455–64.
[14] Sward K, Valsson F, Sellgren J, Ricksten SE. Differential effects of human atrial natri-
Authors' contributions uretic peptide and furosemide on glomerular filtration rate and renal oxygen con-
sumption in humans. Intensive Care Med 2005;31(1):79–85.
SMB designed the trial, obtained funding for the trial, obtained data, [15] Aravindan N, Aravindan S, Riedel BJ, Weng HR, Shaw AD. Furosemide prevents apo-
ptosis and associated gene expression in a rat model of surgical ischemic acute renal
analyzed the data, interpreted data, drafted the manuscript and provid- failure. Ren Fail 2007;29(4):399–407.
ed critical revision of the manuscript. FAM designed the trial, obtained [16] Aravindan N, Shaw A. Effect of furosemide infusion on renal hemodynamics and an-
funding for the trial, interpreted data, and provided critical revision of giogenesis gene expression in acute renal ischemia/reperfusion. Ren Fail 2006;
28(1):25–35.
the manuscript. RB designed the trial, obtained data, obtained funding [17] Silbert BI, Ho KM, Lipman J, Roberts JA, Corcoran TB, Morgan DJ, et al. Does furose-
for the trial, interpreted data, and provided critical revision of the man- mide increase oxidative stress in acute kidney injury? Antioxid Redox Signal
uscript. RTNG obtained funding for the trial, interpreted data, obtained 2016;26(5):221–6.
[18] Cantarovich F, Rangoonwala B, Lorenz H, Verho M, Esnault VL. High-dose
data, and provided critical revision of the manuscript. IH analyzed and
flurosemide in acute renal failure study G: high-dose furosemide for established
interpreted data, and provided critical revision of the manuscript. PK ARF: a prospective, randomized, double-blind, placebo-controlled, multicenter
interpreted data, and provided critical revision of the manuscript. All trial. Am J Kidney Dis 2004;44(3):402–9.
[19] Kleinknecht D, Ganeval D, Gonzalez-Duque LA, Fermanian J. Furosemide in acute
authors have ready and approved the final manuscript.
oliguric renal failure. A controlled trial. Nephron 1976;17(1):51–8.
[20] Shilliday IR, Quinn KJ, Allison ME. Loop diuretics in the management of acute renal
Acknowledgements failure: a prospective, double-blind, placebo-controlled, randomized study. Nephrol
Dial Transplant 1997;12(12):2592–6.
[21] Vargas Hein O, Staegemann M, Wagner D, von Heymann C, Martin M, Morgera S,
SMB was supported by a Clinical Investigator Award from Alberta In- et al. Torsemide versus furosemide after continuous renal replacement therapy
novates – Health Solutions and a Canada Research Chair in Critical Care due to acute renal failure in cardiac surgery patients. Ren Fail 2005;27(4):385–92.
146 S.M. Bagshaw et al. / Journal of Critical Care 42 (2017) 138–146

[22] van der Voort PH, Boerma EC, Koopmans M, Zandberg M, de Ruiter J, Gerritsen RT, [33] Wu VC, Lai CF, Shiao CC, Lin YF, Wu PC, Chao CT, et al. Effect of diuretic use on 30-day
et al. Furosemide does not improve renal recovery after hemofiltration for acute postdialysis mortality in critically ill patients receiving acute dialysis. PLoS One
renal failure in critically ill patients: a double blind randomized controlled trial. 2012;7(3):e30836.
Crit Care Med 2009;37(2):533–8. [34] Grams ME, Estrella MM, Coresh J, Brower RG, Liu KD, National Heart L. Blood Insti-
[23] Faubel S, Chawla LS, Chertow GM, Goldstein SL, Jaber BL, Liu KD. Acute Kidney Injury tute Acute Respiratory Distress Syndrome N: fluid balance, diuretic use, and mortal-
Advisory Group of the American Society of N: ongoing clinical trials in AKI. Clin J Am ity in acute kidney injury. Clin J Am Soc Nephrol 2011;6(5):966–73.
Soc Nephrol 2012;7(5):861–73. [35] Bouchard J, Soroko SB, Chertow GM, Himmelfarb J, Ikizler TA, Paganini EP, et al. Pro-
[24] Molitoris BA, Okusa MD, Palevsky PM, Chawla LS, Kaufman JS, Devarajan P, et al. Design gram to Improve Care in Acute Renal Disease Study G: fluid accumulation, survival
of clinical trials in AKI: a report from an NIDDK workshop. Trials of patients with sepsis and recovery of kidney function in critically ill patients with acute kidney injury.
and in selected hospital settings. Clin J Am Soc Nephrol 2012;7(5):856–60. Kidney Int 2009;76(4):422–7.
[25] Bagshaw SM, Gibney RT, McAlister FA, Bellomo R. The SPARK Study: a phase II ran- [36] Goldstein S, Bagshaw SM, Cecconi M, Okusa M, Wang H, Kellum J, et al. Pharmaco-
domized blinded controlled trial of the effect of furosemide in critically ill patients logical management of fluid overload. Br J Anaesth 2014;113(5):756–63.
with early acute kidney injury. Trials 2010;11:50. [37] Gaudry S, Hajage D, Schortgen F, Martin-Lefevre L, Pons B, Boulet E, et al. Initiation
[26] Ostermann M, Alvarez G, Sharpe MD, Martin CM. Frusemide administration in crit- strategies for renal-replacement therapy in the intensive care unit. N Engl J Med
ically ill patients by continuous compared to bolus therapy. Nephron Clin Pract 2016;375(2):122–33.
2007;107(2):c70–76. [38] Wald R, Adhikari NK, Smith OM, Weir MA, Pope K, Cohen A, et al. Comparison of
[27] Devine DJ. Gentimicin therapy. Drug Intell Clin Pharm 1974;8(11):650–5. standard and accelerated initiation of renal replacement therapy in acute kidney in-
[28] Bellomo R, Ronco C, Kellum JA, Mehta RL, Palevsky P. Acute Dialysis Quality Initiative jury. Kidney Int 2015;88(4):897–904.
w: Acute renal failure - definition, outcome measures, animal models, fluid therapy [39] Bagshaw SM, Delaney A, Jones D, Ronco C, Bellomo R. Diuretics in the management
and information technology needs: the Second International Consensus Conference of acute kidney injury: a multinational survey. Contrib Nephrol 2007;156:236–49.
of the Acute Dialysis Quality Initiative (ADQI) Group. Crit Care 2004;8(4):R204-12. [40] Jones SL, Martensson J, Glassford NJ, Eastwood GM, Bellomo R. Loop diuretic therapy
[29] Bone RC, Sprung CL, Sibbald WJ. Definitions for sepsis and organ failure. Crit Care in the critically ill: a survey. Crit Care Resusc 2015;17(3):223–6.
Med 1992;20(6):724–6. [41] Chawla LS, Davison DL, Brasha-Mitchell E, Koyner JL, Arthur JM, Shaw AD, et al. De-
[30] Hoste EA, Clermont G, Kersten A, Venkataraman R, Angus DC, De Bacquer D, et al. velopment and standardization of a furosemide stress test to predict the severity of
RIFLE criteria for acute kidney injury are associated with hospital mortality in criti- acute kidney injury. Crit Care 2013;17(5):R207.
cally ill patients: a cohort analysis. Crit Care 2006;10(3):R73. [42] Silbert BI, Ho KM, Lipman J, Roberts JA, Corcoran TB, Morgan DJ, et al. Determinants
[31] Levi TM, Rocha MS, Almeida DN, Martins RT, Silva MG, Santana NC, et al. Furosemide of urinary output response to IV furosemide in acute kidney injury: a pharmacoki-
is associated with acute kidney injury in critically ill patients. Braz J Med Biol Res netic/pharmacodynamic study. Crit Care Med 2016;44(10):e923-29.
2012;45(9):827–33. [43] Wald R, Bagshaw SM. The timing of renal replacement therapy initiation in acute
[32] Nisula S, Kaukonen KM, Vaara ST, Korhonen AM, Poukkanen M, Karlsson S, et al. In- kidney injury. Semin Nephrol 2016;36(1):78–84.
cidence, risk factors and 90-day mortality of patients with acute kidney injury in
Finnish intensive care units: the FINNAKI study. Intensive Care Med 2013;39(3):
420–8.

You might also like