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

CE: A.G.

; ANNSURG-D-16-02308; Total nos of Pages: 9;


ANNSURG-D-16-02308

ORIGINAL ARTICLE

Effects of Intraoperative Fluid Management on


Postoperative Outcomes
A Hospital Registry Study
Christina H. Shin, MD,  Dustin R. Long, MD,  Duncan McLean, MBChB,  y
Stephanie D. Grabitz, Cand. Med,  Karim Ladha, MD, MSc,z Fanny P. Timm, Cand. Med, 
Tharusan Thevathasan, Cand. Med,  Alberto Pieretti, MD,§ Cristina Ferrone, MD,§
Andreas Hoeft, MD, PhD,ô Thomas W. L. Scheeren, MD, PhD,jj Boyd Taylor Thompson, MD, 
Tobias Kurth, MD, ScD,yyzz and Matthias Eikermann, MD, PhD 

length of stay and total cost of care. Our models were adjusted for patient-,
Objective: Evaluate the dose-response relationship between intraoperative
procedure-, and anesthesia-related factors.
fluid administration and postoperative outcomes in a large cohort of surgical
Results: A U-shaped association was observed between the volume of fluid
patients.
administered intraoperatively and 30-day mortality, costs, and postoperative
Background: Healthy humans may live in a state of fluid responsiveness
length of stay. Liberal fluid volumes (highest quintile of fluid administration
without the need for fluid supplementation. Goal-directed protocols driven by
practice) were significantly associated with respiratory complications
such measures are limited in their ability to define the optimal fluid state
whereas both liberal and restrictive (lowest quintile) volumes were signifi-
during surgery.
cantly associated with acute kidney injury. Moderately restrictive volumes
Methods: This analysis of data on file included 92,094 adult patients under-
(second quintile) were consistently associated with optimal postoperative
going noncardiac surgery with endotracheal intubation between 2007 and
outcomes and were characterized by volumes approximately 40% less than
2014 at an academic tertiary care hospital and two affiliated community
traditional textbook estimates: infusion rates of approximately 6–7 mL/kg/hr
hospitals. The primary exposure variable was total intraoperative volume of
or 1 L of fluid for a 3-hour case.
crystalloid and colloid administered. The primary outcome was 30-day
Conclusions: Intraoperative fluid dosing at the liberal and restrictive margins
survival. Secondary outcomes were respiratory complications within three
of observed practice is associated with increased morbidity, mortality, cost,
postoperative days (pulmonary edema, reintubation, pneumonia, or respirat-
and length of stay.
ory failure) and acute kidney injury. Exploratory outcomes were postoperative
Keywords: acute kidney injury, cost, healthcare utilization, intraoperative
fluid management, length of stay, mortality, outcomes, postoperative
respiratory complications
From the Department of Anesthesia, Critical Care, and Pain Medicine, Massa-
chusetts General Hospital, Boston, MA; yDepartment of Anesthesiology, (Ann Surg 2017;xx:xxx–xxx)
University of Rochester Medical Center, Rochester, NY; zDepartment of
Anesthesia, Toronto General Hospital and University of Toronto, Toronto,
Canada; §Department of Surgery, Massachusetts General Hospital, Boston,
MA; ôDepartment of Anesthesiology and Intensive Care Medicine, University
Hospital Bonn, Bonn, Germany; jjDepartment of Anesthesiology, University
D uring surgery, maintenance of intravascular volume by intra-
venous fluid is necessary to mitigate hypovolemia caused by
osmotic loss, evaporation, and bleeding. However, excessive intra-
of Groningen, University Medical Center Groningen, Groningen, the Nether-
lands; Department of Medicine, Massachusetts General Hospital, Boston,
operative fluid administration can expose patients to the risks of acid-
MA; yyInstitute of Public Health, Charité Universitätsmedizin, Berlin, base disturbances and edema formation, increasing the distance
Germany; and zzDivision of Preventive Medicine, Brigham and Women’s between capillaries and cells and impairing gas exchange. Although
Hospital, Harvard Medical School, Boston, MA. adverse respiratory outcomes have generally been associated with
Disclosure: This study was funded by Jeff and Judy Buzen in an unrestricted grant
to Dr. Matthias Eikermann. The authors declare no conflicts of interest.
liberal fluid administration, fluid restriction may confer different but
Author Contributions: equally important risks such as organ hypoperfusion with successive
C.H.S., D.R.L., and D.McL. contributed equally in this study. C.H.S., D.R.L., and dysfunction and failure. Renal function is particularly susceptible to
D.McL. contributed to the design and conception of the study, the analysis and changes in volume status1 and hypovolemia may increase risk of
interpretation of data, and drafting of the work. K.L. and T.W.L.S. contributed
to the design and conception of the study. F.P.T. and S.D.G. contributed to the
postoperative kidney injury.2
analysis and the acquisition of data for the work. T.T. and A.P. contributed to The formulaic approach to balancing perioperative fluids
the acquisition of data. C.F., A.H., T.W.L.S., and B.T.T. contributed to the classically described in textbooks (based on fasting, urine output,
critical review and interpretation of the work. T.K. contributed to design and blood loss, evaporative loss, and so on) has since been supplanted by
conception of the study, data analysis and interpretation of data. M.E. is the
guarantor of the paper; he takes responsibility for the integrity of the work as a
a notion that more restrictive approaches may yield improved out-
whole, from inception to published article. M.E. served as clinical research comes.3 –5 However, prospective trials of empiric dosing strategies
mentor to C.H.S., D.R.L., and D.McL. All authors revised the work critically have utilized varying definitions of ‘‘liberal’’ versus ‘‘restrictive,’’
for important intellectual content. been limited by small sample sizes,6 and yielded conflicting results.7
Supplemental digital content is available for this article. Direct URL citations
appear in the printed text and are provided in the HTML and PDF versions of
‘‘Goal-directed fluid therapy’’ (GDFT) has emerged as an approach
this article on the journal’s Web site (www.annalsofsurgery.com). to monitoring fluid-recruitable cardiac output with the potential
Reprints: Matthias Eikermann, MD, PhD, Department of Anesthesia, Critical Care, advantage of identifying patients who may benefit from additional
and Pain Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, fluid administration.8,9 However, healthy humans have significant
MA, 02114. E-mail: meikermann@partners.org.
Copyright ß 2017 Wolters Kluwer Health, Inc. All rights reserved.
preload reserve and may be fluid responsive at baseline, functioning
ISSN: 0003-4932/16/XXXX-0001 on the ascending limb of the Frank-Starling curve without the need
DOI: 10.1097/SLA.0000000000002220 for fluid supplementation10 Therefore, as demonstrated in one recent

Annals of Surgery  Volume XX, Number XX, Month 2017 www.annalsofsurgery.com | 1

Copyright © 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: A.G.; ANNSURG-D-16-02308; Total nos of Pages: 9;
ANNSURG-D-16-02308

Shin et al Annals of Surgery  Volume XX, Number XX, Month 2017

FIGURE 1. Study flow diagram illustrat-


ing surgical cohort used for data analysis.
ASA indicates American Society of Anes-
thesiologists physical classification score;
BMI, body mass index.

trial,11 fluid administration to all patients who are fluid responsive representing incremental quintiles of the exposure variable: restric-
may still increase the risks of fluid overload without conferring tive (quintile 1, Q1), moderately restrictive (quintile 2, Q2), moderate
additional benefit. Although lack of fluid responsiveness may mark (quintile 3, Q3), moderately liberal (quintile 4, Q4), and liberal
the absolute upper bound of safe practice, the optimal state, and the (quintile 5, Q5).
safe lower limit of fluid restriction in this setting remain unclear. For
these reasons, great variability persists in the perioperative dosing of Outcome Measures
intravenous fluid,12 which cannot be explained by patient- or case- The primary outcome was mortality within 30 days after
specific factors. Such high variance between providers normally surgery. Secondary outcomes were PRCs and AKI. PRCs were
indicates uncertainty and implies a high potential for improvement. defined as the occurrence of pneumonia, pulmonary edema, respir-
We hypothesized that variation in intraoperative fluid admin- atory failure, and/or reintubation within three postoperative days.
istration within the range of standard clinical practice would be AKI was defined as an increase in serum creatinine by at least 0.3 mg/
associated with differences in 30-day mortality, postoperative respir- dL or 50% from preoperative levels within 48 postoperative hours14
atory complications (PRCs), and acute kidney injury (AKI). We or the existence of an AKI diagnostic code within seven postoper-
sought to quantify the dose-response relationship between fluid ative days. Exploratory outcomes included postoperative length of
volume and each of these outcomes across 5 quintiles of practice stay (PLOS) and cost. PLOS was defined as the number of hospi-
spanning the range of restrictive to liberal. talized days after surgery. Hospital costs were derived from an
internal financial tracking system and represent the sum of direct
METHODS and indirect expenses incurred by the institution. To arrive at general-
izable cost estimates, our cohort was matched to the 2012 National
Study Design and Setting Inpatient Sample.15
We performed a retrospective analysis of data on file in
surgeries performed at Massachusetts General Hospital and two Statistical Analysis
affiliates between January 2007 and August 2014. Local treatment Quintile-based analysis was performed for all outcomes with
guidelines for intraoperative fluid management were not in place at any the best-performing quintile serving as the reference group. A
of the study sites during the data acquisition period. The study was survival analysis using a multivariable Cox proportional hazards
approved by the Partners Institutional Review Board (2013P001704) model was performed for 30-day mortality. The proportional hazards
and registered at clinicaltrials.gov (NCT02105298). All adult patients assumption was met based on the parallel nature of the –lng[–
undergoing noncardiac surgery with intraoperative endotracheal intu- ln(survival)] curves for each category of the exposure variable versus
bation and extubation were enrolled (Fig. 1). Demographic, intra- ln(analysis time). PRCs and AKI were analyzed using logistic
operative, and outcomes data were extracted from clinical and regression models whereas PLOS and cost were analyzed using
administrative databases, as described in the online supplement. zero-truncated negative binomial regression models. Confounders
were included based on a priori knowledge of predictors of respir-
Exposure Variable atory failure,16 AKI,17 and factors used by clinicians to estimate fluid
Total fluid volume was derived from the anesthesia record and requirements.18 The following were treated as categorical variables:
defined as the volumes of crystalloid plus one-and-a-half times13 sex, admission type, emergency status, surgical body region, use of
colloid administered between initiation of anesthesia care and arrival neuromuscular blocking agents, crystalloid type, and medical comor-
in the postanesthesia care unit. Patients were classified into 5 groups bidities.19 Covariates demonstrating linear correlation with the

2 | www.annalsofsurgery.com ß 2017 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: A.G.; ANNSURG-D-16-02308; Total nos of Pages: 9;
ANNSURG-D-16-02308

Annals of Surgery  Volume XX, Number XX, Month 2017 Intraoperative Fluid Management

primary outcome [age, body mass index (BMI), ASA score, age- are presented as hazard ratios (HR), odds ratios (OR), or incidence
adjusted anesthetic doses20] were included as continuous, whereas rate ratios (IRR) with 95% confidence intervals (95% CI). A 2-tailed
those demonstrating nonlinear associations with the primary out- P < 0.05 was considered statistically significant.
come (vasopressor equivalent dose,21 morphine equivalent dose,22
procedure duration, estimated blood loss, urine output, hypotension, RESULTS
and procedure relative value units23) were categorized into quintiles.
To quantify the distribution of interprovider variation with regard to Patient Selection
the exposure variable, we plotted the probability of each individual A total of 137,102 noncardiac surgical cases occurred at our
anesthesia practitioner providing intraoperative fluid volumes within institution between January 2007 and August 2014. Of these, a total
a specified optimal range, based on the results of the main analyses. of 92,094 met our inclusion criteria for analysis (Fig. 1). A total of
For each provider identified in fifty or more cases in the dataset, the 18,790 (20.4%) patients received intraoperative fluid volumes of
probability of administering fluids in the moderate range (quintiles 900 mL (Q1), 18,825 (20.4%) received volumes of >900 to 1100
2–4) versus restrictive (quintile 1) or liberal (quintile 5) range mL (Q2), 19,292 (20.9%) received volumes of >1100 to 1750 mL
was estimated after propensity matching of cases using the same (Q3), 16,769 (18.2%) received volumes of >1750 to 2700 mL (Q4)
covariates. and 18,418 (20.0%) received volumes of > 2700 mL (Q5). The
We performed several sensitivity analyses to assess the robust- clinical characteristics of the cohort including information about
ness of the findings to analytic approach, most notably, an alternate utilization various fluid types are summarized in Table S2 of the
definition of the primary exposure variable using a formula for fluid Supplemental Digital Content, http://links.lww.com/SLA/B202.
balance to account for individual differences in fasting status, Descriptive data derived using the alternate exposure definition of
maintenance fluid requirement, blood loss, and urine output. This fluid balance, based on an individualized estimate of fluid require-
formula and a full presentation of sensitivity analyses are described ment, are presented Table S3.
in the Supplemental Digital Content, http://links.lww.com/SLA/
B202. We also performed a series of analyses examining the potential Primary Outcome
impact of fluid type and colloid utilization in the cohort. Individual Approximately, 429 (0.5%) patients died within 30 days
subgroup and sensitivity analyses were also performed to assess the (Table 1). Survival analysis demonstrated a U-shaped curve of risk
influences of colloid: crystalloid ratio, categorization of exposure, which was lowest in the moderately restrictive range of quintile 2
and covariate variables, imputation of missing data, adjustment of (Fig. 2). Compared with this group, the risk for 30-day mortality was
PLOS for mortality, anatomic surgical site, emergency status, hos- significantly increased at restrictive (Q1: HR 1.41, 95% CI 1.03–
pital site, and several patient-oriented factors, further described in the 1.93, P ¼ 0.034) and liberal (Q5: HR 1.65, 95% CI 1.04–2.60, P ¼
Supplemental Digital Content, http://links.lww.com/SLA/B202. 0.032) practices.
Variables defined using International Statistical Classification
of Diseases and Related Health Problems, ninth revision codes (ICD- Secondary Outcomes
9) and Current Procedural Terminology (CPT) codes are described in A total of 3657 (3.9%) patients experienced PRCs. Increasing
the Supplemental Digital Content (see Table S1, Supplemental fluid volumes were found to increase the risk of PRCs (Q5: OR 1.27,
Digital Content, http://links.lww.com/SLA/B202). Analyses were 95% CI 1.08–1.48, P ¼ 0.003) (Fig. 2, Table 1). Within 48 hours of
conducted using Stata (versions 13 and 14, StataCorp, TX). Results surgery, 2669 (2.9%) patients met criteria for AKI. The risk for

TABLE 1. Effect of Total Intraoperative Fluid Volume on Postoperative Outcomes


Total Intraoperative Fluid Volume in Quintiles
Quintile 1 Quintile 2 Quintile 3 Quintile 4 Quintile 5
(n ¼ 18,790) (n ¼ 18,825) (n ¼ 19,292) (n ¼ 16,769) (n ¼ 18,418) Total Population
900 mL >900 to 1100 mL >1100 to 1750 mL >1750 to 2700 mL >2700 mL (N ¼ 92,094)
Primary Outcome
Mortality N (%) 154 (0.8) 62 (0.3) 66 (0.3) 65 (0.4) 82 (0.5) 429 (0.5)
HR (95% 1.41 (1.03–1.93), 1 1.17 (0.81–1.68), 1.47 (0.99–2.19), 1.65 (1.04–2.60),
CI), P P ¼ 0.034 P ¼ 0.403 P ¼ 0.056 P ¼ 0.032
Secondary Outcomes
Postoperative N (%) 527 (2.8) 491 (2.6) 685 (3.6) 707 (4.2) 1247 (6.8) 3657 (3.9)
Respiratory OR (95% 1.05 (0.90–1.21), 1 1.08 (0.94–1.23), 1.07 (0.92–1.24), 1.27 (1.08–1.48),
Complication CI), P P ¼ 0.600 P ¼ 0.300 P ¼ 0.38 P ¼ 0.003
Acute Kidney Injury N (%) 348 (1.9) 275 (1.5) 398 (2.1) 438 (2.9) 1165 (6.3) 2669 (2.9)
OR (95% 1.66 (1.37–2.01), 1.18 (0.99–1.41), 1.16 (1.00–1.34), 1 1.29 (1.14–1.46),
CI), P P < 0.001 P ¼ 0.061 P ¼ 0.054 P < 0.001
Exploratory Outcomes
Postoperative Mean (SD) 3.1 (4.7) 3.1 (4.0) 3.7 (4.1) 4.6 (4.9) 6.2 (5.8) 4.1 (4.9)
Length of IRR (95% 1.03 (1.00–1.06), 1 1.00 (0.98–1.02), 1.03 (1.01–1.06), 1.15 (1.12–1.18),
Stay, days CI), P P ¼ 0.076 P ¼ 0.965 P ¼ 0.005 P < 0.001
Total Hospital NIS Matched $12,385.70 $12,230.52 $12,245.16 $12,343.66 $13,140.87 ($9755.53)
Care Costs, Mean (SD) ($9194.90) ($9079.70) ($9090.57) ($9163.70)
dollars IRR (95% 1.01 (1.00–1.03), 1 1.00 (0.99–1.01), 1.01 (0.99–1.02), 1.07 (1.05–1.09),
CI), P P ¼ 0.084 P ¼ 0.842 P ¼ 0.230 P < 0.001
Results of multivariable Cox, logistic regression, and negative binomial regression modeling are presented as adjusted HR, OR or IRR, 95% CI, and P.
CI indicates confidence intervals; HR, hazard ratio; IRR, incidence rate ratios; NIS, national inpatient sample; OR, odds ratio.

ß 2017 Wolters Kluwer Health, Inc. All rights reserved. www.annalsofsurgery.com | 3

Copyright © 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: A.G.; ANNSURG-D-16-02308; Total nos of Pages: 9;
ANNSURG-D-16-02308

Shin et al Annals of Surgery  Volume XX, Number XX, Month 2017

FIGURE 2. Effect of intraoperative fluid volume on postoperative outcomes. Total volume of intravenous fluids administered from
initiation of anesthesia care to delivery in post-anesthesia care unit was analyzed as quintiles: quintile 1 (Q1) ¼ restrictive (900 mL);
quintile 2 (Q2) ¼ moderately restrictive (>900 to 1100 mL); quintile 3 (Q3) ¼ moderate (>1100 to 1750 mL); quintile 4
(Q4) ¼ moderately liberal (>1750 to 2700 mL); and quintile 5 (Q5) ¼ liberal (>2700 mL). The association between total intra-
operative fluid volume and postoperative outcomes was analyzed using multivariable models adjusted for age, sex, BMI, ASA,
emergency status, admission type, surgical body region, age-adjusted inhalational anesthestic dose, neuromuscular blocking agent
use, morphine equivalent dose, vasopressor equivalent dose, duration of surgery, estimated blood loss, urine output, duration of
hypotension, chloride composition of crystalloid, procedure relative value units, and medical comorbidities. A, Restrictive and
liberal fluid administration are associated with increased 30-day mortality according to multivariable Cox proportional hazard
model (P ¼ 0.034, #P ¼ 0.032). B, Liberal fluid administration is associated with increased risk of postoperative respiratory
complications according to multivariable negative binomial regression model (P ¼ 0.003). C, Restrictive and liberal fluid
administration are associated with increased risk of acute kidney injury according to multivariable negative binomial regression
model (P < 0.001, #P < 0.001). D, Liberal fluid administration is associated with increased postoperative length of stay according to
multivariable negative binomial regression model (P ¼ 0.005, #P < 0.001). E, Liberal fluid administration is associated with
increased hospital costs according to multivariable negative binomial regression model (P < 0.001). ASA indicates American
Society of Anesthesiologists physical classification score; BMI, body mass index.

postoperative AKI was lowest in the moderately liberal group (Q4), as a single point on the horizontal axis and the probability of
whereas AKI was increased with both restrictive (Q1: OR 1.66, 95% delivering a fluid volume in the ‘‘desirable’’ moderate range is
CI 1.37–2.01, P < 0.001) and liberal (Q5: OR 1.29, 95% CI 1.14– shown on the vertical axis. Only a small group (left) consistently
1.46, P < 0.001) fluid practices (Fig. 2, Table 1). administers fluids within this optimal range, whereas another group
(right) routinelys provide doses which appear excessively restrictive
Exploratory Analyses or liberal with regard to the outcomes considered in this study.
The mean PLOS in the cohort was 4.1 days. Increasing fluid
volumes were predictive of longer PLOS (compared with Q2, Q4: Sensitivity Analyses
IRR 1.03, 95% CI 1.01–1.06, P ¼ 0.005; Q5: IRR 1.15, 95% CI Defining the primary exposure variable as fluid balance rather
1.12–1.18, P < 0.001) (Fig. 2, Table 1). Total hospital costs were also than total volume had modest effects on confidence intervals but
lowest in the second quintile and highest in the fifth quintile (Q5: IRR demonstrated no overall change in patterns of observed association
1.07, 95% CI 1.05–1.09, P < 0.001) (Fig. 2). Based on the IRR (Fig. 3; Table 2). Mortality, PLOS, and cost continued to demonstrate
derived from our cost data, we estimate an average savings of $910 in U-shaped curves of association optimized in the moderately restric-
total hospital cost per case (from $13,140 to $12,230) in patients tive fluid group. PRCs climbed with increasing fluid doses whereas
receiving moderately restrictive versus liberal fluid volumes. the risk for AKI gradually fell to a nadir in the moderate quintiles
before rising again.
Provider Variability Our subgroup and sensitivity analyses collectively demon-
The practice patterns of individual anesthesia practitioners strated the robust nature of the reported associations. A complete
varied with respect to fluid administration even after accounting for presentation of the following analyses is reported in the Supple-
patient- and case-specific factors which may influence fluid require- mental Digital Content (http://link-s.lww.com/SLA/B202): use of
ments (Fig. 4). In this figure, each anesthesia provider is represented alternate number of quantiles for fluid exposure (Fig. S1A-C),

4 | www.annalsofsurgery.com ß 2017 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: A.G.; ANNSURG-D-16-02308; Total nos of Pages: 9;
ANNSURG-D-16-02308

Annals of Surgery  Volume XX, Number XX, Month 2017 Intraoperative Fluid Management

FIGURE 3. The effect of intraoperative fluid balance on postoperative outcomes. The exposure variable represents the net change in
fluid balance from initiation of anesthesia care to delivery in postanesthesia care unit. Fluid balance accounts for fasting status, blood
loss, urine output, maintenance requirement, and insensible losses. The net change in fluid balance additionally is adjusted for
duration of surgery and patient weight, and is analyzed by quintiles: quintile 1 (Q1) ¼ restrictive fluid balance (–6 mL/kg/hr);
quintile 2 (Q2) ¼ moderately restrictive (>–6 to –3.6 mL/kg/hr); quintile 3 (Q3) ¼ moderate (>–3.6 to –1.5 mL/kg/hr); quintile 4
(Q4) ¼ moderately liberal (>–1.5 to þ1.2 mL/kg/hr); quintile 5 (Q5) ¼ liberal (> þ1.2 mL/kg/hr). The association between fluid
balance and postoperative outcomes was analyzed using multivariable models adjusted for age, sex, BMI, ASA, emergency status,
admission type, surgical body region, age-adjusted inhalational anesthestic dose, neuromuscular blocking agent use, morphine
equivalent dose, vasopressor equivalent dose, duration of surgery, estimated blood loss, urine output, duration of hypotension,
chloride composition of crystalloid, procedure relative value units, and medical comorbidities. A, Liberal fluid administration is
associated with the highest risk of mortality according to multivariable Cox proportional hazard model (P ¼ 0.018). B, Moderately
liberal and liberal fluid administration are associated with increased risk of postoperative respiratory complications according to
multivariable logistic regression model (P ¼ 0.014; #P ¼ 0.003). C, Restrictive and liberal fluid administration are associated with
increased risk of acute kidney injury according to multivariable logistic regression model (P < 0.001, #P < 0.001. D, Moderately
liberal and liberal fluid administration are associated with increased postoperative length of stay according to multivariable negative
binomial regression model (P < 0.001, #P < 0.001). E, A moderately restrictive administration is associated with lowest hospital
costs according to multivariable negative binomial regression model (P < 0.001, #P < 0.001, yP < 0.001). ASA indicates American
Society of Anesthesiologists physical classification score; BMI, body mass index.

expanded use of continuous covariates (Fig. S1D-F), abdominal intraoperative fluid administration and 30-day mortality. PRC and
surgery only (Fig. S2A-C), exclusion of lung and liver resections AKI, 2 established predictors of surgical morbidity and mortality,24
(Fig. S2D-F), stratification by admission type (Fig. S3), stratification were also independently associated with fluid dosing. These fluid-
by ASA classification (Fig. S4), preoperative risk score for pulmon- associated outcome differences were further reflected as differences
ary complications (Fig. S5), baseline creatinine clearance (Fig. S6A- in the cost and duration of hospitalization.
B), duration of intraoperative hypotension (Fig. S6C-D), intraoper-
ative vasopressor use (Fig. S6E-F), adjustments for death prior to Mortality
discharge (Fig. S7), emergency status (Fig. S8), imputation of Existing knowledge about the effects of intraoperative fluid on
missing data (Fig. S9), exclusion of hydroxyethyl starch (Fig. outcomes has been drawn from trials comparing two distinct fluid
S10A-C), exclusion of all colloids (Fig. S10D-F), exclusion of administration strategies (eg, restrictive vs liberal, GDFT vs liberal,
non-standard crystalloids (Fig. S10G-I), statistical adjustment for and so on). This binary approach to comparison conflicts with
colloid utilization (Fig. S11), alternative colloid: crystalloid ratios physiologic predictions of the perioperative risk attributable to fluid
(Fig. S12), hospital site and provider effects (Fig. S13), and changes administration, which has repeatedly been theorized to fall and then
in fluid administration practice over time (Fig. S14-S16). rise with increasing fluid volumes, forming a U-shaped curve.7,25
Previous efforts to characterize the influence of fluid admin-
istration on survival have targeted critically ill populations in which
DISCUSSION the expected incidence of postoperative mortality is higher. Both
In an analysis of 92,094 surgeries performed at the Massa- randomized controlled trials and retrospective studies in the intensive
chusetts General Hospital and two community hospitals over a 7-year care unit have found significant associations between positive fluid
period, we observed a robust U-shaped association between balance and mortality.26,27 However, the influence of intraoperative

ß 2017 Wolters Kluwer Health, Inc. All rights reserved. www.annalsofsurgery.com | 5

Copyright © 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: A.G.; ANNSURG-D-16-02308; Total nos of Pages: 9;
ANNSURG-D-16-02308

Shin et al Annals of Surgery  Volume XX, Number XX, Month 2017

TABLE 2. Effect of Fluid Balance on Postoperative Outcomes


Fluid Balance in Quintiles
Quintile 2 Quintile 3 Quintile 4
Quintile 1 (n ¼ 18,419) (n ¼ 18,419) (n ¼ 18,419) Quintile 5 Total
(n ¼ 18,419) >6 to 3.6 >3.7 to 1.5 >1.5 to þ1.2 (n ¼ 18,418) Population
 6 mL/kg/hr mL/kg/hr mL/kg/hr mL/kg/hr >þ1.2 mL/kg/hr (N ¼ 92,094)
Primary Outcome
30-day Mortality N (%) 185 (1.0) 77 (0.4) 62 (0.3) 47 (0.3) 56 (0.3) 429 (0.5)
HR (95% CI), P 1.20 (0.90–1.61), 1 1.08 (0.77–1.52), 1.21 (0.82–1.77), 1.59 (1.08–2.34),
P ¼ 0.0.210 P ¼ 0.663 P ¼ 0.335 P ¼ 0.018
Secondary Outcomes
Postoperative N (%) 1017 (5.5) 875 (4.8) 787 (4.3) 617 (3.4) 361 (2.0) 3657 (4.0)
Respiratory OR (95% CI), P 0.98 (0.87–1.09), 1 1.09 (0.97–1.22), 1.17 (1.03–1.32), 1.26 (1.08–1.46),
Complication P ¼ 0.669 P ¼ 0.138 P ¼ 0.014 P ¼ 0.003
Acute Kidney Injury N (%) 674 (3.7) 538 (2.9) 529 (2.9) 522 (2.8) 406 (2.2) 2669 (2.9)
OR (95% CI), P 1.41 (1.21–1.64), 1.13 (0.98–1.30), 0.98 (0.85–1.12), 1 1.48 (1.27–1.71),
P < 0.001 P ¼ 0.105 P ¼ 0.718 P < 0.001
Exploratory Outcomes
Postoperative Mean (SD) 5.6 (6.1) 4.6 (4.8) 4.2 (4.6) 3.5 (4.3) 2.6 (4.0) 4.1 (4.9)
Length of IRR (95% CI), P 0.99 (0.98–1.01), 1 1.01 (0.99–1.03), 1.05 (1.03–1.07), 1.16 (1.13–1.19),
Stay, days P ¼ 0.615 P ¼ 0.345 P < 0.001 P < 0.001
Total Hospital NIS Matched $17,705.38 $16,872.74 $17,028.44 $17,539.62 $17,403.76
Mean (SD) ($13,646.70) ($13,004.93) ($13,124.94) ($13,518.94) ($13,414.22)
Care Costs, IRR (95% CI), P 1.05 (1.03–1.07), 1 1.01 (1.00–1.02), 1.04 (1.03–1.05), 1.03 (1.01–1.05),
dollars P < 0.001 P ¼ 0.165 P < 0.001 P < 0.001
Results of multivariable Cox, logistic regression, and negative binomial regression modeling are presented as adjusted HR, OR or IRR, 95% CI, and P.
CI indicates confidence intervals; HR, hazard ratio; IRR, incidence rate ratios; NIS, national inpatient sample; OR, odds ratio.

fluid management on survival in noncritically ill patients may pulmonary consequences of fluid overload, which calls into question
be distinct. the generalizability of the reported trend.
The large size of our cohort uniquely enabled us to study this Our findings support the view that liberal fluid volumes have
endpoint in relatively healthier populations and to examine the role of harmful effects on postoperative lung function and add to the current
fluid administered intraoperatively, before the onset of inflammation understanding in several ways. First, we confirm that the association
and organ dysfunction characteristic of critical illness. Our study is between liberal fluid administration and PRCs is a generalizable
unique in that it quantifies the impact of incremental variations in phenomenon conserved across a wide range of surgical types and
fluid administration on patient outcomes across this spectrum, patient classes. Second, we quantify the magnitude of fluid-attribu-
includes a range of surgeries, and examines associations with table risk for PRCs on a population level. Third, the size of the study
mortality and on complications known to predict mortality. We allowed us to examine incremental differences in fluid dosing and
found the risk of 30-day mortality to be increased with both demonstrate that risk for PRCs continuously increases with fluid
restrictive and liberal intraoperative fluid strategies. Moderate fluid dosing with no clear threshold effect.
restriction, representing the second most restrictive quintile of
practice, was associated with the highest rate of survival. Acute Kidney Injury
The understanding that excessive intraoperative fluid admin-
Respiratory Complications istration may impair respiratory and gastrointestinal function35 has
The harmful effects of fluid excess are frequently manifested already exerted an influence on clinical norms, driving more restric-
in the lungs. Pulmonary edema can impair gas exchange, placing tive strategies. However, the safe lower limit of fluid restriction
patients at heightened risk for infection, respiratory failure, and remains unclear. Renal function is often considered uniquely
reintubation. PRCs are the most common non-surgical compli- susceptible to hypovolemic states and postoperative AKI has been
cation24,28,29 and are known to increase PLOS, mortality, and the implicated as a driver of morbidity and mortality in surgical popu-
financial costs associated with an episode of care.29,30 lations.36,37 Evidence-based methods of estimating renal perfusion
Small randomized trials of various fluid dosing strategies and are lacking: urine output is a poor indicator of volume status during
respiratory outcomes have reached differing conclusions, even when surgery38 and is not predictive of postoperative AKI.17,38 A recent
performed by the same group.31,32 It is difficult to determine whether observational study of resuscitation practices in burn patients found
this heterogeneity is the result of sampling error, differences in an increase in the incidence of AKI in the most restrictive forms of
population, study design, or outcome definition. A meta-analysis practice.2 However, the generalizability of these findings to more
of several trials suggested that larger fluid volumes increase the risks general surgical populations is limited and thus, despite the clinical
of postoperative pneumonia and pulmonary edema.33 However, these importance of AKI as an outcome, the relationship between intra-
findings are limited by marked differences in the definitions of operative fluid management and postoperative kidney function is not
‘‘liberal’’ and ‘‘restrictive,’’ which make the results of individual well defined.
studies difficult to harmonize.7 A subsequent study examining fluid We found both restrictive and liberal extremes of intraoper-
dosing as a continuous variable in patients undergoing lung resection ative fluid administration to be deleterious to kidney function. This
described an inflection point in the risk for complications at 6 mL/kg/ finding contradicts the dogma that intravascular volume expansion is
hr,34 however this population is known to be susceptible to the uniformly renal-protective, but has strong biologic plausibility. On

6 | www.annalsofsurgery.com ß 2017 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: A.G.; ANNSURG-D-16-02308; Total nos of Pages: 9;
ANNSURG-D-16-02308

Annals of Surgery  Volume XX, Number XX, Month 2017 Intraoperative Fluid Management

protocols,46 which show similar reductions in costs and length of


stay associated with changes in fluid management. Of note, our
results are derived from a cohort that predates the implementation of
ERAS protocols at our institution.

Provider Variability
We observed marked variability in the management of intra-
operative fluid administration across individual anesthesia providers.
Our data confirm the findings of another large study of fluid
administration practice12 suggesting that this variability is driven
more by individual preference than by patient and procedural
characteristics. Institution-specific protocols for fluid therapy should
be developed and rigorously implemented based on our data and
other data recently published in the Annals of Surgery.42,47,48

Strengths and Limitations


Our study derives its strength from the size and diversity of
surgical cohort analyzed, increasing the finding’s generalizability
and permitting the rigorous analysis of uncommon but important
postoperative complications. Distinct from most existing studies, we
implemented robust confounder control using data extracted directly
from perioperative records. This methodology and the PRC endpoint
FIGURE 4. Provider-level practice variation in intraoperative have been utilized and validated by our research group.16,49,50
fluid dosing. Fluid administration practices ranged widely Limitations include the use of diagnostic and procedure codes which
across practitioners within our cohort. Each anesthesia provider may be user-dependent, the observational nature of the study design,
identified in 50 or more cases in the dataset is represented as a and the potential for residual confounding of the reported associ-
single point on the horizontal axis. The vertical axis represents ations. To address these challenges, we conducted quality control
the individual probability of that provider administering a fluid checks of codes through a randomized audit of medical records and
dose within the moderate range (quintiles 2, 3, or 4) versus a performed sensitivity analyses to assess the impact of several vari-
very liberal (quintile 5) or restrictive (quintile 1) range. A small ables on the reported associations
number of anesthesia providers consistently administer fluid The diverse nature of the cohort and the stability of the findings
volumes within the range found to optimize outcomes in this across several subgroups support the generalizability of the findings.
cohort (left). The majority deliver fluid volumes in the optimal However, the study population included only patients extubated at the
range in 50% to 65% cases (middle). A smaller group of end of surgery and different approaches to fluid management may be
providers consistently dose fluids in a range that appears required in patients who are not extubated at the end of the case but
excessively restrictive or liberal with regard to the outcomes remain mechanically ventilated. Similarly, the predominance of elec-
considered in this study (right). A similar distribution is tive outpatients and non-urgent surgical inpatients in this cohort limits
observed both with (black circles) and without (grey circles) the applicability of the findings to emergency situations and the
propensity-matched adjustment for patient- and case-specific comparatively small number of such cases the precluded meaningful
factors which may influence fluid requirements (including analysis of emergency cases as an independent subgroup. Fluid deficits
fasting status, patient weight, age, blood loss, urine output, in emergency surgery may be greater and optimal fluid management in
surgical body site, procedure duration, and hypotension). such cases may require larger volumes for resuscitation. Finally,
because only a small fraction of patients received any colloid, we
one hand, untreated hypovolemia and prerenal azotemia may prog- were not adequately powered to independently reproduce the associ-
ress to acute tubular necrosis and renal dysfunction.39 Conversely, ation between various colloid products and outcomes (eg, HES on
excess fluid may impair renal function by mechanisms similar to mortality and renal failure rate).
those observed in the pulmonary and gastrointestinal systems:
increases in central venous pressure elevate renal subcapsular press- Implications for Clinicians and Researchers
ures, reduce renal blood flow and glomerular filtration, and promote Differences in intraoperative fluid dosing within the range of
renal parenchymal edema1,40,41 routine clinical practice are associated with significant differences in
postoperative outcomes. Mortality, cost, and PLOS are each opti-
Cost and Length of Stay mized with moderate fluid restriction. These differences appear to
We sought to quantify the summative economic impact of arise from the distinct effects of fluid administration on various organ
differing practices in fluid administration by examining associations systems. Liberal fluid administration is strongly predictive of pul-
with PLOS and total cost of hospitalization. PLOS and cost were monary complications. Fluid restriction increases the risk of AKI, but
significantly increased in patients receiving liberal volumes, pre- extreme efforts to drive urine output with intravenous volume
sumably because of the need to treat complications arising from expansion may paradoxically promote renal dysfunction via an
hypervolemia and await the resolution of edema. A recent analysis of independent mechanism.
colorectal and lower extremity joint replacement surgeries included These trends persist across a range of subgroups, suggesting
in the Premier Research Database suggested that, compared mod- that the optimum range for intraoperative fluid therapy may be
erate volumes, both high and low fluid volumes on the day of surgery influenced more modestly by the preoperative medical comorbidities
were associated with increased cost and length of stay.42 Our findings and procedure-specific factors than is commonly believed. Recent
are generally in agreement with these studies and economic studies population-based studies in colorectal, gynecologic, arthroplastic,
of GDFT43– 45 and Enhanced Recovery After Surgery (ERAS) and endovascular surgery have also demonstrated the adverse effects

ß 2017 Wolters Kluwer Health, Inc. All rights reserved. www.annalsofsurgery.com | 7

Copyright © 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: A.G.; ANNSURG-D-16-02308; Total nos of Pages: 9;
ANNSURG-D-16-02308

Shin et al Annals of Surgery  Volume XX, Number XX, Month 2017

of both restrictive and liberal perioperative fluid administration on randomized clinical trial and systematic review. Crit Care Med. 2016;44:724–
733.
postoperative outcomes42,47 and are in keeping with the results of a
9. Rahbari NN, Zimmermann JB, Schmidt T, et al. Meta-analysis of standard,
large meta-analysis of fluid therapy protocols in abdominal pro- restrictive and supplemental fluid administration in colorectal surgery. Br J
cedures which highlighted the importance of a ‘‘balanced’’ approach Surg. 2009;96:331–341.
to fluid therapy.6 Taken together, these findings suggest that, in the 10. Marik PE. Fluid responsiveness and the six guiding principles of fluid
absence of compelling case-specific considerations or the use of resuscitation. Crit Care Med. 2016;44:1920–1922.
well-validated GDFT protocols, a moderately restrictive intraoper- 11. Challand C, Struthers R, Sneyd JR, et al. Randomized controlled trial of
ative fluid administration strategy should be the default standard of intraoperative goal-directed fluid therapy in aerobically fit and unfit patients
having major colorectal surgery. Br J Anaesth. 2012;108:53–62.
care. In our population, optimum dose consistently fell in the second
12. Lilot M, Ehrenfeld JM, Lee C, et al. Variability in practice and factors
quintile of clinical practice. In absolute terms, this range was predictive of total crystalloid administration during abdominal surgery: retro-
characterized by an infusion rate of approximately 6–7 mL/kg/hr, spective two-centre analysis. Br J Anaesth. 2015;114:767–776.
a fluid balance of 40% less than traditional textbook estimates 13. Cortés DO, Barros TG, Njimi H, et al. Crystalloids versus colloids: exploring
(Supplementary Equation 1, http://links.lww.com/SLA/B202), or differences in fluid requirements by systematic review and meta-regression.
roughly 1 L for the average three-hour case in the absence of excess Anesth Analg. 2015;120:389–402.
blood loss or other extenuating circumstances. 14. KDIGO Clinical Practice Guideline for acute kidney injury. Kidney Int Suppl.
2012;2:1–138.
Our findings have several practical implications for ongoing
15. National Inpatient Sample, Healthcare Cost and Utilization Project, Agency
work in the field. Based on the U-shaped distribution of risk observed for Healthcare Research and Quality. Rockville, MD, 2012.
in this study, we suggest that quality improvement and enhanced 16. McLean DJ, Diaz-Gil D, Farhan HN, et al. Dose-dependent association
recovery efforts might achieve the greatest benefit by first working to between intermediate-acting neuromuscular-blocking agents and postopera-
identify and understand extremes of practice among providers. tive respiratory complications. Anesthesiology. 2015;122:1201–1213.
Because both restrictive and liberal fluid management appear to 17. Kheterpal S, Tremper KK, Englesbe MJ, et al. Predictors of postoperative
yield suboptimal outcomes, future prospective trials should consider acute renal failure after noncardiac surgery in patients with previously normal
renal function. Anesthesiology. 2007;107:892–902.
including multiple comparison arms. Finally, the value of moderately
18. Miller TE, Bunke M, Nisbet P, et al. Fluid resuscitation practice patterns in
restrictive, outcome-oriented dosing strategies versus cardiac output- intensive care units of the USA: a cross-sectional survey of critical care
driven GDFT protocols remains an important question. A recent physicians. Perioper Med (Lond). 2016;5:15.
meta-analysis in major abdominal surgery has suggested that simpler 19. Quan H, Sundararajan V, Halfon P, et al. Coding algorithms for defining
protocols for standardization of fluid management (as part of an comorbidities in ICD-9-CM and ICD-10 administrative data. Med Care.
ERAS pathway) may be equivalent to GDFT strategies relying on 2005;43:1130–1139.
physiologic measures.48 20. Lerou JGC. Nomogram to estimate age-related MAC. Br J Anaesth.
2004;93:288–291.
In summary, we identified a U-shaped distribution of risk
21. Russell JA, Walley KR, Singer J, et al. Vasopressin versus norepinephrine
associated with intravenous fluid dosing during surgery. Although infusion in patients with septic shock. N Engl J Med. 2008;358:877–887.
the intraoperative period comprises a relatively brief portion of the 22. Miller RD, Eriksson LI, Fleisher LA, et al. Miller’s Anesthesia. 8th Edition,
surgical encounter, it represents a uniquely vulnerable physiologic Philadelphia: Elsevier Saunders; 2015.
state and is paradoxically a period in which the surgical team may be 23. Merkow RP, Bentrem DJ, Cohen ME, et al. Effect of cancer surgery complex-
less directly involved in fluid management. As an easily modifiable ity on short-term outcomes, risk predictions, and hospital comparisons. J Am
factor that significantly influences surgical outcomes, we submit that Coll Surg. 2013;217:685–693.
intraoperative fluid management strategy should routinely be dis- 24. Khuri SF, Henderson WG, DePalma RG, et al. Determinants of long-term
survival after major surgery and the adverse effect of postoperative compli-
cussed preoperatively in consultation with the anesthesia team. For cations. Ann Surg. 2005;242:326–341. discussion341-3.
most patients, a moderately restrictive approach to intraoperative 25. Doherty M, Buggy DJ. Intraoperative fluids: how much is too much? Br J
fluid management may yield improved postoperative outcomes. Anaesth. 2012;109:69–79.
26. Silva JM, de Oliveira AMRR, Nogueira FAM, et al. The effect of excess fluid
REFERENCES balance on the mortality rate of surgical patients: a multicenter prospective
study. Crit Care. 2013;17:R288.
1. Prowle JR, Kirwan CJ, Bellomo R. Fluid management for the prevention and 27. Semler MW, Wheeler AP, Thompson BT, et al. Impact of initial central venous
attenuation of acute kidney injury. Nat Rev Nephrol. 2014;10:37–47. pressure on outcomes of conservative versus liberal fluid management in acute
2. Mason SA, Nathens AB, Finnerty CC, et al. Hold the pendulum: rates respiratory distress syndrome. Critical Care Med. 2016;44:782–789.
of acute kidney injury are increased in patients who receive resuscitation 28. Brueckmann B, Villa-Uribe JL, Bateman BT, et al. Development and vali-
volumes less than predicted by the Parkland equation. Ann Surg. 2016; dation of a score for prediction of postoperative respiratory complications.
264:1142–1147. Anesthesiology. 2013;118:1276–1285.
3. Brandstrup B, Tønnesen H, Beier-Holgersen R, et al. Effects of intravenous 29. Dimick JB, Chen SL, Taheri PA, et al. Hospital costs associated with surgical
fluid restriction on postoperative complications: comparison of two perioper- complications: a report from the private-sector National Surgical Quality
ative fluid regimens: a randomized assessor-blinded multicenter trial. Ann Improvement Program. J Am Coll Surg. 2004;199:531–537.
Surg. 2003;238:641–648.
30. Johnson RG, Arozullah AM, Neumayer L, et al. Multivariable predictors of
4. McArdle GT, McAuley DF, McKinley A, et al. Preliminary results of a postoperative respiratory failure after general and vascular surgery: results
prospective randomized trial of restrictive versus standard fluid regime in from the patient safety in surgery study. J Am Coll Surg. 2007;204:1188–1198.
elective open abdominal aortic aneurysm repair. Ann Surg. 2009;250:28–34.
31. Holte K, Foss NB, Andersen J, et al. Liberal or restrictive fluid administration
5. Kiefer N, Theis J, Putensen-Himmer G, et al. Peristaltic pneumatic com- in fast-track colonic surgery: a randomized, double-blind study. Br J Anesth.
pression of the legs reduces fluid demand and improves hemodynamic 2007;99:500–508.
stability during surgery: a randomized, prospective study. Anesthesiology. 32. Holte K, Klarskov B, Christensen DS, et al. Liberal versus restrictive fluid
2011;114:536–544. administration to improve recovery after laparoscopic cholecystectomy: a
6. Varadhan KK, Lobo DN. A meta-analysis of randomised controlled trials of randomized, double-blind study. Ann Surg. 2004;240:892–899.
intravenous fluid therapy in major elective open abdominal surgery: getting 33. Corcoran T, Rhodes JEJ, Clarke S, et al. Perioperative fluid management
the balance right. Proc Nutr Soc. 2010;69:488–498. strategies in major surgery: a stratified meta-analysis. Anesth Analg. 2012;
7. Bundgaard-Nielsen M, Secher NH, Kehlet H. ‘Liberal’ vs. ‘‘restrictive’’ 114:640–651.
perioperative fluid therapy—a critical assessment of the evidence. Acta 34. Arslantas MK, Kara HV, Tuncer BB, et al. Effect of the amount of intra-
Anesthesiol Scand. 2009;53:843–851. operative fluid administration on postoperative pulmonary complications
8. Osawa EA, Rhodes A, Landoni G, et al. Effect of perioperative goal-directed following anatomic lung resections. J Thorac Cardiovasc Surg. 2015;149.
hemodynamic resuscitation therapy on outcomes following cardiac surgery: a 314-20-321.e1.

8 | www.annalsofsurgery.com ß 2017 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: A.G.; ANNSURG-D-16-02308; Total nos of Pages: 9;
ANNSURG-D-16-02308

Annals of Surgery  Volume XX, Number XX, Month 2017 Intraoperative Fluid Management

35. Lobo DN, Bostock KA, Neal KR, et al. Effect of salt and water 43. Grocott MPW, Dushianthan A, Hamilton MA, et al. Perioperative increase in
balance on recovery of gastrointestinal function after elective colonic global blood flow to explicit defined goals and outcomes after surgery: a
resection: a randomised controlled trial. Lancet. 2002;359:1812– Cochrane Systematic Review. Br J Anesth. 2013;111:535–548.
1818. 44. Benes J, Giglio M, Brienza N, et al. The effects of goal-directed fluid therapy
36. Korenkevych D, Ozrazgat-Baslanti T, Thottakkara P, et al. The pattern of based on dynamic parameters on post-surgical outcome: a meta-analysis of
longitudinal change in serum creatinine and 90-day mortality after major randomized controlled trials. Crit Care. 2014;18:584.
surgery. Ann Surg. 2015;1. 45. Khoo CK, Vickery CJ, Forsyth N, et al. A prospective randomized controlled
37. Goren O, Matot I. Perioperative acute kidney injury. Br J Anesth. 2015; trial of multimodal perioperative management protocol in patients undergoing
115(Suppl 2):ii3–ii14. elective colorectal resection for cancer. Ann Surg. 2007;245:867–872.
38. Matot I, Dery E, Bulgov Y, et al. Fluid management during video-assisted 46. Miller TE, Thacker JK, White WD, et al. Reduced length of hospital stay in
thoracoscopic surgery for lung resection: a randomized, controlled trial of colorectal surgery after implementation of an enhanced recovery protocol.
effects on urinary output and postoperative renal function. J Thorac Cardi- Anesth Analg. 2014;118:1052–1061.
ovasc Surg. 2013;146:461–466. 47. Regenbogen SE, Shah NJ, Collins SD, et al. Population-based assessment of
39. Bellomo R, Kellum JA, Ronco C. Acute kidney injury. Lancet. 2012;380: intraoperative fluid administration practices across three surgical specialties.
756–766. Ann Surg. 2016;1.
40. Prowle JR, Echeverri JE, Ligabo EV, et al. Fluid balance and acute kidney 48. Rollins KE, Lobo DN. Intraoperative goal-directed fluid therapy in elective
injury. Nature Publishing Group. 2010;6:107–115. major abdominal surgery: a meta-analysis of randomized controlled trials. Ann
41. Prowle JR, Chua H-R, Bagshaw SM, et al. Clinical review: volume of fluid Surg. 2016;263:465–476..
resuscitation and the incidence of acute kidney injury. A systematic review. 49. Ladha K, Vidal Melo MF, McLean DJ, et al. Intraoperative protective
Crit Care. 2012;16:230. mechanical ventilation and risk of postoperative respiratory complications:
42. Thacker JKM, Mountford WK, Ernst FR, et al. Perioperative fluid utilization hospital based registry study. BMJ. 2015;351:h3646.
variability and association with outcomes: considerations for enhanced 50. de Jong MA, Ladha KS, Melo MF, et al. Differential effects of intraoperative
recovery efforts in sample US surgical populations. Ann Surg. 2016;263: positive end-expiratory pressure (PEEP) on respiratory outcome in major
502–510. abdominal surgery versus craniotomy. Ann Surg. 2016;264:362–369.

ß 2017 Wolters Kluwer Health, Inc. All rights reserved. www.annalsofsurgery.com | 9

Copyright © 2017 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

You might also like